home *** CD-ROM | disk | FTP | other *** search
Text File | 1997-05-02 | 39.8 MB | 941,512 lines |
Text Truncated. Only the first 1MB is shown below. Download the file for the complete contents.
- *RECORD*
- *FIELD* NO
- 100050
- *FIELD* TI
- 100050 AARSKOG SYNDROME
- SHAWL SCROTUM, INCLUDED;;
- HYPERTELORISM, INCLUDED
- *FIELD* TX
- Grier et al. (1983) reported father and 2 sons with typical Aarskog
- syndrome, including short stature, hypertelorism, and shawl scrotum.
- They tabulated the findings in 82 previous cases. X-linked recessive
- inheritance has been repeatedly suggested (see 305400). The family
- reported by Welch (1974) had affected males in 3 consecutive
- generations. Thus, there is either genetic heterogeneity or this is an
- autosomal dominant with strong sex-influence and possibly ascertainment
- bias resulting from use of the shawl scrotum as a main criterion.
- Stretchable skin was present in the cases of Grier et al. (1983). Teebi
- et al. (1993) reported the case of an affected mother and 4 sons
- (including a pair of monozygotic twins) by 2 different husbands. They
- suggested that the manifestations were as severe in the mother as in the
- sons and that this suggested autosomal dominant inheritance. Actually,
- the mother seemed less severely affected, compatible with X-linked
- inheritance.
-
- *FIELD* RF
- 1. Grier, R. E.; Farrington, F. H.; Kendig, R.; Mamunes, P.: Autosomal
- dominant inheritance of the Aarskog syndrome. Am. J. Med. Genet. 15:
- 39-46, 1983.
-
- 2. Teebi, A. S.; Rucquoi, J. K.; Meyn, M. S.: Aarskog syndrome: report
- of a family with review and discussion of nosology. Am. J. Med.
- Genet. 46: 501-509, 1993.
-
- 3. Welch, J. P.: Elucidation of a 'new' pleiotropic connective tissue
- disorder. Birth Defects Orig. Art. Ser. X(10): 138-146, 1974.
-
- *FIELD* CS
-
- Growth:
- Mild to moderate short stature
-
- Head:
- Normocephaly
-
- Hair:
- Widow's peak
-
- Facies:
- Maxillary hypoplasia;
- Broad nasal bridge;
- Anteverted nostrils;
- Long philtrum;
- Broad upper lip;
- Curved linear dimple below the lower lip
-
- Eyes:
- Hypertelorism;
- Ptosis;
- Down-slanted palpebral fissures;
- Ophthalmoplegia;
- Strabismus;
- Hyperopic astigmatism;
- Large cornea
-
- Ears:
- Floppy ears;
- Lop-ears
-
- Mouth:
- Cleft lip/palate
-
- GU:
- Shawl scrotum;
- Saddle-bag scrotum;
- Cryptorchidism
-
- Limbs:
- Brachydactyly;
- Digital contractures;
- Clinodactyly;
- Mild syndactyly;
- Transverse palmar crease;
- Lymphedema of the feet
-
- Joints:
- Ligamentous laxity;
- Osteochondritis dissecans;
- Proximal finger joint hyperextensibility;
- Flexed distal finger joints;
- Genu recurvatum;
- Flat feet
-
- Skin:
- Stretchable skin
-
- Spine:
- Cervical spine hypermobility;
- Odontoid anomaly
-
- Heme:
- Macrocytic anemia;
- Hemochromatosis
-
- GI:
- Hepatomegaly;
- Portal cirrhosis;
- Imperforate anus;
- Rectoperineal fistula
-
- Pulmonary:
- Interstitial pulmonary disease
-
- Thorax:
- Sternal deformity
-
- Inheritance:
- Sex-influenced autosomal dominant form;
- also X-linked form
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- carol: 7/7/1993
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 100070
- *FIELD* TI
- 100070 ABDOMINAL AORTIC ANEURYSM
- AORTIC ANEURYSM, ABDOMINAL;;
- ANEURYSM, ABDOMINAL AORTIC
- *FIELD* TX
- Tilson and Seashore (1984) reported 50 families in which abdominal
- aortic aneurysm had occurred in 2 or more first-degree relatives, mainly
- males. In 29 families, multiple sibs (up to 4) were affected; in 2
- families, 3 generations were affected; and in 15 families, persons in 2
- generations were affected. Three complex pedigrees were observed: one in
- which both parents and 3 sons were affected; one in which a man and his
- paternal uncle were affected; and one in which a man and his father and
- maternal great-uncle were affected. In the 'one-generation' families,
- there were 3 with only females affected, including a set of identical
- twins. The authors concluded that if a single gene is responsible, it is
- likely to be autosomal but that a multigenic mechanism cannot be
- excluded. Clifton (1977) reported 3 affected brothers. In North
- Carolina, Johnson et al. (1985) found that white males have a frequency
- of abdominal aortic aneurysm about 3 times that in black males, black
- females, or white females; all 3 of the latter groups had about
- comparable frequencies. Frequency was ascertained by a survey of
- autopsies and a survey of abdominal computed tomographic scans in
- subjects over the age of 50 years. Johansen and Koepsell (1986) compared
- the family histories of 250 patients with abdominal aortic aneurysm with
- those of 250 control subjects. Among the control subjects, 2.4% reported
- a first-degree relative with an aneurysm, compared with 19.2% of the
- patients with abdominal aortic aneurysm. This was taken to represent an
- estimated 11.6-fold increase in abdominal aortic aneurysm risk among
- persons with an affected first-degree relative. The authors suggested
- that noninvasive screening to detect early abdominal aortic aneurysm may
- be warranted in the relatives of affected persons. Borkett-Jones et al.
- (1988) brought to 4 the number of reported sets of identical twins
- concordant for abdominal aortic aneurysm. In a 9-year prospective study
- of 542 consecutive patients undergoing operation for abdominal aortic
- aneurysm, Darling et al. (1989) found that 82 (15.1%) had a first-degree
- relative with an aneurysm as compared to 9 (1.8%) of the control group
- of 500 patients of similar age and sex without aneurysmal disease.
- Patients with familial abdominal aortic aneurysm were more likely to be
- women (35% vs 14%), and men with familial abdominal aortic aneurysm
- tended to be about 5 years younger than the women. No significant
- difference was found between the patients with nonfamilial and familial
- abdominal aortic aneurysms in anatomic extent, multiplicity, associated
- occlusive disease, or blood type. The risk of rupture was strongly
- correlated with familial disease and the presence of a female member
- with aneurysm (63% vs 37%). Darling et al. (1989) suggested the term
- 'black widow syndrome' because of the grim significance of the presence
- of an affected female in the family. Abdominal aortic aneurysm is, of
- course, a common disorder; by ultrasound screening, Collin et al. (1988)
- found an abdominal aortic aneurysm in 5.4% of men aged 65 to 74, and in
- 2.3% of men in this age group the aneurysm was 4 cm or more in diameter.
- On the basis of a study of first-degree relatives of 91 probands,
- Majumder et al. (1991) rejected the nongenetic model and concluded that
- the most parsimonious genetic model was that susceptibility to abdominal
- aortic aneurysm is determined by a recessive gene at an autosomal
- diallelic major locus. Loosemore et al. (1988) described 2 brothers with
- abdominal aortic aneurysm at ages 58 and 62 years, whose father died of
- ruptured abdominal aortic aneurysm at the age of 72 years. Four other
- sibs died of myocardial infarction at ages 47 to 61 years. Fitzgerald et
- al. (1995) assessed the incidence of abdominal aortic aneurysm (AAA) in
- the siblings of 120 patients known to have AAA. Twelve percent of the
- siblings were found to have an aneurysm, including 22% of male siblings
- but only 3% of female siblings. Male siblings with hypertension were
- more likely to have AAA.
-
- Ward (1992) looked for association of dilated peripheral arteries with
- aortic aneurysmal disease by measuring the diameters of the common
- femoral, popliteal, brachial, common carotid, internal carotid, and
- external carotid arteries by color-flow duplex scan in 30 control
- subjects and 36 patients with aortic aneurysm matched for age, sex,
- smoking habits, and hypertension. Mean peripheral artery diameter was
- significantly greater in patients with aortic aneurysms than in controls
- at all measurement sites. Peripheral artery dilatation was identified at
- sites that are seldom, if ever, involved in atherosclerosis. Ward (1992)
- concluded that there is a generalized dilating diathesis in aortic
- aneurysmal disease that may be unrelated to atherosclerosis.
-
- Loosemore et al. (1988) suggested that a deficiency of type III collagen
- might be the basis for the aneurysm formation. The proportion of type
- III collagen in forearm skin biopsies was cited as accurately reflective
- of the proportion in the aorta and was said to have been low in the
- brothers. Kontusaari et al. (1989) and Kontusaari et al. (1990)
- incriminated mutation in the COL3A1 gene (120180.0004) in the causation
- of familial aortic aneurysms. See review of Kuivaniemi et al. (1991).
- Tromp et al. (1993) carried out detailed DNA sequencing of the
- triple-helical domain of type III procollagen on cDNA prepared from 54
- patients with aortic aneurysms. In the case of 43 patients, at least 1
- additional blood relative had aneurysms. The 43 males and 11 females
- originated from 50 different families and 5 different nationalities.
- Only one amino acid substitution likely to have functional significance,
- a gly136-to-arg mutation, was found (see 120180.0018). Results indicated
- that mutations in type III procollagen are the cause of only about 2% of
- aortic aneurysms.
-
- As part of a review of abdominal aortic aneurysm as a multifactorial
- process, Henney (1993) reviewed family studies and the molecular
- genetics. In a review focused on surgical aspects, Ernst (1993)
- commented that 'there is little support for atherosclerosis as the
- unitary cause...several factors appear to have an important role,
- including familial clustering...'
-
- Through questionnaire and telephone inquiries, Verloes et al. (1995)
- collected family data on 324 probands with abdominal aortic aneurysm and
- determined multigenerational pedigrees on 313 families, including 39
- with multiple affected patients. There were 276 sporadic cases (264 men;
- 12 women); 81 cases belonged to multiplex pedigrees (76 men; 5 women).
- The familial male cases showed a significantly earlier age at rupture
- and a greater rupture rate as compared with sporadic male cases, as well
- as a tendency (p less than 0.05) towards earlier age of diagnosis.
- Relative risk for male sibs of a male patient was 18. Segregation
- analysis with the mixed model gave single gene effect with dominant
- inheritance as the most likely explanation for the familial occurrence.
- The frequency of the morbid allele was 1:250, and its age-related
- penetrance was not higher than 0.4.
-
- Baird et al. (1995) collected information from 126 probands with
- abdominal aortic aneurysm and 100 controls (cataract surgery patients)
- concerning AAA. Of 427 sibs of probands, 19 (4.4%) had probable or
- definite AAA, compared with 5 (1.1%) of 451 sibs of controls. The
- lifetime cumulative risks of AAA at age 83 were 11.7% and 7.5%,
- respectively. The risk of AAA began at an earlier age and increased more
- rapidly for probands' sibs than for controls' sibs. The risk comparison,
- based on the results of ultrasound screening of 54 geographically
- accessible sibs probands and the 100 controls, showed a similar pattern.
- AAA on ultrasound was found in 10 sibs of probands, or 19%, compared to
- 8% of controls.
-
- *FIELD* SA
- Gatalica et al. (1992); Norrgard et al. (1985); Norrgard et al. (1984)
- *FIELD* RF
- 1. Baird, P. A.; Sadovnick, A. D.; Yee, I. M. L.; Cole, C. W.; Cole,
- L.: Sibling risks of abdominal aortic aneurysm. Lancet 346: 601-604,
- 1995.
-
- 2. Borkett-Jones, H. J.; Stewart, G.; Chilvers, A. S.: Abdominal
- aortic aneurysms in identical twins. J. Roy. Soc. Med. 81: 471-472,
- 1988.
-
- 3. Clifton, M. A.: Familial abdominal aortic aneurysms. Brit. J.
- Surg. 64: 765-766, 1977.
-
- 4. Collin, J.; Araujo, L.; Walton, J.; Lindsell, D.: Oxford screening
- programme for abdominal aortic aneurysm in men aged 65 to 74 years. Lancet II:
- 613-615, 1988.
-
- 5. Darling, R. C., III; Brewster, D. C.; Darling, R. C.; LaMuraglia,
- G. M.; Moncure, A. C.; Cambria, R. P.; Abbott, W. M.: Are familial
- abdominal aortic aneurysms different?. J. Vasc. Surg. 10: 39-43,
- 1989.
-
- 6. Ernst, C. B.: Abdominal aortic aneurysm. New Eng. J. Med. 328:
- 1167-1172, 1993.
-
- 7. Fitzgerald, P.; Ramsbottom, D.; Burke, P.; Grace, P.; McAnen, O.;
- Croke, D. T.; Collins, P.; Johnson, A.; Bouchier-Hayes, D.: Abdominal
- aortic aneurysm in the Irish population. Br. J. Surg. 82: 483-486,
- 1995.
-
- 8. Gatalica, Z.; Gibas, Z.; Martinez-Hernandez, A.: Dissecting aortic
- aneurysm as a complication of generalized fibromuscular dysplasia. Hum.
- Path. 23: 586-588, 1992.
-
- 9. Henney, A. M.: Abdominal aortic aneurysm: molecular genetics. Lancet 341:
- 216-217, 1993.
-
- 10. Johansen, K.; Koepsell, T.: Familial tendency for abdominal aortic
- aneurysms. J.A.M.A. 256: 1934-1936, 1986.
-
- 11. Johnson, G., Jr.; Avery, A.; McDougal, E. G.; Burnham, S. J.;
- Keagy, B. A.: Aneurysms of the abdominal aorta: incidence in blacks
- and whites in North Carolina. Arch. Surg. 120: 1138-1140, 1985.
-
- 12. Kontusaari, S.; Kuivaniemi, H.; Tromp, G.; Grimwood, R.; Prockop,
- D. J.: A single base mutation in the type III procollagen gene (COL3A1)
- on chromosome 2q that causes familial aneurysms. (Abstract) Cytogenet.
- Cell Genet. 51: 1024-1025, 1989.
-
- 13. Kontusaari, S.; Tromp, G.; Kuivaniemi, H.; Romanic, A. M.; Prockop,
- D. J.: A mutation in the gene for type III procollagen (COL3A1) in
- a family with aortic aneurysms. J. Clin. Invest. 86: 1465-1473,
- 1990.
-
- 14. Kuivaniemi, H.; Tromp, G.; Prockop, D. J.: Genetic causes of
- aortic aneurysms: unlearning at least part of what the textbooks say. J.
- Clin. Invest. 88: 1441-1444, 1991.
-
- 15. Loosemore, T. M.; Child, A. H.; Dormandy, J. A.: Familial abdominal
- aortic aneurysms. J. Roy. Soc. Med. 81: 472-473, 1988.
-
- 16. Majumder, P. P.; St. Jean, P. L.; Ferrell, R. E.; Webster, M.
- W.; Steed, D. L.: On the inheritance of abdominal aortic aneurysm. Am.
- J. Hum. Genet. 48: 164-170, 1991.
-
- 17. Norrgard, O.; Angquist, K.-A.; Johnson, O.: Familial aortic aneurysms:
- serum concentrations of triglyceride, cholesterol, HDL-cholesterol
- and (VLDL + LDL)-cholesterol. Brit. J. Surg. 72: 113-116, 1985.
-
- 18. Norrgard, O.; Rais, O.; Angquist, K. A.: Familial occurrence
- of abdominal aortic aneurysms. Surgery 95: 650-656, 1984.
-
- 19. Tilson, M. D.; Seashore, M. R.: Fifty families with abdominal
- aortic aneurysms in two or more first-order relatives. Am. J. Surg. 147:
- 551-553, 1984.
-
- 20. Tromp, G.; Wu, Y.; Prockop, D. J.; Madhatheri, S. L.; Kleinert,
- C.; Earley, J. J.; Zhuang, J.; Norrgard, O.; Darling, R. C.; Abbott,
- W. M.; Cole, C. W.; Jaakkola, P.; Ryynanen, M.; Pearce, W. H.; Yao,
- J. S. T.; Majamaa, K.; Smullens, S. N.; Gatalica, Z.; Ferrell, R.
- E.; Jimenez, S. A.; Jackson, C. E.; Michels, V. V.; Kaye, M.; Kuivaniemi,
- H.: Sequencing of cDNA from 50 unrelated patients reveals that mutations
- in the triple-helical domain of type III procollagen are an infrequent
- cause of aortic aneurysms. J. Clin. Invest. 91: 2539-2545, 1993.
-
- 21. Verloes, A.; Sakalihasan, N.; Koulischer, L.; Limet, R.: Aneurysms
- of the abdominal aorta: familial and genetic aspects in three hundred
- thirteen pedigrees. J. Vas. Surg. 21: 646-655, 1995.
-
- 22. Ward, A. S.: Aortic aneurysmal disease: a generalized dilating
- diathesis?. Arch. Surg. 127: 990-991, 1992.
-
- *FIELD* CS
-
- Vascular:
- Abdominal aortic aneurysm;
- Generalized dilating diathesis
-
- Misc:
- Estimated 11.6-fold increase among persons with an affected first-degree
- relative
-
- Inheritance:
- Autosomal dominant vs. recessive at an autosomal major locus or multifactorial;
- COL3A1 gene (120180.0004) mutations cause about 2%
-
- *FIELD* CN
- Clair A. Francomano - updated: 5/12/1995
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 10/02/1996
- terry: 10/24/1995
- mark: 7/11/1995
- warfield: 4/6/1994
- mimadm: 3/11/1994
- carol: 7/13/1993
-
- *RECORD*
- *FIELD* NO
- 100100
- *FIELD* TI
- 100100 ABDOMINAL MUSCLES, ABSENCE OF, WITH URINARY TRACT ABNORMALITY AND
- CRYPTORCHIDISM
- PRUNE BELLY SYNDROME
- *FIELD* TX
- This condition was first described by Frolich (1839). The appellation
- 'prune belly syndrome' is descriptive because the intestinal pattern is
- evident through the thin, lax, protruding abdominal wall in the infant
- (Osler, 1901). (Osler did not use the term 'prune belly.' His article on
- this subject and one 'on a family form of recurring epistaxis,
- associated with multiple telangiectases of the skin and mucous
- membranes'--see 187300--appeared successively in the November 1901 issue
- of the Johns Hopkins Hospital Bulletin. Osler wrote: 'In the summer of
- 1897 a case of remarkable distension of the abdomen was admitted to the
- wards, with greatly distended bladder, and on my return in September,
- Dr. Futcher, knowing that I would be interested in it, sent for the
- child.') The full syndrome probably occurs only in males (Williams and
- Burkholder, 1967). Multiple cases (of the full syndrome) in families
- have rarely been reported, and the mode of inheritance, indeed whether
- this is a mendelian condition, is still unclear. Autosomal recessive
- inheritance is suggested by some reports. In Lebanon, where the rate of
- consanguinity is high, Afifi et al. (1972) described an affected
- offspring of first-cousin parents. Garlinger and Ott (1974) described 2
- affected brothers in 1 family and 2 affected male cousins in a second,
- and found 3 other reports of affected sibs, 2 of affected cousins and 1
- of concordant male twins. In the first family the parents were
- nonconsanguineous. In the second family the affected boys' mothers were
- half-sisters; they had different maternal grandmothers. If this is an
- X-linked recessive, multiple affected brothers should be observed. If
- the disorder is due to fresh dominant mutation in each case, the
- male-limitation would be unexpected but not impossible. In British
- Columbia, Baird and MacDonald (1981) found a frequency of 1 in 29,231
- live births. This malformation syndrome is similar to Poland syndrome
- (173800) in being rather consistently reproduced in many cases but
- having no clearly demonstrable mendelian basis. A possibly related
- syndrome was described in a single patient by Texter and Murphy (1968).
- The triad consisted of absence of the right testis, kidney, and rectus
- abdominis muscle. King and Prescott (1978) presented evidence to support
- the suggestion that the maldevelopment of the abdominal musculature and
- abdominal laxity are secondary phenomena, the primary event being marked
- distension of the abdomen in the fetal period because of obstruction of
- the urinary tract. Likewise, Pagon et al. (1979) suggested that the
- abdominal muscle deficiency is secondary to fetal abdominal distension
- of various causes, most often perhaps, urethral obstruction with
- enlarged bladder. 'Prune belly' occurs, in the main, as a consequence of
- posterior urethral valves; thus the predominance as a male-limited
- multifactorial trait. Gaboardi et al. (1982) reported 2 brothers and a
- sister with prune belly syndrome with bilateral hydronephrosis,
- megaureter and megabladder, but no urethral stenosis. A better prognosis
- than is usually thought to obtain was suggested by the series of 19
- patients reported by Burke et al. (1969). Greskovich and Nyberg (1988)
- gave a review in which they stated incorrectly that the term prune belly
- syndrome was coined by Osler.
-
- *FIELD* SA
- Burton and Dillard (1984); Harley et al. (1972); Lee (1977); Monie
- and Monie (1979); Riccardi and Grum (1977); Roberts (1956); Welch
- and Kearney (1974); Woodhouse et al. (1982)
- *FIELD* RF
- 1. Afifi, A. K.; Rebeiz, J.; Mire, J.; Andonian, S. J.; Der Kaloustian,
- V. M.: The myopathology of the prune belly syndrome. J. Neurol.
- Sci. 15: 153-166, 1972.
-
- 2. Baird, P. A.; MacDonald, E. C.: An epidemologic study of congenital
- malformations of the anterior abdominal wall in more than half a million
- consecutive live births. Am. J. Hum. Genet. 33: 470-478, 1981.
-
- 3. Burke, E. C.; Shin, M. H.; Kelalis, P. P.: Prune belly syndrome:
- clinical findings and survival. Am. J. Dis. Child. 117: 668-671,
- 1969.
-
- 4. Burton, B. K.; Dillard, R. G.: Prune belly syndrome: observations
- supporting the hypothesis of abdominal overdistention. Am. J. Med.
- Genet. 17: 669-672, 1984.
-
- 5. Frolich, F.: Der Mangel der Muskeln, insbesondere der Seitenbauchmuskeln.
- Dissertation: Wurzburg (pub.) 1839.
-
- 6. Gaboardi, F.; Sterpa, A.; Thiebat, E.; Cornali, R.; Manfredi, M.;
- Bianchi, C.; Giacomoni, M. A.; Bertagnoli, L.: Prune-belly syndrome:
- report of three siblings. Helv. Paediat. Acta 37: 283-288, 1982.
-
- 7. Garlinger, P.; Ott, J.: Prune belly syndrome: possible genetic
- implications. Birth Defects Orig. Art. Ser. X(8): 173-180, 1974.
-
- 8. Greskovich, F. J., III; Nyberg, L. M., Jr.: The prune belly syndrome:
- a review of its etiology, defects, treatment and prognosis. J. Urol. 140:
- 707-712, 1988.
-
- 9. Harley, L. M.; Chen, Y.; Rattner, W. H.: Prune belly syndrome. J.
- Urol. 108: 174-176, 1972.
-
- 10. King, C. R.; Prescott, G.: Pathogenesis of the prune-belly anomaly. J.
- Pediat. 93: 273-274, 1978.
-
- 11. Lee, S. M.: Prune-belly syndrome in a 54-year-old man. J.A.M.A. 237:
- 2216-2217, 1977.
-
- 12. Monie, I. W.; Monie, B. J.: Prune-belly syndrome and fetal ascites. Teratology 19:
- 111-117, 1979.
-
- 13. Osler, W.: Congenital absence of the abdominal muscles with distended
- and hypertrophied urinary bladder. Bull. Johns Hopkins Hosp. 12:
- 331-333, 1901.
-
- 14. Pagon, R. A.; Smith, D. W.; Shepard, T. H.: Urethral obstruction
- malformation complex: a cause of abdominal deficiency and the 'prune
- belly.'. J. Pediat. 94: 900-906, 1979.
-
- 15. Riccardi, V. M.; Grum, C. M.: The prune belly anomaly: heterogeneity
- and superficial X-linkage mimicry. J. Med. Genet. 14: 266-270, 1977.
-
- 16. Roberts, P.: Congenital absence of the abdominal muscles with
- associated abnormalities of the genito-urinary tract. Arch. Dis.
- Child. 31: 236-239, 1956.
-
- 17. Texter, J. H.; Murphy, G. P.: The right-sided syndrome: congenital
- absence of the right testis, kidney and rectus: urologic diagnosis
- and treatment. Johns Hopkins Med. J. 122: 224-228, 1968.
-
- 18. Welch, K. J.; Kearney, G. P.: Abdominal musculature deficiency
- syndrome: prune belly. J. Urol. 111: 693-700, 1974.
-
- 19. Williams, D. I.; Burkholder, G. V.: The prune belly syndrome. J.
- Urol. 98: 244-251, 1967.
-
- 20. Woodhouse, C. R. J.; Ransley, P. G.; Innes-Williams, D.: Prune
- belly syndrome--report of 47 cases. Arch. Dis. Child. 57: 856-859,
- 1982.
-
- *FIELD* CS
-
- Abdomen:
- Absent abdominal musculature;
- Visible intestinal pattern;
- Thin, lax, protruding abdominal wall
-
- Skin:
- Wrinkled abdominal skin
-
- GU:
- Distended bladder;
- Fetal urinary tract obstruction;
- Posterior urethral valves;
- Hydronephrosis;
- Hydroureter;
- Cryptorchidism
-
- GI:
- Imperforate anus
-
- Thorax:
- Flared ribs;
- Pectus excavatum/carinatum
-
- Limbs:
- Club foot
-
- Joints:
- Congenital hip dislocation
-
- Misc:
- Oligohydramnios
-
- Cardiac:
- Congenital heart defect;
- Patent ductus arteriosus
-
- Inheritance:
- ? Autosomal dominant;
- Autosomal recessive suggested by some reports
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 02/13/1997
- terry: 2/13/1997
- carol: 7/12/1996
- mimadm: 4/18/1994
- carol: 2/13/1994
- carol: 8/25/1992
- supermim: 3/16/1992
- carol: 9/4/1990
-
- *RECORD*
- *FIELD* NO
- 100200
- *FIELD* TI
- 100200 ABDUCENS PALSY
- *FIELD* TX
- This is a form of hereditary strabismus. Affected persons in 2 or more
- generations have been reported (Chavasse, 1938; Francois, 1961). Nuclear
- aplasia has been found in some cases (Phillips et al., 1932). Abducens
- palsy also occurs as part of the Moebius syndrome (157900).
-
- *FIELD* RF
- 1. Chavasse, F. B.: The ocular palsies. Trans. Ophthal. Soc. U.K. 58:
- 493 only, 1938.
-
- 2. Francois, J.: Heredity in Ophthalmology. St. Louis: C. V. Mosby
- (pub.) 1961. Pp. 280 only.
-
- 3. Phillips, W. H.; Dirion, J. K.; Graves, G. O.: Congenital bilateral
- palsy of abducens. Arch. Ophthal. 8: 355-364, 1932.
-
- *FIELD* CS
-
- Eyes:
- Abducens palsy;
- Strabismus
-
- Neuro:
- Abducens nucleus aplasia
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- davew: 8/15/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 100300
- *FIELD* TI
- *100300 ABSENCE DEFECT OF LIMBS, SCALP, AND SKULL
- ADAMS-OLIVER SYNDROME;;
- CONGENITAL SCALP DEFECTS WITH DISTAL LIMB REDUCTION ANOMALIES
- *FIELD* TX
- The proband described by Adams and Oliver (1945) had absence of the
- lower extremities below the mid-calf region and absence of all digits
- and some of the metacarpals of the right hand; a denuded ulcerated area
- on the vertex of the scalp present at birth; and a bony defect of the
- skull underlying the scalp defect. The skin and skull lesions were
- similar to those of aplasia cutis congenita (107600, 207700). The
- proband had 4 unaffected brothers and a sister and brother with
- identical defects of limb, scalp and skull. The father was born with
- absence of toes 2-5 on the left foot, with short terminal phalanges of
- all fingers, and with a scalp defect. The father was 1 of 10 children of
- whom 3 others had defects of the extremities. The father's father was
- said to have had short fingers. The proband's parents were not related.
- In a family described by Scribanu and Temtamy (1975), variable
- expressivity and reduced penetrance were evident; cutis marmorata was
- striking in the proband, a 3-year-old male. Toriello et al. (1988) also
- described cutis marmorata telangiectatica congenita (CMTC; 219250) in a
- child with the Adams-Oliver syndrome. The mother also had CMTC without
- the other features of the Adams-Oliver syndrome. These vascular changes
- in the skin may indicate that the features of Adams-Oliver syndrome are
- 'vascular disruption sequences.' The family reported by Bonafede and
- Beighton (1979) added substantial support to dominant inheritance, with
- one instance of male-to-male transmission. Kuster et al. (1988)
- described 10 cases, 7 of them in 2 families and 3 sporadic cases. They
- found 11 families and 19 sporadic cases reported in the literature. They
- suggested that the important differential diagnoses are the syndrome of
- scalp defect and postaxial polydactyly (181250), the amniotic band
- sequence (which is usually nonmendelian), and the Bart type of
- epidermolysis bullosa dystrophica (132000). Koiffmann et al. (1988)
- recorded an experience which suggested autosomal recessive inheritance
- of a disorder identical to the autosomal dominant form. Their patient
- had the congenital scalp defect with hypoplastic fingers and toes. The
- parents were unaffected first cousins. Among 7 sibs, 3 sisters and 2
- brothers were normal, whereas 2 brothers born with the same scalp defect
- died as a consequence of bleeding from this abnormal area. Sybert (1989)
- concluded that 'most, if not all, instances of isolated ACC of the scalp
- are the result of an autosomal dominant gene, that ACC of the body wall
- + limb defects is an extremely heterogeneous group among which there may
- be inherited disorders of all Mendelian types as well as sporadic and
- nongenetic causes, and that ACC limited to the scalp in association with
- limb defects is most often inherited as an autosomal dominant.' Sybert
- (1985) and Frieden (1986) gave comprehensive reviews. Jaeggi et al.
- (1990) reported an affected mother and child as well as a third sporadic
- case. They discussed the probable pathogenesis of the disorder by
- vascular disruption as suggested by Toriello et al. (1988). Cutis
- marmorata and dilated scalp veins further point to a vascular disorder.
- Jaeggi et al. (1990) stated that among the 31 reported patients with the
- full syndrome, major hemorrhage from the scalp defect occurred in 10,
- with 2 fatalities. Local infection was noted in 7 babies, with 1 case of
- fatal meningitis. Only 30% of the patients had surgical treatment of
- their scalp defects by skin grafting. Der Kaloustian et al. (1991)
- described 2 families having members affected with the Poland anomalad
- and the Adams-Oliver syndrome. They hypothesized that the Poland
- anomalad and the Adams-Oliver syndrome result from the interruption of
- early embryonic blood supply in the subclavian arteries, and that the
- gene predisposing to this interruption follows an autosomal dominant
- pattern of inheritance. Hoyme et al. (1992) reported that 2 additional
- individuals in family 2 of Der Kaloustian et al. (1991) had the Poland
- sequence with no findings suggesting Adams-Oliver syndrome. Whitley and
- Gorlin (1991) provided a follow-up on the family studied by Adams and
- Oliver (1945); the gene had been transmitted to a member of a fourth
- generation. They found reports of 81 cases in 32 families with
- approximately equal distribution between males and females. Vertical
- transmission in at least 8 families was consistent with autosomal
- dominant inheritance. Despite large defects of the cranium, central
- nervous system abnormalities have not been found in this disorder and
- intellectual development appears to be normal. On the basis of the case
- of a 10-year-old male, Chitayat et al. (1992) suggested that acrania is
- a severe form of aplasia cutis congenita and is within the spectrum of
- Adams-Oliver syndrome. In acrania, the flat bones of the cranial vault
- are absent, whereas the bones at the base of the skull are normal. The
- patient was a sporadic case. Bamforth et al. (1994) found this syndrome
- in a mother and her 3 children with variable scalp defects and limb
- defects. Other anomalies included congenital heart disease,
- microcephaly, epilepsy, mental retardation, arrhinencephaly,
- hydrocephaly, anatomic bronchial anomalies, and renal anomalies. The 3
- children were by 2 different fathers.
-
- Zapata et al. (1995) reported 2 patients with Adams-Oliver syndrome and
- congenital cardiac malformations. A literature review demonstrated that
- 13.4% of individuals with this syndrome have congenital heart anomalies.
-
- *FIELD* SA
- Burton et al. (1976); Fryns (1987); McMurray et al. (1977)
- *FIELD* RF
- 1. Adams, F. H.; Oliver, C. P.: Hereditary deformities in man due
- to arrested development. J. Hered. 36: 3-7, 1945.
-
- 2. Bamforth, J. S.; Kaurah, P.; Byrne, J.; Ferreira, P.: Adams Oliver
- syndrome: a family with extreme variability in clinical expression.
- Am. J. Med. Genet. 49: 393-396, 1994.
-
- 3. Bonafede, R. P.; Beighton, P.: Autosomal dominant inheritance
- of scalp defects with ectrodactyly. Am. J. Med. Genet. 3: 35-41,
- 1979.
-
- 4. Burton, B. K.; Hauser, L.; Nadler, H. L.: Congenital scalp defects
- with distal limb anomalies: report of a family. J. Med. Genet. 13:
- 466-468, 1976.
-
- 5. Chitayat, D.; Meunier, C.; Hodgkinson, K. A.; Robb, L.; Azouz,
- M.: Acrania: a manifestation of the Adams-Oliver syndrome. Am.
- J. Med. Genet. 44: 562-566, 1992.
-
- 6. Der Kaloustian, V. M.; Hoyme, H. E.; Hogg, H.; Entin, M. A.; Guttmacher,
- A. E.: Possible common pathogenetic mechanisms for Poland sequence
- and Adams-Oliver syndrome. Am. J. Med. Genet. 38: 69-73, 1991.
-
- 7. Frieden, I.: Aplasia cutis congenita: a clinical review and proposal
- for classification. J. Am. Acad. Derm. 14: 646-660, 1986.
-
- 8. Fryns, J. P.: Congenital scalp defects with distal limb reduction
- anomalies. J. Med. Genet. 24: 493-496, 1987.
-
- 9. Hoyme, H. E.; Entin, M. A.; Der Kaloustian, V. M.; Hogg, H.; Guttmacher,
- A. E.: Possible common pathogenetic mechanisms for Poland sequence
- and Adams-Oliver syndrome: an additional clinical observation. (Letter) Am.
- J. Med. Genet. 42: 398-399, 1992.
-
- 10. Jaeggi, E.; Kind, C.; Morger, R.: Congenital scalp and skull
- defects with terminal transverse limb anomalies (Adams-Oliver syndrome):
- report of three additional cases. Europ. J. Pediat. 149: 565-566,
- 1990.
-
- 11. Koiffmann, C. P.; Wajntal, A.; Huyke, B. J.; Castro, R. M.: Congenital
- scalp skull defects with distal limb anomalies (Adams-Oliver syndrome--McKusick
- 10030): further suggestion of autosomal recessive inheritance. Am.
- J. Med. Genet. 29: 263-268, 1988.
-
- 12. Kuster, W.; Lenz, W.; Kaariainen, H.; Majewski, F.: Congenital
- scalp defects with distal limb anomalies (Adams-Oliver syndrome):
- report of ten cases and review of the literature. Am. J. Med. Genet. 31:
- 99-115, 1988.
-
- 13. McMurray, B. R.; Martin, L. W.; Dignan, P. S. J.; Fogelson, M.
- H.: Hereditary aplasia cutis congenita and associated defects: three
- instances in one family and a survey of reported cases. Clin. Pediat. 16:
- 610-614, 1977.
-
- 14. Scribanu, N.; Temtamy, S. A.: Syndrome of aplasia cutis congenita
- with terminal transverse defects of limbs. J. Pediat. 87: 79-82,
- 1975.
-
- 15. Sybert, V. P.: Aplasia cutis congenita: a report of 12 new families
- and review of the literature. Pediat. Derm. 3: 1-14, 1985.
-
- 16. Sybert, V. P.: Congenital scalp defects with distal limb anomalies
- (Adams-Oliver Syndrome--McKusick 10030): further suggestion of autosomal
- recessive inheritance. (Letter) Am. J. Med. Genet. 32: 266-267,
- 1989.
-
- 17. Toriello, H. V.; Graff, R. G.; Florentine, M. F.; Lacina, S.;
- Moore, W. D.: Scalp and limb defects with cutis marmorata telangiectatica
- congenita: Adams-Oliver syndrome?. Am. J. Med. Genet. 29: 269-276,
- 1988.
-
- 18. Whitley, C. B.; Gorlin, R. J.: Adams-Oliver syndrome revisited.
- Am. J. Med. Genet. 40: 319-326, 1991.
-
- 19. Zapata, H. H.; Sletten, L. J.; Pierpont, M. E. M.: Congenital
- cardiac malformations in Adams-Oliver syndrome. Clin. Genet. 47:
- 80-84, 1995.
-
- *FIELD* CS
-
- Limbs:
- Absent lower leg below mid-calf;
- Absent fingers;
- Absent metacarpals;
- Absent toes;
- Short finger terminal phalanges
-
- Skin:
- Congenital scalp defect;
- Cutis marmorata;
- Dilated scalp veins
-
- Skull:
- Skull defect underlying scalp defect
-
- Heme:
- Hemorrhage from scalp defect
-
- Misc:
- Scalp defect local infection;
- Fatal meningitis
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 6/8/1995
- mimadm: 3/11/1994
- carol: 3/7/1994
- carol: 12/18/1992
- supermim: 3/16/1992
- carol: 2/27/1992
-
- *RECORD*
- *FIELD* NO
- 100500
- *FIELD* TI
- *100500 ACANTHOCYTOSIS WITH NEUROLOGIC DISORDER
- NEUROACANTHOCYTOSIS;;
- CHOREOACANTHOCYTOSIS;;
- LEVINE-CRITCHLEY SYNDROME
- *FIELD* TX
- In addition to the form of acanthocytosis that accompanies
- abetalipoproteinemia (200100), Critchley et al. (1967) described an
- adult form of acanthocytosis associated with neurologic abnormalities
- and apparently normal serum lipoproteins. The neurologic manifestations
- resembled those of the Gilles de la Tourette syndrome (137580) or
- Huntington disease (143100). Five of 10 sibs had neurologic
- manifestations. A niece had acanthocytes and a neurologic disorder
- suggesting Friedreich ataxia. The same disorder was probably reported by
- Estes et al. (1967) in a family in which 15 persons in 3 generations had
- some degree of neuronal impairment and 9 of these had acanthocytosis.
- Levine et al. (1968) concluded that the predominant neurologic
- involvement is neuronal.
-
- Critchley et al. (1970) reported a single case from England, a woman who
- showed self-mutilation of the tongue, lips and cheeks. Another family
- was reported by Aminoff (1972). Wasting of girdle and proximal limb
- muscles, absent tendon reflexes, and disturbance of bladder function
- were other features.
-
- Bird et al. (1978) described a family in which 3 offspring (2 males, 1
- female) of unaffected consanguineous parents had a progressive
- neurologic disorder characterized primarily by chorea, which led to
- death in the fourth or fifth decades. No malabsorption or abnormalities
- of serum beta-lipoprotein were found, but erythrocyte acanthocytosis was
- present. At postmortem examination, marked neuronal loss and gliosis of
- the caudate and putamen were demonstrated. The disorder in this family
- seems to have been recessive, whereas that in the family of Estes et al.
- (1967) and Levine et al. (1968) was seemingly dominant. Thus,
- heterogeneity may exist in the category of neurologic disease and
- acanthocytosis. Vance et al. (1987) reviewed the literature and
- concluded that out of 9 families in which there were 2 or more affected
- members, 2 were probably autosomal dominant and 7 were autosomal
- recessive (see 200150).
-
- In a patient with acanthocytosis and degeneration of the basal ganglia,
- Copeland et al. (1982) found an abnormally high level of a protein in
- the 100,000 MW range on 2-D O'Farrell gel electrophoresis of red cell
- membranes. This patient was from the family reported by Bird et al.
- (1978) (Motulsky, 1982).
-
- In 3 patients with neuroacanthocytosis, Rinne et al. (1994) demonstrated
- reduced neuronal density in the substantia nigra. As in Parkinson
- disease, the ventral lateral region was most severely affected, but with
- a slightly more diffuse distribution.
-
- Kartsounis and Hardie (1996) reviewed the clinical features of 19
- previously reported cases of neuroacanthocytosis and found that the most
- consistent neurologic findings were impairment of frontal lobe function
- and psychiatric morbidity, in a pattern suggesting subcortical dementia.
-
- Sakai et al. (1985) urged Levine-Critchley syndrome as the best
- designation for this disorder. They felt that choreoacanthocytosis is
- inappropriate because tics, dystonia, or parkinsonism may dominate the
- clinical picture (Spitz et al., 1985). Neuroacanthocytosis is also
- inappropriate because it might include the Bassen-Kornzweig syndrome
- (200100). Jankovic et al. (1985) suggested that there are 2 other
- neuroacanthocytoses: that associated with hypobetalipoproteinemia
- (107730) and that which is part of the McLeod syndrome, an X-linked
- disorder (314850).
-
- See Kay (1991) for a discussion of band 3 protein (109270) as the site
- of the mutation in choreoacanthocytosis.
-
- *FIELD* SA
- Betts et al. (1970); Kito et al. (1980)
- *FIELD* RF
- 1. Aminoff, M. J.: Acanthocytosis and neurological disease. Brain 95:
- 749-760, 1972.
-
- 2. Betts, J. J.; Nicholson, J. T.; Critchley, E. M. R.: Acanthocytosis
- with normolipoproteinaemia: biophysical aspects. Postgrad. Med.
- J. 46: 702-707, 1970.
-
- 3. Bird, T. D.; Cederbaum, S.; Valpey, R. W.; Stahl, W. L.: Familial
- degeneration of the basal ganglia with acanthocytosis: a clinical,
- neuropathological and neurochemical study. Ann. Neurol. 3: 253-258,
- 1978.
-
- 4. Copeland, B. R.; Todd, S. A.; Furlong, C. E.: High resolution
- two-dimensional gel electrophoresis of human erythrocyte membrane
- proteins. Am. J. Hum. Genet. 34: 15-31, 1982.
-
- 5. Critchley, E. M. R.; Betts, J. J.; Nicholson, J. T.; Weatherall,
- D. J.: Acanthocytosis, normolipoproteinaemia and multiple tics. Postgrad.
- Med. J. 46: 698-701, 1970.
-
- 6. Critchley, E. M. R.; Clark, D. B.; Wikler, A.: An adult form of
- acanthocytosis. Trans. Am. Neurol. Assoc. 92: 132-137, 1967.
-
- 7. Estes, J. W.; Morley, T. J.; Levine, I. M.; Emerson, C. P.: A
- new hereditary acanthocytosis syndrome. Am. J. Med. 42: 868-881,
- 1967.
-
- 8. Jankovic, J.; Killian, J. M.; Spitz, M. C.: Neuroacanthocytosis
- syndrome and choreoacanthocytosis (Levine-Critchley syndrome). (Letter) Neurology 35:
- 1679, 1985.
-
- 9. Kartsounis, L. D.; Hardie, R. J.: The pattern of cognitive impairments
- in neuroacanthocytosis: a frontosubcortical dementia. Arch. Neurol. 53:
- 77-80, 1996.
-
- 10. Kay, M. M. B.: Band 3 in aging and neurological disease. Ann.
- N.Y. Acad. Sci. 621: 179-204, 1991.
-
- 11. Kito, S.; Itoga, E.; Hiroshige, Y.; Matsumoto, N.; Miwa, S.:
- A pedigree of amyotrophic chorea with acanthocytosis. Arch. Neurol. 37:
- 514-517, 1980.
-
- 12. Levine, I. M.; Estes, J. W.; Looney, J. M.: Hereditary neurological
- disease with acanthocytosis: a new syndrome. Arch. Neurol. 19:
- 403-409, 1968.
-
- 13. Motulsky, A. G.: Personal Communication. Seattle, Washington
- 4/21/1982.
-
- 14. Rinne, J. O.; Daniel, S. E.; Scaravilli, F.; Harding, A. E.; Marsden,
- C. D.: Nigral degeneration in neuroacanthocytosis. Neurology 44:
- 1629-1632, 1994.
-
- 15. Sakai, T.; Iwashita, H.; Kakugawa, M.: Neuroacanthocytosis syndrome
- and choreoacanthocytosis (Levine-Critchley syndrome). (Letter) Neurology 35:
- 1679, 1985.
-
- 16. Spitz, M. C.; Jankovic, J.; Killian, J. M.: Familial tic disorder,
- parkinsonism, motor neuron disease, and acanthocytosis: a new syndrome.
- Neurology 35: 366-370, 1985.
-
- 17. Vance, J. M.; Pericak-Vance, M. A.; Bowman, M. H.; Payne, C. S.;
- Fredane, L.; Siddique, T.; Roses, A. D.; Massey, E. W.: Chorea-acanthocytosis:
- a report of three new families and implications for genetic counselling.
- Am. J. Med. Genet. 28: 403-410, 1987.
-
- *FIELD* CS
-
- Neuro:
- Chorea;
- Tics;
- Dystonia;
- Parkinsonism;
- Absent tendon reflexes;
- Abnormal bladder function;
- Self-mutilation of tongue, lips and cheeks
-
- Muscle:
- Myopathy;
- Girdle and proximal limb muscle wasting
-
- Misc:
- Adult form of acanthocytosis
-
- Lab:
- Acanthocytosis;
- Normal serum lipoproteins;
- Neuronal loss and gliosis of the caudate and putamen
-
- Inheritance:
- Autosomal dominant;
- also autosomal recessive form
-
- *FIELD* CN
- Orest Hurko - updated: 4/1/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 04/15/1996
- mark: 4/1/1996
- terry: 4/1/1996
- terry: 2/15/1996
- carol: 12/12/1994
- warfield: 4/7/1994
- mimadm: 3/11/1994
- carol: 3/10/1993
- supermim: 3/16/1992
- carol: 8/23/1990
-
- *RECORD*
- *FIELD* NO
- 100600
- *FIELD* TI
- *100600 ACANTHOSIS NIGRICANS
- *FIELD* TX
- Acanthosis nigricans consists of thickening and hyperpigmentation of the
- skin of the entire body but especially in flexural areas. In 26 patients
- with malignant acanthosis nigricans (secondary to visceral carcinoma),
- Curth and Aschner (1959) found no other affected persons in the family.
- On the other hand, benign acanthosis nigricans may be inherited as a
- mendelian dominant. Curth and Aschner (1959) had families with
- acanthosis nigricans in successive generations, 3 in 1 family and 2 in 2
- others, including instances of male-to-male transmission. Jung et al.
- (1965) observed affected mother and daughter. Lawrence et al. (1971)
- described a patient with acanthosis nigricans inherited from the father
- and telangiectasia (187300) inherited from the mother. Tasjian and
- Jarratt (1984) observed affected mother and daughter. Skin lesions were
- first noted in infancy. In addition to the association with insulin
- resistance (147670), Seip syndrome (269700), and malignancy, acanthosis
- nigricans can be drug-induced; nicotinic acid, diethylstilbestrol, oral
- contraceptives, and exogenous glucocorticoids have been incriminated.
- Clear mendelian inheritance is seen when acanthosis nigricans is part of
- syndromes, e.g., Seip syndrome. Autosomal dominant acanthosis nigricans
- should be studied for insulin resistance. Schwenk et al. (1986) studied
- a white family in which acanthosis nigricans occurred in a mother and 3
- daughters; insulin binding was normal but insulin response was reduced,
- consistent with a postbinding defect (see 147670). Perhaps one should
- speak of types A1 and A2 of acanthosis nigricans, A1 being the form with
- a defect in the insulin receptor and A2 representing a postbinding
- defect. Seemanova et al. (1992) investigated a family in which at least
- 4 men in 3 generations had a syndrome of obesity, mild mental
- retardation, delayed puberty, macroorchidism, acanthosis nigricans,
- hyperinsulinemia, and, later, overt insulin-resistant diabetes mellitus
- (noninsulin-dependent diabetes mellitus; NIDDM). The patients had
- markedly curly scalp hair and deficient hair of the face and body. Teeth
- were normal. There was normal insulin binding to fibroblasts; however,
- insulin-stimulated RNA synthesis was decreased as compared to that of
- normal control individuals, suggesting a postbinding defect in insulin
- action. The pedigree showed an autosomal dominant pattern of
- inheritance.
-
- Acanthosis nigricans in association with insulin resistance behaves as
- either a dominant (e.g., 147670.0001) or a recessive (e.g.,
- 147670.0004). The polycystic ovary syndrome is sometimes reported. The
- autosomal dominant mutations in the insulin receptor gene are 'dominant
- negatives'; the mutant receptor protein interferes with the function of
- the normal receptor.
-
- Chuang et al. (1995) reported familial acanthosis nigricans affecting a
- 35-year-old woman, her 7-year-old son, and 5-year-old daughter. Absence
- of the eyebrows and eyelashes was also present in the affected members
- of this family. The mother had no axillary hair and her pubic hair was
- sparse. The boy also suffered from congenital heart disease and a
- congenital cataract in the left eye. Chuang et al. (1995) suggested that
- the combination of acanthosis nigricans and ectodermal defects in this
- family may represent a distinct nosologic entity. They referred to the
- hair problem as madarosis (loss of the eyebrows or of the eyelashes).
-
- *FIELD* SA
- Hermann (1955)
- *FIELD* RF
- 1. Chuang, S.-D.; Jee, S.-H.; Chiu, H.-C.; Chen, J.-S.; Lin, J.-T.
- : Familial acanthosis nigricans with madarosis. Brit. J. Derm. 133:
- 104-108, 1995.
-
- 2. Curth, H. O.; Aschner, B. M.: Genetic studies on acanthosis nigricans. Arch.
- Derm. 79: 55-66, 1959.
-
- 3. Hermann, H.: Zur Erbpathologie der Acanthosis nigricans. Z. Menschl.
- Vererb. Konstitutionsl. 33: 193-202, 1955.
-
- 4. Jung, H. D.; Bruns, W.; Wulfert, P.; Mieler, W.: Ein Beitrag zum
- Krankheitsbild der Acanthosis nigricans benigna familiaris. Dtsch.
- Med. Wschr. 90: 1669-1673, 1965.
-
- 5. Lawrence, G.; Thurston, C.; Shultz, K.; Mengel, M. C.: Acanthosis
- nigricans, telangiectasia and diabetes mellitus. Birth Defects Orig.
- Art. Ser. VII(8): 322-323, 1971.
-
- 6. Schwenk, W. F.; Rizza, R. A.; Mandarino, L. J.; Gerich, J. E.;
- Hayles, A. B.; Haymond, M. W.: Familial insulin resistance and acanthosis
- nigricans: presence of a postbinding defect. Diabetes 35: 33-37,
- 1986.
-
- 7. Seemanova, E.; Rudiger, H. W.; Dreyer, M.: Autosomal dominant
- insulin resistance syndrome due to a postbinding defect. Am. J. Med.
- Genet. 44: 705-712, 1992.
-
- 8. Tasjian, D.; Jarratt, M.: Familial acanthosis nigricans. Arch.
- Derm. 120: 1351-1354, 1984.
-
- *FIELD* CS
-
- Skin:
- Benign acanthosis nigricans;
- Thick hyperpigmented flexural area skin
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 04/03/1997
- mark: 10/13/1995
- mimadm: 6/26/1994
- carol: 3/12/1994
- supermim: 3/16/1992
- carol: 2/29/1992
- supermim: 3/20/1990
-
- *RECORD*
- *FIELD* NO
- 100640
- *FIELD* TI
- *100640 ACETALDEHYDE DEHYDROGENASE-1
- ALDEHYDE DEHYDROGENASE-1; ALDH1;;
- ALDH, LIVER CYTOSOLIC
- *FIELD* TX
- Acetaldehyde dehydrogenase (EC 1.2.1.3) is the next enzyme after alcohol
- dehydrogenase (103700) in the major pathway of alcohol metabolism. On
- the basis of population studies of isozyme patterns, Harada et al.
- (1978) proposed that there are 3 loci determining acetaldehyde
- dehydrogenase. They suggested that the rarer of the alleles at the
- postulated ALDH1, ALDH2 and ALDH3 loci have a frequency of 0.022, 0.029
- and 0.151, respectively. The sample numbered only 68 specimens, however.
-
- Harada et al. (1980) presented evidence that aldehyde dehydrogenase is
- polymorphic in Japanese. As in previous studies in Europeans, they found
- 2 isozymes of ALDH in liver specimens of Japanese, but unlike the study
- of specimens of Europeans, they found that 52% of Japanese specimens
- showed absence of the faster migrating isozyme (which has a low Km for
- acetaldehyde). The authors suggested that the intoxicating symptoms
- after alcohol drinking in many Japanese may be due to delayed oxidation
- of acetaldehyde. The lack of ALDH isozyme I with a low Km for aldehyde
- is apparently responsible for the higher blood acetaldehyde levels in
- mongoloid peoples, leading to facial flushing and other vasomotor
- symptoms after alcohol intake. Agarwal et al. (1981) performed a
- population genetic study in Orientals of several different extractions.
- They investigated ALDH isozymes in hair root lysates with a sensitive
- isoelectric focusing method. Between 40 and 80% of the several Oriental
- groups were found to be deficient in isozyme I of ALDH, whereas not a
- single European individual was deficient. Deficiency was invariably
- associated with sensitivity to alcohol. Family studies suggested
- autosomal recessive inheritance of the deficiency. Harada et al. (1981)
- found the deficiency in 43% of Japanese; all deficient persons had
- flushing symptoms and, after alcohol drinking, showed a mean
- concentration of acetaldehyde of 37.3 micromoles as compared with 2.1
- micromoles in nondeficient persons.
-
- Thomas et al. (1982) found low cytosolic acetaldehyde dehydrogenase in
- the liver of alcoholic patients with fatty liver; mitochondrial ALDH was
- normal. Abstaining alcoholics showed persistently low cytosolic ALDH.
- Isoelectric focusing showed that the cytosolic and mitochondrial ALDHs
- are distinct isozymes. Impraim et al. (1982) investigated the basis of
- the lack in about 50% of Orientals of 1 of the 2 major liver ALDH
- isozymes. Consistent with a convention of nomenclature adopted by the
- HGM workshops, ALDH1 is cytosolic and ALDH2 is mitochondrial. It is the
- latter that is missing in Orientals. Inoue et al. (1979) purified and
- partially characterized aldehyde dehydrogenase from human erythrocytes.
- This is the cytosolic form, present in only low concentration in red
- cells. Goedde et al. (1979) proposed that the high frequency of acute
- alcoholic intoxication in Orientals is related to the high frequency of
- persons with absence of ALDH2 liver isozyme. On the other hand,
- Stamatoyannopoulos et al. (1975) suggested that the racial difference in
- alcohol intoxication is due to rapid acetaldehyde formation as a result
- of the highly active atypical alcohol dehydrogenase isozyme found in
- high frequency in Orientals. ALDH1 and ALDH2 have molecular weights of
- 245,000 and 225,000, respectively, and both are tetramers. Structural
- and genetic interrelationships are unknown; e.g., does each consist of a
- single type subunit or do they share a common subunit? Impraim et al.
- (1982) found that the ALDH2 in an 'atypical' Japanese liver was
- enzymatically inactive but immunologically cross-reactive. Thus, a
- structural mutation at the ALDH2 locus is presumably the genetic basis.
-
- Goedde et al. (1983) pointed to the existence of 4 isozymes of
- NAD-dependent aldehyde dehydrogenase, designated ALDH I, II, III, or IV
- according to their decreasing electrophoretic mobility and increasing
- isoelectric point. The frequency of absent ALDH I isozyme varied from
- 69% in Indians of the Ecuador Highlands to 44% in Japanese and 35% in
- Chinese to 0% in Egyptians, Liberians, Kenyans, and Europeans. They
- suggested that deficiency is related to flushing and a slower metabolism
- of acetaldehyde, and in turn a lower frequency of alcoholism and
- alcohol-related problems.
-
- Yoshida et al. (1989) demonstrated that among Caucasians alcohol
- flushing can be related to abnormalities of ALDH1. In 9 unrelated
- Caucasian alcohol flushers, they found 1 who exhibited low activity
- (10-20% of normal) and another who exhibited moderately low activity
- (60%) and altered kinetic properties. The electrophoretic mobilities of
- these 2 samples were not altered. Immunologic quantitation indicated
- that the amount of protein in the 2 samples was not reduced in parallel
- with the enzyme deficiency. In the first case, the daughter of the
- proposita also had very low enzyme activity and alcohol flushing.
-
- ALDH1 is cytosolic, is associated with a low Km for NAD and a high Km
- for acetaldehyde, and is strongly inactivated by disulfiram. ALDH2
- (100650) is mitochondrial, has a high Km for NAD and a low Km for
- acetaldehyde, and is insensitive to disulfiram. About 50% of Orientals
- lack ALDH2 activity but have defective enzyme immunologically related to
- ALDH2 (Yoshida et al., 1984). In some Orientals absence of ALDH1
- activity and the presence of an enzymatically inactive protein is
- demonstrable (Yoshida et al., 1983). Yoshida (1984) concluded that one
- can substitute hair roots for liver biopsy specimens if sample size for
- isoelectric focusing is adjusted using MDH or IDH as an internal
- reference. The liver of humans and other mammals contains 2 major and
- several minor aldehyde dehydrogenase isozymes. The major isozymes are
- ALDH1 of cytosolic origin and ALDH2 of mitochondrial origin. (The
- confusion in the numerology of the aldehyde dehydrogenases is evident.
- ALDH I and ALDH II of Goedde and colleagues is ALDH2 and ALDH1 of other
- workers.) In contrast to the wide prevalence of deficiency of ALDH2
- (called ALDH I by Goedde), variants of ALDH1 (called ALDH II by Goedde)
- are rare; Eckey et al. (1986) described one such variant.
-
- With cDNA probes for Southern blot analysis of somatic cell hybrids, Hsu
- et al. (1985, 1986) assigned the ALDH1 locus to 9q and the ALDH2 locus
- to chromosome 12. Hsu et al. (1989) found that the ALDH1 gene is about
- 53 kb long and is divided into 13 exons which encode 501 amino acid
- residues. A similar intron-exon organization is found in ALDH2 which
- also has 13 exons with 9 of the 12 introns interrupting the coding
- sequence at positions homologous to those in ALDH1. Thus, the 2 isozymes
- appear to have evolved after duplication of a common ancestral gene.
-
- *FIELD* SA
- Harada et al. (1981); Hsu et al. (1985)
- *FIELD* RF
- 1. Agarwal, D. P.; Meier-Tackmann, D.; Harada, S.; Goedde, H. W.;
- Du, R.: Mechanism of biological sensitivity to alcohol: inherited
- deficiency of aldehyde dehydrogenase isoenzyme I in Mongoloids. (Abstract) Sixth
- Int. Cong. Hum. Genet., Jerusalem 102 only, 1981.
-
- 2. Eckey, R.; Agarwal, D. P.; Saha, N.; Goedde, H. W.: Detection
- and partial characterization of a variant form of cytosolic aldehyde
- dehydrogenase isozyme. Hum. Genet. 72: 95-97, 1986.
-
- 3. Goedde, H. W.; Agarwal, D. P.; Harada, S.; Meier-Tackmann, D.;
- Ruofu, D.; Bienzle, U.; Kroeger, A.; Hussein, L.: Population genetic
- studies on aldehyde dehydrogenase isozyme deficiency and alcohol sensitivity.
- Am. J. Hum. Genet. 35: 769-772, 1983.
-
- 4. Goedde, H. W.; Harada, S.; Agarwal, D. P.: Racial differences
- in alcohol sensitivity: a new hypothesis. Hum. Genet. 51: 331-334,
- 1979.
-
- 5. Harada, S.; Agarwal, D. P.; Goedde, H. W.: Isozyme variations
- in acetaldehyde dehydrogenase (E.C. 1.2.1.3) in human tissues. Hum.
- Genet. 44: 181-185, 1978.
-
- 6. Harada, S.; Agarwal, D. P.; Goedde, H. W.: Aldehyde metabolism
- and polymorphism of aldehyde dehydrogenase in Japanese. (Abstract) Sixth
- Int. Cong. Hum. Genet., Jerusalem 103 only, 1981.
-
- 7. Harada, S.; Agarwal, D. P.; Goedde, H. W.: Aldehyde dehydrogenase
- deficiency as cause of facial flushing reaction to alcohol in Japanese.
- (Letter) Lancet II: 982 only, 1981.
-
- 8. Harada, S.; Misawa, S.; Agarwal, D. P.; Goedde, H. W.: Liver alcohol
- dehydrogenase and aldehyde dehydrogenase in the Japanese: isozyme
- variation and its possible role in alcohol intoxication. Am. J.
- Hum. Genet. 32: 8-15, 1980.
-
- 9. Hsu, L. C.; Chang, W.-C.; Yoshida, A.: Genomic structure of the
- human cytosolic aldehyde dehydrogenase gene. Genomics 5: 857-865,
- 1989.
-
- 10. Hsu, L. C.; Tani, K.; Fujiyoshi, T.; Kurachi, K.; Yoshida, A.
- : Cloning of cDNAs for human aldehyde dehydrogenases 1 and 2. Proc.
- Nat. Acad. Sci. 82: 3771-3775, 1985.
-
- 11. Hsu, L. C.; Yoshida, A.; Mohandas, T.: Chromosomal assignment
- of the genes for human aldehyde dehydrogenase 1 (ALDH1) and aldehyde
- dehydrogenase 2 (ALDH2). (Abstract) Cytogenet. Cell Genet. 40:
- 656-657, 1985.
-
- 12. Hsu, L. C.; Yoshida, A.; Mohandas, T.: Chromosomal assignment
- of the genes for human aldehyde dehydrogenase-1 and aldehyde dehydrogenase-2.
- Am. J. Hum. Genet. 38: 641-648, 1986.
-
- 13. Impraim, C.; Wang, G.; Yoshida, A.: Structural mutation in a
- major human aldehyde dehydrogenase gene results in loss of enzyme
- activity. Am. J. Hum. Genet. 34: 837-841, 1982.
-
- 14. Inoue, K.; Nishimukai, H.; Yamasawa, K.: Purification and partial
- characterization of aldehyde dehydrogenase from human erythrocytes.
- Biochim. Biophys. Acta 569: 117-123, 1979.
-
- 15. Stamatoyannopoulos, G.; Chen, S.-H.; Fukui, M.: Liver alcohol
- dehydrogenase in Japanese: high population frequency of atypical form
- and its possible role in alcohol sensitivity. Am. J. Hum. Genet. 27:
- 789-796, 1975.
-
- 16. Thomas, M.; Halsall, S.; Peters, T. J.: Role of hepatic acetaldehyde
- dehydrogenase in alcoholism: demonstration of persistent reduction
- of cytosolic activity in abstaining patients. Lancet II: 1057-1059,
- 1982.
-
- 17. Yoshida, A.: Determination of aldehyde dehydrogenase phenotypes
- using hair roots: re-examination. Hum. Genet. 66: 296-299, 1984.
-
- 18. Yoshida, A.; Dave, V.; Ward, R. J.; Peters, T. J.: Cytosolic
- aldehyde dehydrogenase (ALDH1) variants found in alcohol flushers.
- Ann. Hum. Genet. 53: 1-7, 1989.
-
- 19. Yoshida, A.; Huang, I.-Y.; Ikawa, M.: Molecular abnormality of
- an inactive aldehyde dehydrogenase variant commonly found in Orientals.
- Proc. Nat. Acad. Sci. 81: 258-261, 1984.
-
- 20. Yoshida, A.; Wang, G.; Dave, V.: Determination of genotypes of
- human aldehyde dehydrogenase ALDH2 locus. Am. J. Hum. Genet. 35:
- 1107-1116, 1983.
-
- *FIELD* CS
-
- Metabolic:
- Increased intoxicating symptoms after alcohol drinking
-
- Skin:
- Facial flushing after alcohol intake
-
- Misc:
- Caucasian type alcohol flushing with abnormal ALDH1
-
- Lab:
- Cytosolic acetaldehyde dehydrogenase;
- Delayed oxidation of acetaldehyde;
- Low Km for NAD;
- High Km for acetaldehyde;
- Disulfiram sensitive
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 2/29/1992
- supermim: 3/20/1990
- carol: 12/14/1989
- carol: 11/2/1989
-
- *RECORD*
- *FIELD* NO
- 100650
- *FIELD* TI
- *100650 ALDEHYDE DEHYDROGENASE-2; ALDH2
- ALDH, LIVER MITOCHONDRIAL;;
- ACETALDEHYDE DEHYDROGENASE-2
- ALCOHOL SENSITIVITY, INCLUDED
- *FIELD* TX
- See 100640. Almost all Caucasians have 2 major ALDH isozymes in the
- liver: a cytosolic ALDH1 and a mitochondrial ALDH2 (EC 1.2.1.3). On the
- other hand, about 50% of Orientals are missing the ALDH2 isozyme.
- Impraim et al. (1982) showed that the livers of such persons show an
- enzymatically inactive but immunologically cross-reactive material (CRM)
- corresponding to the ALDH2 isozyme. The remarkable difference in
- Orientals of 2 alcohol-metabolizing enzymes, ADH2 (103720) and ALDH2,
- cannot have been coincidence. Ikuta et al. (1986) suggested that the
- explanation is coadaptation to an environment, such as particular diet,
- to which Orientals were exposed, since ADH and ALDH are complementary in
- the metabolic pathway of various alcohols. Fenna et al. (1971) concluded
- that ethanol is metabolized significantly faster in whites than in
- Eskimos or American Indians, but Bennion and Li (1976) could find no
- evidence that this is the case. Wolff (1972) demonstrated that members
- of the Mongoloid race, after drinking amounts of alcohol that have no
- detectable effect on Caucasoids, respond with marked facial flushing and
- mild to moderate symptoms of intoxication. Wolff (1972) believed that
- group differences, which are present at birth, were attributable to
- differences in autonomic reactivity. Absence of the enzyme coded by
- ALDH2, frequent in Mongoloid persons, 'causes' alcohol intolerance
- (Goedde et al., 1979). Individuals lacking the enzyme suffer the
- alcohol-flush reaction when they drink alcoholic beverages. The reaction
- is the result of excessive acetaldehyde accumulation, and the unpleasant
- symptoms tend to reduce alcohol consumption. The lower incidence of
- alcoholism in certain Mongoloid groups may have its basis in these
- observations.
-
- Hsu et al. (1985) assigned the ALDH2 locus to chromosome 12 by means of
- a cDNA probe and Southern blot analysis of somatic cell hybrids. With a
- cDNA fragment corresponding to the 3-prime-coding part of human ALDH-1
- mRNA, Braun et al. (1986) studied human-rodent somatic cell hybrids to
- confirm the assignment to chromosome 12. (The cytosolic form is on
- chromosome 9; see 100640.) The mitochondrial and cytosolic forms of ALDH
- are coded by mouse chromosomes 4 and 19, respectively (Mather and
- Holmes, 1984). Comparative mapping in man, mouse, and bovine led Womack
- (1990) to suggest that ALDH2 is in the distal part of 12q, distal to
- IFNG (147570), a conclusion consistent with other information on the
- mapping of these 2 loci.
-
- The ALDH2 alleles encoding the active and inactive subunits are termed
- 'ALDH2*1' and 'ALDH2*2,' respectively; see 100650.0001. It had been
- thought that the 2 alleles were expressed codominantly, and that only
- individuals homozygous for ALDH2*2 were ALDH2-deficient. However,
- studies of the inheritance of alcohol-induced flushing in families
- suggested that the trait is dominant (Schwitters et al., 1982). Crabb et
- al. (1989) did genotyping on the liver from 24 Japanese individuals,
- using the PCR technique for amplification of genomic DNA. In correlating
- genotype with phenotype, they found that both heterozygotes and
- homozygotes for ALDH2*2 are deficient in ALDH2 activity; that is, the
- ALDH2*2 allele is dominant. Since ALDH2 is a homotetrameric enzyme,
- random association of active and inactive subunits, equally expressed,
- should generate about 6% normal tetramers; the remainder would contain
- at least 1 mutant subunit. Thus, if all tetramers containing at least 1
- mutant subunit were inactive, there would be only 6% activity in
- heterozygotes. This low amount of activity is likely to be below the
- detection limit of activity staining of the gels. Hsu et al. (1987)
- developed a method for distinguishing the 2 main alleles by means of
- allele-specific 21-base synthetic oligonucleotides. Shibuya et al.
- (1988) studied 23 Japanese with alcoholic liver disease. No difference
- was found in the genotypes at the ADH2 locus. However, at the ALDH2
- locus, 20 of the 23 patients were homozygous for the Caucasian type,
- only 3 were heterozygous, and none of the patients was homozygous for
- the Oriental type. The results were interpreted as indicating that
- Japanese with the atypical allele are at a much lower risk for alcoholic
- liver disease, presumably due to their sensitivity to alcohol
- intoxication. By means of a pair of synthetic oligonucleotides, 1
- complementary to the usual ALDH2 allele and the other complementary to
- the atypical ALDH2 allele, Shibuya and Yoshida (1988) determined the
- genotypes of 49 unrelated Japanese persons and 12 Caucasians. The
- frequency of the atypical allele was found to be 0.35 in the Japanese
- samples examined. The atypical gene was not found in the Caucasians.
- Using allele-specific oligonucleotides for ALDH2*2, Singh et al. (1989)
- studied phenotypically deficient individuals in the Chinese, Japanese,
- and South Korean families to determine heterozygous or homozygous
- status. All individuals with a heterozygous genotype were found to be
- deficient, thus demonstrating that only the normal homotetrameric enzyme
- is catalytically active. As suggested by other workers, a random
- tetramerization of the 2 allele products will result in a residual
- enzyme activity of 6.25% of the normal value in heterozygotes if both
- normal and mutant subunits are produced in the same proportions. In
- these studies ALDH phenotype was determined in hair roots, and DNA was
- prepared from peripheral blood. Exon 12 of the gene was amplified by PCR
- for subsequent allele-specific hybridization.
-
- Crabb (1990) pointed out that the single base mutation in ALDH2,
- responsible for acute alcohol-flushing reaction in Asians, is the
- best-characterized genetic factor influencing alcohol drinking behavior.
- He raised the possibility that polymorphism in the several alcohol
- dehydrogenase genes might be related to risk of fetal alcohol syndrome
- (FAS). It is noteworthy that a genetic influence in fetal alcohol
- syndrome is suggested by twin studies: Streissguth and Dehaene (1993)
- established that the rate of concordance for the diagnosis of fetal
- alcohol syndrome was 5 out of 5 for monozygotic and 7 out of 11 for
- dizygotic twins. In 2 DZ pairs, one twin had FAS, while the other had
- fetal alcohol effects (FAE). In 2 other DZ pairs, one twin had no
- evident abnormality, while the other had FAE. IQ scores were most
- similar within pairs of MZ twins and least similar within pairs of DZ
- twins discordant for diagnosis. Johnson et al. (1996) documented the
- central nervous system anomalies of FAS by magnetic resonance imaging
- (MRI). CNS and craniofacial abnormalities were predominantly symmetric
- and central or midline. The authors stated that the association
- emphasized the concept of the midline as a special developmental field,
- vulnerable to adverse factors during embryogenesis and fetal growth and
- development.
-
- Thomasson et al. (1991) hypothesized that the polymorphisms of both of
- the liver enzymes responsible for the oxidative metabolism of ethanol
- may modify the predisposition to development of alcoholism. Using
- leukocyte DNA amplified by PCR and allele-specific oligonucleotides in a
- study of Chinese men living in Taiwan, they demonstrated that alcoholics
- had significantly lower frequencies not only of ALDH2*2 but also of
- ADH2*2 and ADH3*1 (103730). Goedde et al. (1992) gave extensive
- population frequency data on ALDH2 as well as on ADH2. They again showed
- that the atypical ALDH2 gene (ALDH2*2) is extremely rare in Caucasoids,
- Negroids, Papua New Guineans, Australian Aborigines, and Aurocanians
- (South Chile), but widely prevalent among Mongoloids. They cited
- evidence indicating that individuals possessing the ALDH2*2 allele show
- alcohol-related sensitivity responses such as facial flushing, are
- usually not habitual drinkers, and appear to suffer less from alcoholism
- and alcohol-related liver disease.
-
- Muramatsu et al. (1995) used the PCR/RFLP method to determine the
- genotypes of the ADH2 and ALDH2 loci of alcoholic and nonalcoholic
- Chinese living in Shanghai. They found that the alcoholics had
- significantly lower frequencies of the ADH2*2 and ALDH2*2 alleles than
- did the nonalcoholics, suggesting the inhibitory effects of these
- alleles for the development of alcoholism. In the nonalcoholic subjects,
- ADH2*2 had little, if any, effect, despite the significant effect of the
- ALDH2*2 allele in decreasing the alcohol consumption of the individual.
- Taken together, these results were considered consistent with the
- proposed hypothesis for the development of alcoholism, i.e., drinking
- behavior is greatly influenced by the individual's genotype of
- alcohol-metabolizing enzymes and the risk of becoming alcoholic is
- proportionate with the ethanol consumption of the individual.
-
- The ALDH2*2 allele is considered to be a genetic deterrent for
- alcoholism; however, Muramatsu et al. (1996) found that 80 of 655
- Japanese alcoholics had the mutant allele. The authors postulated that
- these alcoholics had some other factor that overcame the adverse effects
- of acetaldehydemia and that such a factor might reside in the brain's
- 'reward system,' in which dopamine plays a crucial role. Muramatsu et
- al. (1996) studied variation at the DRD4 locus (126452) and found in the
- alcoholics a higher frequency of a 5-repeat allele of the DRD4 receptor
- 48-bp repeat polymorphism in alcoholics with ALDH2*2 than in 100 other
- alcoholics and 144 controls. They found that alcoholics with the
- 5-repeat allele also abused other drugs more often.
-
- Data on gene frequencies of allelic variants were tabulated by
- Roychoudhury and Nei (1988).
-
- *FIELD* AV
- .0001
- ALCOHOL INTOLERANCE, ACUTE
- ACETALDEHYDE DEHYDROGENASE-2, ALLELE 2 ALDH2*2
- ALDH2, GLU487LYS
- The ALDH2*2-encoded protein has a change from glutamic acid (glutamate)
- to lysine at residue 487 (Yoshida et al., 1984). Hempel et al. (1985)
- and Hsu et al. (1985) also showed that the catalytic deficiency in
- mitochondrial ALDH in Orientals can be traced to a structural point
- mutation at amino acid position 487 of the polypeptide. A substitution
- of lysine for glutamic acid results from a transition of G-C to A-T.
-
- To study the mechanism by which the ALDH2*2 allele exerts its dominant
- effect in decreasing ALDH2 activity in liver extracts and producing
- cutaneous flushing when the subject drinks alcohol, Xiao et al. (1995)
- cloned ALDH2*1 cDNA and generated the ALDH2*2 allele by site-directed
- mutagenesis. These cDNAs were transduced using retroviral vectors into
- HeLa and CV1 cells, which do not express ALDH2. The normal allele
- directed synthesis of immunoreactive ALDH2 protein with the expected
- isoelectric point and increased aldehyde dehydrogenase activity. The
- ALDH2*2 allele directed synthesis of mRNA and immunoreactive protein,
- but the protein lacked enzymatic activity. When ALDH2*1-expressing cells
- were transduced with ALDH2*2 vectors, both mRNAs were expressed and
- immunoreactive proteins with isoelectric points ranging between those of
- the 2 gene products were present, indicating that the subunits formed
- heteromers. ALDH2 activity in these cells was reduced below that of the
- parental ALDH2*1-expressing cells. Thus, the authors concluded that
- ALDH2*2 allele is sufficient to cause ALDH2 deficiency in vitro.
-
- Xiao et al. (1996) referred to the ALDH2 enzyme encoded by the ALDH2*1
- allele (the wildtype form) as ALDH2E and the enzyme subunit encoded by
- ALDH2*2 as ALDH2K. They found that the ALDH2E enzyme was very stable,
- with a half-life of at least 22 hours. ALDH2K, on the other hand, had an
- enzyme half-life of only 14 hours. In cells expressing both subunits,
- most of the subunits assemble as heterotetramers, and these enzymes had
- a half-life of 13 hours. Thus, the effect of ALDH2K on enzyme turnover
- is dominant. Their studies indicated that ALDH2*2 exerts its dominant
- effect both by interfering with the catalytic activity of the enzyme and
- by increasing its turnover.
-
- *FIELD* SA
- Agarwal et al. (1981); Goedde et al. (1986); Hsu et al. (1985); Reed
- (1977); Wolff (1973); Yoshida et al. (1983)
- *FIELD* RF
- 1. Agarwal, D. P.; Harada, S.; Goedde, H. W.: Racial differences
- in biological sensitivity to ethanol: the role of alcohol dehydrogenase
- and aldehyde dehydrogenase isozymes. Alcoholism 5: 12-16, 1981.
-
- 2. Bennion, L. J.; Li, T.-K.: Alcohol metabolism in American Indians
- and Whites: lack of racial differences in metabolic rate and liver
- alcohol dehydrogenase. New Eng. J. Med. 294: 9-13, 1976.
-
- 3. Braun, T.; Grzeschik, K. H.; Bober, E.; Singh, S.; Agarwal, D.
- P.; Goedde, H. W.: The structural gene for the mitochondrial aldehyde
- dehydrogenase maps to human chromosome 12. Hum. Genet. 73: 365-367,
- 1986.
-
- 4. Crabb, D. W.: Biological markers for increased risk of alcoholism
- and for quantitation of alcohol consumption. J. Clin. Invest. 85:
- 311-315, 1990.
-
- 5. Crabb, D. W.; Edenberg, H. J.; Bosron, W. F.; Li, T.-K.: Genotypes
- for aldehyde dehydrogenase deficiency and alcohol sensitivity: the
- inactive ALDH2*2 allele is dominant. J. Clin. Invest. 83: 314-316,
- 1989.
-
- 6. Fenna, D.; Mix, L.; Schaefer, O.; Gilbert, J. A. L.: Ethanol metabolism
- in various racial groups. Canad. Med. Assoc. J. 105: 472-475, 1971.
-
- 7. Goedde, H. W.; Agarwal, D. P.; Fritze, G.; Meier-Tackmann, D.;
- Singh, S.; Beckmann, G.; Bhatia, K.; Chen, L. Z.; Fang, B.; Lisker,
- R.; Paik, Y. K.; Rothhammer, F.; Saha, N.; Segal, B.; Srivastava,
- L. M.; Czeizel, A.: Distribution of ADH-2 and ALDH2 genotypes in
- different populations. Hum. Genet. 88: 344-346, 1992.
-
- 8. Goedde, H. W.; Agarwal, D. P.; Harada, S.; Rothhammer, F.; Whittaker,
- J. O.; Lisker, R.: Aldehyde dehydrogenase polymorphism in North American,
- South American, and Mexican Indian populations. Am. J. Hum. Genet. 38:
- 395-399, 1986.
-
- 9. Goedde, H. W.; Harada, S.; Agarwal, D. P.: Racial differences
- in alcohol sensitivity: a new hypothesis. Hum. Genet. 51: 331-334,
- 1979.
-
- 10. Hempel, J.; Kaiser, R.; Jornvall, H.: Mitochondrial aldehyde
- dehydrogenase from human liver: primary structure, differences in
- relation to the cytosolic enzyme and functional correlations. Europ.
- J. Biochem. 153: 13-28, 1985.
-
- 11. Hsu, L. C.; Bendel, R. E.; Yoshida, A.: Direct detection of usual
- and atypical alleles on the human aldehyde dehydrogenase-2 (ALDH2)
- locus. Am. J. Hum. Genet. 41: 996-1001, 1987.
-
- 12. Hsu, L. C.; Tani, K.; Fujiyoshi, T.; Kurachi, K.; Yoshida, A.
- : Cloning of cDNAs for human aldehyde dehydrogenases 1 and 2. Proc.
- Nat. Acad. Sci. 82: 3771-3775, 1985.
-
- 13. Hsu, L. C.; Yoshida, A.; Mohandas, T.: Chromosomal assignment
- of the genes for human aldehyde dehydrogenase 1 (ALDH1) and aldehyde
- dehydrogenase 2 (ALDH2).(Abstract) Cytogenet. Cell Genet. 40: 656-657,
- 1985.
-
- 14. Ikuta, T.; Szeto, S.; Yoshida, A.: Three human alcohol dehydrogenase
- subunits: cDNA structure and molecular and evolutionary divergence. Proc.
- Nat. Acad. Sci. 83: 634-638, 1986.
-
- 15. Impraim, C.; Wang, G.; Yoshida, A.: Structural mutation in a
- major human aldehyde dehydrogenase gene results in loss of enzyme
- activity. Am. J. Hum. Genet. 34: 837-841, 1982.
-
- 16. Johnson, V. P.; Swayze, V. W., II; Sato, Y.; Andreasen, N. C.
- : Fetal alcohol syndrome: craniofacial and central nervous system
- manifestations. Am. J. Med. Genet. 61: 329-339, 1996.
-
- 17. Mather, P. B.; Holmes, R. S.: Biochemical genetics of aldehyde
- dehydrogenase isoenzymes in the mouse: evidence for stomach and testis-specific
- isoenzymes. Biochem. Genet. 22: 981-995, 1984.
-
- 18. Muramatsu, T.; Higuchi, S.; Murayama, M.; Matsushita, S.; Hayashida,
- M.: Association between alcoholism and the dopamine D4 receptor gene. J.
- Med. Genet. 33: 113-115, 1996.
-
- 19. Muramatsu, T.; Zu-Cheng, W.; Yi-Ru, F.; Kou-Bao, H.; Heqin, Y.;
- Yamada, K.; Higuchi, S.; Harada, S.; Kono, H.: Alcohol and aldehyde
- dehydrogenase genotypes and drinking behavior of Chinese living in
- Shanghai. Hum. Genet. 96: 151-154, 1995.
-
- 20. Reed, T. E.: Three heritable responses to alcohol in a heterogeneous
- randomly mated mouse strain: inferences for humans. J. Studies Alcohol 38:
- 618-632, 1977.
-
- 21. Roychoudhury, A. K.; Nei, M.: Human Polymorphic Genes: World
- Distribution. New York: Oxford Univ. Press (pub.) 1988.
-
- 22. Schwitters, S. Y.; Johnson, R. C.; Johnson, S. B.; Ahern, F. M.
- : Familial resemblances in flushing following alcohol use. Behav.
- Genet. 12: 349-352, 1982.
-
- 23. Shibuya, A.; Ikuta, T.; Hsu, L. C.; Yoshida, A.: Genotypes of
- alcohol metabolizing enzymes in Japanese with alcoholic liver diseases:
- a strong association of the usual Caucasian type aldehyde dehydrogenase
- allele (ALDH2) with the disease.(Abstract) Am. J. Hum. Genet. 43:
- A201, 1988.
-
- 24. Shibuya, A.; Yoshida, A.: Frequency of the atypical aldehyde
- dehydrogenase-2 gene (ALDH2/2) in Japanese and Caucasians. Am. J.
- Hum. Genet. 43: 741-743, 1988.
-
- 25. Singh, S.; Fritze, G.; Fang, B.; Harada, S.; Paik, Y. K.; Eckey,
- R.; Agarwal, D. P.; Goedde, H. W.: Inheritance of mitochondrial aldehyde
- dehydrogenase: genotyping in Chinese, Japanese and South Korean families
- reveals dominance of the mutant allele. Hum. Genet. 83: 119-121,
- 1989.
-
- 26. Streissguth, A. P.; Dehaene, P.: Fetal alcohol syndrome in twins
- of alcoholic mothers: concordance of diagnosis and IQ. Am. J. Med.
- Genet. 47: 857-861, 1993.
-
- 27. Thomasson, H. R.; Edenberg, H. J.; Crabb, D. W.; Mai, X.-L.; Jerome,
- R. E.; Li, T.-K.; Wang, S.-P.; Lin, Y.-T.; Lu, R.-B.; Yin, S.-J.:
- Alcohol and aldehyde dehydrogenase genotypes and alcoholism in Chinese
- men. Am. J. Hum. Genet. 48: 677-681, 1991.
-
- 28. Wolff, P. H.: Ethnic differences in alcohol sensitivity. Science 175:
- 449-450, 1972.
-
- 29. Wolff, P. H.: Vasomotor sensitivity to alcohol in diverse mongoloid
- populations. Am. J. Hum. Genet. 25: 193-199, 1973.
-
- 30. Womack, J. E.: Personal Communication. College Station, Texas
- 2/26/1990.
-
- 31. Xiao, Q.; Weiner, H.; Crabb, D. W.: The mutation in the mitochondrial
- aldehyde dehydrogenase (ALDH2) gene responsible for alcohol-induced
- flushing increases turnover of the enzyme tetramers in a dominant
- fashion. J. Clin. Invest. 98: 2027-2032, 1996.
-
- 32. Xiao, Q.; Weiner, H.; Johnston, T.; Crabb, D. W.: The aldehyde
- dehydrogenase ALDH2*2 allele exhibits dominance over ALDH2*1 in transduced
- HeLa cells. J. Clin. Invest. 96: 2180-2186, 1995.
-
- 33. Yoshida, A.; Huang, I.-Y.; Ikawa, M.: Molecular abnormality of
- an inactive aldehyde dehydrogenase variant commonly found in Orientals. Proc.
- Nat. Acad. Sci. 81: 258-261, 1984.
-
- 34. Yoshida, A.; Wang, G.; Dave, V.: Determination of genotypes of
- human aldehyde dehydrogenase ALDH-2 locus. Am. J. Hum. Genet. 35:
- 1107-1116, 1983.
-
- *FIELD* CS
-
- Metabolic:
- Increased intoxicating symptoms after alcohol drinking
-
- Skin:
- Facial flushing after alcohol intake
-
- Misc:
- Oriental type alcohol flushing with abnormal ALDH2
-
- Lab:
- Mitochondrial acetaldehyde dehydrogenase;
- Delayed oxidation of acetaldehyde;
- High Km for NAD;
- Low Km foracetaldehyde;
- Disulfiram insensitive
-
- *FIELD* CN
- Mark H. Paalman - updated: 6/12/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 12/06/1996
- mark: 6/12/1996
- mark: 2/26/1996
- terry: 2/20/1996
- mark: 2/2/1996
- terry: 1/26/1996
- mark: 10/15/1995
- warfield: 3/31/1994
- mimadm: 3/11/1994
- carol: 1/26/1994
- carol: 6/9/1992
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 100660
- *FIELD* TI
- *100660 ACETALDEHYDE DEHYDROGENASE-3
- ALDEHYDE DEHYDROGENASE-3; ALDH3;;
- STOMACH ALDH
- *FIELD* TX
- See 100640. In stomach tissue, Teng (1981) described an isozymic form of
- aldehyde dehydrogenase (ALDH). It did not use formaldehyde, acetaldehyde
- or pyruvic aldehyde. Furfuraldehyde and, to a lesser extent,
- propionaldehyde were readily oxidized. Teng (1981) found 1 genetic
- variant among 71 Chinese stomach specimens and a second different
- variant among 33 Asiatic Indian specimens. Unlike liver ALDH, which
- appears to be a tetramer, the electrophoretic pattern in the
- heterozygotes suggested that stomach ALDH is a monomer. ALDH3 is also
- present in lung. By study of somatic cell hybrids, Santisteban et al.
- (1985) assigned the ALDH3 gene to chromosome 17. By in situ
- hybridization, Hiraoka et al. (1995) mapped the ALDH3 gene to 17p11.2.
-
- Data on gene frequencies of allelic variants were tabulated by
- Roychoudhury and Nei (1988).
-
- *FIELD* RF
- 1. Hiraoka, L. R.; Hsu, L.; Hsieh, C.-L.: Assignment of ALDH3 to
- human chromosome 17p11.2 and ALDH5 to human chromosome 9p13. Genomics 25:
- 323-325, 1995.
-
- 2. Roychoudhury, A. K.; Nei, M.: Human Polymorphic Genes: World Distribution.
- New York: Oxford Univ. Press (pub.) 1988.
-
- 3. Santisteban, I.; Povey, S.; West, L. F.; Parrington, J. M.; Hopkinson,
- D. A.: Chromosome assignment, biochemical and immunological studies
- on a human aldehyde dehydrogenase, ALDH3. Ann. Hum. Genet. 49:
- 87-100, 1985.
-
- 4. Teng, Y.-S.: Stomach aldehyde dehydrogenase: report of a new locus.
- Hum. Hered. 31: 74-77, 1981.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 2/7/1995
- mimadm: 2/11/1994
- supermim: 3/16/1992
- carol: 12/6/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 100670
- *FIELD* TI
- *100670 ACETALDEHYDE DEHYDROGENASE-5
- ALDEHYDE DEHYDROGENASE-5; ALDH5
- *FIELD* TX
- The 2 aldehyde dehydrogenase isozymes that play a major role in ethanol
- detoxification, ALDH1 (100640) and ALDH2 (100650), are cytosolic and
- mitochondrial forms, respectively. Their organization is basically
- similar; their sizes are 53 kb and 44 kb, respectively, and both contain
- 13 coding exons interrupted by 12 introns of comparable lengths. Hsu et
- al. (1989) cloned a new ALDH gene from a cosmid human DNA library.
- Although it contains no introns, Northern blot hybridization of human
- liver RNA revealed a unique mRNA component that hybridized with this
- gene probe but with neither the ALDH1 probe or the ALDH2 probe. The new
- gene encoded 517 amino acid residues, suggesting that it is similar to
- ALDH2, and indeed its deduced sequence was 70.6% identical to that of
- ALDH2 and only 62.8% identical to that of ALDH1. Hsu et al. (1989)
- assigned the ALDH5 gene to chromosome 9 by Southern blot analysis of
- rodent-human hybrid cell DNAs. Hsu and Chang (1991) provided a full
- report on this gene which they referred to as ALDHx.
-
- By in situ hybridization, Hiraoka et al. (1995) mapped the ALDH5 gene to
- 9p13.
-
- *FIELD* SA
- Harada et al. (1980)
- *FIELD* RF
- 1. Harada, S.; Agarwal, D. P.; Goedde, H. W.: Electrophoretic and
- biochemical studies of human aldehyde dehydrogenase isozymes in various
- tissues. Life Sci. 26: 1773-1780, 1980.
-
- 2. Hiraoka, L. R.; Hsu, L.; Hsieh, C.-L.: Assignment of ALDH3 to
- human chromosome 17p11.2 and ALDH5 to human chromosome 9p13. Genomics 25:
- 323-325, 1995.
-
- 3. Hsu, L. C.; Chang, W.-C.; Yoshida, A.: Cloning of a new human
- aldehyde dehydrogenase gene and comparison with liver cytosolic ALDH1
- and mitochondrial ALDH2 genes. (Abstract) Am. J. Hum. Genet. 45
- (suppl.): A196 only, 1989.
-
- 4. Hsu, L. C.; Chang, W. C.: Cloning and characterization of a new
- functional human aldehyde dehydrogenase gene. J. Biol. Chem. 266:
- 12257-12265, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 2/7/1995
- carol: 10/22/1992
- carol: 8/28/1992
- supermim: 3/16/1992
- supermim: 3/20/1990
- carol: 11/7/1989
-
- *RECORD*
- *FIELD* NO
- 100675
- *FIELD* TI
- 100675 ACETAMINOPHEN METABOLISM
- *FIELD* TX
- Acetaminophen (paracetamol) is extensively conjugated with glucuronic
- acid and sulfate before renal excretion. A minor metabolic route
- involves microsomal oxidation of acetaminophen to a hepatotoxic reactive
- intermediate, which subsequently undergoes glutathione (GSH)
- conjugation, yielding cysteine and mercapturate conjugates, both of
- which are excreted in the urine (Slattery et al., 1987). Evidence was
- presented by de Morais et al. (1989) that, in comparison with normal
- subjects, glucuronidation of acetaminophen is impaired in subjects with
- Gilbert syndrome (143500), a disorder in which glucuronidation of
- bilirubin is impaired. In studies of 125 Caucasian and 33 Oriental
- subjects, Patel et al. (1992) found no difference in the mean fraction
- of acetaminophen excreted as glucuronide: 51.5% in Caucasians vs 51.8%
- in Orientals. However, bimodality was apparent in both groups, with 20%
- of Caucasians and 33% of Oriental subjects displaying relatively
- extensive glucuronidation. In addition, glucuronidation displayed a
- strong negative correlation with sulfation (r = -0.97), suggesting a
- compensatory or complementary relationship between the 2 metabolic
- pathways. The mean fractional excretions of cysteine and mercapturate
- conjugates between Caucasians and Orientals did show significant
- differences (p = less than 0.005).
-
- *FIELD* RF
- 1. de Morais, S. M. F.; Uetrecht, J. P.; Wells, P. G.: Decreased
- glucuronidation and increased bioactivation of acetaminophen in Gilbert's
- disease. (Abstract) FASEB J. 3: A739 only, 1989.
-
- 2. Patel, M.; Tang, B. K.; Kalow, W.: Variability of acetaminophen
- metabolism in Caucasians and Orientals. Pharmacogenetics 2: 38-45,
- 1992.
-
- 3. Slattery, J. T.; Wilson, J. M.; Kalhorn, T. F.; Nelson, S. D.:
- Dose-dependent pharmacokinetics of acetaminophen: evidence of glutathione
- depletion in humans. Clin. Pharm. Therap. 41: 413-418, 1987.
-
- *FIELD* CS
-
- Skin:
- Jaundice
-
- Lab:
- Impaired acetaminophen glucuronidation in Gilbert syndrome (143500)
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 7/21/1992
-
- *FIELD* ED
- mimadm: 4/14/1994
- carol: 10/13/1992
- carol: 8/10/1992
- carol: 7/21/1992
-
- *RECORD*
- *FIELD* NO
- 100678
- *FIELD* TI
- *100678 ACETYL-CoA ACETYLTRANSFERASE-2; ACAT2
- ACETOCOENZYME A ACETYLTRANSFERASE-2;;
- ACETOACETYL-CoA THIOLASE
- ACAT2 DEFICIENCY, INCLUDED
- *FIELD* TX
- The TCP1 gene (186980) is located on 6p in the vicinity of the major
- histocompatibility complex, and the murine homolog, Tcp-1, is located in
- the t-complex region of mouse chromosome 17. In the mouse, a related
- gene, Tcp-1x, is tightly linked to Tcp-1. Ashworth (1993) showed that 2
- genes located 3-prime to the murine Tcp-1 and Tcp-1x genes code for
- proteins highly homologous to acetyl-CoA acetyltransferases. These Acat
- genes are in opposite orientation to the Tcp-1 genes, and transcription
- results in mRNA species that contain the last exon of Tcp-1 or Tcp-1x
- within the 3-prime untranslated region of the respective Acat mRNA. Both
- Acat genes appear to be transcribed in several mouse tissues. Willison
- et al. (1987) showed (their fig. 2b) that in humans TCP1 and ACAT genes
- also overlap. Retention of this close linkage during mammalian evolution
- suggests the possibility of some functional significance. Transcription
- of both DNA strands at a given locus is common in prokaryotic and viral
- systems. For examples of overlapping transcriptional units in humans,
- see Morel et al. (1989) and Laudet et al. (1991).
-
- It is proposed to use ACAT2 to designate the ACAT gene on human
- chromosome 6; the ACAT1 gene is the one previously mapped to human
- chromosome 11 and found to be mutant in cases of 3-ketothiolase
- deficiency (203750).
-
- Song et al. (1994) cloned cDNA for human cytosolic acetoacetyl-CoA
- thiolase by use of an antibody against the human enzyme. The deduced
- amino acid sequence had a 34 to 57% homology with 4 other human
- thiolases and 4 acetoacetyl-CoA thiolases of microorganisms.
-
- As the human TCP1 gene had been assigned to 6q25-q27 by study of somatic
- cell hybrids and by in situ hybridization, the ACAT2 gene was suspected
- to be localized to the same chromosome region. By fluorescence in situ
- hybridization, Masuno et al. (1996) demonstrated that the ACAT2 gene is
- located on 6q25.3-q26.
-
- Reported patients with ACTA2 deficiency have shown severe mental
- retardation and hypotonus. Laboratory findings, including urinary
- organic acids were not specific (Bennett et al., 1984).
-
- *FIELD* RF
- 1. Ashworth, A.: Two acetyl-CoA acetyltransferase genes located in
- the t-complex region of mouse chromosome 17 partially overlap the
- Tcp-1 and Tcp-1x genes. Genomics 18: 195-198, 1993.
-
- 2. Bennett, M. J.; Hosking, G. P.; Smith, M. F.; Gray, R. G. F.; Middleton,
- B.: Biochemical investigations on a patient with a defect in cytosolic
- acetoacetyl-CoA thiolase, associated with mental retardation. J.
- Inherit. Metab. Dis. 7: 125-128, 1984.
-
- 3. Laudet, V.; Begue, A.; Henry-Duthoit, C.; Joubel, A.; Martin, P.;
- Stehelin, D.; Saule, S.: Genomic organization of the human thyroid
- hormone alpha (c-erbA-1) gene. Nucleic Acids Res. 19: 1105-1112,
- 1991.
-
- 4. Masuno, M.; Fukao, T.; Song, X.-Q.; Yamaguchi, S.; Orii, T.; Kondo,
- N.; Imaizumi, K.; Kuroki, Y.: Assignment of the human cytosolic acetoacetyl-coenzyme
- A thiolase (ACAT2) gene to chromosome 6q25.3-q26. Genomics 36: 217-218,
- 1996.
-
- 5. Morel, Y.; Bristow, J.; Gitelman, S. E.; Miller, W. L.: Transcript
- encoded on the opposite strand of the human steroid 21-hydroxylase/complement
- component C4 gene locus. Proc. Nat. Acad. Sci. 86: 6582-6586, 1989.
-
- 6. Song, X.-Q.; Fukao, T.; Yamaguchi, S.; Miyazawa, S.; Hashimoto,
- T.; Orii, T.: Molecular cloning and nucleotide sequence of complementary
- DNA for human hepatic cytosolic acetoacetyl-coenzyme A thiolase. Biochem.
- Biophys. Res. Commun. 201: 478-485, 1994.
-
- 7. Willison, K.; Kelly, A.; Dudley, K.; Goodfellow, P.; Spurr, N.;
- Groves, V.; Gorman, P.; Sheer, D.; Trowsdale, J.: The human homologue
- of the mouse t-complex gene, TCP1, is located on chromosome 6 but
- is not near the HLA region. EMBO J. 6: 1967-1974, 1987.
-
- *FIELD* CD
- Victor A. McKusick: 12/2/1993
-
- *FIELD* ED
- mark: 09/12/1996
- terry: 9/4/1996
- carol: 10/12/1994
- carol: 12/20/1993
- carol: 12/2/1993
-
- *RECORD*
- *FIELD* NO
- 100680
- *FIELD* TI
- 100680 ACETYLCHOLINESTERASE EXPRESSION; ACEE
- REGULATOR OF ACETYLCHOLINESTERASE; RACH
- *FIELD* TX
- Chen et al. (1978) studied three strains of human fibroblasts that were
- trisomic for chromosome 2 and had an average level of ACE over 28 times
- higher than the average fibroblasts. The mean pseudocholinesterase level
- of the trisomy-2 strains was normal. The 19 control strains comprised 10
- trisomic for other autosomes and 9 euploid strains. The ACE activity of
- control fibroblasts did not differ significantly from zero. Despite the
- unusual elevation of ACE in trisomy-2 fibroblasts, the level, expressed
- in terms of micrograms of DNA, was only 1.5% of that in cerebral cortex.
- Two other enzymes, xanthine oxidase and choline acetyltransferase,
- which, like ACE, have a restricted distribution in human tissues, were
- absent from all 22 strains of fibroblasts. The results were interpreted
- as evidence for a gene on chromosome 2 involved in regulation of ACE.
-
- *FIELD* RF
- 1. Chen, Y.-T.; Worthy, T. E.; Krooth, R. S.: Evidence for a striking
- increase in acetylcholinesterase activity in cultured human fibroblasts
- which are trisomic for chromosome two. Somat. Cell Genet. 4: 265-298,
- 1978.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- carol: 7/13/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 100690
- *FIELD* TI
- *100690 CHOLINERGIC RECEPTOR, NICOTINIC, ALPHA POLYPEPTIDE 1; CHRNA1
- CHRNA;;
- ACETYLCHOLINE RECEPTOR, MUSCLE, ALPHA SUBUNIT; ACHRA
- *FIELD* TX
- The acetylcholine receptor of muscle, like the nicotinic acetylcholine
- receptor of the fish electric organ, has 5 subunits of 4 different
- types: 2 alpha and 1 each of beta, gamma and delta subunits. In the
- electric organ the subunits show conspicuous sequence homology. The
- transmembrane topology of the subunits and the location of functionally
- important regions, such as the acetylcholine binding site and the
- transmembrane segments involved in the ionic channel, have been
- proposed. Noda et al. (1983) cloned cDNA for the alpha subunit precursor
- of the calf skeletal muscle AChR and a human genomic DNA segment
- containing the corresponding gene. Nucleotide sequences showed marked
- homology with the counterpart of Torpedo sp. (electric ray). The
- protein-coding sequence of the human ACHRA gene is divided into 9 exons
- by 8 introns, which correspond to different structural and functional
- domains of the precursor molecule. Analyzing acetylcholine receptor
- clones isolated from a human leg muscle cDNA library, Beeson et al.
- (1990) found that the alpha subunit exists in 2 isoforms. A novel exon,
- coding for 25 amino acids, was inserted into the alpha subunit, giving
- the new isoform 462 amino acids.
-
- Heidmann et al. (1986) analyzed restriction fragment length
- polymorphisms of each of the 4 subunits of muscle nicotinic
- acetylcholine receptor in crosses between 2 mouse species and showed
- that the alpha subunit gene cosegregates with the alpha cardiac actin
- gene on mouse chromosome 17. Taylor and Rowe (1989) concluded that the
- Acra gene in the mouse in fact is located on chromosome 2. Schoepfer et
- al. (1988) showed that a human medulloblastoma cell line expressed a
- muscle type rather than a neuronal type of acetylcholine receptor. They
- succeeded in isolating cDNA clones for the alpha subunit and suggested
- that these should be useful in obtaining large amounts of human
- muscle-type acetylcholine receptor alpha-subunit protein for studies of
- the autoimmune response in myasthenia gravis. Garchon et al. (1994)
- identified 2 stable polymorphic dinucleotide repeats within the first
- intron of the CHRNA gene, designated HB and BB. They found that the
- HB*14 allele conferred a relative risk for myasthenia gravis of 2.5 in
- 81 unrelated patients compared with 100 control subjects. Very
- significantly, family analysis based on haplotype segregation data
- indicated that parental haplotypes associated with HB*14 always
- segregated to the child with myasthenia gravis, whereas their
- transmission to unaffected sibs was as expected ('was equilibrated,' in
- the words of the authors). Myasthenia gravis patients always showed a
- high frequency of microsatellite variants not seen in controls.
-
- By means of somatic cell hybridization, Beeson et al. (1989, 1990)
- assigned the CHRNA gene to chromosome 2; by in situ hybridization, they
- regionalized the gene to 2q24-q32, with the major peak of grains being
- at 2q32. By linkage analysis, Lobos (1993) placed the CHRNA gene about
- 27 cM proximal to the crystallin G pseudogene marker, CRYGP1, located at
- 2q33-q35; the CHRND (100720) and CHRNG (100730) loci were placed about
- 31 cM distal to CRYGP1.
-
- *FIELD* AV
- .0001
- MYASTHENIC SYNDROME, SLOW-CHANNEL CONGENITAL
- SCCMS
- CHRNA1, ASN217LYS
- Engel et al. (1996) described a 30-year-old female patient with ocular
- and limb weakness, scoliosis, and a family history consistent with
- autosomal dominant myasthenia gravis (601462) in 3 generations. The
- mutation leading to pathology in this patient was a heterozygous
- asn217-to-lys substitution in the AChR-alpha subunit. Engel et al.
- (1996) evaluated the pathogenicity of the mutation by engineering the
- mutation into the corresponding cDNA of mouse AChR and coexpressing it
- with the wildtype cDNA in HEK fibroblasts. Receptor function was
- evaluated using patch clamp studies and ACh binding was measured. These
- studies revealed that the mutations resulted in an apparent increased
- affinity for ACh and prolonged AChR activation episodes rendering the
- receptor channel leaky.
-
- *FIELD* SA
- Beeson et al. (1990); Mishina et al. (1986)
- *FIELD* RF
- 1. Beeson, D.; Jeremiah, S.; West, L. F.; Povey, S.; Newsom-Davis,
- J.: Assignment of the human nicotinic acetylcholine receptor genes:
- the alpha and delta subunit genes to chromosome 2 and the beta subunit
- gene to chromosome 17. Ann. Hum. Genet. 54: 199-208, 1990.
-
- 2. Beeson, D.; Jeremiah, S. J.; West, L. F.; Povey, S.; Newsom-Davis,
- J.: Assignment of the human acetylcholine receptor beta subunit gene
- to chromosome 17 and the alpha and delta subunit genes to chromosome
- 2. (Abstract) Cytogenet. Cell Genet. 51: 960 only, 1989.
-
- 3. Beeson, D.; Morris, A.; Vincent, A.; Newsom-Davis, J.: The human
- muscle nicotinic acetylcholine receptor alpha-subunit exists as two
- isoforms: a novel exon. EMBO J. 9: 2101-2106, 1990.
-
- 4. Engel, A. G.; Ohno, K.; Milone, M.; Wang, H.-L.; Nakano, S.; Bouzat,
- C.; Pruitt, J. N., II; Hutchinson, D. O.; Brengman, J. M.; Bren, N.;
- Sieb, J. P.; Sine, S. M.: New mutations in acetylcholine receptor
- subunit genes reveal heterogeneity in the slow-channel congenital
- myasthenic syndrome. Hum. Molec. Genet. 5: 1217-1227, 1996.
-
- 5. Garchon, H.-J.; Djabiri, F.; Viard, J.-P.; Gajdos, P.; Bach, J.-F.
- : Involvement of human muscle acetylcholine receptor alpha-subunit
- gene (CHRNA) in susceptibility to myasthenia gravis. Proc. Nat. Acad.
- Sci. 91: 4668-4672, 1994.
-
- 6. Heidmann, O.; Buonanno, A.; Geoffroy, B.; Robert, B.; Guenet, J.-L.;
- Merlie, J. P.; Changeux, J.-P.: Chromosomal localization of muscle
- nicotinic acetylcholine receptor genes in the mouse. Science 234:
- 866-868, 1986.
-
- 7. Lobos, E. A.: Five subunit genes of the human muscle nicotinic
- acetylcholine receptor are mapped to two linkage groups on chromosomes
- 2 and 17. Genomics 17: 642-650, 1993.
-
- 8. Mishina, M.; Takai, T.; Imoto, K.; Noda, M.; Takahashi, T.; Numa,
- S.; Methfessel, C.; Sakmann, B.: Molecular distinction between fetal
- and adult forms of muscle acetylcholine receptor. Nature 321: 406-411,
- 1986.
-
- 9. Noda, M.; Furutani, Y.; Takahashi, H.; Toyosato, M.; Tanabe, T.;
- Shimizu, S.; Kikyotani, S.; Kayano, T.; Hirose, T.; Inayama, S.; Numa,
- S.: Cloning and sequence analysis of calf cDNA and human genomic
- DNA encoding alpha-subunit precursor of muscle acetylcholine receptor. Nature 305:
- 818-823, 1983.
-
- 10. Schoepfer, R.; Luther, M.; Lindstrom, J.: The human medulloblastoma
- cell line TE671 expresses a muscle-like acetylcholine receptor: cloning
- of the alpha-subunit cDNA. FEBS Lett. 226: 235-240, 1988.
-
- 11. Taylor, B. A.; Rowe, L.: Localization of the gene encoding the
- alpha-subunit of the acetylcholine receptor on chromosome 2 of the
- mouse. Cytogenet. Cell Genet. 52: 102-103, 1989.
-
- *FIELD* CN
- Moyra Smith - updated: 10/09/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 10/09/1996
- carol: 9/19/1994
- mimadm: 4/14/1994
- carol: 10/13/1993
- carol: 9/22/1993
- carol: 2/17/1993
- carol: 1/5/1993
-
- *RECORD*
- *FIELD* NO
- 100700
- *FIELD* TI
- 100700 ACHARD SYNDROME
- *FIELD* TX
- Arachnodactyly, receding lower jaw, and joint laxity limited to the
- hands and feet are features. When Thursfield (1917-18) reviewed the
- literature on Marfan syndrome, he remarked that the skeletal picture in
- the cases described by Achard (1902) differed in that the skull was
- broad and brachycephalic with small mandible; although there was
- arachnodactyly, the body proportions were not altered and the patient
- was not excessively tall. Parish (1960) pictured a case. It is not clear
- what this condition represented or even that it is a distinct entity.
-
- *FIELD* SA
- Parish (1967)
- *FIELD* RF
- 1. Achard, C.: Arachnodactylie. Bull. Mem. Soc. Med. Hop. Paris 19:
- 834-840, 1902.
-
- 2. Parish, J. G.: Heritable disorders of connective tissues with
- arachnodactyly. Proc. Roy. Soc. Med. 53: 515-518, 1960.
-
- 3. Parish, J. G.: Skeletal hand charts in inherited connective tissue
- disease. J. Med. Genet. 4: 227-238, 1967.
-
- 4. Thursfield, H.: Arachnodactyly. St. Bart's Hosp. Rep. 53: 35-40,
- 1917.
-
- *FIELD* CS
-
- Limbs:
- Arachnodactyly
-
- Joints:
- Joint laxity limited to hands and feet
-
- Skull:
- Broad skull
-
- Head:
- Brachycephaly;
- Micrognathia
-
- Misc:
- Normal body proportions
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/16/1986
-
- *FIELD* ED
- pfoster: 8/18/1994
- mimadm: 5/2/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- carol: 3/6/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 100710
- *FIELD* TI
- *100710 CHOLINERGIC RECEPTOR, NICOTINIC, BETA POLYPEPTIDE 1; CHRNB1
- CHRNB;;
- ACETYLCHOLINE RECEPTOR, MUSCLE, BETA SUBUNIT; ACHRB
- *FIELD* TX
- See 100690. In the Torpedo (electric ray), the 4 subunits of the AChR
- show conspicuous sequence homology. Heidmann et al. (1986) analyzed
- restriction fragment length polymorphisms of each of the 4 subunits of
- muscle nicotinic acetylcholine receptor in crosses between 2 mouse
- species. They found that the beta subunit gene is located on mouse
- chromosome 11. The beta subunit gene was found to be tightly linked with
- the locus encoding the different isoforms (embryonic, perinatal and
- adult) of the myosin heavy chain genes which are located on mouse
- chromosome 11. In man these genes are located on chromosome 17p
- (160730), arguing from likely homology of synteny. The beta subunit of
- the acetylcholine receptor may be coded by a gene on human 17p also.
- Using a panel of human-rodent somatic cell hybrids segregating human
- chromosomes, Beeson et al. (1989) demonstrated that the CHRNB locus is
- on human chromosome 17. Beeson et al. (1990) regionalized the CHRNB gene
- to 17p12-p11 by in situ hybridization.
-
- *FIELD* AV
- .0001
- MYASTHENIC SYNDROME, SLOW-CHANNEL CONGENITAL
- SCCMS
- CHRNB1, VAL266MET
- Engel et al. (1996) described a 19-year-old female with myasthenic
- symptoms since birth involving ocular, cranial, and limb muscles. The
- mutation leading to pathology was a heterozygous val266-to-met
- substitution in the transmembrane domain of the AChR-beta subunit.
- Receptor function was evaluated using patch clamp studies and ACh
- binding was measured. These studies revealed that the mutation resulted
- in an apparent increased affinity for ACh and prolonged AChR activation
- episodes rendering the receptor channel leaky. See also 601462.
-
- *FIELD* SA
- Beeson et al. (1989)
- *FIELD* RF
- 1. Beeson, D.; Brydson, M.; Newsom-Davis, J.: Nucleotide sequence
- of human muscle acetylcholine receptor beta-subunit. Nucleic Acids
- Res. 17: 4391 only, 1989.
-
- 2. Beeson, D.; Jeremiah, S.; West, L. F.; Povey, S.; Newsom-Davis,
- J.: Assignment of the human nicotinic acetylcholine receptor genes:
- the alpha and delta subunit genes to chromosome 2 and the beta subunit
- gene to chromosome 17. Ann. Hum. Genet. 54: 199-208, 1990.
-
- 3. Beeson, D.; Jeremiah, S. J.; West, L. F.; Povey, S.; Newsom-Davis,
- J.: Assignment of the human acetylcholine receptor beta subunit gene
- to chromosome 17 and the alpha and delta subunit genes to chromosome
- 2. (Abstract) Cytogenet. Cell Genet. 51: 960 only, 1989.
-
- 4. Engel, A. G.; Ohno, K.; Milone, M.; Wang, H.-L.; Nakano, S.; Bouzat,
- C.; Pruitt, J. N., II; Hutchinson, D. O.; Brengman, J. M.; Bren, N.;
- Sieb, J. P.; Sine, S. M.: New mutations in acetylcholine receptor
- subunit genes reveal heterogeneity in the slow-channel congenital
- myasthenic syndrome. Hum. Molec. Genet. 5: 1217-1227, 1996.
-
- 5. Heidmann, O.; Buonanno, A.; Geoffroy, B.; Robert, B.; Guenet, J.-L.;
- Merlie, J. P.; Changeux, J.-P.: Chromosomal localization of muscle
- nicotinic acetylcholine receptor genes in the mouse. Science 234:
- 866-868, 1986.
-
- *FIELD* CN
- Moyra Smith - updated: 10/09/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 10/09/1996
- mark: 8/29/1996
- carol: 6/30/1992
- carol: 4/7/1992
- supermim: 3/16/1992
- carol: 2/29/1992
- carol: 2/5/1992
- carol: 1/29/1991
-
- *RECORD*
- *FIELD* NO
- 100720
- *FIELD* TI
- *100720 ACETYLCHOLINE RECEPTOR, MUSCLE, DELTA SUBUNIT; ACHRD
- CHOLINERGIC RECEPTOR, NICOTINIC, DELTA POLYPEPTIDE; CHRND
- *FIELD* TX
- See 100690. Heidmann et al. (1986) analyzed restriction fragment length
- polymorphisms of the 4 subunits of muscle nicotinic acetylcholine
- receptor in 2 mouse species and crosses between the two. They found that
- the gamma and delta subunit genes cosegregated with each other and with
- the gene of the fast skeletal muscle isoforms of myosin alkali light
- chain (160780). The acetylcholine receptor genes cosegregated less
- tightly with the gene for isocitrate dehydrogenase-1 (147700). The
- myosin locus and the Idh-1 locus are on mouse chromosome 1. IDH1 in man
- is located on chromosome 2, which carries another locus homologous to
- one on mouse no. 1, namely, the cluster of genes for a gamma polypeptide
- of crystallin (123660-123690). Thus, the gamma and delta subunit genes
- of acetylcholine receptor may be tightly linked to each other and may be
- situated in man on chromosome 2, possibly on the long arm. Lobos et al.
- (1989) found at least 1 RFLP in each of the 4 subunit genes. The delta
- gene was assigned by in situ hybridization to 2q31-q34. All pairs of
- RFLPs were analyzed for linkage disequilibrium. Of the 16 pairs of RFLPs
- from the same gene or from the linked gamma and delta genes, 13 showed
- evidence of significant disequilibrium (P less than 0.05). By Southern
- analysis of a panel of somatic cell hybrids and by in situ
- hybridization, Beeson et al. (1990) assigned the CHRND gene to
- 2q33-qter. Together with the earlier information, this suggests a
- location of 2q33-q34. Work of Pasteris et al. (1993) suggested a more
- distal location; a molecular analysis of a chromosome 2 deletion mapping
- panel suggested the following order: cen--PAX3--COL4A3--CHRND--tel. PAX3
- (193500) is located in band 2q35 and COL4A3 (120070) is located in band
- 2q36.
-
- *FIELD* RF
- 1. Beeson, D.; Jeremiah, S.; West, L. F.; Povey, S.; Newsom-Davis,
- J.: Assignment of the human nicotinic acetylcholine receptor genes:
- the alpha and delta subunit genes to chromosome 2 and the beta subunit
- gene to chromosome 17. Ann. Hum. Genet. 54: 199-208, 1990.
-
- 2. Heidmann, O.; Buonanno, A.; Geoffroy, B.; Robert, B.; Guenet, J.-L.;
- Merlie, J. P.; Changeux, J.-P.: Chromosomal localization of muscle
- nicotinic acetylcholine receptor genes in the mouse. Science 234:
- 866-868, 1986.
-
- 3. Lobos, E. A.; Rudnick, C. H.; Watson, M. S.; Isenberg, K. E.:
- Linkage disequilibrium study of RFLPs detected at the human muscle
- nicotinic acetylcholine receptor subunit genes. Am. J. Hum. Genet. 44:
- 522-533, 1989.
-
- 4. Pasteris, N. G.; Trask, B. J.; Sheldon, S.; Gorski, J. L.: Discordant
- phenotype of two overlapping deletions involving the PAX3 gene in
- chromosome 2q35. Hum. Molec. Genet. 2: 953-959, 1993.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 8/18/1993
- carol: 6/30/1992
- supermim: 3/16/1992
- carol: 10/30/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 100725
- *FIELD* TI
- *100725 CHOLINERGIC RECEPTOR, NICOTINIC, EPSILON POLYPEPTIDE; CHRNE
- ACETYLCHOLINE RECEPTOR, MUSCLE, EPSILON SUBUNIT; ACHRE
- *FIELD* TX
- Acetylcholine receptors at mature mammalian neuromuscular junctions are
- pentameric protein complexes composed of 4 subunits in the ratio of 2
- alpha subunits (100690) to 1 beta (100710), 1 epsilon, and 1 delta
- subunit (100720). Most, if not all, embryonic acetylcholine receptors
- contain a different subunit, gamma (CHRNG; 100730), in place of the
- epsilon subunit. It is likely that this change in subunit composition,
- which occurs during the first 2 weeks after birth, accounts for the
- switch in properties of acetylcholine-activated channels from
- low-conductance, long open time to high-conductance, brief open time
- that occurs over approximately the same time course. In neonatal mouse
- and rat myotubes, epsilon-subunit mRNA is present at low levels, whereas
- gamma-subunit mRNA is present at relatively high levels. During the
- first 2 weeks after birth, the amount of epsilon-subunit mRNA rises
- 10-fold and gamma-subunit mRNA falls to undetectable levels. The
- increase in epsilon-subunit mRNA appears to be confined to the
- developing motor endplate. The switch to the epsilon subunit is mediated
- by ARIA (acetylcholine receptor-inducing activity; 100735).
-
- Lobos (1993) concluded that the CHRNE gene is located about 5 cM from
- the CHRNB1 gene (100710) in the vicinity of TP53 (191170) on 17p13.1.
- Using linkage analysis, the conclusion was confirmed by hybridization of
- CHRNE and CHRNB1 probes to a panel of human/hamster somatic cell
- hybrids. CHRNB1 was previously assigned to 17p12-p11. Beeson et al.
- (1993) isolated cDNA sequences encompassing the full coding region of
- the CHRNE and CHRNG genes. The deduced amino acid sequences indicated
- that the mature epsilon subunit contains 473 amino acids and is preceded
- by a 20-amino acid signal peptide. In common with the human alpha, beta,
- gamma, and delta subunits, the epsilon subunit is highly conserved among
- mammalian species. By PCR analysis of somatic cell hybrids, Beeson et
- al. (1993) demonstrated that the CHRNE gene is located on chromosome 17.
-
- Witzemann et al. (1996) noted that in mammalian muscle the functional
- properties of end plate channels change during postnatal development.
- The length of channel-opening bursts decreases and, as a consequence,
- the duration of miniature end plate current (mEPC) decreases, whereas
- the conductance and the Ca(2+) permeability of end plate channels
- increase. The underlying molecular mechanism is a switch in the
- expression of acetylcholine receptor subunit genes shortly after birth.
- The gamma-subunit (CHRNG) is repressed while the epsilon-subunit gene is
- activated selectively in the myonuclei underlying the synapse. To
- investigate the significance of the CHRNG/CHRNE switch for motor
- behavior, Witzemann et al. (1996) ablated the Chrng gene in mouse
- embryonic stem (es) cells by homologous recombination and injected
- correctly engineered cells of 2 independently isolated clones into
- C57BL/6 blastocyts. Chimeric male mice derived from both clones showed
- germline transmission of the targeted allele. Homozygous mutant animals
- showed that after apparently normal development in early neonatal life,
- neuromuscular transmission was progressively impaired. The lack of
- epsilon subunits caused muscle weakness, defects in motor behavior, and
- premature death 2 to 3 months after birth. Their results demonstrated
- that postnatal incorporation of epsilon subunits in acetylcholine
- receptors into the end plate is essential for normal development of
- skeletal muscle.
-
- *FIELD* AV
- .0001
- MYASTHENIC SYNDROME, SLOW-CHANNEL CONGENITAL
- SCCMS; MYASTHENIA, CONGENITAL
- CHRNE, THR245PRO
- Ohno et al. (1995) demonstrated a mutation in the CHRNE gene in a
- 20-year-old woman who had myasthenic symptoms since the neonatal period,
- a decremental electromyographic response on stimulation of motor nerves,
- negative tests for antiacetylcholine receptor (AChR) antibodies, and no
- history of similarly affected relatives. Studies of an intercostal
- muscle specimen from this patient at age 17 had revealed signs of severe
- end plate myopathy, and patch-clamp studies showed markedly prolonged
- acetylcholine receptor channel openings. The patient was heterozygous
- for an A-to-C transversion at nucleotide 790 in exon 8 of the epsilon
- subunit gene, predicting substitution of proline for threonine at codon
- 264. Genetically engineered mutant AChR expressed in a human embryonic
- kidney fibroblast cell line also exhibited markedly prolonged openings
- in the presence of agonist and even opened in its absence.
-
- .0002
- MYASTHENIC SYNDROME, SLOW-CHANNEL CONGENITAL
- SCCMS
- CHRNE, LEU269PHE
- Engel et al. (1996) described a 16-year-old male patient with myasthenic
- symptoms since early infancy involving ocular, trunkal, and limb
- muscles. He experienced intermittent episodes of respiratory
- insufficiency. SSCP analysis and DNA sequencing revealed that the
- pathological mutation in this patient was a heterozygous leu269-to-phe
- substitution within the transmembrane domain of the AChR-epsilon
- subunit. Engel et al. (1996) evaluated the pathogenicity of the mutation
- by engineering the mutation into the corresponding cDNA of mouse AChR
- and coexpressing it with the wildtype cDNA in HEK fibroblasts. Receptor
- function was evaluated using patch clamp studies and ACh binding was
- measured. These studies revealed that the mutations resulted in an
- apparent increased affinity for ACh and prolonged AChR activation
- episodes rendering the receptor leaky. See also 601462.
-
- *FIELD* SA
- Martinou et al. (1991)
- *FIELD* RF
- 1. Beeson, D.; Brydson, M.; Betty, M.; Jeremiah, S.; Povey, S.; Vincent,
- A.; Newsom-Davis, J.: Primary structure of the human muscle acetylcholine
- receptor cDNA cloning of the gamma and epsilon subunits. Europ. J.
- Biochem. 215: 229-238, 1993.
-
- 2. Engel, A. G.; Ohno, K.; Milone, M.; Wang, H.-L.; Nakano, S.; Bouzat,
- C.; Pruitt, J. N., II; Hutchinson, D. O.; Brengman, J. M.; Bren, N.;
- Sieb, J. P.; Sine, S. M.: New mutations in acetylcholine receptor
- subunit genes reveal heterogeneity in the slow-channel congenital
- myasthenic syndrome. Hum. Molec. Genet. 5: 1217-1227, 1996.
-
- 3. Lobos, E. A.: Five subunit genes of the human muscle nicotinic
- acetylcholine receptor are mapped to two linkage groups on chromosomes
- 2 and 17. Genomics 17: 642-650, 1993.
-
- 4. Martinou, J.-C.; Falls, D. L.; Fischbach, G. D.; Merlie, J. P.
- : Acetylcholine receptor-inducing activity stimulates expression of
- the epsilon-subunit gene of the muscle acetylcholine receptor. Proc.
- Nat. Acad. Sci. 88: 7669-7673, 1991.
-
- 5. Ohno, K.; Hutchinson, D. O.; Milone, M.; Brengman, J. M.; Bouzat,
- C.; Sine, S. M.; Engel, A. G.: Congenital myasthenic syndrome caused
- by prolonged acetylcholine receptor channel openings due to a mutation
- in the M2 domain of the epsilon subunit. Proc. Nat. Acad. Sci. 92:
- 758-762, 1995.
-
- 6. Witzemann, V.; Schwarz, H.; Koenen, M.; Berberich, C.; Villarroel,
- A.; Wernig, A.; Brenner, H. R.; Sakmann, B.: Acetylcholine receptor
- epsilon-subunit deletion causes muscle weakness and atrophy in juvenile
- and adult mice. Proc. Nat. Acad. Sci. 93: 13286-13291, 1996.
-
- *FIELD* CN
- Moyra Smith - updated: 10/9/1996
-
- *FIELD* CD
- Victor A. McKusick: 1/10/1992
-
- *FIELD* ED
- terry: 12/10/1996
- terry: 12/5/1996
- mark: 10/9/1996
- carol: 2/16/1995
- mimadm: 4/14/1994
- carol: 11/9/1993
- carol: 9/22/1993
- supermim: 3/16/1992
- carol: 1/10/1992
-
- *RECORD*
- *FIELD* NO
- 100730
- *FIELD* TI
- *100730 ACETYLCHOLINE RECEPTOR, MUSCLE, GAMMA SUBUNIT; ACHRG
- CHOLINERGIC RECEPTOR, NICOTINIC, GAMMA POLYPEPTIDE; CHRNG
- *FIELD* TX
- See 100690. See also 100720 for a discussion of the probable close
- linkage of the genes for the gamma and delta subunits and their possible
- location on chromosome 2q. Shibahara et al. (1985) showed that the genes
- encoding the gamma and delta subunits of CHRN are contained in an EcoRI
- restriction fragment of approximately 20 kb. Cohen-Haguenauer et al.
- (1989) used a murine full-length 1,900-bp-long cDNA encoding the gamma
- subunit to map the gene to chromosome 2 in human/rodent somatic cell
- hybrids. (They used conditions of low stringency to favor cross-species
- hybridization, and prehybridization with rodent DNA to prevent rodent
- background.) The use of a chromosomal translocation t(X;2)(p22;q32.1)
- served to localize the CHRNG gene to 2q32-qter.
-
- In the first days of life, a switch occurs from the gamma to the epsilon
- subunit (100725) of the acetylcholine receptor. This switch is mediated
- by ARIA (acetylcholine receptor-inducing activity; 100735).
-
- Schurr et al. (1990) mapped this gene to mouse chromosome 1 (symbol
- Acrg) at a position between Vil (193040) proximally and Col6a3 (120250)
- distally.
-
- Two forms of AChR are found in mammalian skeletal muscle cells. The
- mature form is predominant in innervated adult muscle and the embryonic
- form is present in fetal and denervated muscle. Embryonic and mature
- AChR differ by the replacement of the gamma subunit in the pentameric
- glycoprotein complex by its isoform, the epsilon subunit (100725), which
- is specific to the mature AChR subtype. Transient neonatal myasthenia
- gravis occurs in approximately 20% of infants born to mothers with
- myasthenia gravis. Symptoms usually appear within hours after birth and
- disappear after 2 or 3 weeks. The severity of neonatal MG is highly
- variable, ranging from mild hypotonia to respiratory distress requiring
- assisted mechanical ventilation. Antenatal onset leading to multiple
- joint contractures, hydramnios, and decreased fetal movements is rare.
- The disease severity is not correlated to the clinical status of the
- mother. Vernet-der Garabedian et al. (1994) studied 22 mothers with
- myasthenia gravis and their newborns. Twelve mothers had transmitted MG
- to their neonates with, in 3 cases, antenatal injury. A clear
- correlation was found between occurrence of neonatal MG and high overall
- levels of anti-AChR antibodies. However, a strong correlation was also
- found between occurrence of neonatal MG and the ratio of anti-embryonic
- AChR to anti-adult muscle AChR antibodies. Taken together, the data
- suggested that autoantibodies directed against the embryonic form of
- AChR may play a predominant role in the pathogenesis of neonatal MG.
- Paradoxically, the 3 cases with antenatal injury, presumably the most
- severe form of the disorder, were not associated with high ratio of
- anti-embryonic ACh to anti-adult AChR antibodies.
-
- *FIELD* RF
- 1. Cohen-Haguenauer, O.; Barton, P. J.; Buonanno, A.; Cong, N. V.;
- Masset, M.; de Tand, M. F.; Merlie, J.; Frezal, J.: Localization
- of the acetylcholine receptor gamma subunit gene to human chromosome
- 2q32-qter. Cytogenet. Cell Genet. 52: 124-127, 1989.
-
- 2. Schurr, E.; Skamene, E.; Morgan, K.; Chu, M.-L.; Gros, P.: Mapping
- of Col3a1 and Col6a3 to proximal murine chromosome 1 identifies conserved
- linkage of structural protein genes between murine chromosome 1 and
- human chromosome 2q. Genomics 8: 477-486, 1990.
-
- 3. Shibahara, S.; Kubo, T.; Perski, H. J.; Takahashi, H.; Noda, M.;
- Numa, S.: Cloning and sequence analysis of human genomic DNA encoding
- gamma subunit precursor of muscle acetylcholine receptor. Europ.
- J. Biochem. 146: 15-22, 1985.
-
- 4. Vernet-der Garabedian, B.; Lacokova, M.; Eymard, B.; Morel, E.;
- Faltin, M.; Zajac, J.; Sadovsky, O.; Dommergues, M.; Tripon, P.; Bach,
- J.-F.: Association of neonatal myasthenia gravis with antibodies
- against the fetal acetylcholine receptor. J. Clin. Invest. 94:
- 555-559, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 9/29/1994
- supermim: 3/16/1992
- carol: 12/11/1991
- carol: 9/27/1991
- carol: 10/10/1990
- supermim: 3/20/1990
-
- *RECORD*
- *FIELD* NO
- 100735
- *FIELD* TI
- *100735 ACETYLCHOLINE RECEPTOR-INDUCING ACTIVITY; ARIA
- *FIELD* TX
- Martinou et al. (1991) found that acetylcholine receptor-inducing
- activity (ARIA), a 42-kD glycoprotein purified on the basis of its
- ability to increase the synthesis of acetylcholine receptors in chick
- myotubes, increases epsilon-subunit mRNA levels up to 10-fold. Thus,
- ARIA appears to be responsible in a major way for the switch from gamma
- subunits (100730) to epsilon subunits (100725) in the pentameric
- acetylcholine receptor protein complex.
-
- *FIELD* RF
- 1. Martinou, J.-C.; Falls, D. L.; Fischbach, G. D.; Merlie, J. P.
- : Acetylcholine receptor-inducing activity stimulates expression of
- the epsilon-subunit gene of the muscle acetylcholine receptor. Proc.
- Nat. Acad. Sci. 88: 7669-7673, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 9/27/1991
-
- *FIELD* ED
- supermim: 3/16/1992
- carol: 10/10/1991
- carol: 9/27/1991
-
- *RECORD*
- *FIELD* NO
- 100740
- *FIELD* TI
- *100740 ACETYLCHOLINESTERASE
- ACETYLCHOLINE ACETYLHYDROLASE; ACHE
- *FIELD* TX
- Coates and Simpson (1972) concluded that 3 phenotypic variants of
- acetylcholinesterase (EC 3.1.1.7) result from 2 codominant alleles at a
- single locus. Rotundo et al. (1988) showed that all the forms of
- acetylcholinesterase observed in avian nerves and muscle are encoded by
- a single autosomal gene. Differences in assembly and localization of the
- multiple synaptic forms of acetylcholinesterase are thought to arise
- through posttranscriptional events. Lapidot-Lifson et al. (1989)
- referred to the cloning of the gene for acetylcholinesterase. They used
- these clones to study the coamplification of acetylcholinesterase and
- pseudocholinesterase (butyrylcholinesterase; EC 3.1.1.8; 177400). Their
- coamplification in certain leukemias and in disorders of platelet
- formation suggest that the 2 loci may be linked. (The
- pseudocholinesterase gene is located at 3q25.2.) Whereas
- pseudocholinesterase is a soluble plasma enzyme presumed to be produced
- by the liver but also present in muscle and brain, acetylcholinesterase
- or 'true' cholinesterase is involved in the signal transmission at
- neuromuscular junctions and is also intensely expressed in the human
- central nervous system and the erythrocyte membrane.
-
- It has been demonstrated that the Yt erythrocyte blood group antigen
- system (112100) resides on the acetylcholinesterase molecule. Since this
- blood group system has been mapped to the long arm of chromosome 7 in
- the proximity of the COL1A2 (120160) locus, one can conclude that this
- is the site of the acetylcholinesterase locus. That such was the case
- was demonstrated by Getman et al. (1992). By chromosomal in situ
- suppression hybridization analysis, they showed that a single gene is
- located at 7q22 and confirmed the results by PCR analysis of genomic DNA
- from a human/hamster somatic cell hybrid containing a single human
- chromosome 7. Thus the gene maps to the same region that is frequently
- the site of nonrandom deletion in leukemias of myeloid cell precursors
- known to express acetylcholinesterase during normal differentiation.
- Ehrlich et al. (1992) mapped the ACHE gene to 7q22 by fluorescence in
- situ hybridization and by selective PCR amplification from a somatic
- hybrid cell panel and chromosome-sorted DNA libraries. This conforms
- well with the previous assignment of the YT blood group to 7q21-q22.
- Mapping of the ACHE gene to chromosome 3 was convincingly excluded.
- Ehrlich et al. (1992) suggested that the assignment of the gene to 7q22
- may provide an explanation of the in vivo amplification of the ACHE gene
- observed in ovarian tumors and leukemias and the phenomenon of
- tumor-related breakage in 7q. By analysis of a RFLP in recombinant
- inbred (RI) strains, Rachinsky et al. (1992) demonstrated that the Ache
- gene is located on distal mouse chromosome 5.
-
- *FIELD* AV
- .0001
- YT BLOOD GROUP POLYMORPHISM
- ACHE, HIS322ASN
- Bartels et al. (1993) demonstrated that the wildtype sequence of the
- ACHE gene, which corresponds to the YT1 blood group antigen, has
- histidine at codon 322 (CAC) and that the rare variant, the YT2 blood
- group antigen, has asparagine (AAC) at that position.
-
- *FIELD* SA
- Telen and Whitsett (1992)
- *FIELD* RF
- 1. Bartels, C. F.; Zelinski, T.; Lockridge, O.: Mutation at codon
- 322 in the human acetylcholinesterase (ACHE) gene accounts for YT
- blood group polymorphism. Am. J. Hum. Genet. 52: 928-936, 1993.
-
- 2. Coates, P. M.; Simpson, N. E.: Genetic variation in human erythrocyte
- acetylcholinesterase. Science 175: 1466-1467, 1972.
-
- 3. Ehrlich, G.; Viegas-Pequignot, E.; Ginzberg, D.; Sindel, L.; Soreq,
- H.; Zakut, H.: Mapping the human acetylcholinesterase gene to chromosome
- 7q22 by fluorescent in situ hybridization coupled with selective PCR
- amplification from a somatic hybrid cell panel and chromosome-sorted
- DNA libraries. Genomics 13: 1192-1197, 1992.
-
- 4. Getman, D. K.; Eubanks, J. H.; Camp, S.; Evans, G. A.; Taylor,
- P.: The human gene encoding acetylcholinesterase is located on the
- long arm of chromosome 7. Am. J. Hum. Genet. 51: 170-177, 1992.
-
- 5. Lapidot-Lifson, Y.; Prody, C. A.; Ginzberg, D.; Meytes, D.; Zakut,
- H.; Soreq, H.: Coamplification of human acetylcholinesterase and
- butyrylcholinesterase genes in blood cells: correlation with various
- leukemias and abnormal megakaryocytopoiesis. Proc. Nat. Acad. Sci. 86:
- 4715-4719, 1989.
-
- 6. Rachinsky, T. L.; Crenshaw, E. B., III; Taylor, P.: Assignment
- of the gene for acetylcholinesterase to distal mouse chromosome 5. Genomics 14:
- 511-514, 1992.
-
- 7. Rotundo, R. L.; Gomez, A. M.; Fernandez-Valle, C.; Randall, W.
- R.: Allelic variants of acetylcholinesterase: genetic evidence that
- all acetylcholinesterase forms in avian nerves and muscles are encoded
- by a single gene. Proc. Nat. Acad. Sci. 85: 7805-7809, 1988.
-
- 8. Telen, M. J.; Whitsett, C. F.: Erythrocyte acetylcholinesterase
- bears the Cartwright blood group antigens. (Abstract) Clin. Res. 40:
- 170A only, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 12/15/1988
-
- *FIELD* ED
- mark: 11/27/1996
- carol: 4/6/1994
- carol: 5/21/1993
- carol: 4/14/1993
- carol: 11/2/1992
- carol: 10/15/1992
- carol: 8/13/1992
-
- *RECORD*
- *FIELD* NO
- 100790
- *FIELD* TI
- *100790 ACHAETE-SCUTE COMPLEX (DROSOPHILA) HOMOLOG-LIKE 1; ASCL1
- ACHAETE-SCUTE HOMOLOG; ASH1
- *FIELD* TX
- Basic helix-loop-helix transcription factors of the achaete-scute family
- are instrumental in Drosophila neurosensory development and are
- candidate regulators of development in the mammalian central nervous
- system and neural crest. Ball et al. (1993) isolated and characterized a
- human achaete-scute homolog that is highly expressed in 2 neuroendocrine
- cancers, medullary thyroid cancer (155240) and small cell lung cancer
- (182280). The human gene, symbolized ASH1 by them, was cloned from a
- human MTC cDNA library. It encoded a predicted protein of 238 amino
- acids that was 95% homologous to mammalian achaete-scute homolog MASH-1,
- a rodent basic helix-loop-helix factor. The proximal coding region of
- the cDNA contains a striking 14-copy repeat of the triplet CAG that
- exhibits polymorphism in human genomic DNA; thus, ASH1 is a candidate
- locus. By analysis of rodent/human somatic cell hybrids, Ball et al.
- (1993) assigned the gene to human chromosome 12. Northern blots revealed
- ASH1 transcripts in RNA from a human MTC cell line, 2 fresh MTC tumors,
- fetal brain, and 3 lines of human SCLC. In contrast, cultured lines of
- non-SCLC lung cancers and a panel of normal adult human tissues showed
- no detectable ASH1 transcripts. The gene was later symbolized ASCL1.
-
- Achaete-scute homolog-1 was genetically mapped to 12q24.1 by using a CAG
- repeat polymorphism within the gene and a CEPH pedigree DNA panel.
- Twells et al. (1995) subsequently ruled out ASCL1 and NOS1 as candidates
- for spinocerebellar atrophy type 2.
-
- By homologous recombination in embryonic stem cells, Guillemot et al.
- (1993) created a null allele of the mouse Ash-1 gene. Homozygous mice
- died at birth with apparent breathing and feeding defects. The brain and
- spinal cord appeared normal, but the olfactory epithelium and
- sympathetic, parasympathetic, and enteric ganglia were severely
- affected. These observations suggested that the Ash-1 gene, like its
- Drosophila homologs, controls a basic operation in development of
- neuronal progenitors in distinct neural lineages.
-
- Ahmad (1995) found that Mash1 is expressed during development of rat
- retina and interacts specifically with an E-box identified in the
- promoter for the opsin gene during rod photoreceptor differentiation.
-
- Renault et al. (1995) mapped ASCL1 onto a YAC contig distal to PAH
- (261600) and proximal to TRA1 (191175). The authors used fluorescence in
- situ hybridization to determine the cytogenetic assignment of 12q22-q23.
-
- *FIELD* RF
- 1. Ahmad, I.: Mash-1 is expressed during ROD photoreceptor differentiation
- and binds an E-box, E(opsin-1) in the rat opsin gene. Develop. Brain
- Res. 90: 184-189, 1995.
-
- 2. Ball, D. W.; Azzoli, C. G.; Baylin, S. B.; Chi, D.; Dou, S.; Donis-Keller,
- H.; Cumaraswamy, A.; Borges, M.; Nelkin, B. D.: Identification of
- a human achaete-scute homolog highly expressed in neuroendocrine tumors.
- Proc. Nat. Acad. Sci. 90: 5648-5652, 1993.
-
- 3. Guillemot, F.; Lo, L.-C.; Johnson, J. E.; Auerbach, A.; Anderson,
- D. J.; Joyner, A. L.: Mammalian achaete-scute homolog 1 is required
- for the early development of olfactory and autonomic neurons. Cell 75:
- 463-476, 1993.
-
- 4. Renault, B.; Lieman, J.; Ward, D.; Krauter, K.; Kucherlapati, R.
- : Localization of the human achaete-scute homolog gene (ASCL1) distal
- to phenylalanine hydroxylase (PAH) and proximal to tumor rejection
- antigen (TRA1) on chromosome 12q22-q23. Genomics 30: 81-83, 1995.
-
- 5. Twells, R.; Weiming, X.; Ball, D.; Allotey, R.; Williamson, R.;
- Chamberlain, S.: Exclusion of the neuronal nitric oxide synthase
- gene and the human achaete-scute homologue 1 gene as candidate loci
- for spinal cerebellar ataxia. (Letter) Am. J. Hum. Genet. 56:
- 336-337, 1995.
-
- *FIELD* CN
- Orest Hurko - updated: 4/3/1996
- Alan F. Scott - updated: 11/13/1995
-
- *FIELD* CD
- Victor A. McKusick: 7/6/1993
-
- *FIELD* ED
- terry: 04/15/1996
- mark: 4/3/1996
- terry: 3/22/1996
- mark: 1/21/1996
- pfoster: 6/2/1995
- carol: 2/9/1994
- carol: 12/9/1993
- carol: 7/6/1993
-
- *RECORD*
- *FIELD* NO
- 100800
- *FIELD* TI
- #100800 ACHONDROPLASIA; ACH
- *FIELD* MN
- Achondroplasia is the most frequent form of short-limb dwarfism.
- Affected individuals have rhizomelic shortening of the limbs, a
- characteristic facies with frontal bossing and mid-face hypoplasia,
- exaggerated lumbar lordosis, limitation of elbow extension, genu varum,
- and trident hand.
-
- The phenotype is distinctive and easily identified clinically and
- radiologically at birth. In children, caudad narrowing of the
- interpediculate distance, rather than the normal caudad widening, and a
- notchlike sacroiliac groove are typical radiologic features, and
- epiphyseal ossification centers show a circumflex or chevron seat on the
- metaphysis ( Langer et al., 1967). True megalencephaly occurs (Dennis et
- al., 1961). Disproportion between the base of the skull and the brain
- results in internal hydrocephalus in some cases. Obesity in
- achondroplasia is a major problem, which aggravates the morbidity
- associated with lumbar stenosis and contributes to the nonspecific joint
- problems and to the possible early cardiovascular mortality in this
- condition (Hecht et al., 1988).
-
- The large head of the achondroplastic fetus creates an increased risk of
- intracranial bleeding during delivery (Hall et al., 1982). The authors
- recommended that ultrasonography be done at birth and at 2, 4, and 6
- months of age to establish ventricular size, the presence or absence of
- hydrocephalus, and possible intracranial bleed. That some achondroplasts
- have only brainstem compression is common and may contribute to antral
- apnea (Nelson et al., 1988). Pyeritz et al. (1987) reported the results
- of laminectomy for spinal stenosis and made recommendations on the
- optimal extent of surgery.
-
- Homozygosity for the achondroplasia gene results in a severe disorder of
- the skeleton (Hall et al., 1969). Hypochondroplasia (146000) may be
- caused by an allele at the achondroplasia locus (Sommer et al., 1987).
- The delineation from severe hypochondroplasia may be arbitrary.
-
- Achondroplasia is inherited as an autosomal dominant with essentially
- complete penetrance. About seven-eighths of cases are the result of new
- mutation, there being a considerable reduction of effective reproductive
- fitness. There is a paternal age effect (Penrose, 1955). Gonadal
- mosaicism (or spermatogonial mutation) is a possible explanation for the
- occasional report of affected sibs from normal parents (Philip et al.,
- 1988).
-
- The gene for achondroplasia, assigned to 4p16.3 Velinov et al., 1994,
- turns out to be the FGFR3 gene for fibroblast growth factor receptor-3
- (134934). Almost all achondroplasts have a substitution at nucleotide
- 1138, in the transmembrane domain of the FGFR3 gene, the most mutable
- nucleotide discovered to date Bellus et al., 1995. The
- glycine-to-arginine substitution would have a major effect on the
- structure and/or function of the hydrophobic transmembrane domain and
- most likely would have a significant effect on the function of the
- receptor. In embryonic mouse tissues, the highest level of FGFR3 mRNA
- outside of the developing central nervous system was found in the
- prebone cartilage rudiments of all bones. During endochondrial
- ossification, FGFR3 was detected in resting but not hypertrophic
- cartilage Peters et al., 1993. FGFR3 codes for at least 2 isoforms of
- the gene product by alternate use of 2 different exons that encode the
- last half of the third immunoglobulin domain (IgIII), which is primarily
- responsible for the ligand-binding specificity. The isoforms are
- preferentially activated by the various fibroblast growth factors.
-
- Prenatal diagnosis by mid-trimester ultrasonography is feasible
- (Elejalde et al., 1983). The demonstration of a very limited number of
- mutations causing achondroplasia and the ease with which they can be
- detected (1 PCR and 1 restriction digest) provides a simple method for
- prenatal diagnosis of ACH homozygotes Shiang et al., 1994.
-
- The prevalence of achondroplasia is uncertain; most previous estimates
- are undoubtedly incorrect because of misdiagnosis. More recent estimates
- of frequency range from 0.13 per 10,000 births in Denmark (Andersen and
- Hauge, 1989) to 0.5-1.5 per 10,000 births in Latin America (Orioli et
- al., 1986).
-
- *FIELD* ED
- jenny: 02/04/1997 jamie: 12/20/1996
-
- *FIELD* TX
-
- DESCRIPTION
-
- A number sign is used with this entry because of evidence that
- achondroplasia is caused by mutation in the fibroblast growth factor
- receptor-3 gene (FGFR3; 134934), which is located at 4p16.3.
-
- Achondroplasia is the most frequent form of short-limb dwarfism.
- Affected individuals exhibit short stature caused by rhizomelic
- shortening of the limbs, characteristic facies with frontal bossing and
- mid-face hypoplasia, exaggerated lumbar lordosis, limitation of elbow
- extension, genu varum, and trident hand.
-
- Achondroplasia is an autosomal dominant disorder; a majority of cases
- are sporadic, the result of a de novo mutation.
-
- CLINICAL FEATURES
-
- Whereas many conditions that cause short stature have inappropriately
- been called achondroplasia in the past, the phenotype of this
- osteochondrodysplasia is so distinctive and so easily identified
- clinically and radiologically at birth that confusion should not occur.
- It is characterized by a long, narrow trunk, short extremities,
- particularly in the proximal (rhizomelic) segments, a large head with
- frontal bossing, hypoplasia of the midface and a trident configuration
- of the hands. Hyperextensibility of most joints, especially the knees,
- is common, but extension and rotation are limited at the elbow. A
- thoracolumbar gibbus is typically present at birth, but usually gives
- way to exaggerated lumbar lordosis when the child begins to ambulate.
- Mild to moderate hypotonia is common, and motor milestones are usually
- delayed. Intelligence is normal unless hydrocephalus or other central
- nervous system complications arise. In 13 achondroplastic infants, Hecht
- et al. (1991) found that cognitive development was average and did not
- correlate with motor development which typically was delayed. It was
- noteworthy that reduced mental capacity correlated with evidence of
- respiratory dysfunction detected by polysomnography.
-
- In children, caudad narrowing of the interpediculate distance, rather
- than the normal caudad widening, and a notchlike sacroiliac groove are
- typical radiologic features. Also in children, epiphyseal ossification
- centers show a circumflex or chevron seat on the metaphysis. Limb
- shortening is especially striking in the proximal segments, e.g., the
- humerus; hence the description rhizomelic ('root limb'). The radiologic
- features of true achondroplasia and much concerning the natural history
- of the condition were presented by Langer et al. (1967) on the basis of
- a study of 101 cases and by Hall (1988).
-
- True megalencephaly occurs in achondroplasia and has been speculated to
- indicate effects of the gene other than those on the skeleton alone
- (Dennis et al., 1961). Disproportion between the base of the skull and
- the brain results in internal hydrocephalus in some cases. The
- hydrocephalus may be caused by increased intracranial venous pressure
- due to stenosis of the sigmoid sinus at the level of the narrowed
- jugular foramina (Pierre-Kahn et al., 1980). Hall et al. (1982) pointed
- out that the large head of the achondroplastic fetus creates an
- increased risk of intracranial bleeding during delivery. They
- recommended that in the management of achondroplastic infants
- ultrasonography be done at birth and at 2, 4 and 6 months of age to
- establish ventricular size, the presence or absence of hydrocephalus,
- and possible intracranial bleed. They stated the impression that some
- achondroplasts have only megalencephaly, others have true communicating
- hydrocephalus, and yet others have dilated ventricles without
- hydrocephalus. Nelson et al. (1988) concluded that brainstem compression
- is common in achondroplasia and may account in part for the abnormal
- respiratory function.
-
- Pauli et al. (1984) focused attention on the risk of sudden unexpected
- death in infants with achondroplasia. While uncontrolled and
- retrospective, their study demonstrated an excess of deaths in the first
- year of life, most or all of which were attributable to abnormalities at
- the craniocervical junction. Hecht et al. (1987) showed that the excess
- risk of death in infants with achondroplasia may approach 7.5%, largely
- because of cervical cord compression. Pauli et al. (1995) performed a
- prospective assessment of risk for cervical medullary-junction
- compression in 53 infants, 5 of whom were judged to have sufficient
- craniocervical junction compression to require surgical decompression.
- Intraoperative observation showed marked abnormality of the cervical
- spinal cord, and all operated-on children showed marked improvement of
- neurologic function. The best predictors of need for suboccipital
- decompression included lower-limb hyperreflexia or clonus on
- examination, central hypopnea demonstrated by polysomnography, and
- foramen magnum measures below the mean for children with achondroplasia.
-
- Hecht et al. (1988) reviewed the subject of obesity in achondroplasia,
- concluding that it is a major problem which, whatever its underlying
- cause, aggravates the morbidity associated with lumbar stenosis and
- contributes to the nonspecific joint problems and to the possible early
- cardiovascular mortality in this condition. Using data about 409
- Caucasian patients with achondroplasia from different countries (1,147
- observations), Hunter et al. (1996) developed weight for height (W/H)
- curves for these patients. They showed that to a height of about 75 cm,
- the mean W/H curves are virtually identical for normal and
- achondroplastic children. After this height, the W/H curves for
- achondroplastic patients rise above those for the general population.
- Hunter et al. (1996) contended that the best estimation of weight excess
- for achondroplastic patients aged 3 to 6 years is given by the Quetelet
- index, whereas that for patients aged 6 to 18 years is the Rohrer index.
-
- Homozygosity for the achondroplasia gene results in a severe disorder of
- the skeleton with radiologic changes qualitatively somewhat different
- from those of the usual heterozygous achondroplasia; early death results
- from respiratory embarrassment from the small thoracic cage and
- neurologic deficit from hydrocephalus (Hall et al., 1969). Yang et al.
- (1977) reported upper cervical myelopathy in a homozygote.
-
- Horton et al. (1988) found that the epiphyseal and growth plate
- cartilages have a normal appearance histologically, and the major matrix
- constituents exhibit a normal distribution by immunostaining; however,
- morphometric investigations have indicated that the growth plate is
- shorter than normal and that the shortening is greater in homozygous
- than in heterozygous achondroplasia, suggesting a gene dosage effect.
- Stanescu et al. (1990) reported histochemical, immunohistochemical,
- electron microscopic, and biochemical studies on upper tibial cartilage
- from a case of homozygous achondroplasia. No specific abnormality was
- defined. Aterman et al. (1983) expressed puzzlement at the striking
- histologic changes in homozygous achondroplasia despite the virtual
- absence of changes in the heterozygote. They pointed out that histologic
- studies in the heterozygote at a few weeks or months of age have not
- been done. They suggested that because of similarities between what they
- called PHA (presumed homozygous achondroplasia) and thanatophoric
- dwarfism (187600), some cases of the latter condition may be due to a
- particularly severe mutation at the achondroplasia locus.
-
- Hypochondroplasia (146000) may be caused by an allele at the
- achondroplasia locus. The evidence comes from observations of a presumed
- genetic compound in the offspring of an achondroplastic father and a
- hypochondroplastic mother who exhibited growth deficiency and
- radiographic abnormalities of the skeleton that were much more severe
- than those typically seen in achondroplasia (McKusick et al., 1973;
- Sommer et al., 1987) and somewhat less severe than those of the ACH
- homozygote. Young et al. (1992) described lethal short-limb dwarfism in
- the offspring of a father with spondyloepiphyseal dysplasia congenita
- (SEDC; 183900) and a mother with achondroplasia. Young et al. (1992)
- suggested that the infant was a double heterozygote for the 2 dominant
- genes rather than a compound heterozygote. It was considered unlikely
- that SEDC and achondroplasia are allelic because of the evidence that
- most, if not all, cases of SEDC result from mutation in the type II
- collagen gene (COL2A1; 120140), whereas this gene has been excluded as
- the site of the mutation in achondroplasia.
-
- In a presentation of adult genetic skeletal dysplasias found in the
- Museum of Pathological Anatomy in Vienna, Beighton et al. (1993)
- pictured the skeleton of a 61-year-old man with achondroplasia who died
- of transverse myelitis. Randolph et al. (1988) reported an
- achondroplastic patient who developed classic ankylosing spondylitis
- (106300). There is no fundamental connection between the 2 disorders.
- The importance of the observation is mainly to indicate that back
- problems in achondroplasts can be due to causes other than the
- underlying disease.
-
- INHERITANCE
-
- Achondroplasia is inherited as an autosomal dominant with essentially
- complete penetrance. About seven-eighths of cases are the result of new
- mutation, there being a considerable reduction of effective reproductive
- fitness.
-
- Paternal age effect on mutation was noted by Penrose (1955). Stoll et
- al. (1982) reported advanced paternal age in sporadic cases ascertained
- through the French counterpart of LPA (Little People of America), APPT
- (Association des Personnes de Petite Taille). Thompson et al. (1986)
- found that, on average, the severity of achondroplasia tends to be
- reduced with increasing parental age. It is doubtful that a recessive
- form of achondroplasia, indistinguishable from the dominant form,
- exists. Documentation of the diagnosis is inadequate in most reports of
- possible recessive inheritance.
-
- Cohn and Weinberg (1956) reported affected twins with an affected sib.
- (This may have been achondrogenesis, e.g., 200600.) Chiari (1913)
- reported affected half-sibs whose father had achondroplasia. Two first
- cousins, whose mothers were average-statured sisters, had undoubted
- achondroplasia (Wadia, 1969). Most dominants show sufficient variability
- to account for observations such as these on the basis of reduced
- penetrance but such is not the case with achondroplasia.
-
- Gonadal mosaicism (or spermatogonial mutation) is a possible explanation
- for affected sibs from normal parents. Bowen (1974) described a possible
- instance of gonadal mosaicism; 2 daughters of normal parents had
- achondroplasia. One of the daughters had 2 children, one of whom was
- also achondroplastic. Fryns et al. (1983) reported 3 achondroplastic
- sisters born to normal parents. Philip et al. (1988) described the case
- of a man who had 3 daughters with classic achondroplasia, by 2 different
- women.
-
- Affected cousins could be due to the coincidence of two independent
- mutations. Such was probably the case, in McKusick's opinion, in the
- second cousins once removed reported by Fitzsimmons (1985). Reiser et
- al. (1984) reviewed 6 families with unexpected familial recurrence and
- hypothesized that these recurrences were simply the result of two
- independent chance events. Dodinval and Le Marec (1987) reported 2
- families, each with 2 cases of achondroplasia. In 1 family, a girl and
- her great aunt were affected; in the other, male and female first
- cousins. Both germinal mosaicism and paternal age effect appear to have
- their basis in the way spermatogonia are replenished, a feature that
- distinguishes gametogenesis in the male from that in the female. As
- outlined by Clermont (1966), spermatogonia go through a few mitotic
- divisions before embarking on the meiotic divisions that lead to mature
- sperm. Some of the products of the mitotic divisions are returned to the
- 'cell bank' to replenish the supply of spermatogonia. Mutations
- occurring during DNA replication can, therefore, accumulate, providing a
- basis for paternal age effect and for germinal mosaicism. Hoo (1984)
- suggested a small insertional translocation as a possible mechanism for
- recurrent achondroplasia in sibs with normal parents.
-
- The severe phenotype of the homozygote for the ACH gene and the
- possibility that hypochondroplasia represents an allelic disorder were
- discussed in connection with the discussion of clinical features of
- achondroplasia.
-
- Langer et al. (1993) described a patient who was doubly heterozygous for
- achondroplasia and pseudoachondroplasia (177170). Woods et al. (1994)
- described a family in which the father had pseudoachondroplasia and the
- mother had achondroplasia, and 2 daughters were doubly affected and a
- son had achondroplasia only. At birth, the 2 daughters appeared to have
- achondroplasia. Later, the development of a fixed lumbar gibbus, unusual
- radiographic changes in the spine, increasing joint laxity of the hands,
- and characteristic gait and hand posture made the appearance of
- pseudoachondroplasia apparent.
-
- MAPPING
-
- Strom (1984) and Eng et al. (1985) purported to find abnormality of the
- type II collagen gene in achondroplasia. If such a defect is present,
- one might expect ocular abnormality in achondroplasia inasmuch as type
- II collagen is present in vitreous. SED congenita was a more plausible
- candidate for a structural defect of type II collagen because it is a
- dominant disorder that combines skeletal dysplasia with vitreous
- degeneration and deafness (experimental studies with antibodies to type
- II collagen indicate that this collagen type is represented in the
- middle ear); subsequently, defects were in fact found in the COL2A1 gene
- in SEDC. The report by Eng et al. (1985) was withdrawn in 1986 because
- figures, 'which were generated in the laboratory of C. Strom and C. Eng,
- were improperly assembled and therefore cannot be used to support the
- conclusions of the article.' Francomano and Pyeritz (1988) excluded
- COL2A1 as the site of the mutation in achondroplasia by use of probes
- spanning the gene in an analysis of genomic DNA from 49 affected persons
- and 2 multiplex families. No gross rearrangements were seen on Southern
- blot analysis, and linkage studies in the multiplex families
- demonstrated discordant inheritance of achondroplasia and COL2A1
- alleles. Evidence against linkage to COL2A1 has been presented before by
- Ogilvie et al. (1986). From their studies, Finkelstein et al. (1991)
- concluded that mutations at the chondroitin sulfate proteoglycan core
- protein (CSPGP) locus do not cause achondroplasia or
- pseudoachondroplasia (177170).
-
- Edwards et al. (1988) commented on a report, made at the national
- meeting of the Neurofibromatosis Foundation, of 2 individuals with
- achondroplasia and neurofibromatosis (162200) who had translocations
- involving the long arm of chromosome 17. In both cases the breakpoint
- was at the region consistent with localization of the neurofibromatosis
- gene by linkage studies; a third case of coincident achondroplasia and
- neurofibromatosis was also mentioned. Korenberg et al. (1989) and Pulst
- et al. (1990) demonstrated by linkage analysis that the achondroplasia
- locus does not map between the 2 groups of markers flanking the gene for
- neurofibromatosis-1 on human chromosome 17. Verloes et al. (1991)
- observed connatal neuroblastoma in an infant with achondroplasia and
- suggested that the achondroplasia gene may be located on the short arm
- of chromosome 1 where the neuroblastoma gene (256700) appears to be
- situated.
-
- By linkage studies using DNA markers, Velinov et al. (1994) and Le
- Merrer et al. (1994) mapped the gene for achondroplasia and
- hypochondroplasia to the distal area of the short arm of chromosome 4
- (4p16.3). Francomano et al. (1994) likewise mapped the ACH gene to
- 4p16.3, using 18 multigenerational families with achondroplasia and 8
- anonymous dinucleotide repeat polymorphic markers from this region. No
- evidence of genetic heterogeneity was found. Analysis of a recombinant
- family localized the ACH locus to the 2.5-Mb region between D4S43 and
- the telomere.
-
- MOLECULAR GENETICS
-
- Once the gene for achondroplasia was assigned to 4p16.3 by linkage
- analysis (Le Merrer et al., 1994; Velinov et al., 1994; Francomano et
- al., 1994), causative mutations were identified by the candidate gene
- approach and reported within 6 months of the first mapping report.
- Mutations in the gene for fibroblast growth factor receptor-3 (134934)
- were identified by Shiang et al. (1994) and independently by Rousseau et
- al. (1994). The FGFR3 gene had previously been mapped to the same
- region, 4p16.3, as the ACH gene and the Huntington disease gene. The
- mutation in 15 of the 16 achondroplasia-affected chromosomes studied by
- Shiang et al. (1994) was the same, a G-to-A transition at nucleotide
- 1138 (134934.0001) of the cDNA. The mutation on the only other
- ACH-affected chromosome 4 without the G-to-A transition at nucleotide
- 1138 had a G-to-C transversion at this same position (134934.0002). Both
- mutations resulted in the substitution of an arginine residue for a
- glycine at position 380 of the mature protein, which is in the
- transmembrane domain of FGFR3. The mutation was located in a CpG
- dinucleotide. Rousseau et al. (1994) found the G380R mutation in all
- cases studied: 17 sporadic cases and 6 unrelated familial cases. Because
- of the high mutation rate, it might have been predicted that the
- achondroplasia gene is large and that any one of many mutations could
- lead to the same or a similar (hypochondroplasia) phenotype. Such is
- apparently not the case. The fact that there are no reports of
- Wolf-Hirschhorn syndrome (194190) patients with stigmata of
- achondroplasia may indicate that the phenotype is due to some mechanism
- other than haploinsufficiency, i.e., represents a dominant negative
- effect. (The independent work of Shiang et al. (1994) and Rousseau et
- al. (1994) was reported in the 29 July issue of Cell and the 15
- September issue of Nature, respectively.)
-
- Bellus et al. (1995) found that 150 of 154 unrelated achondroplasts had
- the G-to-A transition (134934.0001) and 3 had the G-to-C transversion
- (134934.0002) at nucleotide 1138 of the FGFR3 gene. All 153 had the
- gly380-to-arg substitution; in one individual, an atypical case, the
- gly380-to-arg substitution was missing. Nucleotide 1138 of the FGFR3
- gene is the most mutable nucleotide discovered to date. Superti-Furga et
- al. (1995) reported the case of a newborn with achondroplasia who did
- not carry the mutation at nucleotide 1138 changing glycine-380 to
- arginine but had a mutation causing substitution of a nearby glycine
- with a cysteine (134934.0003).
-
- The FGFR3 gene was isolated and studied in connection with a search for
- the Huntington disease gene. The distribution of FGFR3 mRNA in embryonic
- mouse tissues was found to be more restricted than that of FGFR1
- (136350) and FGFR2 (176943) mRNA. Outside of the developing central
- nervous system, the highest level of FGFR3 mRNA was found to be in the
- prebone cartilage rudiments of all bones, and during endochondral
- ossification, FGFR3 was detected in resting but not hypertrophic
- cartilage (Peters et al., 1993). The glycine-to-arginine substitution
- would have a major effect on the structure, function, or both of the
- hydrophobic transmembrane domain and most likely would have a
- significant effect on the function of the receptor. Five of 6 ACH
- homozygotes were homozygous for the G-to-A transition and each of 6
- sporadic cases, including the parents of 2 of the homozygotes, were
- heterozygous for the 1138A allele and the wildtype allele. The fact that
- FGFR3 transcripts are present in fetal and adult brain (which has the
- highest levels of any tissue) may have relevance in connection with the
- megalencephaly which is thought to occur in achondroplasia (Dennis et
- al., 1961).
-
- FGFR3 codes for at least 2 isoforms of the gene product by alternate use
- of 2 different exons that encode the last half of the third
- immunoglobulin domain (IgIII), which is primarily responsible for the
- ligand-binding specificity. The isoforms are preferentially activated by
- the various fibroblast growth factors.
-
- DIAGNOSIS
-
- The diagnosis is based on the typical clinical and radiologic features;
- the delineation from severe hypochondroplasia may be arbitrary.
-
- The demonstration of a very limited number of mutations causing
- achondroplasia and the ease with which they can be detected (1 PCR and 1
- restriction digest) provides a simple method for prenatal diagnosis of
- ACH homozygotes in families at risk and in which the parents are
- heterozygous for either the 1138A or 1138C allele (Shiang et al., 1994).
- Shiang et al. (1994) expressed the opinion that other than the screening
- of at-risk pregnancies for homozygous ACH fetuses, any 'other
- application of the diagnostic test for ACH mutations should be
- prohibited.' Bellus et al. (1994) practiced prenatal diagnosis by
- chorionic villus sampling at 10 weeks and 4 days of gestation, both
- parents having achondroplasia. Both parents and the fetus were shown to
- be heterozygous for the more common G-to-A transition. Homozygous
- achondroplasia was excluded.
-
- CLINICAL MANAGEMENT
-
- Recommendations for follow-up and management were reviewed at the first
- international symposium on achondroplasia (Nicoletti et al., 1988) and
- by Horton and Hecht (1993). The recommendations included: measurements
- of growth and head circumference using growth curves standardized for
- achondroplasia (Horton et al., 1978); careful neurologic examinations
- (including CT, MRI, somatosensory evoked potentials and polysomnography)
- and surgical enlargement of the foramen magnum in cases of severe
- stenosis; management of frequent middle ear infections and dental
- crowding; measures to control obesity starting in early childhood;
- growth hormone therapy (Horton et al., 1992), which is still
- experimental, and lengthening of the limb bones; tibial osteotomy or
- epiphysiodesis of the fibular growth plate to correct bowing of the
- legs; lumbar laminectomy for spinal stenosis which typically manifests
- in early adulthood; delivery of pregnant women with achondroplasia by
- cesarean section; and prenatal detection of affected fetuses by
- ultrasound.
-
- Shohat et al. (1996) investigated the effect of recombinant human growth
- hormone (hGH) treatment on the growth rate and proportion of individuals
- with achondroplasia and hypochondroplasia. They studied 15 individuals
- over 24 months including 6 months of observation, 12 months of hGH
- therapy (0.04 mg/kg.day), and 6 months of posttreatment growth rate
- determination. The mean growth rate during hGH treatment (5.3 +/- 1.6
- cm) of achondroplasts was significantly increased compared to
- pretreatment (4.0 +/- 1.0 cm/year, P less than 0.01) and posttreatment
- periods (3.1 +/- 1.3 cm; P less than 0.001). In the 4 children with
- hypochondroplasia, the growth rate during hGH treatment was 7.0 +/- 2.4
- cm/year and 4.9 +/- 1.5 cm/year during the pre- and posttreatment
- periods, respectively. In achondroplasts, there was a significant
- increase in growth rate of only the lower segment (from 1.1 +/- 1.6
- cm/year to 3.1 +/- 1.2 cm/year, P less than 0.02). Unexpectedly, this
- treatment does not seem to have a lesser effect on limbs than on trunk
- growth rate and, therefore, during 1 year of treatment, does not
- increase body disproportion.
-
- Hunter et al. (1996) recommended that achondroplastic children stay
- within 1 SD of the mean weight for height curves for achondroplasts.
-
- Waters et al. (1995) studied the results of treatment of obstructive
- sleep apnea in achondroplasia. Treatment included adenotonsillectomy,
- weight loss, and nasal-mask continuous positive airway pressure (CPAP).
- They observed improvements in measurements of disturbed sleep
- architecture and some evidence of improvement in neurologic function.
-
- Weber et al. (1996) studied the effects of recombinant human growth
- hormone treatment in 6 prepubertal children with achondroplasia, ranging
- in age from 2 to 8 years. They were given a GH dose of 0.1 IU/kg/day
- subcutaneously. During the year of treatment the growth velocity
- increased from 1.1 to 2.6 cm/year in 3 patients while in the others no
- variation was detected. No side effects were observed during the trial
- apart from the slight advancement of bone age in 2 patients. Their
- findings confirmed the individual variability in the response to GH
- treatment.
-
- POPULATION GENETICS
-
- The prevalence of achondroplasia is uncertain; previous estimates are
- undoubtedly incorrect because of misdiagnosis. For example, Wallace et
- al. (1970) reported 2 female sibs as examples of achondroplasia; both
- died in the neonatal period and showed, in addition to chondrodystrophy,
- central harelip, hypoplastic lungs, and hydrocephalus. Without
- radiographic studies it is impossible to identify the nature of this
- condition, but it is certainly not true achondroplasia; Jeune
- asphyxiating thoracic dystrophy (208500), thanatophoric dwarfism, and
- achondrogenesis are each possibilities.
-
- Using modern diagnostic criteria, Gardner (1977) estimated the mutation
- rate at 0.000014. Orioli et al. (1986) reported on the frequency of
- skeletal dysplasias among 349,470 births (live and stillbirths). The
- prevalence rate for achondroplasia was between 0.5 and 1.5/10,000
- births. The mutation rate was estimated to be between 1.72 and 5.57 x
- 10(-5) per gamete per generation. The stated range is a consequence of
- the uncertainty of diagnosis in some cases. (The thanatophoric
- dysplasia/achondrogenesis group had a prevalence between 0.2 and
- 0.5/10,000 births. Osteogenesis imperfecta had a prevalence of
- 0.4/10,000 births. Only 1 case of diastrophic dysplasia was identified.)
- In the county of Fyn in Denmark, Andersen and Hauge (1989) determined
- the prevalence of generalized bone dysplasias by study of all children
- born in a 14-year period. The figures, which they referred to as
- 'point-prevalence at birth,' showed that achondroplasia was less common
- than generally thought (1.3 per 100,000), while osteogenesis imperfecta
- (21.8), multiple epiphyseal dysplasia tarda (9.0), achondrogenesis
- (6.4), osteopetrosis (5.1), and thanatophoric dysplasia (3.8) were found
- to be more frequent. Stoll et al. (1989) found a mutation rate of 3.3 x
- 10(-5) per gamete per generation. In Spain, Martinez-Frias et al. (1991)
- found a frequency of achondroplasia of 2.53 per 100,000 live births.
- Total prevalence of autosomal dominant malformation syndromes was 12.1
- per 100,000 live births.
-
- HISTORY
-
- It is of historic interest that Weinberg (1912), of Hardy-Weinberg law
- fame, noted in the data collected by Rischbieth and Barrington that
- sporadic cases were more often last-born than first-born. The studies by
- Morch (1941) in Denmark and by Hobaek (1961) were early examples of full
- population studies.
-
- *FIELD* SA
- Beighton and Bathfield (1981); Cohen et al. (1967); Durr (1968);
- Elejalde et al. (1983); Fremion et al. (1984); Hall et al. (1979);
- Maroteaux and Lamy (1964); Morgan and Young (1980); Murdoch et al.
- (1970); Oberklaid et al. (1979); Opitz (1984); Pauli et al. (1983);
- Penrose (1957); Pyeritz et al. (1987); Rimoin et al. (1970); Siebens
- et al. (1978)
- *FIELD* RF
- 1. Andersen, P. E., Jr.; Hauge, M.: Congenital generalised bone dysplasias:
- a clinical, radiological, and epidemiological survey. J. Med. Genet. 26:
- 37-44, 1989.
-
- 2. Aterman, K.; Welch, J. P.; Taylor, P. G.: Presumed homozygous
- achondroplasia: a review and report of a further case. Path. Res.
- Pract. 178: 27-39, 1983.
-
- 3. Beighton, P.; Bathfield, C. A.: Gibbal achondroplasia. J. Bone
- Joint Surg. 63: 328-329, 1981.
-
- 4. Beighton, P.; Sujansky, E.; Patzak, B.; Portele, K. A.: Genetic
- skeletal dysplasias in the Museum of Pathological Anatomy, Vienna. Am.
- J. Med. Genet. 47: 843-847, 1993.
-
- 5. Bellus, G. A.; Escallon, C. S.; de Luna, R. O.; Shumway, J. B.;
- Blakemore, K. J.; McIntosh, I.; Francomano, C. A.: First-trimester
- prenatal diagnosis in couple at risk for homozygous achondroplasia.
- (Letter) Lancet 344: 1511-1512, 1994.
-
- 6. Bellus, G. A.; Hefferon, T. W.; Ortiz de Luna, R. I.; Hecht, J.
- T.; Horton, W. A.; Machado, M.; Kaitila, I.; McIntosh, I.; Francomano,
- C. A.: Achondroplasia is defined by recurrent G380R mutations of
- FGFR3. Am. J. Hum. Genet. 56: 368-373, 1995.
-
- 7. Bowen, P.: Achondroplasia in two sisters with normal parents. Birth
- Defects Orig. Art. Ser. X(12): 31-36, 1974.
-
- 8. Chiari, H.: Ueber familiaere Chondrodystrophia foetalis. Muench.
- Med. Wschr. 60: 248-249, 1913.
-
- 9. Clermont, Y.: Renewal of spermatogonia in man. Am. J. Anat. 118:
- 509-524, 1966.
-
- 10. Cohen, M. E.; Rosenthal, A. D.; Matson, D. D.: Neurological abnormalities
- in achondroplastic children. J. Pediat. 71: 367-376, 1967.
-
- 11. Cohn, S.; Weinberg, A.: Identical hydrocephalic achondroplastic
- twins. Subsequent delivery of single sibling with same abnormality. Am.
- J. Obstet. Gynec. 72: 1346-1348, 1956.
-
- 12. Dennis, J. P.; Rosenberg, H. S.; Alvord, E. C., Jr.: Megalencephaly,
- internal hydrocephalus and other neurological aspects of achondroplasia. Brain 84:
- 427-445, 1961.
-
- 13. Dodinval, P.; Le Marec, B.: Genetic counselling in unexpected
- familial recurrence of achondroplasia. Am. J. Med. Genet. 28: 949-954,
- 1987.
-
- 14. Durr, D. K.: Eine neue Dysostoseform mit Mikromelie bei zwei
- Geschwistern. Helv. Paediat. Acta 23: 184-194, 1968.
-
- 15. Edwards, J. H.; Huson, S.; Ponder, B.: Neurofibromatosis. (Letter) Lancet II:
- 330, 1988.
-
- 16. Elejalde, B. R.; Elejalde, M. M.; Hamilton, P. R.; Lombardi, J.
- N.: Prenatal diagnosis in two pregnancies of an achondroplastic woman. Am.
- J. Med. Genet. 15: 437-439, 1983.
-
- 17. Eng, C. E. L.; Pauli, R. M.; Strom, C. M.: Nonrandom association
- of a type II procollagen genotype with achondroplasia. Proc. Nat.
- Acad. Sci. 82: 5465-5469, 1985. Note: Retraction: Proc. Nat. Acad.
- Sci. 83:5354 only, 1986.
-
- 18. Finkelstein, J. E.; Doege, K.; Yamada, Y.; Pyeritz, R. E.; Graham,
- J. M., Jr.; Moeschler, J. B.; Pauli, R. M.; Hecht, J. T.; Francomano,
- C. A.: Analysis of the chondroitin sulfate proteoglycan core protein
- (CSPGP) gene in achondroplasia and pseudoachondroplasia. Am. J. Hum.
- Genet. 48: 97-102, 1991.
-
- 19. Fitzsimmons, J. S.: Familial recurrence of achondroplasia. Am.
- J. Med. Genet. 22: 609-613, 1985.
-
- 20. Francomano, C. A.; Ortiz de Luna, R. I.; Hefferon, T. W.; Bellus,
- G. A.; Turner, C. E.; Taylor, E.; Meyers, D. A.; Blanton, S. H.; Murray,
- J. C.; McIntosh, I.; Hecht, J. T.: Localization of the achondroplasia
- gene to the distal 2.5 Mb of human chromosome 4p. Hum. Molec. Genet. 3:
- 787-792, 1994.
-
- 21. Francomano, C. A.; Pyeritz, R. E.: Achondroplasia is not caused
- by mutation in the gene for type II collagen. Am. J. Med. Genet. 29:
- 955-961, 1988.
-
- 22. Fremion, A. S.; Garg, B. P.; Kalsbeck, J.: Apnea as the sole
- manifestation of cord compression in achondroplasia. J. Pediat. 104:
- 398-401, 1984.
-
- 23. Fryns, J. P.; Kleczkowska, A.; Verresen, H.; van den Berghe, H.
- : Germinal mosaicism in achondroplasia: a family with 3 affected siblings
- of normal parents. Clin. Genet. 24: 156-158, 1983.
-
- 24. Gardner, R. J. M.: A new estimate of the achondroplasia mutation
- rate. Clin. Genet. 11: 31-38, 1977.
-
- 25. Hall, J. G.: The natural history of achondroplasia.In: Nicoletti,
- B.; Kopits, S. E.; Ascani, E.; McKusick, V. A.: Human Achondroplasia:
- A Multidisciplinary Approach. New York: Plenum Press (pub.) 1988.
- Pp. 3-10.
-
- 26. Hall, J. G.; Dorst, J. P.; Taybi, H.; Scott, C. I., Jr.; Langer,
- L. O., Jr.; McKusick, V. A.: Two probable cases of homozygosity for
- the achondroplasia gene. Birth Defects Orig. Art. Ser. V(4): 24-34,
- 1969.
-
- 27. Hall, J. G.; Golbus, M. S.; Graham, C. B.; Pagon, R. A.; Luthy,
- D. A.; Filly, R. A.: Failure of early prenatal diagnosis in classic
- achondroplasia. Am. J. Med. Genet. 3: 371-375, 1979.
-
- 28. Hall, J. G.; Horton, W.; Kelly, T.; Scott, C. I.: Head growth
- in achondroplasia: use of ultrasound studies. (Letter) Am. J. Med.
- Genet. 13: 105, 1982.
-
- 29. Hecht, J. T.; Francomano, C. A.; Horton, W. A.; Annegers, J. F.
- : Mortality in achondroplasia. Am. J. Hum. Genet. 41: 454-464, 1987.
-
- 30. Hecht, J. T.; Hood, O. J.; Schwartz, R. J.; Hennessey, J. C.;
- Bernhardt, B. A.; Horton, W. A.: Obesity in achondroplasia. Am.
- J. Med. Genet. 31: 597-602, 1988.
-
- 31. Hecht, J. T.; Thompson, N. M.; Weir, T.; Patchell, L.; Horton,
- W. A.: Cognitive and motor skills in achondroplastic infants: neurologic
- and respiratory correlates. Am. J. Hum. Genet. 41: 208-211, 1991.
-
- 32. Hobaek, A.: Problems of Hereditary Chondrodysplasia. Oslo:
- Oslo Univ. Press (pub.) 1961.
-
- 33. Hoo, J. J.: Alternative explanations for recurrent achondroplasia
- in siblings with normal parents. Clin. Genet. 25: 553-554, 1984.
-
- 34. Horton, W. A.; Hecht, J. T.: The chondrodysplasias.In: Royce,
- P. M.; Steinmann, B.: Connective Tissue and Its Heritable Disorders:
- Molecular, Genetic, and Medical Aspects. New York: Wiley-Liss (pub.)
- 1993. Pp. 641-675.
-
- 35. Horton, W. A.; Hecht, J. T.; Hood, O. J.; Marshall, R. N.; Moore,
- W. V.; Hollowell, J. G.: Growth hormone therapy in achondroplasia. Am.
- J. Med. Genet. 42: 667-670, 1992.
-
- 36. Horton, W. A.; Hood, O. J.; Machado, M. A.; Campbell, D.: Growth
- plate cartilage studies in achondroplasia.In: Nicoletti, B.; Kopits,
- S. E.; Ascani, E.; McKusick, V. A.: Human Achondroplasia: A Multidisciplinary
- Approach. New York: Plenum Press (pub.) 1988. Pp. 81-89.
-
- 37. Horton, W. A.; Rotter, J. I.; Rimoin, D. L.; Scott, C. L.; Hall,
- J. G.: Standard growth curves for achondroplasia. J. Pediat. 93:
- 435-438, 1978.
-
- 38. Hunter, A. G. W.; Hecht, J. T.; Scott, Jr., C. I.: Standard weight
- for height curves in achondroplasia. Am. J. Med. Genet. 62: 255-261,
- 1996.
-
- 39. Korenberg, J. R.; Barker, D.; Fain, P.; Graham, J.; Pribyl, T.;
- Pulst, S.-M.: Achondroplasia is not tightly linked to the locus for
- neurofibromatosis 1. (Abstract) Cytogenet. Cell Genet. 51: 1025,
- 1989.
-
- 40. Langer, L. O., Jr.; Baumann, P. A.; Gorlin, R. J.: Achondroplasia. Am.
- J. Roentgen. 100: 12-26, 1967.
-
- 41. Langer, L. O., Jr.; Schaefer, G. B.; Wadsworth, D. T.: Patient
- with double heterozygosity for achondroplasia and pseudoachondroplasia,
- with comments on these conditions and the relationship between pseudoachondroplasia
- and multiple epiphyseal dysplasia, Fairbank type. Am. J. Med. Genet. 47:
- 772-781, 1993.
-
- 42. Le Merrer, M.; Rousseau, F.; Legeai-Mallet, L.; Landais, J.-C.;
- Pelet, A.; Bonaventure, J.; Sanak, M.; Weissenbach, J.; Stoll, C.;
- Munnich, A.; Maroteaux, P.: A gene for achondroplasia--hypochondroplasia
- maps to chromosome 4p. Nature Genet. 6: 314-317, 1994.
-
- 43. Maroteaux, P.; Lamy, P.: Achondroplasia in man and animals. Clin.
- Orthop. 33: 91-103, 1964.
-
- 44. Martinez-Frias, M. L.; Cereijo, A.; Bermejo, E.; Lopez, M.; Sanchez,
- M.; Gonzalo, C.: Epidemiological aspects of mendelian syndromes in
- a Spanish population sample: I. Autosomal dominant malformation syndromes. Am.
- J. Med. Genet. 38: 622-625, 1991.
-
- 45. McKusick, V. A.; Kelly, T. E.; Dorst, J. P.: Observations suggesting
- allelism of the achondroplasia and hypochondroplasia genes. J. Med.
- Genet. 10: 11-16, 1973.
-
- 46. Morch, E. T.: Chondrodystrophic dwarfs in Denmark. Op. Ex Domo
- Biol. Hered. Hum. U. Hafniensis 3: 1-200, 1941.
-
- 47. Morgan, D. F.; Young, R. F.: Spinal neurological complications
- of achondroplasia: results of surgical treatment. J. Neurosurg. 52:
- 463-472, 1980.
-
- 48. Murdoch, J. L.; Walker, B. A.; Hall, J. G.; Abbey, H.; Smith,
- K. K.; McKusick, V. A.: Achondroplasia--a genetic and statistical
- survey. Ann. Hum. Genet. 33: 227-244, 1970.
-
- 49. Nelson, F. W.; Hecht, J. T.; Horton, W. A.; Butler, I. J.; Goldie,
- W. D.; Miner, M.: Neurological basis of respiratory complications
- in achondroplasia. Ann. Neurol. 24: 89-93, 1988.
-
- 50. Nicoletti, B.; Kopits, S. E.; Ascani, E.; McKusick, V. A.: Human
- Achondroplasia: A Multidisciplinary Approach. New York: Plenum Press
- (pub.) 1988. Pp. 3-9.
-
- 51. Oberklaid, F.; Danks, D. M.; Jensen, F.; Stace, L.; Rosshandler,
- S.: Achondroplasia and hyperchondroplasia: comments on frequency,
- mutation rate, and radiological features in skull and spine. J. Med.
- Genet. 16: 140-146, 1979.
-
- 52. Ogilvie, D.; Wordsworth, P.; Thompson, E.; Sykes, B.: Evidence
- against the structural gene encoding type II collagen (COL2A1) as
- the mutant locus in achondroplasia. J. Med. Genet. 23: 19-22, 1986.
-
- 53. Opitz, J. M.: 'Unstable premutation' in achondroplasia: penetrance
- vs phenotrance. (Editorial) Am. J. Med. Genet. 19: 251-254, 1984.
-
- 54. Orioli, I. M.; Castilla, E. E.; Barbosa-Neto, J. G.: The birth
- prevalence rates for the skeletal dysplasias. J. Med. Genet. 23:
- 328-332, 1986.
-
- 55. Pauli, R. M.; Conroy, M. M.; Langer, L. O., Jr.; McLone, D. G.;
- Naidich, T.; Franciosi, R.; Ratner, I. M.; Copps, S. C.: Homozygous
- achondroplasia with survival beyond infancy. Am. J. Med. Genet. 16:
- 459-473, 1983.
-
- 56. Pauli, R. M.; Horton, V. K.; Glinski, L. P.; Reiser, C. A.: Prospective
- assessment of risks for cervicomedullary-junction compression in infants
- with achondroplasia. Am. J. Hum. Genet. 56: 732-744, 1995.
-
- 57. Pauli, R. M.; Scott, C. I.; Wassman, E. R., Jr.; Gilbert, E. F.;
- Leavitt, L. A.; Ver Hoeve, J.; Hall, J. G.; Partington, M. W.; Jones,
- K. L.; Sommer, A.; Feldman, W.; Langer, L. O.; Rimoin, D. L.; Hecht,
- J. T.; Lebovitz, R.: Apnea and sudden unexpected death in infants
- with achondroplasia. J. Pediat. 104: 342-348, 1984.
-
- 58. Penrose, L. S.: Parental age in achondroplasia and mongolism. Am.
- J. Hum. Genet. 9: 167-169, 1957.
-
- 59. Penrose, L. S.: Parental age and mutation. Lancet II: 312-313,
- 1955.
-
- 60. Peters, K.; Ornitz, D.; Werner, S.; Williams, L.: Unique expression
- pattern of the FGF receptor 3 gene during mouse organogenesis. Dev.
- Biol. 155: 423-430, 1993.
-
- 61. Philip, N.; Auger, M.; Mattei, J. F.; Giraud, F.: Achondroplasia
- in sibs of normal parents. J. Med. Genet. 25: 857-859, 1988.
-
- 62. Pierre-Kahn, A.; Hirsch, J. F.; Renier, D.; Metzger, J.; Maroteaux,
- P.: Hydrocephalus and achondroplasia: a study of 25 observations. Child's
- Brain 7: 205-219, 1980.
-
- 63. Pulst, S.-M.; Graham, J. M., Jr.; Fain, P.; Barker, D.; Pribyl,
- T.; Korenberg, J. R.: The achondroplasia gene is not linked to the
- locus for neurofibromatosis 1 on chromosome 17. Hum. Genet. 85:
- 12-14, 1990.
-
- 64. Pyeritz, R. E.; Sack, G. H., Jr.; Udvarhelyi, G. B.: Thoracolumbosacral
- laminectomy in achondroplasia: long-term results in 22 patients. Am.
- J. Med. Genet. 28: 433-444, 1987.
-
- 65. Randolph, L. M.; Shohat, M.; Miller, D.; Lachman, R.; Rimoin,
- D. L.: Achondroplasia with ankylosing spondylitis. Am. J. Med. Genet. 31:
- 117-121, 1988.
-
- 66. Reiser, C. A.; Pauli, R. M.; Hall, J. G.: Achondroplasia: unexpected
- familial recurrence. Am. J. Med. Genet. 19: 245-250, 1984.
-
- 67. Rimoin, D. L.; Hughes, G. N.; Kaufman, R. L.; Rosenthal, R. E.;
- McAlister, W. H.; Silberberg, R.: Endochondral ossification in achondroplastic
- dwarfism. New Eng. J. Med. 283: 728-735, 1970.
-
- 68. Rousseau, F.; Bonaventure, J.; Legeai-Mallet, L.; Pelet, A.; Rozet,
- J.-M.; Maroteaux, P.; Le Merrer, M.; Munnich, A.: Mutations in the
- gene encoding fibroblast growth factor receptor-3 in achondroplasia. Nature 371:
- 252-254, 1994.
-
- 69. Shiang, R.; Thompson, L. M.; Zhu, Y.-Z.; Church, D. M.; Fielder,
- T. J.; Bocian, M.; Winokur, S. T.; Wasmuth, J. J.: Mutations in the
- transmembrane domain of FGFR3 cause the most common genetic form of
- dwarfism, achondroplasia. Cell 78: 335-342, 1994.
-
- 70. Shohat, M.; Tick, D.; Barakat, S.; Bu, X.; Melmed, S.; Rimoin,
- D.L.: Short-term recombinant human growth hormone treatment increases
- growth rate in achondroplasia. J. Clin. Endocr. Metab. 81: 4033-4037,
- 1996.
-
- 71. Siebens, A. A.; Hungerford, D. S.; Kirby, N. A.: Curves of the
- achondroplastic spine: a new hypothesis. Johns Hopkins Med. J. 142:
- 205-210, 1978.
-
- 72. Sommer, A.; Young-Wee, T.; Frye, T.: Achondroplasia-hypochondroplasia
- complex. Am. J. Med. Genet. 26: 949-957, 1987.
-
- 73. Stanescu, R.; Stanescu, V.; Maroteaux, P.: Homozygous achondroplasia:
- morphologic and biochemical study of cartilage. Am. J. Med. Genet. 37:
- 412-421, 1990.
-
- 74. Stoll, C.; Dott, B.; Roth, M.-P.; Alembik, Y.: Birth prevalence
- rates of skeletal dysplasias. Clin. Genet. 35: 88-92, 1989.
-
- 75. Stoll, C.; Roth, M.-P.; Bigel, P.: A reexamination of parental
- age effect on the occurrence of new mutations for achondroplasia.In:
- Papadatos, C. J.; Bartsocas, C. S.: Skeletal Dysplasias. New York:
- Alan R. Liss (pub.) 1982. Pp. 419-426.
-
- 76. Strom, C. M.: Achondroplasia due to DNA insertion into the type
- II collagen gene. (Abstract) Pediat. Res. 18: 226A, 1984.
-
- 77. Superti-Furga, A.; Eich, G.; Bucher, H. U.; Wisser, J.; Giedion,
- A.; Gitzelmann, R.; Steinmann, B.: A glycine 375-to-cysteine substitution
- in the transmembrane domain of the fibroblast growth factor receptor-3
- in a newborn with achondroplasia. Europ. J. Pediat. 154: 215-219,
- 1995.
-
- 78. Thompson, J. N., Jr.; Schaefer, G. B.; Conley, M. C.; Mascie-Taylor,
- C. G. N.: Achondroplasia and parental age. (Letter) New Eng. J.
- Med. 314: 521-522, 1986.
-
- 79. Velinov, M.; Slaugenhaupt, S. A.; Stoilov, I.; Scott, C. I., Jr.;
- Gusella, J. F.; Tsipouras, P.: The gene for achondroplasia maps to
- the telomeric region of chromosome 4p. Nature Genet. 6: 318-321,
- 1994.
-
- 80. Verloes, A.; Massart, B.; Jossa, V.; Langhendries, J. P.; Hainaut,
- H.; Paquot, J. P.; Koulischer, L.: Neuroblastoma in a dwarfed newborn:
- possible clue to the chromosomal localization of the gene for achondroplasia?. Ann.
- Genet. 34: 25-26, 1991.
-
- 81. Wadia, R.: Achondroplasia in two first cousins. Birth Defects
- Orig. Art. Ser. V(4): 227-230, 1969.
-
- 82. Wallace, D. C.; Exton, L. A.; Pritchard, D. A.; Leung, Y.; Cooke,
- R. A.: Severe achondroplasia: demonstration of probable heterogeneity
- within this clinical syndrome. J. Med. Genet. 7: 22-26, 1970.
-
- 83. Waters, K. A.; Everett, F.; Sillence, D. O.; Fagan, E. R.; Sullivan,
- C. E.: Treatment of obstructive sleep apnea in achondroplasia: evaluation
- of sleep, breathing, and somatosensory-evoked potentials. Am. J.
- Med. Genet. 59: 460-466, 1995.
-
- 84. Weber, G.; Prinster, C.; Meneghel, M.; Russo, F.; Mora, S.; Puzzovio,
- M.; Del Maschio, M.; Chiumello, G.: Human growth hormone treatment
- in prepubertal children with achondroplasia. Am. J. Med. Genet. 61:
- 396-400, 1996.
-
- 85. Weinberg, W.: Zur Vererbung des Zwergwuchses. Arch. Rass. Ges.
- Biol. 9: 710-717, 1912.
-
- 86. Woods, C. G.; Rogers, J. G.; Mayne, V.: Two sibs who are double
- heterozygotes for achondroplasia and pseudoachondroplastic dysplasia. J.
- Med. Genet. 31: 565-569, 1994.
-
- 87. Yang, S. S.; Corbett, D. P.; Brough, A. J.; Heidelberger, K. P.;
- Bernstein, J.: Upper cervical myelopathy in achondroplasia. Am.
- J. Clin. Path. 68: 68-72, 1977.
-
- 88. Young, I. D.; Ruggins, N. R.; Somers, J. M.; Zuccollo, J. M.;
- Rutter, N.: Lethal skeletal dysplasia owing to a double heterozygosity
- for achondroplasia and spondyloepiphyseal dysplasia congenita. J.
- Med. Genet. 29: 831-833, 1992.
-
- *FIELD* CS
- Skel:
- Osteochondrodysplasia
-
- Growth:
- Short-limb dwarfism identifiable at birth;
- Mean male adult height: 131 cm;
- Mean female height: 124 cm;
- Obesity, tendency to
-
- Head:
- Frontal bossing;
- Megalencephaly
-
- Facies:
- Midfacial hypoplasia;
- Low nasal bridge
-
- Eyes:
- Strabismus
-
- Ears:
- Recurrent otitis media in infancy and childhood;
- Conductive hearing loss
-
- Resp:
- Respiratory insufficiency;
- Upper airway obstruction
-
- Spine:
- Lumbar gibbus in infancy;
- Exaggerated lumbar lordosis during childhood and adulthood
-
- Joints:
- Limited elbow and hip extension
-
- Limbs:
- Trident hand;
- Brachydactyly;
- Limited extension at elbows;
- Genu varum;
- Bowleg;
- Rhizomelia
-
- Neuro:
- Hydrocephalus, occasional;
- Mild hypotonia in infancy and early childhood;
- Lumbar spinal stenosis common;
- Occasional thoracic or cervical spinal stenosis;
- Radiculopathy;
- Brain stem compression
-
- Misc:
- Paternal age mutation effect
-
- Radiology:
- Cuboidal vertebral bodies;
- Progressive lumbar interpediculate narrowing after first year;
- Vertebral canal narrows in cranio-caudal direction;
- Notch-like sacroiliac groove;
- Metaphyseal flaring;
- Circumflex or chevron seated epiphyseal ossification centers on the
- metaphysis;
- Short narrow femoral neck;
- Vertebral scalloping;
- Wide intervertebral discs;
- Foraminal narrowing;
- Flat roofed acetabula;
- Small foramen magnum;
- Short cranial base;
- Early sphenooccipital closure
-
- Inheritance:
- Autosomal dominant with complete penetrance;
- most (7/8) cases new mutations
-
- *FIELD* CN
- Victor A. McKusick - edited: 02/04/1997
-
- *FIELD* ED
- joanna: 02/04/1997
-
- *FIELD* CN
- John A. Phillips, III - updated: 4/1/1997
- Victor A. McKusick - updated: 2/4/1997
- Iosif W. Lurie - updated: 7/1/1996
- Beat Steinmann - updated: 2/4/1994
-
- *FIELD* CD
- Victor A. McKusick: 6/16/1986
-
- *FIELD* ED
- jenny: 04/04/1997
- jenny: 4/1/1997
- joanna: 2/14/1997
- joanna: 2/4/1997
- terry: 12/17/1996
- carol: 7/1/1996
- mark: 4/11/1996
- mark: 2/26/1996
- terry: 2/20/1996
- mark: 1/17/1996
- terry: 1/16/1996
- mark: 7/19/1995
- terry: 2/27/1995
- carol: 1/18/1995
- mimadm: 6/8/1994
- warfield: 3/31/1994
-
- *RECORD*
- *FIELD* NO
- 100820
- *FIELD* TI
- *100820 ACHOO SYNDROME
- AUTOSOMAL DOMINANT COMPELLING HELIOOPHTHALMIC OUTBURST SYNDROME;;
- PHOTIC SNEEZE REFLEX;;
- SNEEZING FROM LIGHT EXPOSURE;;
- PEROUTKA SNEEZE
- *FIELD* TX
- Collie et al. (1978) described a 'disorder' characterized by nearly
- uncontrollable paroxysms of sneezing provoked in a reflex fashion by the
- sudden exposure of a dark-adapted subject to intensely bright light,
- usually sunlight. The number of successive sneezes was usually 2 or 3,
- but could be as many as 43. The 4 authors were the probands of the 4
- families they reported. Several instances of male-to-male transmission
- were noted. Sneezing in response to bright light was said by Peroutka
- and Peroutka (1984) to be a common yet poorly understood phenomenon.
- Photic sneeze reflex was suggested as the appropriate designation by
- Everett (1964), who found it in 23% of Johns Hopkins medical students.
- In a poll of 25 neurologists at Johns Hopkins, Peroutka and Peroutka
- (1984) found the phenomenon in 9, but only 2 of the respondents knew
- that such a specific reflex exists. The Peroutkas (father and daughter)
- reported the reflex in 3 generations of their family: grandfather, the
- father (the proband), his brother and his daughter. The index subject
- (S.J.P.) invariably sneezes twice when he moves from indoors into bright
- sunlight. Lewkonia (1969) described sneezing as a complication of slit
- lamp examination. Katz et al. (1990) found light-induced sneezing in 5
- of 19 patients with nephropathic cystinosis (219800). This was
- presumably related to the crystal deposition in the cornea. Lerner
- (1991) took Hunter (1990) to task for referring to the photic sneeze
- reflex as a 'comic syndrome.' He cited reports by Beckman and Nordenson
- (1983), Forrester (1985), Morris (1987), and Lang and Howland (1987), in
- addition to those already cited here. Benbow (1991) reported that he had
- suffered from photic sneezing for over 20 years and having just learned
- of its existence found that the 'symptoms are more easily tolerated if
- you can put a name to them, even if that produces only an illusory
- understanding of their significance.' He commented on the potential
- hazards of photic sneezing if it occurs while one is driving a car on a
- sunny day. He said that he found that 'sudden exposure to sunlight when
- emerging from a road tunnel of sufficient length is sure to induce a
- sneeze.' Furthermore, 'driving through sunlit gaps in otherwise dense
- forest or past blocks of buildings can bring on a sneeze.'
-
- Duncan (1995) pointed out public awareness of the ACHOO syndrome is much
- more widespread than one might guess, to the point that it has entered
- into the popular wisdom conveyed to preschoolers. In a best-selling
- children's book by Berenstain and Berenstain (1981), Papa and Mama bear
- are taking sister bear and brother bear to their pediatrician, Dr.
- Grizzly, for a check-up. The cubs are expressing their apprehension
- about the possibility of injections when Papa bear suddenly cuts loose
- with an explosive sneeze. 'Bless you!' said Mama.' 'It's just this
- bright sunlight,' sniffed Papa. 'I never get sick.'
-
- *FIELD* RF
- 1. Beckman, L.; Nordenson, I.: Individual differences with respect
- to the sneezing reflex: an inherited physiological trait in man?.
- Hum. Hered. 33: 390-391, 1983.
-
- 2. Benbow, E. W.: Practical hazards of photic sneezing. (Letter) Brit.
- J. Ophthal. 75: 447 only, 1991.
-
- 3. Berenstain, S.; Berenstain, J.: The Berenstain Bears Go to the
- Doctor. New York: Random House (pub.) 1981.
-
- 4. Collie, W. R.; Pagon, R. A.; Hall, J. G.; Shokeir, M. H. K.: ACHOO
- syndrome (helio-ophthalmic outburst syndrome). Birth Defects Orig.
- Art. Ser. XIV(6B): 361-363, 1978.
-
- 5. Duncan, R.: Personal Communication. Los Angeles, Calif. 2/1/1995.
-
- 6. Everett, H. C.: Sneezing in response to light. Neurology 14:
- 483-490, 1964.
-
- 7. Forrester, J. M.: Sneezing on exposure to bright light as an inherited
- response. Hum. Hered. 35: 113-114, 1985.
-
- 8. Hunter, K. M.: An N of one: syndrome letters in the New England
- Journal of Medicine. Perspect. Biol. Med. 33: 237-251, 1990.
-
- 9. Katz, B.; Melles, R. B.; Swenson, M. R.; Schneider, J. A.: Photic
- sneeze reflex in nephropathic cystinosis. Brit. J. Ophthal. 74:
- 706-708, 1990.
-
- 10. Lang, D. M.; Howland, W. C., III: Solar sneeze reflex. (Letter) J.A.M.A. 257:
- 1330-1331, 1987.
-
- 11. Lerner, D. L.: Letter to the editor. Perspect. Biol. Med. 34:
- 469-470, 1991.
-
- 12. Lewkonia, I.: An infrequent response to slit lamp examination.
- Brit. J. Ophthal. 53: 493-495, 1969.
-
- 13. Morris, H. H., III: ACHOO syndrome: prevalence and inheritance.
- Cleveland Clin. J. Med. 54: 431-433, 1987.
-
- 14. Peroutka, S. J.; Peroutka, L. A.: Autosomal dominant transmission
- of the 'photic sneeze reflex.'. (Letter) New Eng. J. Med. 310:
- 599-600, 1984.
-
- *FIELD* CS
-
- Neuro:
- Paroxysmal sneezing;
- Light-induced sneezing
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 2/20/1995
- davew: 7/19/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 9/18/1991
- carol: 7/22/1991
-
- *RECORD*
- *FIELD* NO
- 100850
- *FIELD* TI
- *100850 ACONITASE, MITOCHONDRIAL; ACO2
- *FIELD* TX
- Slaughter et al. (1975) reported that an electrophoretic survey had
- demonstrated 2 alleles at this locus. From the findings in
- heterozygotes, they concluded that both aconitases are monomeric.
- Sparkes et al. (1978) assigned this locus to chromosome 22 by study of
- Chinese hamster-human hybrid cells. See also Meera Khan et al. (1978)
- and Slaughter et al. (1978). From study of human-rodent hybrid clones,
- Geurts van Kessel et al. (1980) concluded that ACO2 is located between
- 22q11 and 22q13.
-
- *FIELD* SA
- Slaughter et al. (1977); Sparkes et al. (1978)
- *FIELD* RF
- 1. Geurts van Kessel, A. H. M.; Westerveld, A.; de Groot, P. G.; Meera
- Khan, P.; Hagemeijer, A.: Regional localization of the genes coding
- for human ACO2, ARSA, and NAGA on chromosome 22. Cytogenet. Cell
- Genet. 28: 169-172, 1980.
-
- 2. Meera Khan, P.; Wijnen, L. M. M.; Pearson, P. L.: Assignment of
- the mitochondrial aconitase gene (ACON-M) to human chromosome 22.
- Cytogenet. Cell Genet. 22: 212-214, 1978.
-
- 3. Slaughter, C. A.; Hopkinson, D. A.; Harris, H.: Aconitase polymorphism
- in man. Ann. Hum. Genet. 39: 193-202, 1975.
-
- 4. Slaughter, C. A.; Hopkinson, D. A.; Harris, H.: The distribution
- and properties of aconitase isozymes in man. Ann. Hum. Genet. 40:
- 385-401, 1977.
-
- 5. Slaughter, C. A.; Povey, S.; Carritt, B.; Solomon, E.; Bobrow,
- M.: Assignment of the locus ACON-M to chromosome 22. Cytogenet.
- Cell Genet. 22: 223-225, 1978.
-
- 6. Sparkes, R. S.; Mohandas, T.; Sparkes, M. C.; Shulkin, J. D.:
- Assignment of the aconitase (EC 4.2.1.3) mitochondrial locus (ACON-M)
- to human chromosome 22. Biochem. Genet. 16: 751-756, 1978.
-
- 7. Sparkes, R. S.; Mohandas, T.; Sparkes, M. C.; Shulkin, J. D.:
- Aconitase (E. C. 4.2.1.3) mitochondrial locus (ACON-M) mapped to human
- chromosome 22. Cytogenet. Cell Genet. 22: 226-227, 1978.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
-
- *RECORD*
- *FIELD* NO
- 100880
- *FIELD* TI
- *100880 ACONITASE, SOLUBLE; ACO1
- *FIELD* TX
- Slaughter et al. (1975) reported that an electrophoretic survey had
- demonstrated 7 alleles at this locus. Among the populations studied,
- Nigerians showed polymorphism for ACON-S. Aconitase catalyzes the
- conversion of cis-aconitate to isocitrate. In studies of man-Chinese
- hamster somatic cell hybrids, Westerveld et al. (1975) showed that human
- gal-1-p uridyl transferase (GALT; 230400) and aconitase are syntenic.
- Povey et al. (1976) assigned ACO1 to chromosome 9. ACO1 and GALT are on
- 9p in man and on chromosome 4 in the mouse (Nadeau and Eicher, 1982).
- The location in the mouse was predicted from the human linkage. The
- smallest region of overlap (SRO) for ACO1 was estimated to be 9p22-p13
- (Robson and Meera Khan, 1982).
-
- Aconitase-1 and aconitase-2 (ACO2; 100850) are isozymes present in the
- cytosol and mitochondria, respectively. Other pairs of cytosolic and
- mitochondrial isozymes are ALDH1 (100640) and ALDH2 (100650), GOT1
- (138180) and GOT2 (138150), IDH1 (147700) and IDH2 (147650), MDH1
- (154200) and MDH2 (154100), SOD1 (147450) and SOD2 (147460), and TK1
- (188300) and TK2 (188250). In all these cases, the 2 isozymes of
- different subcellular localization, although similar in structure and
- function, are encoded by genes on different chromosomes, i.e., are
- nonsyntenic. The presumption is that in each case both originated from a
- common ancestral gene in a primordial genome, but that whereas the
- cytosolic isozyme is encoded by a gene that is a direct descendant from
- a nuclear progenitor gene, the mitochondrial isozyme, although now
- encoded by a nuclear gene, is descended from a gene in the
- bacterium-like progenitor of the mitochondrion. When this primitive
- organism took up intracellular existence, most of its genes were
- transferred to the nuclear genome and since they inserted more or less
- at random into the nuclear genome, it was to be expected that the
- cytosolic and mitochondrial forms of the enzyme would end up being
- encoded by genes on different chromosomes. That mitochondrial DNA can be
- inserted into the nuclear genome is indicated by work such as that of
- Shay and Werbin (1992) who characterized in detail 2 instances of
- mitochondrial DNA fragments that had been inserted into the nucleus of
- HeLa cells. In one of these cases, the mitochondrial sequence encoding
- cytochrome c oxidase subunit III was contiguous with and 5-prime of
- exons 2 and 3 of the MYC oncogene (190080) and the chimeric gene was
- transcribed. Shay and Werbin (1992) discussed possible mechanisms for
- the transfer of mitochondrial DNA into the nucleus.
-
- Data on gene frequencies of allelic variants were tabulated by
- Roychoudhury and Nei (1988).
-
- *FIELD* SA
- Azevedo et al. (1979); Mohandas et al. (1979); Robson et al. (1977);
- Shows and Brown (1977); Teng et al. (1978)
- *FIELD* RF
- 1. Azevedo, E. S.; Da Silva, M. C. B. O.; Lima, A. M. V.; Fonseca,
- E. F.; Conseicao, M. M.: Human aconitase polymorphism in three samples
- from northeastern Brazil. Ann. Hum. Genet. 43: 7-10, 1979.
-
- 2. Mohandas, T.; Sparkes, R. S.; Sparkes, M. C.; Shulkin, J. D.; Toomey,
- K. E.; Funderburk, S. J.: Regional localization of human gene loci
- on chromosome 9: studies of somatic cell hybrids containing human
- translocations. Am. J. Hum. Genet. 31: 586-600, 1979.
-
- 3. Nadeau, J. H.; Eicher, E. M.: Conserved linkage of soluble aconitase
- and galactose-1-phosphate uridyl transferase in mouse and man: assignment
- of these genes to mouse chromosome 4. Cytogenet. Cell Genet. 34:
- 271-281, 1982.
-
- 4. Povey, S.; Slaughter, C. A.; Wilson, D. E.; Gormley, I. P.; Buckton,
- K. E.; Perry, P.; Bobrow, M.: Evidence for the assignment of the
- loci AK 1, AK 3 and ACON to chromosome 9 in man. Ann. Hum. Genet. 39:
- 413-422, 1976.
-
- 5. Robson, E. B.; Cook, P. J. L.; Buckton, K. E.: Family studies
- with the chromosome 9 markers ABO, AK-1, ACON-S and 9qh. Ann. Hum.
- Genet. 41: 53-60, 1977.
-
- 6. Robson, E. B.; Meera Khan, P.: Report of the committee on the
- genetic constitution of chromosomes 7, 8, and 9. Cytogenet. Cell
- Genet. 32: 144-152, 1982.
-
- 7. Roychoudhury, A. K.; Nei, M.: Human Polymorphic Genes: World Distribution.
- New York: Oxford Univ. Press (pub.) 1988.
-
- 8. Shay, J. W.; Werbin, H.: New evidence for the insertion of mitochondrial
- DNA into the human genome: significance for cancer and aging. Mutat.
- Res. 275: 227-235, 1992.
-
- 9. Shows, T. B.; Brown, J. A.: Mapping AK-1, ACON-S, and AK-3 to
- chromosome 9 in man employing an X-9 translocation and somatic cell
- hybrids. Cytogenet. Cell Genet. 19: 26-37, 1977.
-
- 10. Slaughter, C. A.; Hopkinson, D. A.; Harris, H.: Aconitase polymorphism
- in man. Ann. Hum. Genet. 39: 193-202, 1975.
-
- 11. Teng, Y. S.; Tan, S. G.; Lopez, C. G.: Red cell glyoxalase I
- and placental soluble aconitase polymorphisms in the three major ethnic
- groups of Malaysia. Jpn. J. Hum. Genet. 23: 211-215, 1978.
-
- 12. Westerveld, A.; van Henegouwen, B. H. M. A.; Van Someren, H.:
- Evidence for synteny between the human loci for galactose-1-phosphate
- uridyl transferase and aconitase in man-Chinese hamster somatic cell
- hybrids. Cytogenet. Cell Genet. 14: 453-454, 1975.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 2/11/1994
- carol: 2/17/1993
- carol: 2/2/1993
- carol: 8/25/1992
- supermim: 3/16/1992
- carol: 12/6/1990
-
- *RECORD*
- *FIELD* NO
- 100900
- *FIELD* TI
- *100900 ACONITATE HYDRATASE, SOLUBLE
- *FIELD* TX
- Aconitate hydratase (citrate, or isocitrate, hydrolyase, EC 4.2.1.3)
- exists in structurally distinct soluble and mitochondrial forms. Schmitt
- and Ritter (1974) found electrophoretic variants of the soluble form in
- human placenta. No mitochondrial variants were found.
-
- *FIELD* RF
- 1. Schmitt, J.; Ritter, H.: Genetic variation of aconitate hydratase
- in man. Humangenetik 22: 263-264, 1974.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 101000
- *FIELD* TI
- *101000 NEUROFIBROMATOSIS, TYPE II
- NEUROFIBROMATOSIS, CENTRAL TYPE;;
- ACOUSTIC SCHWANNOMAS, BILATERAL;;
- BILATERAL ACOUSTIC NEUROFIBROMATOSIS; BANF
- NEUROFIBROMIN 2; NF2, INCLUDED;;
- ACOUSTIC NEURINOMA, BILATERAL; ACN, INCLUDED;;
- MERLIN, INCLUDED;;
- SCHWANNOMIN; SCH, INCLUDED
- *FIELD* MN
-
- The central form of neurofibromatosis is characterized by tumors of the
- eighth cranial nerve (usually bilateral), meningiomas of the brain, and
- schwannomas of the dorsal roots of the spinal cord. There is a high
- frequency of presenile posterior subcapsular, capsular, or peripheral
- cortical cataracts which sometimes require surgery and may predate the
- symptoms of bilateral acoustic neurofibromatosis (Bouzas et al., 1993).
- Other causes of decreased vision were damage in the optic pathways,
- macular hamartomas, and corneal opacities. Most patients with the
- central form have no cafe-au-lait spots or peripheral neurofibromata.
- Acoustic neuroma is almost always unilateral (Nager, 1969). Bilateral
- tumours, in addition to their autosomal dominant inheritance and
- association with neurofibromatosis, differ from unilateral ones in that
- they can reach a remarkably large size with extensive involvement of the
- temporal bone and the nerves therein. According to an NIH Consensus
- Development Conference (1988) the criteria for NF2 are (1) bilateral
- eighth nerve masses seen with appropriate imaging techniques (e.g., CT
- or MRI); or (2) a first-degree relative with NF2 and either unilateral
- eighth nerve mass, or two of the following: neurofibroma, meningioma,
- glioma, schwannoma, or juvenile posterior subcapsular lenticular
- opacity. Small (less than 8 mm) acoustic neuromas can be detected in
- asymptomatic individuals by the use of gadolinium-enhanced MRI (Pastores
- et al.,1991).
-
- The natural history of the condition was described by Evans et al.
- (1992). The mean age at onset was 21.6 years and no patient presented
- after 55 years of age. Patients presented with symptoms attributable to
- vestibular schwannomas (acoustic neuromas), cranial meningiomas, and
- spinal tumors. Forty-four percent presented with deafness, unilateral in
- 35%. Muscle weakness or wasting was the first symptom in 12%. A
- generalized and isolated neuropathy appears to be a relatively common
- feature. Cafe-au-lait spots occurred in 43% of the patients but only 1
- of 150 had as many as 6 spots. Cataract was detected in 39%. Of three
- types of skin tumors, the least common (20% of patients) was similar to
- the intradermal papillary skin neurofibroma with violaceous coloring
- occurring in NF1. The second type (33%) comprised subcutaneous
- well-circumscribed, often spherical, tumors that appeared to be located
- on peripheral nerves. The most frequent type (47%) were discrete
- well-circumscribed, slightly raised, roughened areas of skin often
- pigmented and accompanied by excess hair. Cases of NF2 can be divided
- into the Wishart type, with early onset, rapid course, and multiple
- other tumors in addition to bilateral vestibular schwannomas, and the
- Gardner type with late onset, more benign course, and usually only
- bilateral vestibular schwannomas. Birth incidence of NF2 was estimated
- to be 1 in 33,000-40,562. Half of the cases were new mutations. There
- was a maternal effect on severity (age of onset) and a preponderance of
- maternally inherited cases.
-
- Loss of heterozygosity of alleles from chromosome 22 has been found in
- acoustic neuromas, neurofibromas, and meningiomas from patients with
- bilateral acoustic neurofibromatosis (Wolff et al.,1992). Rouleau et al.
- (1993) found a gene, designated Schwannomin (symbol=SCH), bearing
- homology to erythrocyte protein 4.1 and the ezrin/moesin/talin family of
- genes, and showed that this gene is the site of the mutations causing
- NF2 by demonstrating germline and somatic SCH mutations in NF2 patients
- and in NF2-related tumors. Most of the variants were nonsense,
- frameshift, or splice mutations predicted to lead to the synthesis of a
- truncated SCH protein.
-
- Loss of heterozygosity for polymorphic DNA markers flanking NF2 on
- chromosome 22 was found in 60% of 170 primary sporadic meningiomas
- (Ruttledge et al.,1994), and of 30 vestibular schwannomas (Sainz et
- al.,1994). It appears that loss of NF2 protein function is a necessary
- step in schwannoma pathogenesis and that the NF2 gene functions as a
- recessive tumor suppressor gene.Using polymorphic DNA markers it is
- possible to determine, with a high degree of certainty, the carrier
- status of about 85% of persons at risk (Ruttledge et al., 1993).
-
- *FIELD* TX
- The central form of neurofibromatosis, characterized by tumors of the
- eighth cranial nerve (usually bilateral), meningiomas of the brain and
- schwannomas of the dorsal roots of the spinal cord, has few of the
- hallmarks of the peripheral form of neurofibromatosis (162200). Most
- patients with the central form have no cafe-au-lait spots or peripheral
- neurofibromata and no patients in one large series had 6 or more
- cafe-au-lait spots (Eldridge, 1981). The term von Recklinghausen disease
- should be reserved for the peripheral form of neurofibromatosis. Gardner
- and Frazier (1933) reported a family of 5 generations in which 38
- members were deaf because of acoustic neuromas; of these, 15 later
- became blind. The average age of onset of deafness was 20 years. The
- average age at death of affected persons in the second generation was
- 72, in the third generation 63, in the fourth 42, and in the fifth 28.
- Follow-up of this family (Gardner and Turner, 1940; Young et al., 1970)
- revealed no evidence of the systemic manifestations of von
- Recklinghausen disease. Other families with no evidence of the latter
- disease were reported by Worster-Drought et al. (1937), Feiling and Ward
- (1920), and Moyes (1968). Worster-Drought et al. (1937) pointed out that
- Wishart reported the first case of bilateral acoustic neuroma in 1822.
- Wishart's patient, Michael Blair, was 21 years old when he consulted Mr.
- Wishart, president of the Royal College of Surgeons of Edinburgh,
- because of bilateral deafness. He had a peculiarly shaped head from
- infancy, and blindness in the right eye was discovered at about 4 months
- after birth. He became completely blind and deaf toward the end of his
- life. Autopsy revealed tumors of the dura mater and brain and also a
- 'tumour of the size of a small nut, and very hard, being attached to
- each of them (auditory nerves), just where they enter the meatus
- auditorius internus.'
-
- Nager (1969) showed that in about 4% of cases acoustic neuroma is
- bilateral. In addition to their autosomal dominant inheritance and
- association with neurofibromatosis, bilateral tumors differ from
- unilateral ones in that they can reach a remarkably large size with
- extensive involvement of the temporal bone and the nerves therein. More
- than 30 kindreds with 'central neurofibromatosis' have been reported
- (Fabricant et al., 1979). Kanter et al. (1980), who reviewed 9
- personally studied kindreds and 15 reported ones, with a total of 130
- cases, showed an increase only in antigenic activity of nerve growth
- factor (NGF) in central neurofibromatosis and only in functional
- activity in peripheral neurofibromatosis. Thus, these disorders may
- involve different defects in NGF synthesis and/or regulation. In a
- review of NF2, Martuza and Eldridge (1988) defined criteria for the
- diagnosis of both NF1 and NF2. An NIH Consensus Development Conference
- (1988) concluded that the criteria for NF2 are met if a person is found
- to have '(1) bilateral eighth nerve masses seen with appropriate imaging
- techniques (e.g., CT or MRI); or (2) a first-degree relative with NF2
- and either unilateral eighth nerve mass, or two of the following:
- neurofibroma, meningioma, glioma, schwannoma, or juvenile posterior
- subcapsular lenticular opacity.' Pearson-Webb et al. (1986) pointed out
- that Lisch nodules, which are iris hamartomas, are not found in NF2.
- They found, however, an apparently high frequency of presenile posterior
- subcapsular and nuclear cataracts which sometimes required surgery
- and/or predated the symptoms of bilateral acoustic neurofibromatosis.
- Kaiser-Kupfer et al. (1989) found posterior capsular lens opacities in
- 20 NF2 patients in 11 families. Parry et al. (1991) extended these
- observations. In 26 persons who were first-degree relatives of an
- affected individual, they found posterior capsular cataracts in 21. Of
- 14 at-risk individuals, i.e., persons with mild changes of NF but not
- NF1, persons under age 40 with unilateral acoustic neuroma, a child with
- meningioma and/or schwannoma, and a person with multiple meningioma,
- they found posterior capsular lens opacities in 13. These patients
- probably represented new mutations. The presence of posterior capsular
- opacities in a relative of persons with NF2 was suggestive of NF2.
- Furthermore, NF2 should be considered in young persons without NF1 but
- with mild skin findings of NF or CNS tumors with posterior capsular
- opacities. Bouzas et al. (1993) found posterior subcapsular/capsular
- cataracts in 36 (80%) of 45 affected individuals in 29 families. In
- addition, the association of peripheral cortical lens opacities with NF2
- was found to be statistically significant: such cataracts were found in
- 17 of the patients (37.8%) but in none of the unaffected family members
- (p less than 0.0001). In 3 patients, peripheral cortical opacities were
- present despite the absence of posterior subcapsular/capsular cataracts.
- Bouzas et al. (1993), reporting further on the NIH experience, reviewed
- visual impairment in 54 NF2 patients, 51 of whom had bilateral
- vestibular schwannomas. Causes of decreased vision were cataracts,
- damage in the optic pathways, macular hamartomas, and corneal opacities.
- Although lens opacities are an important marker for NF2, they usually do
- not interfere with vision; some progress, requiring cataract extraction.
- In 6 patients, decreased visual acuity was due to corneal opacifications
- secondary to either seventh or fifth cranial nerve damage, or both.
- Damage to the seventh cranial nerve caused lagophthalmos and decreased
- lacrimal secretion; damage to the fifth cranial nerve caused corneal
- hypesthesia. The nerves were damaged by the growth of vestibular tumors
- in 1 patient, but in most patients they were damaged during
- neurosurgical procedures.
-
- Pastores et al. (1991) demonstrated that small (less than 8 mm) acoustic
- neuromas can be detected in asymptomatic individuals by the use of
- gadolinium-enhanced MRI. They demonstrated such neuromas in 2
- asymptomatic children, aged 7 and 11 years, one of whom had normal
- audiometric and brainstem-evoked response testing. Landau et al. (1990)
- described combined pigment epithelial and retinal hamartoma (CEPRH) in
- NF2. In a series reported by Mrazek et al. (1988), 1 of 41 acoustic
- neurinoma cases was bilateral. This was in a 10-year-old girl with von
- Recklinghausen neurofibromatosis, whose first tumor had been diagnosed
- at age 6. Mayfrank et al. (1990) studied 10 patients with NF2 and found
- that all were sporadic cases, each presumably the result of a new
- mutational event. From a survey of these patients and those in the
- literature, they concluded that sporadic cases are characterized by a
- high incidence of multiple meningiomas and spinal tumors in addition to
- bilateral acoustic neurinomas. Pulst et al. (1991) described a family
- with spinal neurofibromatosis without cafe-au-lait spots or other
- manifestations of either NF1 or NF2 such as cutaneous tumors, Lisch
- nodules, or acoustic tumors. Mutation at the NF1 locus was excluded with
- odds greater than 100,000:1. Markers with the NF2 locus were
- uninformative in this family.
-
- Evans et al. (1992) studied 150 patients. The mean age at onset was
- 21.57 years (n = 110) and no patient presented after 55 years of age.
- Patients presented with symptoms attributable to vestibular schwannomas
- (acoustic neuroma), cranial meningiomas, and spinal tumors. In 100
- patients studied personally by the authors, 44 presented with deafness,
- which was unilateral in 35. Deafness was accompanied by tinnitus in 10.
- Muscle weakness or wasting was the first symptom in 12%. In 3 of the 100
- patients, there was a distal symmetrical sensorimotor neuropathy,
- confirmed by nerve conduction studies and electromyography. Although
- similar features may result from the multiple spinal and intracranial
- tumors that occur in this condition, a generalized and isolated
- neuropathy appears to be a relatively common feature of NF2.
- Cafe-au-lait spots occurred in 43 of the 100 patients but only 1 had as
- many as 6 spots. Cataract was detected in 34 of 90 patients. Cataracts
- were probably congenital in 4 patients in this study. Three types of
- skin tumors were recognized. The first and least common was similar to
- the intradermal papillary skin neurofibroma with violaceous coloring
- occurring in NF1. The second type comprised subcutaneous
- well-circumscribed, often spherical, tumors that appeared to be located
- on peripheral nerves; the thickened nerve could often be palpated at
- either end of the tumor, the skin being mobile and separate from the
- tumor. The third and most frequent type, first described by Martuza and
- Eldridge (1988), was represented by discrete well-circumscribed,
- slightly raised, roughened areas of skin often pigmented and accompanied
- by excess hair. Skin tumors of some kind were found in 68% of patients,
- type 1 being present in 20%, type 2 in 33%, and type 3 in 47%. Evans et
- al. (1992) divided their 120 cases of NF2 into 2 types: the Wishart
- (1822) type, with early onset, rapid course, and multiple other tumors
- in addition to bilateral vestibular schwannomas, and the Gardner type
- (1930, 1933, 1940), with late onset, more benign course, and usually
- only bilateral vestibular schwannomas. This classification had been
- suggested by Eldridge et al. (1991). Evans et al. (1992) found no
- evidence for the existence of a third type of generalized
- meningiomatosis that might be designated the Lee-Abbott type (Lee and
- Abbott, 1969). They could find no evidence that either pregnancy or
- contraceptive pill has adverse effects on vestibular schwannomas or
- other manifestations. Evans et al. (1992) provided useful advice on the
- follow-up of persons identified as having NF2 and the management of
- persons at risk of developing NF2. The age at onset of deafness and the
- age at diagnosis were almost identical in the 2 sexes. Birth incidence
- of NF2 was estimated to be 1 in 33,000-40,562. Evans et al. (1992)
- considered 49% of the 150 cases to represent new mutations. The mutation
- rate was estimated to be 6.5 x 10(-6). A maternal effect on severity was
- noted in that age of onset was 18.17 years in 36 maternally inherited
- cases and 24.5 years in 20 paternally inherited cases (p = 0.027). A
- preponderance of maternally inherited cases was also significant (p =
- 0.03). (A maternal effect on severity had been noted also for
- neurofibromatosis, type I (NF1; 162200).)
-
- Parry et al. (1994) assessed possible heterogeneity in NF2 by evaluating
- 63 affected members of 32 families. In addition to skin and neurologic
- examinations, workup included audiometry, complete ophthalmologic
- examination with slit-lamp biomicroscopy of the lens and fundus, and
- gadolinium-enhanced MRI of the brain and, in some, of the spine. Mean
- age-at-onset in 58 individuals was 20.3 years; initial symptoms were
- related to vestibular schwannomas (44.4%), other CNS tumors (22.2%),
- skin tumors (12.7%), and ocular manifestations including cataracts and
- retinal hamartomas (12.7%). Screening uncovered 5 affected but
- asymptomatic family members; vestibular schwannomas were demonstrated in
- 62 (98.4%). Other findings included cataracts (81.0%), skin tumors
- (67.7%), spinal tumors (67.4%), and meningiomas (49.2%). As a rule,
- clinical manifestations and clinical course were similar within families
- but differed among families. Parry et al. (1994) concluded that 2
- subtypes but not 3 can be defined.
-
- Ragge et al. (1995) concluded that the most common ocular abnormalities
- in NF2 are posterior subcapsular or capsular, cortical, or mixed lens
- opacities, found in 33 of 49 patients (67%), and retinal hamartomas
- found in 11 of 49 patients (22%). The types of cataract that were most
- suggestive of NF2 were plaque-like posterior subcapsular or capsular
- cataract and cortical cataract with onset under the age of 30 years.
-
- Seizinger et al. (1986) found loss of genes on chromosome 22 in acoustic
- neuromas; i.e., whereas normal tissue was heterozygous, tumor tissue was
- hemizygous (or homozygous) for the polymorphic markers SIS (190040),
- IGLC (147220), and the anonymous DNA locus D22S1. They were prompted to
- undertake the study by analogy to retinoblastoma and Wilms tumor and by
- the facts that meningioma occurs in association with familial acoustic
- neuroma and that cytologic change in chromosome 22 is frequent in
- meningioma (see 156100). Seizinger et al. (1987) found specific loss of
- alleles from chromosome 22 in 2 acoustic neuromas, 2 neurofibromas, and
- 1 meningioma from patients with bilateral acoustic neurofibromatosis. In
- each case, a partial deletion occurred with a breakpoint distal to the
- D22S9 locus in band 22q11. Wertelecki et al. (1988) confirmed
- localization of the gene on chromosome 22 (22q11.21-q13.1) by
- demonstration of linkage in family studies to markers on chromosome 22.
- Wertelecki et al. (1988) also presented the clinical data on 15 affected
- male and 8 affected female members of the 1 large kindred they studied
- for linkage data. Rouleau et al. (1990) identified markers bracketing
- the NF2 gene which are therefore useful for accurate presymptomatic and
- prenatal diagnosis, as well as for isolating the defective gene. Narod
- et al. (1992) concluded that there is no evidence of genetic
- heterogeneity in NF2. They indicated that the presence of bilateral
- vestibular schwannomas, as they termed the acoustic neuromas, is
- sufficient for the diagnosis. Using 8 polymorphic loci on chromosome 22
- to study tumor and constitutional DNAs isolated from 39 unrelated
- patients with sporadic or NF2-associated acoustic neuromas, meningiomas,
- schwannomas, and ependymomas, Wolff et al. (1992) found 2 tumors with
- loss of heterozygosity (LOH) patterns consistent with the presence of
- chromosome 22 terminal deletions. By use of additional polymorphic
- markers, the terminal deletion breakpoint in one of the tumors, an
- acoustic neuroma from an NF2 patient, was mapped within the previously
- defined NF2 region. In addition, they identified a sporadic acoustic
- neuroma with an LOH pattern consistent with mitotic recombination or
- deletion and reduplication. The findings lent further support to the
- recessive tumor-suppressor model for the NF2 gene. Arai et al. (1992)
- described a patient with bilateral acoustic neurinomas and other tumors
- in the central nervous system and a constitutional translocation
- t(4;22)(q12;q12.2). Thus, 22q12.2 is a refined localization for the NF2
- gene. The same karyotype that was seen in cultured peripheral
- lymphocytes was found in a paraspinal neurinoma. The patient's father
- was also a carrier of the translocation but he had no clinical symptoms
- of NF2, nor did other relatives. As explanation for the failure of
- expression in the father, Arai et al. (1992) suggested various
- possibilities including nonpenetrance, mosaicism, or genetic imprinting.
- They quoted Kanter et al. (1980) as demonstrating earlier onset of
- symptoms when NF2 is transmitted by the mother. In a family with the
- mild or so-called Gardner type of neurofibromatosis type 2, Watson et
- al. (1993) defined a submicroscopic deletion which involved the
- neurofilament heavy chain locus (NEFH; 162230) but did not extend as far
- as the Ewing sarcoma region (EWS; 133450) proximally or the leukemia
- inhibitory factor locus (LIF; 159540) distally. They estimated that the
- deletion was about 700 kb long.
-
- Claudio et al. (1994) demonstrated that the mouse homolog of the NF2
- gene is located in the proximal region of chromosome 11. The
- localization was achieved by analysis of allele distribution in
- recombinant inbred strains using a simple sequence repeat polymorphism
- in the 3-prime untranslated region of the mouse NF2 cDNA. The region of
- chromosome 11 also contains genes for leukemia inhibitory factor (LIF;
- 159540) and neurofilament heavy chain polypeptide (NFH; 162230), both of
- which map to the same region of human chromosome 22 as does NF2.
-
- Trofatter et al. (1993) identified a candidate gene for the NF2 tumor
- suppressor that had suffered nonoverlapping deletions in DNA from 2
- independent NF2 families as well as alterations in the meningiomas from
- 2 unrelated NF2 patients. The candidate gene encoded a 587-amino acid
- protein with striking similarity to several members of a family of
- proteins proposed to link cytoskeletal components with proteins in the
- cell membrane; these included moesin (309845), ezrin (123900), and
- radixin (179410). Because of the resemblance to these 3 proteins (45-47%
- identity), Trofatter et al. (1993) called the NF2 gene product merlin.
- The NF2 gene may represent a novel class of tumor suppressor genes.
- Schwannomin (symbol = SCH) was the designation used by Rouleau et al.
- (1993), who likewise isolated a gene bearing homology to erythrocyte
- protein 4.1 and the ezrin/moesin/talin family of genes. They provided
- incontrovertible evidence that this gene is the site of the mutations
- causing NF2 by demonstrating germline and somatic SCH mutations in NF2
- patients and in NF2-related tumors. To isolate the gene, they cloned the
- region between 2 flanking polymorphic markers in which they found
- several genes, only one of which carried mutations in NF2. Rouleau et
- al. (1993) found 16 mutations, 15 of which were predicted to result in
- truncated proteins. Consistent with the classic Knudson theory of tumor
- suppressor genes, loss of the wildtype allele at the NF2 locus was
- demonstrated in 6 of 8 tumors containing NF2 mutations (Trofatter et
- al., 1993; Rouleau et al., 1993). For example, in a meningioma in a
- patient without features of NF2, they found deletion of 2 nucleotides,
- TC, from codon 61 resulting in a frameshift; the normal allele on the
- other chromosome had been lost. In 2 instances of schwannoma occurring
- in patients without evidence of NF2, Rouleau et al. (1993) found
- nonsense mutations that were absent in the patient's blood DNA; in these
- instances also the normal allele had been lost.
-
- Using polymorphic DNA markers in a study of 13 NF2 kindreds, Ruttledge
- et al. (1993) concluded that it is possible to determine, with a high
- degree of certainty, the carrier status of about 85% of persons at risk.
- Risk prediction was possible in every case in which DNA was available
- from both parents. In 76% of informative individuals, it was possible to
- assign a decreased risk of being carriers. Thus, the use of probes for
- construction of chromosome 22 haplotypes for risk assessment should
- result in a greatly reduced number of individuals who will require
- periodic screening.
-
- Bianchi et al. (1994) described a novel isoform of the NF2 transcript
- that shows differential tissue expression and encodes a modified C
- terminus of the predicted protein. Mutations affecting both isoforms of
- the NF2 transcript were detected in multiple tumor types including
- melanoma and breast carcinoma. These findings provided evidence that
- alterations in the NF2 transcript occurred not only in the hereditary
- brain neoplasms typically associated with NF, but also as somatic
- mutations in their sporadic counterparts.
-
- By November 1993, 24 mutations, including both germline and somatic
- mutations, had been detected in schwannomin (Thomas, 1993). Most of the
- mutations cause the synthesis of a truncated schwannomin protein. After
- examining 8 of the 16 known NF2 exons in 151 meningiomas, Ruttledge et
- al. (1994) characterized 24 inactivating mutations. Significantly, these
- aberrations were exclusively detected in tumors that lost the other
- chromosome 22 allele. These results provided strong evidence that the
- suppressor gene on chromosome 22, frequently inactivated in meningioma,
- is the NF2 gene. The same group had found loss of heterozygosity (LOH)
- for polymorphic DNA markers flanking NF2 on chromosome 22 in 102 (60%)
- of 170 primary sporadic meningiomas. Thus, another gene may be involved
- in the development of 40% of meningiomas. It is probably noteworthy that
- all 24 of the inactivating mutations found by Ruttledge et al. (1994) in
- sporadic meningiomas were nonsense, frameshift (due to small deletions),
- or splice site mutations; there were no missense mutations.
- Wellenreuther et al. (1995) likewise concluded that NF2 represents the
- meningioma locus on chromosome 22. There was a significant association
- of loss of heterozygosity on chromosome 22 with mutations in the NF2
- gene. They analyzed the entire coding region of the NF2 gene in 70
- sporadic meningiomas and identified 43 mutations in 41 patients. These
- resulted predominantly in immediate truncation, splicing abnormalities,
- or an altered reading frame of the predicted protein product. All
- mutations occurred in the first 13 exons, the region of homology with
- the filopodial proteins moesin, ezrin, and radixin.
-
- Parry et al. (1996) used SSCP analysis to screen for mutations in DNA
- from 32 unrelated NF2 patients. Mutations were identified in 66% of
- patients and 20 different mutations were found in 21 patients. They
- reported that their results confirm the association between nonsense and
- frameshift mutations and clinical manifestations compatible with severe
- disease. Parry et al. (1996) stated that their data raise questions
- regarding the role of factors, other than the intrinsic properties of
- individual mutations, that might influence the phenotype.
-
- Sainz et al. (1994) performed mutational analysis in 30 vestibular
- schwannomas and found 18 mutations, 7 of which contained loss or
- mutation of both NF2 alleles. Most mutations predicted a truncated
- protein. Mutational hot spots were not identified. Only 1 of the
- mutations was in a tumor from a patient with NF2. Immunocytochemical
- studies using antibodies to the NF2 protein showed complete absence of
- staining in tumor Schwann cells, whereas staining was observed in normal
- vestibular nerve. These data indicated that loss of NF2 protein function
- is a necessary step in schwannoma pathogenesis and that the NF2 gene
- functions as a recessive tumor suppressor gene. In studies of 34
- vestibular schwannomas and 14 schwannomas at other locations, Bijlsma et
- al. (1994) found that the SCH gene is implicated in the development of
- these tumors in all locations of the nervous system. Using a screening
- method based on denaturing gradient gel electrophoresis, which allows
- the detection of mutations in 95% of the coding sequence, Merel et al.
- (1995) observed mutations in 17 of 57 meningiomas and in 30 of 89
- schwannomas. All of the meningiomas and half of the schwannomas with
- identified NF2 mutations demonstrated chromosome 22 allelic losses. No
- mutations were observed in 17 ependymomas, 70 gliomas, 23 primary
- melanomas, 24 pheochromocytomas, 15 neuroblastomas, 6 medulloblastomas,
- 15 colon cancers, and 15 breast cancers. This led Merel et al. (1995) to
- conclude that the involvement of the NF2 gene is restricted to
- schwannomas and meningiomas, where it is frequently inactivated by a
- 2-hit process.
-
- Neurilemmomatosis, first reported by Niimura (1973) as neurofibromatosis
- type 3, is characterized by multiple cutaneous neurilemmomas and spinal
- schwannomas, without acoustic tumors or other signs of NF1 or NF2. In
- neurilemmomas, the tumor consists of Schwann cells. Honda et al. (1995)
- analyzed the peripheral leukocytes and tissue from cutaneous
- neurilemmomas of 7 patients with neurilemmomatosis using DNA markers for
- different regions of chromosome 22. They detected allele losses in 3 of
- 7 tumors from 7 patients with a probe for the NF2 region and the
- germline mutations in 2 of 3 tumors from the same 3 patients. They
- concluded that neurilemmomatosis is a form of NF2. The mutations they
- described included a deletion from codon 334 to 579 (at least) and a G
- insertion at codon 42.
-
- Ruttledge et al. (1996) reported that when individuals harboring
- protein-truncating mutations are compared with patients having single
- codon alterations, a significant correlation (p less than 0.001) with
- clinical outcome is observed. They noted that 24 of 28 patients with
- mutations that cause premature truncation of the NF2 protein present
- with severe phenotypes. In contrast, all 16 cases from 3 families with
- mutations that affect only a single amino acid have mild NF2.
-
- Malignant mesotheliomas (MMs) are aggressive tumors that develop most
- frequently in the pleura of patients exposed to asbestos. In contrast to
- many other cancers, relatively few molecular alterations had been
- described in MMs. The most frequent numerical cytogenetic abnormality in
- MMs is loss of chromosome 22. This prompted Bianchi et al. (1995) to
- investigate the status of the NF2 gene in these tumors. In studies of
- cDNAs from 15 MM cell lines and genomic DNAs from 7 matched primary
- tumors, NF2 mutations predicting either interstitial inframe deletions
- or truncation of the NF2-encoded protein (merlin) were detected in 8
- cell lines (53%), 6 of which were confirmed in primary tumor DNAs. In 2
- samples that showed NF2 gene transcript alterations, no genomic DNA
- mutations were detected, suggesting that aberrant splicing may
- constitute an additional mechanism for merlin inactivation. Unlike
- previously described NF2-related tumors, MM derived from the mesoderm;
- malignancies of this origin had not previously been associated with
- frequent alterations of the NF2 gene. In a commentary in the same
- journal issue, Knudson (1995) wrote: 'We are left wondering why
- mesothelioma is not a feature of the hereditary disease NF2.'
-
- *FIELD* AV
- .0001
- NEUROFIBROMATOSIS, TYPE 2
- NF2, LEU360PRO
- After isolating a candidate gene for neurofibromatosis type 2 by cloning
- the region of chromosome 22 between 2 flanking markers, Rouleau et al.
- (1993) succeeded in demonstrating that the gene is indeed the site of
- germline mutations in NF2 patients and of somatic mutations in
- NF2-related tumors. The search was initiated by first determining the
- exons and intron-exon boundaries within the coding sequence of the gene
- they referred to as schwannomin (SCH). Specific exons were amplified by
- polymerase chain reaction (PCR) and the resulting products were analyzed
- using denaturing gradient gel electrophoresis as described by Myers et
- al. (1985). A total of 15 genetic variants were identified. With the
- exception of a leu360-to-pro mutation due to a T-to-C transition, all
- the variants were nonsense, frameshift, or splice mutations predicted to
- lead to the synthesis of a truncated SCH protein. Whenever it was
- possible to investigate several family members in 2 generations, the SCH
- mutations were found to segregate with the disease. In 3 instances, the
- DNA variants were present only in the patient's constitutional DNA and
- not in either of the unaffected parents, providing strong evidence for a
- causal relationship between the occurrence of a new mutation and the
- development of the disease.
-
- .0002
- NEUROFIBROMATOSIS, TYPE 2
- NF2, IVS2DS, G-T, +1
- In a patient with hereditary neurofibromatosis type 2, Rouleau et al.
- (1993) identified a change from AGgt to AGtt at the junction between
- codons 80 and 81 (presumably the splice donor site of intron 2).
-
- .0003
- MENINGIOMA, SPORADIC
- NF2, 1BP DEL, A993
- Among the 24 inactivating mutations in the NF2 gene found by Ruttledge
- et al. (1994) in sporadic meningiomas were 7 instances of deletion of 1
- bp. One of these was deletion of adenine at position 993 resulting in
- frameshift. An LOH pattern consistent with monosomy for chromosome 22,
- i.e., loss of the homologous NF2 locus, was found in this as well as in
- most of the other 23 tumors.
-
- .0004
- MENINGIOMA, SPORADIC
- NF2, ARG57TER
- Papi et al. (1995) analyzed 61 sporadic meningiomas for loss of
- heterozygosity of 22q and for mutations in the NF2 gene. LOH was
- detected in 36 of the 60 informative tumors. They used single-strand
- conformational polymorphism analysis to identify 9 mutations in 5 of the
- 8 exons of the NF2 gene studied. The 9 tumors with an altered NF2 gene
- also showed LOH for 22q markers, supporting the hypothesis that the NF2
- gene acts as a tumor suppressor. Papi et al. (1995) found no germline
- mutations in these cases. One of the fibroblastic meningiomas in a
- 62-year-old female had a C-to-T transition at codon 57 in exon 2,
- resulting in a premature stop codon.
-
- .0005
- NEUROFIBROMATOSIS, TYPE 2
- NF2, LEU535PRO
- Evans et al. (1995) reported a family with type 2 neurofibromatosis and
- late-onset tumors. Hearing loss developed late in life in 5 members of
- the family, 2 of whom were first shown to have NF2 in their 70s. Three
- other obligate gene carriers died undiagnosed at ages 64, 72, and 78
- years of age. Evans et al. (1995) demonstrated a missense mutation at
- the the C-terminal end of the NF2 protein; a T-to-C transition at
- nucleotide 1604 caused a leu535-to-pro amino acid substitution.
-
- .0006
- NEUROFIBROMATOSIS, TYPE 2
- NF2, GLN538PRO
- In a family with 4 affected members, Kluwe and Mautner (1996) found a
- gln538-to-pro mutation in exon 15 of the NF2 gene by studying lymphocyte
- DNA. They suggested that missense mutations such as this were rare.
- Although both of the 2 affected members of the family who were studied
- developed bilateral vestibular schwannomas, the first showed onset of
- the disease at the age of 31 years and presented with various central,
- peripheral, and abdominal tumors, while the second patient showed later
- onset of clinical symptoms (at age 52 years) and presented with only 2
- additional small spinal tumors.
-
- .0007
- NEUROFIBROMATOSIS, TYPE 2
- NF2, PHE96DEL
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- an inframe deletion of 3 basepairs corresponding to codon 96 (CTT) in
- exon 3. The mutation causes a deletion of phenylalanine at position 96.
-
- .0008
- NEUROFIBROMATOSIS, TYPE 2
- NF2, GLU182TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a G-to-T substitution at nucleotide 544 in exon 6, resulting in a stop
- codon at position 182.
-
- .0009
- NEUROFIBROMATOSIS, TYPE 2
- NF2, ARG262TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a C-to-T substitution at nucleotide 784 in exon 8, resulting in a stop
- codon at position 262.
-
- .0010
- NEUROFIBROMATOSIS, TYPE 2
- NF2, GLN320TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a C-to-T substitution at nucleotide 958 in exon 10, resulting in a stop
- codon at position 320.
-
- .0011
- NEUROFIBROMATOSIS, TYPE 2
- NF2, ARG341TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a C-to-T substitution at nucleotide 1021 in exon 11, resulting in a stop
- codon at position 341.
-
- .0012
- NEUROFIBROMATOSIS, TYPE 2
- NF2, GLN407TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a C-to-T substitution at nucleotide 1219 in exon 12, resulting in a stop
- codon at position 407.
-
- .0013
- NEUROFIBROMATOSIS, TYPE 2
- NF2, GLU463TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a G-to-T substitution at nucleotide 1387 in exon 13, resulting in a stop
- codon at position 463.
-
- .0014
- NEUROFIBROMATOSIS, TYPE 2
- NF2, ARG466TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a C-to-T substitution at nucleotide 1396 in exon 13, resulting in a stop
- codon at position 466.
-
- .0015
- NEUROFIBROMATOSIS, TYPE 2
- NF2, GLU527TER
- In a study of 33 unrelated patients, MacCollin et al. (1994) identified
- a G-to-T substitution at nucleotide 1579 in exon 15, resulting in a stop
- codon at position 527.
-
- .0016
- NEUROFIBROMATOSIS, TYPE 2
- NF2, PHE62SER
- Scoles et al. (1996) found a T-to-C transition at nucleotide 185 in exon
- 2 resulting in a substitution of serine for phenylalanine-62 in a family
- with both mild and severe NF2 phenotypes. This mutation had previously
- been reported by Bourn et al. (1994) in a family in which the NF2
- phenotype was uniformly mild.
-
- *FIELD* SA
- Bouzas et al. (1993); Evans et al. (1992); Evans et al. (1992); Gardner
- and Frazier (1930); Martuza and Ojemann (1982); Nager (1964); Perez
- Demoura et al. (1969); Rouleau et al. (1987); Rouleau et al. (1987);
- Siggers et al. (1975)
- *FIELD* RF
- 1. Arai, E.; Ikeuchi, T.; Karasawa, S.; Tamura, A.; Yamamoto, K.;
- Kida, M.; Ichimura, K.; Yuasa, Y.; Tonomura, A.: Constitutional translocation
- t(4;22)(q12;q12.2) associated with neurofibromatosis type 2. Am.
- J. Med. Genet. 44: 163-167, 1992.
-
- 2. Bianchi, A. B.; Hara, T.; Ramesh, V.; Gao, J.; Klein-Szanto, A.
- J. P.; Morin, F.; Menon, A. G.; Trofatter, J. A.; Gusella, J. F.;
- Seizinger, B. R.; Kley, N.: Mutations in transcript isoforms of the
- neurofibromatosis 2 gene in multiple human tumour types. Nature Genet. 6:
- 185-192, 1994.
-
- 3. Bianchi, A. B.; Mitsunaga, S.-I.; Cheng, J. Q.; Klein, W. M.; Jhanwar,
- S. C.; Seizinger, B.; Kley, N.; Klein-Szanto, A. J. P.; Testa, J.
- R.: High frequency of inactivating mutations in the neurofibromatosis
- type 2 gene (NF2) in primary malignant mesotheliomas. Proc. Nat.
- Acad. Sci. 92: 10854-10858, 1995.
-
- 4. Bijlsma, E. K.; Merel, P.; Bosch, D. A.; Westerveld, A.; Delattre,
- O.; Thomas, G.; Hulsebos, T. J. M.: Analysis of mutations in the
- SCH gene in schwannomas. Genes Chromosomes Cancer 11: 7-14, 1994.
-
- 5. Bourn, D.; Carter, S. A.; Mason, S.; Evans, D. G. R.; Strachan,
- T.: Germline mutations in the neurofibromatosis type 2 tumor suppressor
- gene. Hum. Molec. Genet. 3: 813-816, 1994.
-
- 6. Bouzas, E. A.; Freidlin, V.; Parry, D. M.; Eldridge, R.; Kaiser-Kupfer,
- M. I.: Lens opacities in neurofibromatosis 2: further significant
- correlations. Brit. J. Ophthal. 77: 354-357, 1993.
-
- 7. Bouzas, E. A.; Parry, D. M.; Eldridge, R.; Kaiser-Kupfer, M. I.
- : Visual impairment in patients with neurofibromatosis 2. Neurology 43:
- 622-623, 1993.
-
- 8. Claudio, J. O.; Malo, D.; Rouleau, G. A.: The mouse neurofibromatosis
- type 2 gene maps to chromosome 11. Genomics 21: 437-439, 1994.
-
- 9. Eldridge, R.: Central neurofibromatosis with bilateral acoustic
- neuroma. Adv. Neurol. 29: 57-65, 1981.
-
- 10. Eldridge, R.; Parry, D. M.; Kaiser-Kupfer, M. I.: Neurofibromatosis
- 2 (NF2): clinical heterogeneity and natural history based on 39 individuals
- in 9 families and 16 sporadic cases. (Abstract) Am. J. Hum. Genet. 49
- (suppl.): 133, 1991.
-
- 11. Evans, D. G. R.; Bourn, D.; Wallace, A.; Ramsden, R. T.; Mitchell,
- J. D.; Strachan, T.: Diagnostic issues in a family with late onset
- type 2 neurofibromatosis. J. Med. Genet. 32: 470-474, 1995.
-
- 12. Evans, D. G. R.; Huson, S. M.; Donnai, D.; Neary, W.; Blair, V.;
- Newton, V.; Harris, R.: A clinical study of type 2 neurofibromatosis. Quart.
- J. Med. 84: 603-618, 1992.
-
- 13. Evans, D. G. R.; Huson, S. M.; Donnai, D.; Neary, W.; Blair, V.;
- Newton, V.; Strachan, T.; Harris, R.: A genetic study of type 2 neurofibromatosis
- in the United Kingdom. II. Guidelines for genetic counselling. J.
- Med. Genet. 29: 847-852, 1992.
-
- 14. Evans, D. G. R.; Huson, S. M.; Donnai, D.; Neary, W.; Blair, V.;
- Teare, D.; Newton, V.; Strachan, T.; Ramsden, R.; Harris, R.: A genetic
- study of type 2 neurofibromatosis in the United Kingdom. I. Prevalence,
- mutation rate, fitness, and confirmation of maternal transmission
- effect on severity. J. Med. Genet. 29: 841-846, 1992.
-
- 15. Fabricant, R. N.; Todaro, G. J.; Eldridge, R.: Increased levels
- of a nerve-growth factor cross-reacting protein in 'central' neurofibromatosis. Lancet I:
- 4-7, 1979.
-
- 16. Feiling, A.; Ward, E.: A familial form of acoustic tumour. Brit.
- Med. J. 1: 496-497, 1920.
-
- 17. Gardner, W. J.; Frazier, C. H.: Hereditary bilateral acoustic
- tumors. J. Hered. 22: 7-8, 1933.
-
- 18. Gardner, W. J.; Frazier, C. H.: Bilateral acoustic neurofibromas.
- A clinical study and survey of a family of five generations with bilateral
- deafness in 38 members. Arch. Neurol. Psychiat. 23: 266-302, 1930.
-
- 19. Gardner, W. J.; Turner, O.: Bilateral acoustic neurofibromas:
- further clinical and pathologic data on hereditary deafness and Recklinghausen's
- disease. Arch. Neurol. Psychiat. 44: 76-99, 1940.
-
- 20. Honda, M.; Arai, E.; Sawada, S.; Ohta, A.; Niimura, M.: Neurofibromatosis
- 2 and neurilemmomatosis gene are identical. J. Invest. Derm. 104:
- 74-77, 1995.
-
- 21. Kaiser-Kupfer, M. I.; Freidlin, V.; Datiles, M. B.; Edwards, P.
- A.; Sherman, J. L.; Parry, D.; McCain, L. M.; Eldridge, R.: The association
- of posterior capsular lens opacities with bilateral acoustic neuromas
- in patients with neurofibromatosis type 2. Arch. Ophthal. 107: 541-544,
- 1989.
-
- 22. Kanter, W. R.; Eldridge, R.; Fabricant, R.; Allen, J. C.; Koerber,
- T.: Central neurofibromatosis with bilateral acoustic neuroma: genetic,
- clinical and biochemical distinctions from peripheral neurofibromatosis. Neurology 30:
- 851-859, 1980.
-
- 23. Kluwe, L.; Mautner, V.-F.: A missense mutation in the NF2 gene
- results in moderate and mild clinical phenotypes of neurofibromatosis
- type 2. Hum. Genet. 97: 224-227, 1996.
-
- 24. Knudson, A.: Asbestos and mesothelioma: genetic lessons from
- a tragedy. Proc. Nat. Acad. Sci. 92: 10819-10820, 1995.
-
- 25. Landau, K.; Dossetor, F. M.; Hoyt, W. F.; Muci-Mendoza, R.: Retinal
- hamartoma in neurofibromatosis 2. Arch. Ophthal. 108: 328-329, 1990.
-
- 26. Lee, D. K.; Abbott, M. L.: Familial central nervous system neoplasia:
- case report of a family with von Recklinghausen's neurofibromatosis. Arch.
- Neurol. 20: 154-160, 1969.
-
- 27. MacCollin, M.; Ramesh, V.; Jacoby, L. B.; Louis, D. N.; Rubio,
- M.-P.; Pulaski, K.; Trofatter, J. A.; Short, M. P.; Bove, C.; Eldridge,
- R.; Parry, D. M.; Gusella, J. F.: Mutational analysis of patients
- with neurofibromatosis 2. Am. J. Hum. Genet. 55: 314-320, 1994.
-
- 28. Martuza, R. L.; Eldridge, R.: Neurofibromatosis 2 (bilateral
- acoustic neurofibromatosis). New Eng. J. Med. 318: 684-688, 1988.
-
- 29. Martuza, R. L.; Ojemann, R. G.: Bilateral acoustic neuromas:
- clinical aspects, pathogenesis and treatment. Neurosurgery 10: 1-12,
- 1982.
-
- 30. Mayfrank, L.; Wullich, B.; Wolff, G.; Finke, J.; Gouzoulis, E.;
- Gilsbach, J. M.: Neurofibromatosis 2: a clinically and genetically
- heterogeneous disease? Report on 10 sporadic cases. Clin. Genet. 38:
- 362-370, 1990.
-
- 31. Merel, P.; Hoang-Xuan, K.; Sanson, M.; Moreau-Aubry, A.; Bijlsma,
- E. K.; Lazaro, C.; Moisan, J. P.; Resche, F.; Nishisho, I.; Estivill,
- X.; Delattre, J. Y.; Poisson, M.; Theillet, C.; Hulsebos, T.; Delattre,
- O.; Thomas, G.: Predominant occurrence of somatic mutations of the
- NF2 gene in meningiomas and schwannomas. Genes Chromosomes Cancer 13:
- 211-216, 1995.
-
- 32. Moyes, P. D.: Familial bilateral acoustic neuroma affecting 14
- members from four generations. J. Neurosurg. 29: 78-82, 1968.
-
- 33. Mrazek, J.; Fiser, Z.; Mrazkova, D.: Diagnosis, size, and operation
- results in 41 acoustic neurinomas. Neoplasma 35: 467-474, 1988.
-
- 34. Myers, R. M.; Fischer, S. G.; Lerman, L. S.; Maniatis, T.: Nearly
- all single base substitutions in DNA fragments joined to a GC-clamp
- can be detected by denaturing gradient gel electrophoresis. Nucleic
- Acids Res. 13: 3131-3145, 1985.
-
- 35. Nager, G. T.: Association of bilateral VIIIth nerve tumors with
- meningioma in von Recklinghausen's disease. Laryngoscope 74: 1220-1261,
- 1964.
-
- 36. Nager, G. T.: Acoustic neuromas: pathology and differential diagnosis. Arch.
- Otolaryng. 89: 252-279, 1969.
-
- 37. Narod, S. A.; Parry, D. M.; Parboosingh, J.; Lenoir, G. M.; Ruttledge,
- M.; Fischer, G.; Eldridge, R.; Martuza, R. L.; Frontali, M.; Haines,
- J.; Gusella, J. F.; Rouleau, G. A.: Neurofibromatosis type 2 appears
- to be a genetically homogeneous disease. Am. J. Hum. Genet. 51:
- 486-496, 1992.
-
- 38. NIH Consensus Development Conference: Neurofibromatosis: conference
- statement. Arch. Neurol. 45: 475-578, 1988.
-
- 39. Niimura, M.: Neurofibromatosis (3). Rinsho Derma (Tokyo) 15:
- 653-663, 1973.
-
- 40. Papi, L.; De Vitis, L. R.; Vitelli, F.; Ammannati, F.; Mennonna,
- P.; Montali, E.; Bigozzi, U.: Somatic mutations in the neurofibromatosis
- type 2 gene in sporadic meningiomas. Hum. Genet. 95: 347-351, 1995.
-
- 41. Parry, D. M.; Eldridge, R.; Kaiser-Kupfer, M. I.; Bouzas, E. A.;
- Pikus, A.; Patronas, N.: Neurofibromatosis 2 (NF2): clinical characteristics
- of 63 affected individuals and clinical evidence for heterogeneity. Am.
- J. Med. Genet. 52: 450-461, 1994.
-
- 42. Parry, D. M.; Kaiser-Kupfer, M. I.; Eldridge, R.: Neurofibromatosis
- 2 (NF2): lens findings in 40 patients in 5 high risk groups. (Abstract) Am.
- J. Hum. Genet. 49 (suppl.): 155, 1991.
-
- 43. Parry, D. M.; MacCollin, M. M.; Kaiser-Kupfer, M. I.; Pulaski,
- K.; Nicholson, H. S.; Bolesta, M.; Eldridge, R.; Gusella, J. F.:
- Germ-line mutations in the neurofibromatosis 2 gene: correlations
- with disease severity and retinal abnormalities. Am. J. Hum. Genet. 59:
- 529-539, 1996.
-
- 44. Pastores, G. M.; Michels, V. V.; Jack, C. R., Jr.: Early childhood
- diagnosis of acoustic neuromas in presymptomatic individuals at risk
- for neurofibromatosis 2. Am. J. Med. Genet. 41: 325-329, 1991.
-
- 45. Pearson-Webb, M. A.; Kaiser-Kupfer, M. I.; Eldridge, R.: Eye
- findings in bilateral acoustic (central) neurofibromatosis: association
- with presenile lens opacities and cataracts but absence of Lisch nodules.
- (Letter) New Eng. J. Med. 315: 1553-1554, 1986.
-
- 46. Perez Demoura, L. F.; Hayden, R. C., Jr.; Conner, G. H.: Bilateral
- acoustic neurinoma and neurofibromatosis. Arch. Otolaryng. 90: 28-34,
- 1969.
-
- 47. Pulst, S.-M.; Riccardi, V. M.; Fain, P.; Korenberg, J. R.: Familial
- spinal neurofibromatosis: clinical and DNA linkage analysis. Neurology 41:
- 1923-1927, 1991.
-
- 48. Ragge, N. K.; Baser, M. E.; Klein, J.; Nechiporuk, A.; Sainz,
- J.; Pulst, S.-M.; Riccardi, V. M.: Ocular abnormalities in neurofibromatosis
- 2. Am. J. Ophthal. 120: 634-641, 1995.
-
- 49. Rouleau, G. A.; Merel, P.; Lutchman, M.; Sanson, M.; Zucman, J.;
- Marineau, C.; Hoang-Xuan, K.; Demczuk, S.; Desmaze, C.; Plougastel,
- B.; Pulst, S. M.; Lenoir, G.; Bijlsma, E.; Fashold, R.; Dumanski,
- J.; de Jong, P.; Parry, D.; Eldrige, R.; Aurias, A.; Delattre, O.;
- Thomas, G.: Alteration in a new gene encoding a putative membrane-organizing
- protein causes neuro-fibromatosis type 2. Nature 363: 515-521, 1993.
-
- 50. Rouleau, G. A.; Seizinger, B. R.; Wertelecki, W.; Haines, J. L.;
- Superneau, D. W.; Martuza, R. L.; Gusella, J. F.: Flanking markers
- bracket the neurofibromatosis type 2 (NF2) gene on chromosome 22. Am.
- J. Hum. Genet. 46: 323-328, 1990.
-
- 51. Rouleau, G. A.; Wertelecki, W.; Haines, J. L.; Hobbs, W. J.; Trofatter,
- J. A.; Seizinger, B.; Martuza, R. L.; Superneau, D. W.; Conneally,
- P. M.; Gusella, J. F.: Genetic linkage of bilateral acoustic neurofibromatosis
- to DNA markers on chromosome 22. (Abstract) Cytogenet. Cell Genet. 46:
- 684-685, 1987.
-
- 52. Rouleau, G. A.; Wertelecki, W.; Haines, J. L.; Hobbs, W. J.; Trofatter,
- J. A.; Seizinger, B. R.; Martuza, R. L.; Superneau, D. W.; Conneally,
- P. M.; Gusella, J. F.: Genetic linkage of bilateral acoustic neurofibromatosis
- to a DNA marker on chromosome 22. Nature 329: 246-248, 1987.
-
- 53. Ruttledge, M. H.; Andermann, A. A.; Phelan, C. M.; Claudio, J.
- O.; Han, F.; Chretien, N.; Rangaratnam, S.; MacCollin, M.; Short,
- P.; Parry, D.; Michels, V.; Riccardi, V. M.; Weksberg, R.; Kitamura,
- K.; Bradburn, J. M.; Hall, B. D.; Propping, P.; Rouleau, G. A.: Type
- of mutation in the neurofibromatosis type 2 gene (NF2) frequently
- determines severity of disease. Am. J. Hum. Genet. 59: 331-342,
- 1996.
-
- 54. Ruttledge, M. H.; Narod, S. A.; Dumanski, J. P.; Parry, D. M.;
- Eldridge, R.; Wertelecki, W.; Parboosingh, J.; Faucher, M.-C.; Lenoir,
- G. M.; Collins, V. P.; Nordenskjold, M.; Rouleau, G. A.: Presymptomatic
- diagnosis for neurofibromatosis 2 with chromosome 22 markers. Neurology 43:
- 1753-1760, 1993.
-
- 55. Ruttledge, M. H.; Sarrazin, J.; Rangaratnam, S.; Phelan, C. M.;
- Twist, E.; Merel, P.; Delattre, O.; Thomas, G.; Nordenskjold, M.;
- Collins, V. P.; Dumanski, J. P.; Rouleau, G. A.: Evidence for the
- complete inactivation of the NF2 gene in the majority of sporadic
- meningiomas. Nature Genet. 6: 180-184, 1994.
-
- 56. Sainz, J.; Huynh, D. P.; Figueroa, K.; Ragge, N. K.; Baser, M.
- E.; Pulst, S.-M.: Mutations of the neurofibromatosis type 2 gene
- and lack of the gene product in vestibular schwannomas. Hum. Molec.
- Genet. 3: 885-891, 1994.
-
- 57. Scoles, D. R.; Baser, M. E.; Pulst, S.-M.: A missense mutation
- in the neurofibromatosis 2 gene occurs in patients with mild and severe
- phenotypes. Neurology 47: 544-546, 1996.
-
- 58. Seizinger, B. R.; Martuza, R. L.; Gusella, J. F.: Loss of genes
- on chromosome 22 in tumorigenesis of human acoustic neuroma. Nature 322:
- 644-647, 1986.
-
- 59. Seizinger, B. R.; Rouleau, G.; Ozelius, L. J.; Lane, A. H.; St.
- George-Hyslop, P.; Huson, S.; Gusella, J. F.; Martuza, R. L.: Common
- pathogenetic mechanism for three tumor types in bilateral acoustic
- neurofibromatosis. Science 236: 317-319, 1987.
-
- 60. Siggers, D. C.; Boyer, S. H.; Eldridge, R.: Nerve-growth factor
- in disseminated neurofibromatosis. (Letter) New Eng. J. Med. 292:
- 1134, 1975.
-
- 61. Thomas, G.: Personal Communication. Paris, France 11/17/1993.
-
- 62. Trofatter, J. A.; MacCollin, M. M.; Rutter, J. L.; Murrell, J.
- R.; Duyao, M. P.; Parry, D. M.; Eldridge, R.; Kley, N.; Menon, A.
- G.; Pulaski, K.; Haase, V. H.; Ambrose, C. M.; Munroe, D.; Bove, C.;
- Haines, J. L.; Martuza, R. L.; MacDonald, M. E.; Seizinger, B. R.;
- Short, M. P.; Buckler, A. J.; Gusella, J. F.: A novel moesin-, ezrin-,
- radixin-like gene is a candidate for the neurofibromatosis 2 tumor
- suppressor. Cell 72: 791-800, 1993.
-
- 63. Watson, C. J.; Gaunt, L.; Evans, G.; Patel, K.; Harris, R.; Strachan,
- T.: A disease-associated germline deletion maps the type 2 neurofibromatosis
- (NF2) gene between the Ewing sarcoma region and the leukaemia inhibitory
- factor locus. Hum. Molec. Genet. 2: 701-704, 1993.
-
- 64. Wellenreuther, R.; Kraus, J. A.; Lenartz, D.; Menon, A. G.; Schramm,
- J.; Louis, D. N.; Ramesh, V.; Gusella, J. F.; Wiestler, O. D.; von
- Deimling, A.: Analysis of the neurofibromatosis 2 gene reveals molecular
- variants of meningioma. Am. J. Path. 146: 827-832, 1995.
-
- 65. Wertelecki, W.; Rouleau, G. A.; Superneau, D. W.; Forehand, L.
- W.; Williams, J. P.; Haines, J. L.; Gusella, J. F.: Neurofibromatosis
- 2: clinical and DNA linkage studies of a large kindred. New Eng.
- J. Med. 319: 278-283, 1988.
-
- 66. Wishart, J. H.: Case of tumours in the skull, dura mater, and
- brain. Edinburgh Med. Surg. J. 18: 393-397, 1822.
-
- 67. Wolff, R. K.; Frazer, K. A.; Jackler, R. K.; Lanser, M. J.; Pitts,
- L. H.; Cox, D. R.: Analysis of chromosome 22 deletions in neurofibromatosis
- type 2-related tumors. Am. J. Hum. Genet. 51: 478-485, 1992.
-
- 68. Worster-Drought, C.; Dickson, W. E. C.; McMenemey, W. H.: Multiple
- meningeal and perineural tumors with analogous changes in the glia
- and ependyma (neurofibroblastomatosis). Brain 60: 85-117, 1937.
-
- 69. Young, D. F.; Eldridge, R.; Gardner, W. J.: Bilateral acoustic
- neuroma in a large kindred. J.A.M.A. 214: 347-353, 1970.
-
- *FIELD* CS
-
- Neuro:
- Bilateral acoustic neuroma;
- Meningioma;
- Glioma;
- Schwannoma;
- Generalized and isolated neuropathy
-
- Eyes:
- Visual loss;
- Juvenile posterior subcapsular or nuclear cataract;
- No Lisch nodules;
- Macular hamartoma;
- Lagophthalmos;
- Decreased lacrimal secretion;
- Corneal hypesthesia
-
- Ears:
- Hearing loss;
- Tinnitus
-
- Skin:
- Usually less than 6 cafe-au-lait spots;
- Often no peripheral neurofibromata;
- Discrete well-circumscribed, slightly raised, roughened skin areas
- often pigmented and hairy;
- Spherical subcutaneous tumors on peripheral nerves;
- Intradermal violaceous papillary skin neurofibroma
-
- Inheritance:
- Autosomal dominant (22q12.2)
-
- *FIELD* CN
- Orest Hurko - updated: 11/6/1996
- Moyra Smith - updated: 10/1/1996
- Moyra Smith - updated: 9/13/1996
- Stylianos E. Antonarakis - updated: 7/4/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 03/31/1997
- mark: 11/6/1996
- terry: 10/23/1996
- mark: 10/1/1996
- mark: 9/13/1996
- carol: 7/4/1996
- terry: 7/1/1996
- mark: 6/7/1996
- joanna: 5/6/1996
- mark: 3/3/1996
- terry: 2/26/1996
- mark: 2/16/1996
- mark: 2/13/1996
- mark: 12/12/1995
- terry: 12/11/1995
- mark: 9/10/1995
- terry: 5/25/1995
- carol: 2/17/1995
- jason: 7/25/1994
- mimadm: 6/26/1994
- warfield: 4/7/1994
-
- *RECORD*
- *FIELD* NO
- 101120
- *FIELD* TI
- 101120 ACROCEPHALOPOLYSYNDACTYLY TYPE III
- ACPS III;;
- ACPS WITH LEG HYPOPLASIA;;
- SAKATI-NYHAN SYNDROME
- *FIELD* TX
- This designation may be appropriate for the malformation syndrome
- described by Sakati et al. (1971) in a single male. The calvaria was
- large and the face disproportionately small. All cranial sutures were
- fused. The ears were dysplastic and low-set. Maxillary hypoplasia,
- dental crowding, prognathism and short neck with low hairline were
- features. A sixth digit had been removed from the right hand. The feet
- were adducted and showed polysyndactyly with 7 toes on the right and 6
- toes on the left. The tibias were hypoplastic and the fibulas were
- deformed and displaced. The chromosomes were normal. Advanced parental
- age supported new dominant mutation as the cause. No other cases have,
- it seems, been reported.
-
- *FIELD* RF
- 1. Sakati, N.; Nyhan, W. L.; Tisdale, W. K.: A new syndrome with
- acrocephalopolydactyly, cardiac disease, and distinctive defects of
- the ear, skin and lower limbs. J. Pediat. 79: 104-109, 1971.
-
- *FIELD* CS
-
- Skull:
- Craniosynostosis;
- Acrocephaly
-
- Facies:
- Flat facies;
- Small facies;
- Prognathism;
- Maxillary hypoplasia
-
- Eyes:
- Shallow orbits;
- Hypertelorism
-
- Ears:
- Dysplastic ears;
- Low-set ears
-
- Teeth:
- Dental crowding
-
- Limbs:
- Preaxial polydactyly;
- Syndactyly;
- Broad thumbs and broad great toes;
- Hypoplastic legs
-
- Neck:
- Short neck with low hairline
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
-
- *RECORD*
- *FIELD* NO
- 101200
- *FIELD* TI
- #101200 ACROCEPHALOSYNDACTYLY TYPE I; ACS1
- ACS I;;
- APERT SYNDROME
- APERT-CROUZON DISEASE; ACS II, INCLUDED;;
- VOGT CEPHALODACTYLY, INCLUDED
- *FIELD* TX
- A number sign (#) is used with this entry because of evidence (Wilkie et
- al., 1995) that Apert syndrome results from mutations in the gene
- encoding fibroblast growth factor receptor-2 (176943).
-
- Apert (1906) defined a syndrome characterized by skull malformation
- (acrocephaly of brachysphenocephalic type) and syndactyly of the hands
- and feet of a special type (complete distal fusion with a tendency to
- fusion also of the bony structures). The hand, when all the fingers are
- webbed, has been compared to a spoon and, when the thumb is free, to an
- obstetric hand. Blank (1960) assembled case material on 54 patients born
- in Great Britain. Two clinical categories were distinguished: (1)
- 'typical' acrocephalosyndactyly, to which Apert's name is appropriately
- applied; and (2) other forms lumped together as 'atypical'
- acrocephalosyndactyly. The feature distinguishing the two types is a
- middigital hand mass with a single nail common to digits 2-4, found in
- Apert syndrome and lacking in the others. Thirty-nine of the 54 were of
- Apert type. Six of 12 autopsies showed visceral anomalies but in none
- were these identical. A frequency of Apert syndrome of 1 in 160,000
- births was estimated.
-
- Varying degrees of mental deficiency are associated with the syndrome;
- however, individuals with normal intelligence have been reported.
- Individuals who have craniectomy early in life may have improved
- intelligence. Patton et al. (1988) did a longterm follow-up on 29
- patients of whom 14 (48%) had a normal or borderline IQ, 9 had mild
- mental retardation (IQ, 50-70), 4 were moderately retarded (IQ, 35-49),
- and 2 (7%) were severely retarded (IQ less than 35). Early craniectomy
- did not appear to improve intellectual outcome. Six of 7 school
- drop-outs with normal or borderline intelligence were in full-time
- employment or vocational training. Contrary to early conclusions such as
- that of Park and Powers (1920), Cohen and Kreiborg (1990) concluded that
- many patients are mentally retarded. They had information on 30 patients
- with malformations of the corpus callosum, the limbic structures, or
- both. A variety of other malformations were observed. The authors
- suggested that these malformations may be responsible for mental
- retardation. Progressive hydrocephalus seemed to be uncommon and was
- frequently confused with nonprogressive ventriculomegaly.
-
- Cinalli et al. (1995) found that only 4 of their series of 65 patients
- with Apert syndrome required shunting for progressive hydrocephalus.
- Only 1.9% of their Apert's patients had chronic herniation of the
- cerebellar tonsils, the finding present in 72.7% in Crouzon (123500)
- patients. In reviewing their series of 70 children with Apert syndrome,
- Reiner et al. (1996) found an IQ greater than 70 in 50% of the children
- who had a skull decompression before 1 year of age versus only 7.1% in
- those operated on later in life. Malformations of the corpus callosum
- and ventricular size did not correlate with the final IQ whereas
- anomalies of the septum pellucidum did. The third significant factor in
- intellectual achievement was the setting in which the children were
- raised. Only 12.5% of institutionalized children had a normal IQ,
- whereas 39.3% were from those living with their families.
-
- Pelz et al. (1994) reported an 18-month-old girl who had distal
- esophageal stenosis in addition to typical manifestations of Apert
- syndrome.
-
- Schauerte and St-Aubin (1966) pointed out that progressive synostosis
- occurs in the feet, hands, carpus, tarsus, cervical vertebrae, and
- skull, and proposed 'progressive synosteosis with syndactyly' as a more
- appropriate designation.
-
- Most cases of Apert syndrome are sporadic, but there are at least 2
- reported instances of parent-to-child transmission. Roberts and Hall
- (1971) observed affected mother and daughter. Van den Bosch (quoted by
- Blank, 1960) observed the typical deformity in mother and son, and Weech
- (1927) reported mother and daughter. Low frequency of consanguinity and
- failure to observe multiple sibs make recessive inheritance unlikely.
- The evidence strongly suggests dominant inheritance, presumably
- autosomal in view of the equal sex ratio. Paternal age effect is
- demonstrable. Allanson (1986) described 2 sisters with Apert syndrome,
- born to normal, unrelated parents. Paternity appeared to be legitimate.
- Germinal mosaicism was proposed. Rollnick (1988) described what is
- purportedly the first example of male transmission; a father and
- daughter were affected. Dodson et al. (1970) described
- deletion-translocation of the short arm of a chromosome 2 to the long
- arm of a chromosome 11 or 12 in a patient with Apert syndrome. They
- found reports of chromosomal abnormalities (all involving the A group)
- in 3 other cases of Apert syndrome. Cohen (1973) provided a review of
- all the 'craniosynostosis syndromes.' Cohen et al. (1992) studied the
- birth prevalence of Apert syndrome in Denmark, Italy, Spain, and 4 areas
- of the United States. A total of 57 cases gave a birth prevalence
- calculated to be approximately 15.5 per million births, which is twice
- the rate determined in earlier studies. The mutation rate was calculated
- to be 7.8 x 10(-6) per gene per generation. Apert syndrome accounted for
- about 4.5% of all cases of craniosynostosis. Czeizel et al. (1993)
- reported a validated birth prevalence of Apert syndrome in Hungary to be
- 9.9 per million live births. The mutation rate was calculated to be 4.6
- x 10(-5) per gene per generation. Data on 14 other 'sentinel' anomalies
- observed between 1980 and 1989 were given.
-
- Kreiborg et al. (1992) found fusion of cervical vertebrae in 68% of
- patients with Apert syndrome: single fusions in 37% and multiple fusions
- in 31%. C5-C6 fusion was most common. In contrast, cervical fusion
- occurs in 25% of patients with Crouzon disease (123500) and most
- commonly involves C2-C3 only. Kreiborg et al. (1992) concluded that when
- fusions are present, C5-C6 involvement in the Apert syndrome and C2-C3
- involvement in Crouzon disease separate the 2 conditions in most cases.
- Radiographic study of the cervical spine is imperative before
- undertaking anesthesia for surgery in these patients.
-
- Cohen and Kreiborg (1995) commented on the cutaneous manifestations in a
- series of 136 cases of Apert syndrome (Cohen and Kreiborg, 1993).
- Hyperhidrosis was found in all patients. At adolescence and thereafter
- the skin was oily. Acniform lesions were particularly prevalent on the
- face, chest, back, and upper arms. They commented on and illustrated the
- phenomenon of 'interrupted eyebrows.' The explanation probably involves
- the underlying bony defect. The orbital plate of the frontal bone is
- very short, resulting in early fusion of the sphenoparietal suture. This
- leads to marked retrusion and elevation of the supraorbital wings, most
- pronounced laterally. Interruption of the eyebrows corresponds to this
- defect. Several patients had excessive skin wrinkling of the forehead.
-
- Vogt (1933) described cases presenting the hand and foot malformations
- characteristic of Apert disease, together with the facial
- characteristics of Crouzon disease, caused by a very hypoplastic
- maxilla. The syndactyly was less severe than in Apert disease and the
- thumbs and little fingers were usually free. Nager and de Reynier (1948)
- gave this deformity the name of Vogt cephalodactyly, while other authors
- called it Apert-Crouzon disease, indicating the similarity to both
- abnormalities. Temtamy and McKusick (1969) called it ACS II in an
- earlier classification. There were no reported instances of hereditary
- transmission of this specific phenotype, but this could be due simply to
- low reproductive fitness. In a report on Crouzon disease, Dodge et al.
- (1959) described 2 sporadic cases of Crouzon-type craniofacial changes
- with syndactyly of both hands and feet. Most conclude that this disorder
- is actually Apert syndrome with unusually marked facial features
- (Temtamy and McKusick, 1978). Maroteaux and Fonfria (1987) described
- seemingly typical Apert syndrome except that postaxial polydactyly was
- present in the hands, and polydactyly of the feet was apparently
- preaxial. Maroteaux and Fonfria (1987) could not discern whether this
- represented a low frequency finding of Apert syndrome or a distinct
- syndrome. Sidhu and Deshmukh (1988) reported a somewhat similar case in
- the child of a first-cousin couple. Gorlin (1989) doubted the existence
- of a separate recessive entity because polysyndactyly in the feet,
- especially replication of metatarsals, is not rare in Apert syndrome and
- because parental consanguinity is probably frequent in the population
- studied by Sidhu and Deshmukh (1988).
-
- In a study of mutations in the FGFR2 gene in Apert syndrome, Wilkie et
- al. (1995) scored the severity of the syndactyly according to a modified
- version of the classification of Upton (1991). In the Apert hand, the
- central 3 digits are always syndactylous; in the least severe instance
- (type 1), the thumb and part of the finger are separate from the
- syndactylous mass; in type 2, the little finger is not separate; and in
- type 3, the thumb and all fingers are included. Similarly, syndactyly in
- the foot may involve mainly the 3 lateral digits (type 1) or digits 2-5
- with a separate big toe (type 2), or be continuous (type 3).
-
- Cohen and Kreiborg (1995) studied 44 pairs of hands and 37 pairs of feet
- in Apert syndrome, using clinical, dermatoglyphic, and radiographic
- methods. They also studied histologic sections of the hand from a
- 31-week stillborn fetus. They suggested that acrocephalosyndactyly vs.
- acrocephalopolysyndactyly represents a pseudodistinction and that use of
- these terms should be discontinued. As generalizations, they pointed out
- that in Apert syndrome, the upper limb is more severely affected than
- the lower limb. Coalition of distal phalanges and synonychia found in
- the hands is never present in the feet.
-
- Park et al. (1995) performed a phenotype/genotype survey of 36 Apert
- syndrome patients. In all but one patient, an FGFR2 mutation, either
- S252W (176943.0010) or P253R (176943.0011), was found in exon IIIa (exon
- U or 7). The frequency was 71% and 26% for these 2 mutations,
- respectively. These mutations occur in the linker region between
- immunoglobulin-like domains II and III, which are involved in activation
- of the receptor by ligand binding and dimerization. The fact that one
- patient did not have a mutation in this region suggests further genetic
- heterogeneity in Apert syndrome. Study of 29 different clinical features
- demonstrated no statistically significant differences between the 2
- subgroups defined by the 2 major mutations. Since these mutations
- involve adjacent amino acids, Park et al. (1995) reasoned that they
- might be expected to have similar biologic and phenotypic consequences.
-
- Moloney et al. (1996) provided information on the mutational spectrum
- and the parental origin of the Apert mutation. Their analysis of 118
- unrelated patients with new mutations revealed that the mutational
- spectrum in Apert syndrome is remarkably narrow. The ser252to-trp
- (934C-G) mutation occurred in 74 patients and the pro253-to-arg (937C-G)
- mutation in 44 patients. To determine the parental origin of the new
- mutations in these sporadic cases of Apert syndrome, Moloney et al.
- (1996) carried out sequence analysis of the upstream and downstream
- introns that flanked the mutation-prone exon. Sequence analysis on 48
- normal individuals led to the identification of common sequence
- polymorphisms. They then used a novel PCR-based assay, ARMS
- (amplification refractory mutation system), to determine the phase of
- the mutant allele and the natural occurring polymorphisms present in the
- introns flanking the Apert mutation. Based on this assay, Moloney et al.
- (1996) determined that in all 57 informative Apert families, the mutant
- allele was paternal in origin. They noted that a paternal bias for point
- mutations is evident in a number of disorders, but that the extreme
- skewing in favor of paternal mutations observed in Apert syndrome is
- unusual. A paternal age effect was noted. Their data suggested a
- stronger paternal age effect for the 934C-G mutation, which involves a
- CpG dinucleotide, than for the 937C-G mutation, which does not.
-
- Slaney et al. (1996) found differential effects of the 2 FGFR2 mutations
- on syndactyly and cleft palate in Apert syndrome. Among 70 unrelated
- patients with Apert syndrome, 45 had the ser252-to-trp mutation and 25
- had the pro253-to-arg mutation. The syndactyly was more severe with the
- pro253-to-arg mutation, for both the hands and the feet. In contrast,
- cleft palate was significantly more common in the S252W patients. No
- convincing differences were found in the prevalence of other
- malformations associated with Apert syndrome.
-
- *FIELD* SA
- Cohen (1977); Cohen and Kreiborg (1995); Erickson (1974); Hoover
- et al. (1970); Leonard et al. (1982); Solomon et al. (1970)
- *FIELD* RF
- 1. Allanson, J. E.: Germinal mosaicism in Apert syndrome. Clin.
- Genet. 29: 429-433, 1986.
-
- 2. Apert, M. E.: De l'acrocephalosyndactylie. Bull. Mem. Soc. Med.
- Hop. Paris 23: 1310-1330, 1906.
-
- 3. Blank, C. E.: Apert's syndrome (a type of acrocephalosyndactyly):
- observations on a British series of thirty-nine cases. Ann. Hum.
- Genet. 24: 151-164, 1960.
-
- 4. Cinalli, G.; Renier, D.; Sebag, G.; Sainte-Rose, C.; Arnaud, E.;
- Pierre-Kahn, A.: Chronic tonsillar herniation in Crouzon's and Apert's
- syndromes: the role of premature synostosis of the lambdoid suture. J.
- Neurosurg. 83: 575-582, 1995.
-
- 5. Cohen, M. M., Jr.: Genetic perspectives on craniosynostosis and
- syndromes with craniosynostosis. J. Neurosurg. 47: 886-898, 1977.
-
- 6. Cohen, M. M., Jr.: An etiologic and nosologic overview of craniosynostosis
- syndromes. Birth Defects Orig. Art. Ser. XI(2): 137-189, 1973.
-
- 7. Cohen, M. M., Jr.; Kreiborg, S.: Cutaneous manifestations of Apert
- syndrome. (Letter) Am. J. Med. Genet. 58: 94-96, 1995.
-
- 8. Cohen, M. M., Jr.; Kreiborg, S.: Hands and feet in the Apert syndrome. Am.
- J. Med. Genet. 57: 82-96, 1995.
-
- 9. Cohen, M. M., Jr.; Kreiborg, S.: The central nervous system in
- the Apert syndrome. Am. J. Med. Genet. 35: 36-45, 1990.
-
- 10. Cohen, M. M., Jr.; Kreiborg, S.: Visceral anomalies in the Apert
- syndrome. Am. J. Med. Genet. 45: 758-760, 1993.
-
- 11. Cohen, M. M., Jr.; Kreiborg, S.; Lammer, E. J.; Cordero, J. F.;
- Mastroiacovo, P.; Erickson, J. D.; Roeper, P.; Martinez-Frias, M.
- L.: Birth prevalence study of the Apert syndrome. Am. J. Med. Genet. 42:
- 655-659, 1992.
-
- 12. Czeizel, A. E.; Elek, C.; Susanszky, E.: Birth prevalence study
- of Apert syndrome. (Letter) Am. J. Med. Genet. 45: 392, 1993.
-
- 13. Dodge, H. W.; Wood, M. W.; Kennedy, R. L. J.: Craniofacial dysostosis:
- Crouzon's disease. Pediatrics 23: 98-106, 1959.
-
- 14. Dodson, W. E.; Museles, M.; Kennedy, J. L., Jr.; Al-Aish, M.:
- Acrocephalosyndactylia associated with a chromosomal translocation:
- 46,XX,t(2p-;Cq+). Am. J. Dis. Child. 120: 360-362, 1970.
-
- 15. Erickson, J. D.: A study of parental age effects on the occurrence
- of fresh mutations for the Apert syndrome. Ann. Hum. Genet. 38:
- 89-96, 1974.
-
- 16. Gorlin, R. J.: Apert syndrome with polysyndactyly of the feet.
- (Letter) Am. J. Med. Genet. 32: 557, 1989.
-
- 17. Hoover, G. H.; Flatt, A. E.; Weiss, M. W.: The hand and Apert's
- syndrome. J. Bone Joint Surg. 52A: 878-895, 1970.
-
- 18. Kreiborg, A.; Barr, M., Jr.; Cohen, M. M., Jr.: Cervical spine
- in the Apert syndrome. Am. J. Med. Genet. 43: 704-708, 1992.
-
- 19. Leonard, C. O.; Daikoku, N. H.; Winn, K.: Prenatal fetoscopic
- diagnosis of the Apert syndrome. Am. J. Med. Genet. 11: 5-9, 1982.
-
- 20. Maroteaux, P.; Fonfria, M. C.: Apparent Apert syndrome with polydactyly:
- rare pleiotropic manifestation or new syndrome?. Am. J. Med. Genet. 28:
- 153-158, 1987.
-
- 21. Moloney, D. M.; Slaney, S. F.; Oldridge, M.; Wall, S. A.; Sahlin,
- P.; Stenman, G.; Wilkie, A. O. M.: Exclusive paternal origin of new
- mutations in Apert syndrome. Nature Genet. 13: 48-53, 1996.
-
- 22. Nager, F. R.; de Reynier, J. P.: Das Gehoerorgan bei den angeborenen
- Kopfmissbildungen. Pract. Otorhinolaryng. 10 (suppl. 2): 1-128,
- 1948.
-
- 23. Park, E. A.; Powers, G. F.: Acrocephaly and scaphocephaly with
- symmetrically distributed malformations of the extremities. Am. J.
- Dis. Child. 20: 235-315, 1920.
-
- 24. Park, W.-J.; Theda, C.; Maestri, N. E.; Meyers, G. A.; Fryburg,
- J. S.; Dufresne, C.; Cohen, M. M., Jr.; Jabs, E. W.: Analysis of
- phenotypic features and FGFR2 mutations in Apert syndrome. Am. J.
- Hum. Genet. 57: 321-328, 1995.
-
- 25. Patton, M. A.; Goodship, J.; Hayward, R.; Lansdown, R.: Intellectual
- development in Apert's syndrome: a long term follow up of 29 patients. J.
- Med. Genet. 25: 164-167, 1988.
-
- 26. Pelz, L.; Unger, K.; Radke, M.: Esophageal stenosis in acrocephalosyndactyly
- type I. (Letter) Am. J. Med. Genet. 53: 91 only, 1994.
-
- 27. Reiner, D.; Arnaud, E.; Cinalli, G.; Sebag, G.; Zerah, M.; Marchac,
- D.: Prognosis for mental function in Apert's syndrome. J. Neurosurg. 85:
- 66-72, 1996.
-
- 28. Roberts, K. B.; Hall, J. G.: Apert's acrocephalosyndactyly in
- mother and daughter: cleft palate in the mother. Birth Defects Orig.
- Art. Ser. VII(7): 262-264, 1971.
-
- 29. Rollnick, B. R.: Male transmission of Apert syndrome. Clin.
- Genet. 33: 87-90, 1988.
-
- 30. Schauerte, E. W.; St-Aubin, P. M.: Progressive synosteosis in
- Apert's syndrome (acrocephalosyndactyly): with a description of roentgenographic
- changes in the feet. Am. J. Roentgen. 97: 67-73, 1966.
-
- 31. Sidhu, S. S.; Deshmukh, R.: Recessive inheritance of apparent
- Apert syndrome with polysyndactyly? (Letter) Am. J. Med. Genet. 31:
- 179-180, 1988.
-
- 32. Slaney, S. F.; Oldridge, M.; Hurst, J. A.; Morriss-Kay, G. M.;
- Hall, C. M.; Poole, M. D.; Wilkie, A. O. M.: Differential effects
- of FGFR2 mutations on syndactyly and cleft palate in Apert syndrome. Am.
- J. Hum. Genet. 58: 923-932, 1996.
-
- 33. Solomon, L. M.; Fretzin, D. F.; Pruzansky, S.: Pilosebaceous
- abnormalities in Apert's syndrome. Arch. Derm. 102: 381-385, 1970.
-
- 34. Temtamy, S. A.; McKusick, V. A.: Synopsis of hand malformations
- with particular emphasis on genetic factors. Birth Defects Orig.
- Art. Ser. V(3): 125-184, 1969.
-
- 35. Temtamy, S. A.; McKusick, V. A.: The Genetics of Hand Malformations.
- New York: National Foundation-March of Dimes (pub.) 1978.
-
- 36. Upton, J.: Classification and pathologic anatomy of limb anomalies. Clin.
- Plast. Surg. 18: 321-355, 1991.
-
- 37. Vogt, A.: Dyskephalie (dysostosis craniofacialis, maladie De
- Crouzon 1912) und eine neuartige Kombination dieser Krankheit mit
- Syndaktylie der 4 Extremitaeten (Dyskephalodaktylie). Klin. Mbl.
- Augenheilk. 90: 441-454, 1933.
-
- 38. Weech, A. A.: Combined acrocephaly and syndactylism occurring
- in mother and daughter: a case report. Bull. Johns Hopkins Hosp. 40:
- 73-76, 1927.
-
- 39. Wilkie, A. O. M.; Slaney, S. F.; Oldridge, M.; Poole, M. D.; Ashworth,
- G. J.; Hockley, A. D.; Hayward, R. D.; David, D. J.; Pulleyn, L. J.;
- Rutland, P.; Malcolm, S.; Winter, R. M.; Reardon, W.: Apert syndrome
- results from localized mutations of FGFR2 and is allelic with Crouzon
- syndrome. Nature Genet. 9: 165-172, 1995.
-
- *FIELD* CS
-
- Facies:
- Flat facies
-
- Eyes:
- Shallow orbits;
- Hypertelorism
-
- Mouth:
- Narrow palate
-
- Skull:
- Craniosynostosis;
- Brachysphenocephalic acrocephaly
-
- Limbs:
- Syndactyly;
- Broad thumb;
- Broad great toe
-
- Nails:
- Single nail digits 2-4
-
- Neuro:
- Variable mental retardation;
- Corpus callosum and/or limbic malformations
-
- Spine:
- Fused cervical vertebrae
-
- Inheritance:
- Autosomal dominant;
- paternal age effect
-
- *FIELD* CN
- Orest Hurko - updated: 11/05/1996
- Iosif W. Lurie - updated: 8/10/1996
- Moyra Smith - updated: 4/29/1996
- Orest Hurko - updated: 4/1/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/16/1986
-
- *FIELD* ED
- mark: 11/05/1996
- terry: 10/23/1996
- carol: 8/10/1996
- mark: 7/11/1996
- carol: 5/22/1996
- terry: 5/3/1996
- mark: 5/3/1996
- carol: 4/29/1996
- terry: 4/15/1996
- terry: 4/1/1996
- terry: 3/22/1996
- mark: 3/3/1996
- mark: 2/5/1996
- mark: 8/30/1995
- carol: 2/17/1995
- pfoster: 8/18/1994
- warfield: 4/6/1994
- mimadm: 3/11/1994
- carol: 11/3/1993
-
- *RECORD*
- *FIELD* NO
- 101400
- *FIELD* TI
- #101400 ACROCEPHALOSYNDACTYLY TYPE III
- ACS III; ACS3;;
- CHOTZEN SYNDROME;;
- SAETHRE-CHOTZEN SYNDROME; SCS;;
- ACROCEPHALY, SKULL ASYMMETRY, AND MILD SYNDACTYLY
- *FIELD* TX
- A number sign (#) is used with this entry because of evidence that the
- Saethre-Chotzen syndrome is caused by mutations in the TWIST
- transcription factor gene (601622).
-
- In the family described by Saethre (1931), a mother, 2 daughters, and
- probably other maternal relatives showed mild acrocephaly, asymmetry of
- the skull, and partial soft tissue syndactyly of fingers 2 and 3 and
- toes 3 and 4. Chotzen (1932) found identical malformations in a father
- and 2 sons. Bartsocas et al. (1970) described a Lithuanian kindred
- living in the United States in which 10 persons in 3 generations were
- affected, with several instances of male-to-male transmission. In 1961
- Waardenburg reported asymmetry of the skull and orbits (plagiocephaly),
- strabismus, and a thin, long, pointed nose in 6 generations of a
- kindred. Some affected persons had bifid terminal phalanges of digits 2
- and 3 and absence of the first metatarsal. Cleft palate, hydrophthalmos,
- cardiac malformation, and contractures of elbows and knees were present
- in some. Aase and Smith (1970) described a syndrome comprising asymmetry
- of the face (hypoplasia of the left side), unusually shaped ear with
- prominent crus (see their Fig. 2), and Simian crease in 5 members of 3
- generations (with 1 instance of male-to-male transmission). They pointed
- out similarities to and differences from the asymmetry of the face and
- skull with abnormalities of the digits described by Waardenburg et al.
- (1961). Gorlin (1971) thought the syndrome described by Aase and Smith
- (1970) was Chotzen syndrome. Carter et al. (1982) recognized 9 patients,
- including familial cases. Like Aase and Smith (1970), they recognized a
- long and prominent ear crus as a valuable sign. Kurczynski and Casperson
- (1988) described mother and daughter with craniosynostosis and
- symmetrical syndactyly involving the fourth and fifth toes. In addition,
- both had a short columella and small pinnae. Kurczynski and Casperson
- (1988) concluded that this represented a new form of
- acrocephalosyndactyly and suggested the designation
- auralcephalosyndactyly (109050). Legius et al. (1989) described mother
- and son with bilateral symmetrical syndactyly of the third, fourth and
- fifth toes, mild craniosynostosis, and small pinnae. In addition, the
- mother had fusion of 2 cervical vertebrae and partial duplication of the
- first metatarsal. Furthermore, the distal phalanges of both great toes
- were bifid. These skeletal changes in combination with cutaneous
- syndactyly of the toes, abnormal auricles, and acrocephaly have been
- described in the Saethre-Chotzen syndrome (Kopysc et al., 1980) and also
- in the Robinow-Sorauf syndrome (Carter et al., 1982). Legius et al.
- (1989) concluded that the Saethre-Chotzen, auralcephalosyndactyly, and
- Robinow-Sorauf (180750) syndromes may be somewhat different expressions
- of the same dominant gene. Marini et al. (1991) presented a family
- illustrating the mild and easily missed expression of the gene in a
- parent. Niemann-Seyde et al. (1991) observed ACS III in 9 members of 4
- generations of a family; 5 of them were severely affected. Russo et al.
- (1991) described a case of renotubular dysgenesis (267430) in an infant
- who had widely patent cranial fontanels and whose father and sister
- showed acrocephalosyndactyly of the Saethre-Chotzen type. This was
- probably a coincidental association between a recessive disorder and a
- dominant disorder.
-
- See craniosynostosis (123100) for well-established mapping to
- 7p21.3-p21.2 on the basis of structural alterations in that region. The
- gene for Greig cephalopolysyndactyly syndrome (GCPS; 175700) appears to
- be located at 7p13. Brueton et al. (1992) presented molecular genetic
- linkage studies suggesting localization of the gene for the
- Saethre-Chotzen syndrome on distal 7p. Sixteen families with involvement
- in 2 or more generations were available for study. One of their families
- (number 16) had characteristics suggesting the Jackson-Weiss syndrome
- (123150). Excluding this family and pedigree number 15 which had a
- Pfeiffer-like syndrome (101600), Brueton et al. (1992) found tight
- linkage to D7S370 (maximum lod = 3.00 at theta = 0.00) and with D7S10
- (maximum lod = 2.39 at theta = 0.00). The relationship to other forms of
- craniosynostosis with hand anomalies that map to 7p remains to be
- determined. In linkage analysis on 6 ACS III families using 5 CA repeat
- polymorphisms from 7p, Malcolm et al. (1993) found evidence suggesting
- location between D7S493 and D7S516. Two patients, a father and daughter,
- were found with ACS III and a balanced translocation t(7;10)(p21;q21.2).
- Reid et al. (1993) reported 2 additional patients, a male infant and his
- mother, with an apparently balanced translocation t(2;7)(p23;p22).
- Lewanda et al. (1994) confirmed linkage of the Saethre-Chotzen syndrome
- to 7p. The tightest linkage was to D7S493; linkage and haplotype
- analyses refined the location of the gene to the region between D7S513
- and D7S516. On the basis of 4 patients with apparently balanced
- translocations at 7p21.2, Rose et al. (1994) narrowed the localization
- of the ACS3 gene to a 6-cM region. By fluorescence in situ
- hybridization, they showed that the breakpoints were situated within the
- region flanked by genetic markers D7S488 and D7S493 in distal 7p.
- Lewanda et al. (1994) used linkage and haplotype analyses to narrow the
- disease locus to an 8-cM region between D7S664 and D7S507. The tightest
- linkage was to D7S664; maximum lod = 7.16 at theta = 0.00. Studying the
- t(2;7)(p23;p22) in a patient with Saethre-Chotzen syndrome, Lewanda et
- al. (1994) found that the D7S664 locus lay distal to the 7p22
- breakpoint, whereas the D7S507 locus was deleted from the translocation
- chromosome. Wilkie et al. (1995) reported 3 further families, each
- segregating a different reciprocal chromosomal translocation involving
- 7p21. A total of 7 apparently balanced carriers were identified and all
- manifest features of the Saethre-Chotzen syndrome, although only 2 had
- overt craniosynostosis. In one family, the carriers were immediately
- recognized by their unusual ears, and clefts of the hard or soft palate
- were present in all 3 families. The abnormally configured ear was
- pictured in 1 member from each of 3 generations.
-
- Ma et al. (1996) studied 3 further families to provide additional
- support to the localization of a disease gene between D7S493 and D7S664.
- There was a suspicion that at least 2 disease-causing genes may map to
- 7p, 1 distal and 1 proximal to D7S488. The MEOX2 gene (600535) maps to
- the same region of 7p (as does SCS), and is a major candidate gene in
- SCS, as it is expressed in the mesenchyma of craniofacial and limb
- structures during early mouse embryogenesis.
-
- Reardon and Winter (1994) wrote as follows: 'Clinical geneticists are
- inured to anecdotes recounting odd presentations of dysmorphic
- syndromes. Saethre-Chotzen syndrome is a case in point. A consultation
- for schizophrenia led to the first report from the Norwegian
- psychiatrist, Haakon Saethre...' (Saethre, 1931). Chotzen (1932)
- reported a father and 2 sons with the syndrome that came to carry his
- name.
-
- Howard et al. (1997) and El Ghouzzi et al. (1997) demonstrated that the
- Saethre-Chotzen syndrome results from mutations in the TWIST gene
- (601622). They were prompted to evaluate the TWIST gene, which encodes a
- basic helix-loop-helix transcription factor, because its expression
- pattern and mutant phenotypes in Drosophila and mouse are consistent
- with the SCS phenotype in humans. Howard et al. (1997) mapped the human
- TWIST gene by PCR analysis of somatic cell hybrids to 7p22-p21 in a
- region homologous to the region of mouse chromosome 12 where the murine
- TWIST gene had been mapped. They assigned it to a specific YAC which was
- known to contain the breakpoint of a chromosome translocation in 1
- Saethre-Chotzen syndrome case. Bourgeois et al. (1996) had previously
- cloned human TWIST and mapped it to 7p21. Howard et al. (1997)
- identified nonsense, missense, insertion, and deletion mutations in
- TWIST in patients with Saethre-Chotzen syndrome. El Ghouzzi et al.
- (1997) reported 21-bp insertions and nonsense mutations in the TWIST
- gene in 7 probands with SCS.
-
- *FIELD* SA
- Bianchi et al. (1985); Escobar et al. (1977); Kreiborg et al. (1972);
- Lewanda et al. (1994); McKeon-Kern and Mamunes (1977); Pantke et al.
- (1975)
- *FIELD* RF
- 1. Aase, J. M.; Smith, D. W.: Facial asymmetry and abnormalities
- of palms and ears: a dominantly inherited developmental syndrome. J.
- Pediat. 76: 928-930, 1970.
-
- 2. Bartsocas, C. S.; Weber, A. L.; Crawford, J. D.: Acrocephalosyndactyly
- type 3: Chotzen's syndrome. J. Pediat. 77: 267-272, 1970.
-
- 3. Bianchi, E.; Arico, M.; Podesta, A. F.; Grana, M.; Fiori, P.; Beluffi,
- G.: A family with the Saethre-Chotzen syndrome. Am. J. Med. Genet. 22:
- 649-658, 1985.
-
- 4. Bourgeois, P.; Stoetzel, C.; Bolcato-Bellemin, A. L.; Mattei, M.
- G.; Perrin-Schmitt, F.: The human H-twist gene is located at 7p21
- and encodes a B-HLH protein that is 96% similar to its murine M-twist
- counterpart. Mammalian Genome 7: 915-917, 1996.
-
- 5. Brueton, L. A.; van Herwerden, L.; Chotai, K. A.; Winter, R. M.
- : The mapping of a gene for craniosynostosis: evidence for linkage
- of the Saethre-Chotzen syndrome to distal chromosome 7p. J. Med.
- Genet. 29: 681-685, 1992.
-
- 6. Carter, C. O.; Till, K.; Fraser, V.; Coffey, R.: A family study
- of craniosynostosis, with probable recognition of a distinct syndrome. J.
- Med. Genet. 19: 280-285, 1982.
-
- 7. Chotzen, F.: Eine eigenartige familiaere Entwicklungsstoerung
- (Akrocephalosyndaktylie, Dysostosis craniofacialis und Hypertelorismus). Mschr.
- Kinderheilk. 55: 97-122, 1932.
-
- 8. El Ghouzzi, V.; Le Merrer, M.; Perrin-Schmitt, F.; Lajeunie, E.;
- Benit, P.; Renier, D.; Bourgeois, P.; Bolcato-Bellemin, A.-L.; Munnich,
- A.; Bonaventure, J.: Mutations of the TWIST gene in the Saethre-Chotzen
- syndrome. Nature Genet. 15: 42-46, 1997.
-
- 9. Escobar, V.; Brandt, I. K.; Bixler, D.: Unusual association of
- Saethre-Chotzen syndrome and congenital adrenal hyperplasia. Clin.
- Genet. 11: 365-371, 1977.
-
- 10. Gorlin, R. J.: Personal Communication. Minneapolis, Minn.
- 1971.
-
- 11. Howard, T. D.; Paznekas, W. A.; Green, E. D.; Chiang, L. C.; Ma,
- N.; Ortiz De Luna, R. I.; Delgado, C. G.; Gonzalez-Ramos, M.; Kline,
- A. D.; Jabs, E. W.: Mutations in TWIST, a basic helix-loop-helix
- transcription factor, in Saethre-Chotzen syndrome. Nature Genet. 15:
- 36-41, 1997.
-
- 12. Kopysc, Z.; Stanska, M.; Ryzko, J.; Kulczyk, B.: The Saethre-Chotzen
- syndrome with partial bifid of the distal phalanges of the great toes:
- observations of three cases in one family. Hum. Genet. 56: 195-204,
- 1980.
-
- 13. Kreiborg, S.; Pruzansky, S.; Pashayan, H.: The Saethre-Chotzen
- syndrome. Teratology 6: 287-294, 1972.
-
- 14. Kurczynski, T. W.; Casperson, S. M.: Auralcephalosyndactyly:
- a new hereditary craniosynostosis syndrome. J. Med. Genet. 25: 491-493,
- 1988.
-
- 15. Legius, E.; Fryns, J. P.; Van den Berghe, H.: Auralcephalosyndactyly:
- a new craniosynostosis syndrome or a variant of the Saethre-Chotzen
- syndrome?. J. Med. Genet. 26: 522-524, 1989.
-
- 16. Lewanda, A. F.; Cohen, M. M., Jr.; Jackson, C. E.; Taylor, E.
- W.; Li, X.; Beloff, M.; Day, D.; Clarren, S. K.; Ortiz, R.; Garcia,
- C.; Hauselman, E.; Figueroa, A.; Wulfsberg, E.; Wilson, M.; Warman,
- M. L.; Padwa, B. L.; Whiteman, D. A. H.; Mulliken, J. B.; Jabs, E.
- W.: Genetic heterogeneity among craniosynostosis syndromes: mapping
- the Saethre-Chotzen syndrome locus between D7S513 and D7S516 and exclusion
- of Jackson-Weiss and Crouzon syndrome loci from 7p. Genomics 19:
- 115-119, 1994.
-
- 17. Lewanda, A. F.; Green, E. D.; Weissenbach, J.; Jerald, H.; Taylor,
- E.; Summar, M. L.; Phillips, J. A., III; Cohen, M.; Feingold, M.;
- Mouradian, W.; Clarren, S. K.; Jabs, E. W.: Evidence that the Saethre-Chotzen
- syndrome locus lies between D7S664 and D7S507, by genetic analysis
- and detection of a microdeletion in a patient. Am. J. Hum. Genet. 55:
- 1195-1201, 1994.
-
- 18. Ma, H. W.; Lajeunie, E.; de Parseval, N.; Munnich, A.; Renier,
- D.; Le Merrer, M.: Possible genetic heterogeneity in the Saethre-Chotzen
- syndrome. Hum. Genet. 98: 228-232, 1996.
-
- 19. Malcolm, S.; Rose, C. P. S.; van Herwerden, L.; Reardon, W.; Brueton,
- L.; Weissenbach, J.; Winter, R. M.: Mapping of Saethre-Chotzen syndrome
- (ACS III) to 7p21. (Abstract) Am. J. Hum. Genet. 53 (suppl.): A136
- only, 1993.
-
- 20. Marini, R.; Temple, K.; Chitty, L.; Genet, S.; Baraitser, M.:
- Pitfalls in counselling: the craniosynostoses. J. Med. Genet. 28:
- 117-121, 1991.
-
- 21. McKeon-Kern, C.; Mamunes, P.: A case of Saethre-Chotzen syndrome. Med.
- Coll. Va. Quart. 13(4): 186-188, 1977.
-
- 22. Niemann-Seyde, S. C.; Eber, S. W.; Zoll, B.: Saethre-Chotzen
- syndrome (ACS III) in four generations. Clin. Genet. 40: 271-276,
- 1991.
-
- 23. Pantke, O. A.; Cohen, M. M., Jr.; Witkop, C. J., Jr.; Feingold,
- M.; Schaumann, B.; Pantke, H. C.; Gorlin, R. J.: The Saethre-Chotzen
- syndrome. Birth Defects Orig. Art. Ser. XI(2): 190-225, 1975.
-
- 24. Reardon, W.; Winter, R. M.: Saethre-Chotzen syndrome. J. Med.
- Genet. 31: 393-396, 1994.
-
- 25. Reid, C. S.; McMorrow, L. E.; McDonald-McGinn, D. M.; Grace, K.
- J.; Ramos, F. J.; Zackai, E. H.; Cohen, M. M., Jr.; Jabs, E. W.:
- Saethre-Chotzen syndrome with familial translocation at chromosome
- 7p22. Am. J. Med. Genet. 47: 637-639, 1993.
-
- 26. Rose, C. S. P.; King, A. A. J.; Summers, D.; Palmer, R.; Yang,
- S.; Wilkie, A. O. M.; Reardon, W.; Malcolm, S.; Winter, R. M.: Localization
- of the genetic locus for Saethre-Chotzen syndrome to a 6 cM region
- of chromosome 7 using four cases with apparently balanced translocations
- at 7p21.2. Hum. Molec. Genet. 3: 1405-1408, 1994.
-
- 27. Russo, R.; D'Armiento, M.; Vecchione, R.: Renal tubular dysgenesis
- and very large cranial fontanels in a family with acrocephalosyndactyly
- S.C. type. Am. J. Med. Genet. 39: 482-485, 1991.
-
- 28. Saethre, M.: Ein Beitrag zum Turmschaedelproblem (Pathogenese,
- Erblichkeit und Symptomatologie). Dtsch. Z. Nervenheilk. 119: 533-555,
- 1931.
-
- 29. Waardenburg, P. J.; Franceschetti, A.; Klein, D.: Genetics and
- Ophthalmology. Springfield, Ill.: Charles C Thomas (pub.) 1:
- 1961. Pp. 301-354.
-
- 30. Wilkie, A. O. M.; Yang, S. P.; Summers, D.; Poole, M. D.; Reardon,
- W.; Winter, R. M.: Saethre-Chotzen syndrome associated with balanced
- translocations involving 7p21: three further families. J. Med. Genet. 32:
- 174-180, 1995.
-
- *FIELD* CS
-
- Facies:
- Flat facies;
- Thin, long, pointed nose
-
- Eyes:
- Shallow orbits;
- Hypertelorism;
- Plagiocephaly (asymmetry of orbits);
- Strabismus;
- Hydrophthalmos
-
- Ears:
- Long and prominent ear crus
-
- Mouth:
- Cleft palate
-
- Skull:
- Craniosynostosis;
- Acrocephaly;
- Cranial asymmetry
-
- Limbs:
- Mild syndactyly;
- Bifid terminal phalanges digits 2 and 3;
- Absent first metatarsal
-
- Cardiac:
- Congenital heart defect
-
- Joints:
- Contractures of elbows and knees
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- jenny: 01/14/1997
- terry: 1/8/1997
- terry: 12/13/1996
- terry: 4/19/1995
- carol: 1/4/1995
- pfoster: 3/31/1994
- mimadm: 3/28/1994
- carol: 10/29/1993
- carol: 10/20/1993
-
- *RECORD*
- *FIELD* NO
- 101600
- *FIELD* TI
- #101600 ACROCEPHALOSYNDACTYLY TYPE V; ACS5
- ACS V;;
- PFEIFFER TYPE ACROCEPHALOSYNDACTYLY
- NOACK SYNDROME, INCLUDED
- *FIELD* TX
- A number sign (#) is used with this entry because of the evidence
- presented by Muenke et al. (1994) that mutations in the gene for
- fibroblast growth factor receptor-1 (FGFR1; 136350) cause one form of
- familial Pfeiffer syndrome. Other cases are caused by mutation in the
- gene for fibroblast growth factor receptor-2 (FGFR2; 176943); the
- original family reported by Pfeiffer (1964) was of this type (Muenke,
- 1996). Yet other families cannot be related to either the FGFR1 locus on
- chromosome 8 or the FGFR2 locus on chromosome 10 by linkage studies.
-
- Pfeiffer (1964) found 8 affected in 3 generations, with 2 instances of
- male-to-male transmission. The striking feature was broad, short thumbs
- and big toes. The proximal phalanx of the thumb was either triangular or
- trapezoid (and occasionally fused with the distal phalanx) so that the
- thumb pointed outward (i.e., away from the other digits). Martsolf et
- al. (1971) described the case of an affected boy whose mother and
- maternal half-brother were said to be affected also. Another pedigree
- consistent with autosomal dominant inheritance was reported by Saldino
- et al. (1972).
-
- Acrocephalopolysyndactyly differs from Apert syndrome
- (acrocephalosyndactyly; 101200) in the presence of polydactyly as an
- additional feature. Earlier (Temtamy and McKusick, 1969), 2 types were
- thought to exist: type I, or Noack syndrome, a dominant, and type II, or
- Carpenter syndrome, a recessive (201000). Only the latter is, it seems,
- a valid entity.
-
- Robin et al. (1994) demonstrated linkage of markers from chromosome 8 in
- some Pfeiffer syndrome families. By performing fluorescence in situ
- hybridization on artificial chromosomes (YACs) that contained the linked
- DNA markers, they localized one gene for Pfeiffer syndrome to the
- pericentromeric region of chromosome 8. Genetic heterogeneity in the
- syndrome was demonstrated by exclusion of close linkage in other
- families. Because FGFR1 had been mapped to 8p12-p11.2, it became a
- strong candidate gene for Pfeiffer syndrome. Muenke et al. (1994)
- identified a specific mutation in this gene in all affected members of 5
- unrelated Pfeiffer syndrome families. Schell et al. (1995) demonstrated
- that Pfeiffer syndrome can also result from point mutations in the gene
- for fibroblast growth factor receptor-2.
-
- The genetic heterogeneity reflected by the linkage studies was also
- indicated by studies of the molecular defect: Lajeunie et al. (1995) and
- Rutland et al. (1995) found mutations in the FGFR2 gene in some patients
- with Pfeiffer syndrome. Crouzon syndrome (CFD1; 123500) had been the
- type of craniosynostosis hitherto related to mutations in the FGFR2
- gene. Lajeunie et al. (1995) described FGFR2 mutations in one sporadic
- case and one familial form of Pfeiffer syndrome. Rutland et al. (1995)
- reported point mutations in FGFR2 in 7 sporadic Pfeiffer syndrome
- patients. Six of the 7 Pfeiffer syndrome patients shared 2 missense
- mutations that had also been reported in Crouzon syndrome. The Crouzon
- and Pfeiffer phenotypes usually 'breed true' within families and the
- finding of identical mutations in unrelated individuals giving different
- phenotypes was a highly unexpected observation.
-
- Noack (1959) reported a 43-year-old man and his 11-month-old daughter,
- both of whom exhibited acrocephaly and polysyndactyly. Enlarged thumbs
- and great toes with duplication of the latter (preaxial polydactyly)
- were described, as well as syndactyly. Intelligence was apparently
- normal. Follow-up of Noack's kindred by Pfeiffer (1964) indicated that
- the disorder is the same as acrocephalosyndactyly type V. Robinow and
- Sorauf (1975) described an extensively affected kindred which
- illustrates the extent to which penetrance can be reduced. The proband
- showed marked valgus of unduly broad great toes, which radiologically
- showed duplication of the phalanges. In commenting on the paper, Temtamy
- (1976) stated that in her view the Noack and Pfeiffer types are one.
- (The disorder in the family reported by Robinow and Sorauf (1975) is
- treated as a separate entity and discussed under 180750.)
-
- Baraitser et al. (1980) reported a kindred particularly instructive as
- to the range of variability. The proband had the full-blown syndrome,
- whereas 8 persons in 4 sibships of the previous 3 generations had large
- halluces and partial syndactyly of the toes (mainly toes 2 and 3). The
- variability of expression was also illustrated by Vanek and Losan
- (1982). Kroczek et al. (1986) described Kleblattschaedel in association
- with Pfeiffer syndrome. Rasmussen and Frias (1988) described a girl with
- severe manifestations of Pfeiffer syndrome. The case was thought to
- represent a new mutation until the mother was examined in detail and
- found to show abnormalities of the right thumb consistent with mild
- expression of the Pfeiffer syndrome. The mother was thought to have mild
- mid-facial hypoplasia. The possibility of mosaicism in the mother seems
- strong. The mother's father was 40 years old at the time of her birth.
-
- Stone et al. (1990) described an infant with the Pfeiffer syndrome in
- whom the trachea showed replacement of the cartilaginous rings by a
- solid cartilaginous plate extending the full length of the trachea and
- beyond the carina. This resulted in tracheal stenosis. Devine et al.
- (1984) described a completely cartilaginous trachea without ring
- formation in a child with Crouzon syndrome (123500) who continued to
- have respiratory distress despite surgical repair of choanal stenosis.
- Death from respiratory problems occurred at the age of 23 months.
- Soekarman et al. (1992) described classic Pfeiffer syndrome in mother
- and son. The infant son had cloverleaf skull anomaly. The development in
- the child after surgery appeared to be normal, indicating that all
- children with the cloverleaf skull abnormality do not have a dire
- prognosis.
-
- Cohen (1993) stated that 7 Pfeiffer syndrome pedigrees (three
- 3-generation and four 2-generation) had been reported, in addition to at
- least a dozen sporadic cases. Cohen (1993) recognized 3 clinical
- subtypes which, he suggested, do not have status as separate entities
- but have important diagnostic and prognostic implications nonetheless.
- The classic syndrome is designated type 1. Type 2 consists of cloverleaf
- skull with Pfeiffer hands and feet, together with ankylosis of the
- elbows. Type 3 is similar to type 2 but without cloverleaf skull. Ocular
- proptosis is severe, and the anterior cranial base is markedly short.
- Various visceral malformations have been found in association with type
- 3. Cohen and Barone (1994) further tabulated the findings in the 3 types
- of Pfeiffer syndrome. Early demise is characteristic of both type 2 and
- type 3, which to date have occurred only as sporadic cases.
-
- Bellus et al. (1996) described a pro250-to-arg mutation in the
- extracellular domain of the FGFR3 gene (134934.0014) in 10 unrelated
- families with dominant craniosynostosis syndromes. This mutation
- (749C-G) occurs precisely at the position in FGFR3 analogous to that of
- mutations in FGFR1 (P252R; 136350.0001) and FGFR2 (P253R; 176943.0011)
- previously reported in Pfeiffer syndrome and Apert syndrome,
- respectively. The FGFR mutations in Pfeiffer syndrome and nonsyndromic
- craniosynostosis were reviewed in detail.
-
- *FIELD* SA
- Cremers (1981); Eastman et al. (1978); Escobar and Bixler (1977);
- Gnamey and Farriaux (1972); Naveh and Friedman (1976)
- *FIELD* RF
- 1. Baraitser, M.; Bowen-Bravery, M.; Saldana-Garcia, P.: Pitfalls
- of genetic counselling in Pfeiffer's syndrome. J. Med. Genet. 17:
- 250-256, 1980.
-
- 2. Bellus, G. A.; Gaudenz, K.; Zackai, E. H.; Clarke, L. A.; Szabo,
- J.; Francomano, C. A.; Muenke, M.: Identical mutations in three different
- fibroblast growth factor receptor genes in autosomal dominant craniosynostosis
- syndromes. Nature Genet. 14: 174-176, 1996.
-
- 3. Cohen, M. M., Jr.: Pfeiffer syndrome update, clinical subtypes,
- and guidelines for differential diagnosis. Am. J. Med. Genet. 45:
- 300-307, 1993.
-
- 4. Cohen, M. M., Jr.; Barone, C. M.: Reply to Dr. Winter. (Letter) Am.
- J. Med. Genet. 49: 358-359, 1994.
-
- 5. Cremers, C. W. R. J.: Hearing loss in Pfeiffer's syndrome. Int.
- J. Pediat. Otorhinolaryng. 3: 343-353, 1981.
-
- 6. Devine, P.; Bhan, M.; Feingold, M.; Leonidas, J.; Wolpert, S.:
- Completely cartilaginous trachea in a child with Crouzon syndrome. Am.
- J. Dis. Child. 138: 40-43, 1984.
-
- 7. Eastman, J. R.; Escobar, V.; Bixler, D.: Linkage analysis in dominant
- acrocephalosyndactyly. J. Med. Genet. 15: 292-293, 1978.
-
- 8. Escobar, V.; Bixler, D.: The acrocephalosyndactyly syndrome: a
- metacarpophalangeal pattern profile analysis. Clin. Genet. 11: 295-305,
- 1977.
-
- 9. Gnamey, D.; Farriaux, J.-P.: Syndrome dominant associant polysyndactylie,
- pouces en spatule, anomalies facials et retard mental (une forme particuliere
- de l'acrocephalo-polysyndactylie de type Noack). J. Genet. Hum. 19:
- 299-316, 1972.
-
- 10. Kroczek, R. A.; Muhlbauer, W.; Zimmermann, I.: Cloverleaf skull
- associated with Pfeiffer syndrome: pathology and management. Europ.
- J. Pediat. 145: 442-445, 1986.
-
- 11. Lajeunie, E.; Ma, H. W.; Bonaventure, J.; Munnich, A.; Le Merrer,
- M.; Renier, D.: FGFR2 mutations in Pfeiffer syndrome. (Letter) Nature
- Genet. 9: 108, 1995.
-
- 12. Martsolf, J. T.; Cracco, J. B.; Carpenter, G. G.; O'Hara, A. E.
- : Pfeiffer syndrome: an unusual type of acrocephalosyndactyly with
- broad thumbs and great toes. Am. J. Dis. Child. 121: 257-262, 1971.
-
- 13. Muenke, M.: Personal Communication. Philadelphia, Pennsylvania
- 2/25/1996.
-
- 14. Muenke, M.; Schell, U.; Hehr, A.; Robin, N. H.; Losken, H. W.;
- Schinzel, A.; Pulleyn, L. J.; Rutland, P.; Reardon, W.; Malcolm, S.;
- Winter, R. M.: A common mutation in the fibroblast growth factor
- receptor 1 gene in Pfeiffer syndrome. Nature Genet. 8: 269-274,
- 1994.
-
- 15. Naveh, Y.; Friedman, A.: Pfeiffer syndrome: report of a family
- and review of the literature. J. Med. Genet. 13: 277-280, 1976.
-
- 16. Noack, M.: Ein Beitrag zum Krankheitsbild der Akrozephalosyndaktylie
- (Apert). Arch. Kinderheilk. 160: 168-171, 1959.
-
- 17. Pfeiffer, R. A.: Dominant erbliche Akrocephalosyndaktylie. Z.
- Kinderheilk. 90: 301-320, 1964.
-
- 18. Rasmussen, S. A.; Frias, J. L.: Mild expression of the Pfeiffer
- syndrome. Clin. Genet. 33: 5-10, 1988.
-
- 19. Robin, N. H.; Feldman, G. J.; Mitchell, H. F.; Lorenz, P.; Wilroy,
- R. S.; Zackai, E. H.; Allanson, J. E.; Reich, E. W.; Pfeiffer, R.
- A.; Clarke, L. A.; Warman, M. L.; Mulliken, J. B.; Brueton, L. A.;
- Winter, R. M.; Price, R. A.; Gasser, D. L.; Muenke, M.: Linkage of
- Pfeiffer syndrome to chromosome 8 centromere and evidence for genetic
- heterogeneity. Hum. Molec. Genet. 3: 2153-2158, 1994.
-
- 20. Robinow, M.; Sorauf, T. J.: Acrocephalopolysyndactyly, type Noack,
- in a large kindred. Birth Defects Orig. Art. Ser. XI(5): 99-106,
- 1975.
-
- 21. Rutland, P.; Pulleyn, L. J.; Reardon, W.; Baraitser, M.; Hayward,
- R.; Jones, B.; Malcolm, S.; Winter, R. M.; Oldridge, M.; Slaney, S.
- F.; Poole, M. D.; Wilkie, A. O. M.: Identical mutations in the FGFR2
- gene cause both Pfeiffer and Crouzon syndrome phenotypes. Nature
- Genet. 9: 173-176, 1995.
-
- 22. Saldino, R. M.; Steinbach, H. L.; Epstein, C. J.: Familial acrocephalosyndactyly
- (Pfeiffer syndrome). Am. J. Roentgen. 116: 609-622, 1972.
-
- 23. Schell, U.; Hehr, A.; Feldman, G. J.; Robin, N. H.; Zackai, E.
- H.; de Die-Smulders, C.; Viskochil, D. H.; Stewart, J. M.; Wolff,
- G.; Ohashi, H.; Price, R. A.; Cohen, M. M., Jr.; Muenke, M.: Mutations
- in FGFR1 and FGFR2 cause familial and sporadic Pfeiffer syndrome. Hum.
- Molec. Genet. 4: 323-328, 1995.
-
- 24. Soekarman, D.; Fryns, J. P.; van den Berghe, H.: Pfeiffer acrocephalosyndactyly
- syndrome in mother and son with cloverleaf skull anomaly in the child. Genetic
- Counseling 3: 217-220, 1992.
-
- 25. Stone, P.; Trevenen, C. L.; Mitchell, I.; Rudd, N.: Congenital
- tracheal stenosis in Pfeiffer syndrome. Clin. Genet. 38: 145-148,
- 1990.
-
- 26. Temtamy, S.: Personal Communication. Cairo, Egypt 1976.
-
- 27. Temtamy, S.; McKusick, V. A.: Synopsis of hand malformations
- with particular emphasis on genetic factors. Birth Defects Orig.
- Art. Ser. V(3): 125-184, 1969.
-
- 28. Vanek, J.; Losan, F.: Pfeiffer's type of acrocephalosyndactyly
- in two families. J. Med. Genet. 19: 289-292, 1982.
-
- *FIELD* CS
-
- Facies:
- Flat facies
-
- Eyes:
- Shallow orbits;
- Hypertelorism
-
- Skull:
- Mild craniosynostosis;
- Acrocephaly
-
- Limbs:
- Broad thumb;
- Broad great toe;
- Polysyndactyly
-
- Radiology:
- Thumb proximal phalanx triangular or trapezoid, occasionally fused
- with distal phalanx
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 10/05/1996
- terry: 10/2/1996
- carol: 8/20/1996
- mark: 3/3/1996
- terry: 2/27/1996
- mark: 8/11/1995
- carol: 2/13/1995
- terry: 1/31/1995
- warfield: 4/7/1994
- mimadm: 3/11/1994
- carol: 10/14/1993
-
- *RECORD*
- *FIELD* NO
- 101800
- *FIELD* TI
- *101800 ACRODYSOSTOSIS
- *FIELD* TX
- Maroteaux and Malamut (1968) suggested that 'peripheral dysostosis'
- (q.v.) is a heterogeneous class. They described acrodysostosis as a
- condition in which peculiar facies (short nose, open mouth and
- prognathism) is associated with the small hands and feet. Mental
- deficiency is frequent. Cone epiphyses occur in this condition. Robinow
- et al. (1971) reported 9 cases and reviewed 11 from the literature. None
- was familial. Jones et al. (1975) found elevated average paternal age in
- this disorder, thus supporting autosomal dominant inheritance. It is
- possible that at least some cases that have been labeled acrodysostosis
- represent the normocalcemic form of pseudohypoparathyroidism (300800).
- Butler et al. (1988) reported an affected 13-year-old boy and reviewed
- the literature. They emphasized the features of nasal and maxillary
- hypoplasia, peripheral dysostosis, decreased interpedicular distance,
- advanced skeletal maturation, and mental retardation. In their review
- they also found that parental age was increased. They suggested that the
- metacarpophalangeal pattern profile is characteristically abnormal and
- that this can be a useful diagnostic tool. The first ray in the foot may
- be relatively hyperplastic. Viljoen and Beighton (1991) reviewed the
- radiologic features in 12 affected children and found that epiphyseal
- stippling is a consistent and prominent characteristic during infancy.
- Butler et al. (1988) found a pattern of autosomal dominant inheritance
- in 2 families (Niikawa et al., 1978; Frey et al., 1982). Niikawa et al.
- (1978) described Japanese brother and sister, aged 7 months and 2 years,
- respectively, with severe nasal hypoplasia, peripheral dysostosis, blue
- eyes, and mental retardation. The mother showed nasal hypoplasia and
- irregular shortening of fingers and toes. Hernandez et al. (1991)
- described an affected mother and daughter. Steiner and Pagon (1992) also
- described an affected mother and daughter. The mother had been diagnosed
- at the age of 4 years and was pictured in the 1982 edition of Smith's
- Recognizable Patterns of Human Malformation. At the age of 20, she
- suffered from recurrent carpal tunnel syndrome. The daughter showed
- cone-shaped epiphyses as in the mother.
-
- Because of the similarity between acrodysostosis and Albright hereditary
- osteodystrophy (AHO; 103580), both of which show shortening of the
- tubular bones of the hands and feet with cone-shaped epiphyses, Wilson
- et al. (1997) looked for abnormalities in the alpha subunit of the
- signal transducing protein, Gs, and in the GNAS1 gene (139320). In 2
- unrelated patients with acrodysostosis, they found that Gs-alpha
- bioactivity in erythrocyte membranes was normal. Mutation analysis of
- the GNAS1 gene showed no sequence variation in 12 of the 13 exons
- examined. The results were interpreted as indicating that, at least in a
- proportion of patients with acrodysostosis, the condition is
- etiologically distinct from AHO.
-
- *FIELD* SA
- Arkless and Graham (1967); Smith (1982)
- *FIELD* RF
- 1. Arkless, R.; Graham, C. B.: An unusual case of brachydactyly. Am.
- J. Roentgen. 99: 724-735, 1967.
-
- 2. Butler, M. G.; Rames, L. J.; Wadlington, W. B.: Acrodysostosis:
- report of a 13-year-old boy with review of literature and metacarpophalangeal
- pattern profile analysis. Am. J. Med. Genet. 30: 971-980, 1988.
-
- 3. Frey, V. G.; Martin, J.; Diefel, K.: Die Akrodysostose--eine autosomal-dominant
- verebte periphere Dysplasie. Kinderarztl. Prax. 3: 149-153, 1982.
-
- 4. Hernandez, R. M.; Miranda, A.; Kofman-Alfaro, S.: Acrodysostosis
- in two generations: an autosomal dominant syndrome. Clin. Genet. 39:
- 376-382, 1991.
-
- 5. Jones, K. L.; Smith, D. W.; Harvey, M. A. S.; Hall, B. D.; Quan,
- L.: Older paternal age and fresh gene mutation: data on additional
- disorders. J. Pediat. 86: 84-88, 1975.
-
- 6. Maroteaux, P.; Malamut, G.: L'acrodysostose. Presse Med. 76:
- 2189-2192, 1968.
-
- 7. Niikawa, N.; Matsuda, I.; Ohsawa, T.; Kajii, T.: Familial occurrence
- of a syndrome with mental retardation, nasal hypoplasia, peripheral
- dysostosis, and blue eyes in Japanese siblings. Hum. Genet. 42:
- 227-232, 1978.
-
- 8. Robinow, M.; Pfeiffer, R. A.; Gorlin, R. J.; McKusick, V. A.; Renuart,
- A. W.; Johnson, G. F.; Summitt, R. L.: Acrodysostosis: a syndrome
- of peripheral dysostosis, nasal hypoplasia, and mental retardation. Am.
- J. Dis. Child. 121: 195-203, 1971.
-
- 9. Smith, D. W.: Recognizable Patterns of Human Malformation: Genetic,
- Embryologic and Clinical Aspects. Philadelphia: W. B. Saunders (pub.)
- (3rd ed.): 1982. Pp. 322-323.
-
- 10. Steiner, R. D.; Pagon, R. A.: Autosomal dominant transmission
- of acrodysostosis. Clin. Dysmorph. 1: 201-206, 1992.
-
- 11. Viljoen, D.; Beighton, P.: Epiphyseal stippling in acrodysostosis. Am.
- J. Med. Genet. 38: 43-45, 1991.
-
- 12. Wilson, L. C.; Oude Luttikhuis, M. E. M.; Baraitser, M.; Kingston,
- H. M.; Trembath, R. C.: Normal erythrocyte membrane Gs-alpha bioactivity
- in two unrelated patients with acrodysostosis. J. Med. Genet. 34:
- 133-136, 1997.
-
- *FIELD* CS
-
- Facies:
- Short nose;
- Nasal hypoplasia;
- Open mouth;
- Maxillary hypoplasia;
- Prognathism
-
- Limbs:
- Small hands and feet
-
- Neuro:
- Mental retardation
-
- Misc:
- Increased average paternal age
-
- Radiology:
- Cone epiphyses;
- Peripheral dysostosis;
- Decreased interpedicular distance;
- Advanced skeletal maturation;
- Abnormal metacarpophalangeal pattern profile;
- Hyperplastic foot first ray;
- Epiphyseal stippling
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CN
- Victor A. McKusick - updated: 03/06/1997
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 03/06/1997
- terry: 3/5/1997
- davew: 8/1/1994
- mimadm: 3/11/1994
- carol: 12/6/1993
- carol: 11/11/1993
- supermim: 3/16/1992
- carol: 5/29/1991
-
- *RECORD*
- *FIELD* NO
- 101805
- *FIELD* TI
- 101805 ACROFACIAL DYSOSTOSIS, CATANIA TYPE
- AFD, CATANIA TYPE
- *FIELD* TX
- Opitz et al. (1993) reported a 'new' form of acrofacial dysostosis in a
- Sicilian woman and her 4 sons. Features included apparent mild
- intrauterine growth retardation and postnatal shortness of stature,
- microcephaly, widow's peak, mandibulofacial dysostosis without cleft
- palate, mild pre- and more conspicuous postaxial upper limb involvement
- with short hands, simian creases, mild interdigital webbing, low total
- ridge count, and facultative preauricular fistulae, cryptorchidism,
- hypospadias, inguinal hernia, and spina bifida occulta of C1. Although
- X-linked dominant inheritance was possible, the authors considered
- autosomal dominant inheritance more likely because the mother was as
- severely affected as her sons. Wulfsberg et al. (1996) described a
- similar association in a 5-year-old girl and her mother. In addition to
- typical manifestations of the syndrome, the mother had an edentulous
- upper jaw and carious teeth in both lower and upper jaw.
-
- *FIELD* RF
- 1. Opitz, J. M.; Mollica, F.; Sorge, G.; Milana, G.; Cimino, G.; Caltabiano,
- M.: Acrofacial dysostoses: review and report of a previously undescribed
- condition: the autosomal or X-linked dominant Catania form of acrofacial
- dysostosis. Am. J. Med. Genet. 47: 660-678, 1993.
-
- 2. Wulfsberg, E. A.; Campbell, A. B.; Lurie, I. W.; Eanet, K. R.:
- Confirmation of the Catania brachydactylous type of acrofacial dysostosis:
- report of a second family. Am. J. Med. Genet. 63: 554-557, 1996.
-
- *FIELD* CN
- Iosif W. Lurie - updated: 08/11/1996
-
- *FIELD* CD
- Victor A. McKusick: 11/4/1993
-
- *FIELD* ED
- carol: 08/11/1996
- carol: 11/4/1993
-
- *RECORD*
- *FIELD* NO
- 101840
- *FIELD* TI
- 101840 ACROKERATODERMA, HEREDITARY PAPULOTRANSLUCENT
- *FIELD* TX
- Onwukwe et al. (1973) described a family in which multiple members of 4
- generations and by inference a fifth, in a pattern consistent with
- autosomal dominant inheritance (including male-to-male transmission),
- had persistent, asymptomatic, yellowish-white, translucent papules and
- plaques on the hands and feet, associated with fine-textured scalp hair
- and atopic diathesis. Histologic study of the translucent lesions showed
- orthohypergranulosis, acanthosis, and a relatively normal dermis.
- Onwukwe et al. (1973) suggested that this might be a new variant of
- familial punctate keratoderma. De Wit and Hulsmans (1986) observed a
- Surinam woman with abnormalities of palmar and plantar skin. Her father
- was reported to have similar changes confined to the feet. The index
- patient was observed to have both classical keratosis punctata palmaris
- et plantaris (175860) and papulotranslucent acrokeratoderma.
-
- *FIELD* RF
- 1. de Wit, F. S.; Hulsmans, R. F. H. J.: Hereditair papulotranslucent
- keratoderma van de acra als variant van en in combinatie met keratosis
- punctata palmaris et plantaris. Nederl. T. Geneesk. 130: 2015 only,
- 1986.
-
- 2. Onwukwe, M. F.; Mihm, M. C., Jr.; Toda, K.: Hereditary papulotranslucent
- acrokeratoderma: a new variant of familial punctate keratoderma?.
- Arch. Derm. 108: 108-110, 1973.
-
- *FIELD* CS
-
- Skin:
- Persistent, asymptomatic, yellowish-white, translucent papules and
- plaques of hands and feet
-
- Hair:
- Fine-textured scalp hair
-
- Immunology:
- Atopic diathesis
-
- Lab:
- Skin lesions show orthohypergranulosis, acanthosis, and a relatively
- normal dermis
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 4/1/1991
-
- *FIELD* ED
- mimadm: 3/11/1994
- carol: 3/31/1992
- supermim: 3/16/1992
- carol: 4/5/1991
- carol: 4/1/1991
-
- *RECORD*
- *FIELD* NO
- 101850
- *FIELD* TI
- *101850 ACROKERATOELASTOIDOSIS; AKE
- COLLAGENOUS PLAQUES OF HANDS
- *FIELD* TX
- This disorder was first described and named by Costa (1953). Jung (1973)
- studied an extensively affected family. The palms and soles are
- primarily affected, but involvement may extend to the dorsum of the
- hands and feet in severe cases. The lesions are nodular and yellow with
- hyperkeratotic surfaces. The histology combines hyperkeratosis and
- disorganization of elastic fibers. No systemic manifestation has been
- detected. The differential diagnosis includes other forms of
- palmoplantar keratosis and palmoplantar xanthomata. Matthews and Harman
- (1977) observed the disorder in 2 brothers whose mother was also
- affected. In a linkage study of the large kindred reported by Jung
- (1973), Greiner et al. (1983) found a suggestion of linkage of AKE to
- ACP1 (171500), Jk (111000) and IGKC (147200). Although the lod scores
- did not reach the level of significance considered to be proof, the fact
- that all three of these markers are on 2p suggests that AKE may be there
- also. Maximum lod scores were as follows: with IGKC, 0.57 at theta 0.16;
- with ACP1, 0.18 at theta 0.22; with Jk, 0.11 at theta 0.31.
-
- Stevens et al. (1996) classified focal acrohyperkeratosis, otherwise
- known as acrokeratoelastoidosis, as type III punctate PPK.
-
- *FIELD* SA
- Costa (1954); Matthews and Harman (1974)
- *FIELD* RF
- 1. Costa, O. G.: Acrokeratoelastoidosis: a hitherto undescribed skin
- disease. Dermatologica 107: 164-167, 1953.
-
- 2. Costa, O. G.: Ackrokeratoelastoidosis. Arch. Derm. Syph. 70:
- 228-231, 1954.
-
- 3. Greiner, J.; Kruger, J.; Palden, L.; Jung, E. G.; Vogel, F.: A
- linkage study of acrokeratoelastoidosis: possible mapping to chromosome
- 2. Hum. Genet. 63: 222-227, 1983.
-
- 4. Jung, E. G.: Acrokeratoelastoidosis. Humangenetik 17: 357-358,
- 1973.
-
- 5. Matthews, C. N. A.; Harman, R. R. M.: Acrokerato-elastoidosis
- (without elastorrhexis). Proc. Roy. Soc. Med. 67: 1237-1238, 1974.
- Derm. 132: 640-651, 1996.
-
- 6. Matthews, C. N. A.; Harman, R. R. M.: Acrokerato-elastoidosis
- in a Somerset mother and her two sons. Brit. J. Derm. 97 (suppl.
- 15): 42-43, 1977.
-
- 7. Stevens, H. P.; Kelsell, D. P.; Bryant, S. P.; Bishop, D. T.; Spurr,
- N. K.; Weissenbach, J.; Marger, D.; Marger, R. S.; Leigh, I. M.:
- Linkage of an American pedigree with palmoplantar keratoderma and
- malignancy (palmoplantar ectodermal dysplasia type III) to 17q24:
- literature survey and proposed updated classification of the keratodermas. Arch.
- Derm. 132: 640-651, 1996.
-
- *FIELD* CS
-
- Skin:
- Acrokeratoelastoidosis;
- Hyperkeratosis;
- Acrokeratosis
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 12/03/1996
- terry: 11/8/1996
- mimadm: 3/11/1994
- carol: 8/25/1992
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- carol: 4/20/1988
-
- *RECORD*
- *FIELD* NO
- 101900
- *FIELD* TI
- *101900 ACROKERATOSIS VERRUCIFORMIS
- HOPF DISEASE
- *FIELD* TX
- Warty hyperkeratotic lesions are found on the dorsal aspect of the hands
- and feet and on the knees and elbows. The pedigree studied by Niedelman
- and McKusick (1962) contained instances of male-to-male transmission as
- well as unaffected daughters of affected males. Herndon and Wilson
- (1966) emphasized the phenotypic overlap between this entity and
- Darier-White disease (124200) and even proposed that they may not be
- separate entities. In the family they studied, 7 persons had typical
- acrokeratosis verruciformis, 1 or possibly 2 had Darier disease, and 3
- had minor disturbances of keratinization (white nails from subungual
- hyperkeratosis, or punctate keratoses of palms or soles). Also see
- benign familial pemphigus (169600).
-
- *FIELD* RF
- 1. Herndon, J. H., Jr.; Wilson, J. D.: Acrokeratosis verruciformis
- (Hopf) and Darier's disease: genetic evidence for a unitary origin.
- Arch. Derm. 93: 305-310, 1966.
-
- 2. Niedelman, M. L.; McKusick, V. A.: Acrokeratosis verruciformis
- (Hopf): a follow-up study. Arch. Derm. 86: 779-782, 1962.
-
- *FIELD* CS
-
- Skin:
- Acrokeratosis;
- Warty hyperkeratosis, dorsal hands, feet, knees and elbows;
- Acrokeratosis verruciformis
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 4/19/1995
- pfoster: 9/2/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
-
- *RECORD*
- *FIELD* NO
- 102000
- *FIELD* TI
- 102000 ACROLEUKOPATHY, SYMMETRIC
- *FIELD* TX
- Sugai et al. (1965) described mother and daughter with symmetric
- depigmentation of the great toes.
-
- *FIELD* RF
- 1. Sugai, T.; Saito, T.; Hamada, T.: Symmetric acroleukopathy in
- mother and daughter. Arch. Derm. 92: 172-173, 1965.
-
- *FIELD* CS
-
- Skin:
- Symmetric great toe depigmentation
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 102100
- *FIELD* TI
- *102100 ACROMEGALOID CHANGES, CUTIS VERTICIS GYRATA, AND CORNEAL LEUKOMA
- ROSENTHAL-KLOEPFER SYNDROME
- *FIELD* TX
- Rosenthal and Kloepfer (1962) described a 'new' syndrome with these
- three features in 13 persons of 4 generations of a Louisiana black
- family. Through the courtesy of Kloepfer, I saw affected members of this
- family in 1971. The corneal leukoma is an epithelial change. The hands,
- feet and chin are very large and the affected persons unusually tall.
- Although growth hormone assays had not been done, other endocrine
- studies and x-ray views of the sella turcica gave no indication of
- pituitary dysfunction. One of the affected females examined had 9 living
- children. The skin of the hands is unusually soft and has an abnormal
- dermal ridge pattern, referred to as 'split ridges,' which permits
- identification of the disorder in children of preclinical age. A
- possible difference from the usual cutis verticis gyrata is a
- longitudinal orientation of the skin folds rather than transverse
- orientation. X-ray features were reported by Harbison and Nice (1971).
-
- *FIELD* RF
- 1. Harbison, J. B.; Nice, C. M., Jr.: Familial pachydermoperiostosis
- presenting as an acromegaly-like syndrome. Am. J. Roentgen. 112:
- 532-536, 1971.
-
- 2. Rosenthal, J. W.; Kloepfer, H. W.: An acromegaloid, cutis verticis
- gyrata, corneal leukoma syndrome. Arch. Ophthal. 68: 722-726, 1962.
-
- *FIELD* CS
-
- Eyes:
- Corneal leukoma
-
- Limbs:
- Large hands and feet
-
- Facies:
- Large chin
-
- Growth:
- Tall stature
-
- Skin:
- Soft skin;
- Split ridge dermal ridge pattern;
- Cutis verticis gyrata with longitudinal folding
-
- Radiology:
- Periostosis
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- warfield: 4/6/1994
- mimadm: 3/11/1994
- carol: 3/24/1992
- supermim: 3/16/1992
- supermim: 5/15/1990
- supermim: 3/20/1990
-
- *RECORD*
- *FIELD* NO
- 102150
- *FIELD* TI
- *102150 ACROMEGALOID FACIAL APPEARANCE SYNDROME
- AFA SYNDROME;;
- THICK LIPS AND ORAL MUCOSA
- *FIELD* TX
- In many members of a kindred through at least 5 generations, Hughes et
- al. (1985) described a syndrome of acromegaloid facial features:
- thickened lips (without a true 'double lip'), overgrowth of the
- intraoral mucosa resulting in exaggerated rugae and frenula, and
- thickened upper eyelids leading to narrow palpebral fissures
- (blepharophimosis). The nose tended to be bulbous. The hands were large
- and doughy without clubbing. Highly arched eyebrows were striking in
- published photographs. There was no evident impairment of general
- health. Pachydermoperiostosis (167100), Ascher syndrome (109900), and
- multiple neuroma syndrome (162300) were considered in the differential
- diagnosis. Low positive lod scores were obtained for linkage between AFA
- and Rh and PGM1 (on 1p), GLO (on 6p), IGHG and PI (on 14q), and HP (on
- 16q). Dallapiccola et al. (1992) reported a family with the disorder in
- 2 generations. Five affected persons, a mother and 4 children, showed a
- striking resemblance to the patients reported by Hughes et al. (1985).
- They had progressively coarsening acromegaloid facial appearance, narrow
- palpebral fissures, bulbous nose, and thickening of the lips and
- intraoral mucosa, resulting in exaggerated rugae of the tongue and
- frenula. The patients had increased birth weight and dull mentality.
- Tapering fingers in the mother and one daughter, somewhat like those in
- the Coffin-Lowry syndrome (303600), were pictured.
-
- *FIELD* RF
- 1. Dallapiccola, B.; Zelante, L.; Accadia, L.; Mingarelli, R.: Acromegaloid
- facial appearance (AFA) syndrome: report of a second family. J.
- Med. Genet. 29: 419-422, 1992.
-
- 2. Hughes, H. E.; McAlpine, P. J.; Cox, D. W.; Philipps, S.: An autosomal
- dominant syndrome with 'acromegaloid' features and thickened oral
- mucosa. J. Med. Genet. 22: 119-125, 1985.
-
- *FIELD* CS
-
- Mouth:
- Thickened lips;
- Intraoral mucosal overgrowth;
- Exaggerated oral rugae and frenula
-
- Eyes:
- Thickened upper eyelids;
- Blepharophimosis;
- Highly arched eyebrows
-
- Nose:
- Bulbous nose
-
- Limbs:
- Large doughy hands;
- Tapering fingers
-
- Growth:
- Increased birth weight
-
- Neuro:
- Dull mentality
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- carol: 7/1/1992
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 102200
- *FIELD* TI
- *102200 ACROMEGALY
- SOMATOTROPHINOMA, INCLUDED
- *FIELD* TX
- Koch and Tiwisina (1959) reviewed 8 examples of affected persons in 2
- successive generations, including 4 instances of father and 1 or more
- sons affected. Some reported instances of familial acromegaly may in
- fact be pachydermoperiostosis (167100), acromegaloid-cutis verticis
- gyrata-leukoma syndrome (102100), or cerebral gigantism (117550).
- Furthermore, familial acromegaly can be a partial expression of the
- multiple endocrine adenomatosis syndrome, specifically multiple
- endocrine neoplasia type I (MEN1; 131100). Levin et al. (1974) reported
- the cases of 2 brothers with acromegaly confirmed by elevated growth
- hormone levels. Both had acanthosis nigricans and pituitary tumors.
- Pestell et al. (1989) described a family in which 5 members over 3
- generations had isolated functional pituitary adenomas. In 4 cases this
- was associated with acromegaly, and in the fifth galactorrhea from
- prolactin excess was the presenting feature. The tumors were
- histologically of either atypical mixed cell or undifferentiated cell
- type. No parent-child transmission was observed. The 5 individuals were
- related as uncle and nephew or uncle and niece or as second cousins.
- There were no consanguineous marriages in the family. Autosomal dominant
- inheritance with reduced penetrance was proposed. Pestell et al. (1989)
- considered the disorder in this family to be distinct from MEN1. Jones
- et al. (1984), Abbassioun et al. (1986), and McCarthy et al. (1990) also
- reported cases of familial acromegaly.
-
- Growth hormone secreting pituitary adenomas (somatotrophinomas) occur in
- families either as an isolated autosomal dominant endocrinopathy (as
- illustrated by the examples cited above) or as part of MEN1. Thakker et
- al. (1993) compared DNA in somatotrophinomas and peripheral leukocytes
- obtained from 13 patients with acromegaly; one patient also suffered
- from MEN1. Five DNA probes identifying RFLPs from 11q demonstrated
- allele loss in pituitary tumors from 5 patients, 4 non-MEN1 and 1 MEN1.
- Deletion mapping revealed that the region of allele loss common to the
- somatotrophinomas involved 11q13. Similar allelic deletions at 12 other
- loci distributed through the genome did not reveal generalized allele
- loss in the somatotrophinomas. Thakker et al. (1993) interpreted these
- results as indicating that a recessive oncogene on 11q13 is specifically
- involved in the monoclonal development of somatotrophinomas; 11q13 is
- also the site of the gene for MEN1 in which somatotrophinomas are a
- feature. (It is a well known phenomenon that tumors that occur as a
- component of a familial neoplasia syndrome also occur as sporadic tumors
- on the basis of somatic mutation. Is it not possible that the findings
- of Thakker et al. (1993) have the same basis as sporadic meningioma due
- to mutation in the NF2 gene (e.g., 101000.0003), cerebellar
- hemangioblastoma, sporadic cerebellar hemangioblastoma, or sporadic
- renal carcinoma due to mutation in the gene for von Hippel-Lindau
- syndrome (e.g., 193300.0002 and 193300.0007, respectively)? VAM.)
-
- In addition, Thakker et al. (1993) found mutations in the GNAS1 gene
- (139320) in 2 non-MEN1 somatotrophinomas, one of which also demonstrated
- allele loss of chromosome 11. (The authors referred to GNAS1 as GSP.)
-
- *FIELD* SA
- Koch (1949)
- *FIELD* RF
- 1. Abbassioun, K.; Fatourehchi, V.; Amirjamshidi, A.; Meibodi, N.
- A.: Familial acromegaly with pituitary adenoma: report of three affected
- siblings. J. Neurosurg. 64: 510-512, 1986.
-
- 2. Jones, M. K.; Evans, P. J.; Jones, I. R.; Thomas, J. P.: Familial
- acromegaly. Clin. Endocr. 20: 355-358, 1984.
-
- 3. Koch, G.: Erbliche Hirngeschwuelste. Z. Menschl. Vererb. Konstitutionsl. 29:
- 400-423, 1949.
-
- 4. Koch, G.; Tiwisina, T.: Beitrag zur Erblichkeit der Akromegalie
- und der Hyperostosis generalisata mit Pachydermie. Aerztl. Forsch. 13:
- 489-504, 1959.
-
- 5. Levin, S. R.; Hafeldt, F. D.; Becker, N.; Wilson, C. B.; Seymour,
- R.; Forsham, P. H.: Hypersomatotropism and acanthosis nigricans in
- two brothers. Arch. Intern. Med. 134: 365-367, 1974.
-
- 6. McCarthy, M. I.; Noonan, K.; Wass, J. A. H.; Monson, J. P.: Familial
- acromegaly: studies in three families. Clin. Endocr. 32: 719-728,
- 1990.
-
- 7. Pestell, R. G.; Alford, F. P.; Best, J. D.: Familial acromegaly. Acta
- Endocr. 121: 286-289, 1989.
-
- 8. Thakker, R. V.; Pook, M. A.; Wooding, C.; Boscaro, M.; Scanarini,
- M.; Clayton, R. N.: Association of somatotrophinomas with loss of
- alleles on chromosome 11 and with gsp mutations. J. Clin. Invest. 91:
- 2815-2821, 1993.
-
- *FIELD* CS
-
- Endocrine:
- Acromegaly;
- Functional pituitary adenoma
-
- Lab:
- Elevated growth hormone levels
-
- Skin:
- Acanthosis nigricans;
- Galactorrhea from prolactin excess
-
- Oncology:
- Somatotrophinoma
-
- Inheritance:
- Autosomal dominant;
- recessive gene loss at 11q13 for somatotrophinoma
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 03/13/1997
- terry: 2/5/1997
- mark: 12/30/1996
- mark: 12/26/1996
- terry: 12/16/1996
- mark: 9/22/1995
- mimadm: 3/11/1994
- carol: 7/9/1993
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
-
- *RECORD*
- *FIELD* NO
- 102300
- *FIELD* TI
- *102300 ACROMELALGIA, HEREDITARY
- RESTLESS LEGS
- *FIELD* TX
- Because of paresthesia when first going to bed or sitting still for a
- time, the affected person cannot resist fidgeting with his or her feet.
- Huizinga (1957) described a family with affected persons in 5
- generations. The condition, which began in adolescence, was relieved by
- cold. Ekbom (1960) and Bornstein (1961) also described familial
- aggregation. Autosomal dominant inheritance was particularly well
- documented by Boghen and Peyronnard (1976), who furthermore described
- myoclonic jerks in 10 of 18 affected persons. The jerks occurred at
- night before sleep and severely interfered with it. The authors referred
- to the 'painful-legs--moving-toes syndrome' in a patient whose relatives
- had the restless legs syndrome and proposed that the disorders are the
- same. Sudden bodily jerking on falling asleep is a frequent finding in
- normal persons (Oswald, 1959).
-
- Trenkwalder et al. (1996) found evidence of anticipation in restless
- legs syndrome in 1 large German pedigree. The disorder had a 30-year
- age-at-onset difference between generations.
-
- *FIELD* RF
- 1. Boghen, D.; Peyronnard, J.-M.: Myoclonus in familial restless
- legs syndrome. Arch. Neurol. 33: 368-370, 1976.
-
- 2. Bornstein, B.: Restless legs. Psychiat. Neurol. 141: 165-201,
- 1961.
-
- 3. Ekbom, K. A.: Restless legs syndrome. Neurology 10: 868-873,
- 1960.
-
- 4. Huizinga, J.: Hereditary acromelalgia (or 'restless legs'). Acta
- Genet. Statist. Med. 7: 121-123, 1957.
-
- 5. Oswald, I.: Sudden bodily jerks on falling asleep. Brain 82:
- 92-103, 1959.
-
- 6. Trenkwalder, C.; Seidel, V. C.; Gasser, T.; Oertel, W. H.: Clinical
- symptoms and possible anticipation in a large kindred with familial
- restless legs syndrome. Mov. Disord. 11: 389-394, 1996.
-
- *FIELD* CS
-
- Neuro:
- Acromelalgia;
- Myoclonus;
- Paresthesia;
- Restless legs
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 12/29/1996
- terry: 12/20/1996
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
-
- *RECORD*
- *FIELD* NO
- 102350
- *FIELD* TI
- *102350 ACROMIAL DIMPLES
- SUPRASPINOUS FOSSAE, CONGENITAL
- *FIELD* TX
- Dimples overlying the acromial process of the scapula, i.e., on the back
- of the shoulders, is a regular feature of the 18q- syndrome. Bianchine
- (1974) described acromial dimples in a 4-year-old girl, her 30-year-old
- mother, and her 65-year-old maternal grandmother. All 3 were generally
- healthy. Gorlin (1974) told me of acromial dimples transmitted through 4
- and probably 5 generations. Halal (1980) observed segregation in 2
- kindreds but found no instance of male-to-male transmission. Mehes and
- Meggyessy (1987) described acromial dimples in a 1-year-old boy and his
- healthy 29-year-old father. In another family, a 3-year-old girl, her
- 31-year-old mother, and her 6-year-old brother had bilateral acromial
- dimples. Wood (1990) and Samlaska (1991) described inherited symmetric
- shoulder dimpling over the acromial process, which they referred to as
- congenital supraspinous fossae. The familial pattern was consistent with
- autosomal dominant inheritance. Acromial dimples occur as a virtually
- constant feature of 18q deletion (Insley, 1967).
-
- *FIELD* RF
- 1. Bianchine, J. W.: Acromial dimples: a benign familial trait. Am.
- J. Hum. Genet. 26: 412-413, 1974.
-
- 2. Gorlin, R. J.: Personal Communication. Minneapolis, Minn. 6/10/1974.
-
- 3. Halal, F.: Dominant inheritance of acromial skin dimples. Am.
- J. Med. Genet. 6: 259-262, 1980.
-
- 4. Insley, J.: Syndrome associated with a deficiency of part of the
- long arm of chromosome no. 18. Arch. Dis. Child. 42: 140-146, 1967.
-
- 5. Mehes, K.; Meggyessy, V.: Autosomal dominant inheritance of benign
- bilateral acromial dimples. Hum. Genet. 76: 206 only, 1987.
-
- 6. Samlaska, C. P.: Congenital supraspinous fossae. J. Am. Acad.
- Derm. 25: 1078-1079, 1991.
-
- 7. Wood, V. E.: Congenital skin fossae about the shoulder. Plast.
- Reconst. Surg. 85: 798-800, 1990.
-
- *FIELD* CS
-
- Skin:
- Acromial dimples
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- warfield: 4/7/1994
- mimadm: 3/11/1994
- carol: 11/20/1992
- carol: 3/31/1992
- supermim: 3/16/1992
- carol: 1/21/1992
-
- *RECORD*
- *FIELD* NO
- 102370
- *FIELD* TI
- 102370 ACROMICRIC DYSPLASIA
- *FIELD* TX
- Maroteaux et al. (1986) described (and named) a 'new' entity on the
- basis of 6 patients. Features were mild facial anomalies, markedly
- shortened hands and feet, and growth retardation that was severe in
- most. The metacarpals and phalanges were short and stubby; the proximal
- portion of the second metacarpal showed a notch on its radial side and
- the fifth metacarpal had a notch on its ulnar side. Similar histologic
- changes were found in biopsy of the proximal tibial growth cartilage in
- 2 cases: disorganization of the growth zone with islands of cells and
- abnormal arrangement of collagen. Both sexes were affected. All 6 cases
- were sporadic (with normal parental age and no parental consanguinity).
- In an addendum, Maroteaux et al. (1986) stated that they had observed
- acromicric dysplasia in mother and son.
-
- *FIELD* RF
- 1. Maroteaux, P.; Stanescu, R.; Stanescu, V.; Rappaport, R.: Acromicric
- dysplasia. Am. J. Med. Genet. 24: 447-459, 1986.
-
- *FIELD* CS
-
- Facies:
- Mild facial anomalies
-
- Limbs:
- Short hands and feet
-
- Growth:
- Severe growth retardation
-
- Radiology:
- Short stubby metacarpals and phalanges;
- Second metacarpal notched proximally on radial side;
- Fifth metacarpal notched on ulnar side
-
- Lab:
- Growth cartilage disorganized, with islands of cells and abnormal
- collagen arrangement
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 10/16/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 10/16/1986
-
- *RECORD*
- *FIELD* NO
- 102400
- *FIELD* TI
- 102400 ACROOSTEOLYSIS
- *FIELD* TX
- Schinz et al. (1951) described dominant inheritance of slowly
- progressive osteolysis of the phalanges in the hands and feet associated
- with recurrent ulcers of the fingers and soles, elimination of bone
- sequestra, and healing with loss of toes or fingers, with onset between
- 8 and 22 years. Lamy and Maroteaux (1961) described a dominant form in
- mother and son. Members of 2 earlier generations were also affected. No
- abnormality of sensation was present. Maroteaux (1970) found no basilar
- impression or other changes in the skull or long bones to suggest that
- this was Cheney syndrome (102500). A phenocopy is produced in men
- working in the polymerization of vinyl chloride (Harris and Adams, 1967;
- Ross, 1970). Reed (1974) told me of other families.
-
- *FIELD* SA
- Harms (1954)
- *FIELD* RF
- 1. Harms, I.: Ueber die familiaere Akro-osteolyse. Fortschr. Roentgenstr. 80:
- 727-733, 1954.
-
- 2. Harris, D. K.; Adams, W. G. F.: Acro-osteolysis occurring in men
- engaged in the polymerization of vinyl chloride. Brit. Med. J. 3:
- 712-714, 1967.
-
- 3. Lamy, M.; Maroteaux, P.: Acro-osteolyse dominante. Arch. Franc.
- Pediat. 18: 693-702, 1961.
-
- 4. Maroteaux, P.: Personal Communication. Paris, France 1970.
-
- 5. Reed, W. B.: Personal Communication. Burbank, Calif. 1974.
-
- 6. Ross, J. A.: An unusual occupational bone change. In: Jelliffe,
- A. M.; Strickland, B.: Symposium Ossium. London: Livingstone (pub.)
- 1970.
-
- 7. Schinz, H. R.; Baensch, W. E.; Friedl, E.; Uehlinger, E.: Roentgen-diagnostics.
- Trans. in English by J. T. Case. New York: Grune and Stratton (pub.)
- 1: 1951. Pp. 734 only. Note: Fig. 969.
-
- *FIELD* CS
-
- Limbs:
- Osteolysis of phalanges;
- Recurrent ulcers, fingers and soles;
- Bone sequestra;
- Loss of toes or fingers
-
- Misc:
- Onset 8 to 22 years;
- Phenocopy in vinyl chloride workers
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/28/1994
- pfoster: 3/25/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 102480
- *FIELD* TI
- *102480 ACROSIN; ACR
- PROACROSIN, INCLUDED;;
- PREPROACROSIN, INCLUDED
- *FIELD* TX
- Acrosin (EC 3.4.21.10) is the major proteinase present in the acrosome
- of mature spermatozoa. It is a typical serine proteinase with
- trypsin-like specificity. It is stored in the acrosome in its precursor
- form, proacrosin. The active enzyme functions in the lysis of the zona
- pellucida, thus facilitating penetration of the sperm through the
- innermost glycoprotein layers of the ovum. In many species, it is shown
- that biosynthesis of acrosin is confined to the haploid phase of
- spermatogenesis. By indirect immunofluorescent techniques,
- Florke-Gerloff et al. (1983) demonstrated that in man (pro)acrosin first
- appears in the haploid spermatids. Adham et al. (1989, 1990) isolated a
- full-length cDNA clone for human proacrosin. The deduced amino acid
- sequence of human proacrosin in the proline-rich domain is different
- from the corresponding sequence of boar proacrosin. This domain may be
- involved in a species-specific binding of spermatozoa to the zona
- pellucida. The mRNA for proacrosin is synthesized only in the
- postmeiotic stages of spermatogenesis. The cDNA sequence indicates that
- acrosin is synthesized as a preproacrosin. Adham et al. (1989) used
- somatic cell hybrid analysis to localize the human proacrosin gene to
- chromosome 22q13-qter. By in situ hybridization, Engel (1990) assigned
- the acrosin gene to mouse chromosome 15 and rat chromosome 7; see
- Kremling et al. (1991). Furthermore, by an immunohistologic method,
- Engel (1990) demonstrated deficiency of acrosin in spermatids of
- infertile males. Keime et al. (1990) used cDNA clones as probes to
- isolate the gene for proacrosin from a human leukocyte genomic library.
- They found that the gene contains 4 introns varying in length from 0.2
- to 4.5 kb. Klemm et al. (1991) provided a review. Vazquez-Levin et al.
- (1992) reported on the sequence and structure of the proacrosin gene and
- pointed to differences from previously reported data. Adham et al.
- (1992) defended the validity of the previous data.
-
- *FIELD* SA
- Adham et al. (1989); Adham et al. (1989)
- *FIELD* RF
- 1. Adham, I. M.; Grzeschik, K.-H.; Geurts van Kessel, A. H. M.; Engel,
- W.: Localization of human preproacrosin to chromosome 22q13-qter
- by somatic cell hybrid analysis. (Abstract) Cytogenet. Cell Genet. 51:
- 948 only, 1989.
-
- 2. Adham, I. M.; Grzeschik, K.-H.; Geurts van Kessel, A. H. M.; Engel,
- W.: The gene encoding the human preproacrosin (ACR) maps to the q13-qter
- region on chromosome 22. Hum. Genet. 84: 59-62, 1989.
-
- 3. Adham, I. M.; Klemm, U.; Maier, W.-M.; Engel, W.: Molecular cloning
- of human preproacrosin cDNA. Hum. Genet. 84: 125-128, 1990.
-
- 4. Adham, I. M.; Klemm, U.; Maier, W.-M.; Tsaousidou, S.; Engel, W.
- : Molecular cloning and expression of boar and human proacrosin cDNA.
- (Abstract) Meeting of Gesellschaft fuer Humangenetik, Munich 149
- only, 4/4/1989.
-
- 5. Adham, I. M.; Spitzer, U.; Schlosser, M.; Kremling, H.; Keime,
- S.; Engel, W.: A reply: the human proacrosin gene. Europ. J. Biochem. 207:
- 27-28, 1992.
-
- 6. Engel, W.: Personal Communication. Goettingen, Germany 5/17/1990.
-
- 7. Florke-Gerloff, S.; Topfer-Petersen, E.; Muller-Esterl, W.; Schill,
- W.-B.; Engel, W.: Acrosin and the acrosome in human spermatogenesis.
- Hum. Genet. 65: 61-67, 1983.
-
- 8. Keime, S.; Adham, I. M.; Engel, W.: Nucleotide sequence and exon-intron
- organization of the human proacrosin gene. Europ. J. Biochem. 190:
- 195-200, 1990.
-
- 9. Klemm, U.; Muller-Esterl, W.; Engel, W.: Acrosin, the peculiar
- sperm-specific serine protease. Hum. Genet. 87: 635-641, 1991.
-
- 10. Kremling, H.; Keime, S.; Wilhelm, K.; Adham, I. M.; Hameister,
- H.; Engel, W.: Mouse proacrosin gene: nucleotide sequence, diploid
- expression and chromosomal localization. Genomics 11: 828-834,
- 1991.
-
- 11. Vazquez-Levin, M. H.; Reventos, J.; Gordon, J. W.: Molecular
- cloning, sequencing and restriction mapping of the genomic sequence
- encoding human proacrosin. Europ. J. Biochem. 207: 23-26, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 5/5/1989
-
- *FIELD* ED
- warfield: 4/7/1994
- carol: 9/1/1992
- supermim: 3/16/1992
- carol: 12/5/1991
- carol: 11/25/1991
- carol: 9/7/1990
-
- *RECORD*
- *FIELD* NO
- 102490
- *FIELD* TI
- 102490 ACRORENOOCULAR SYNDROME
- *FIELD* TX
- Halal et al. (1984) reported a French-Canadian family in which 7 persons
- in 3 generations had various combinations of acral, renal, and ocular
- defects. The acral anomalies varied from mild hypoplasia of the distal
- part of the thumb with limitation of motion at the interphalangeal joint
- to severe thumb hypoplasia and preaxial polydactyly. Renal anomalies
- varied from mild malrotation to crossed renal ectopia without fusion;
- other urinary tract anomalies were vesicoureteral reflux and bladder
- diverticula. Ocular features included 'complete' coloboma, coloboma of
- the optic nerve, ptosis, and Duane anomaly (126800). The disorder
- behaved as an autosomal dominant (with 1 instance of male-to-male
- transmission) with high penetrance but variable expressivity.
- Dermatoglyphic abnormalities were described. Temtamy and McKusick (1978)
- described father and son with some combination of Duane anomaly, radial
- defects, and kidney anomalies. The father had Duane anomaly, bilateral
- thenar and thumb hypoplasia with syndactyly of the index finger and
- unilateral clubhand deformity, and malrotation of both kidneys with
- partial horseshoe anomaly. The son had apparently normal eyes, bilateral
- clubhand with absent thumbs and absent right kidney with malrotation of
- the left kidney. Halal et al. (1984) thought that the disorder in the
- Temtamy-McKusick family might be different because extensive pectoral
- and upper limb involvement present in those cases was absent in all the
- Halal cases.
-
- Naito et al. (1989) and Pierquin et al. (1991) described 3 more cases of
- acrorenoocular syndrome. Aalfs et al. (1996) reported an affected family
- from the Dutch Antilles. Hypoplasia of the right thumb and absence of
- the left thumb, hypoplastic left forearm, microphthalmia, microcornea,
- coloboma of iris and choroidea, cataract, and left-crossed renal ectopia
- with fusion were the main manifestations in the proband. His mother had
- hypoplastic left thumb and cataract (possibly due to diabetes mellitus).
- The sister of the proband demonstrated absence of both thumbs, radii and
- ulnae, and bilateral chorioretinal scars between optic disc and fovea.
- Urologic investigations could not be done in the proband's mother and
- sister. The clinical picture in this family fit all criteria for
- acrorenoocular syndrome.
-
- *FIELD* SA
- Temtamy (1986); Temtamy et al. (1975)
- *FIELD* RF
- 1. Aalfs, C. M.; van Schooneveld, M. J.; van Keulen, E. M.; Hennekem,
- R. C. M.: Further delineation of the acro-renal-ocular syndrome.
- Am. J. Med. Genet. 62: 276-281, 1996.
-
- 2. Halal, F.; Homsy, M.; Perreault, G.: Acro-renal-ocular syndrome:
- autosomal dominant thumb hypoplasia, renal ectopia, and eye defect.
- Am. J. Med. Genet. 17: 753-762, 1984.
-
- 3. Naito, T.; Kida, H.; Yokoyama, H.; Abe, T.; Takeda, S.; Uno, D.;
- Hattori, N.: Nature of renal involvement in the acro-renal-ocular
- syndrome. Nephron 51: 115-118, 1989.
-
- 4. Pierquin, G.; Hall, M.; Vanhelleputte, C.; Van Regemorter, N.:
- A new case of acro-renal-ocular (radio-renal-ocular) syndrome with
- cleft palate and costo-vertebral defects? A brief clinical report. Ophthal.
- Paediat. Genet. 12: 183-186, 1991.
-
- 5. Temtamy, S. A.: The DR syndrome or the Okihiro syndrome?. (Letter) Am.
- J. Med. Genet. 25: 173-174, 1986.
-
- 6. Temtamy, S. A.; McKusick, V. A.: The Genetics of Hand Malformations.
- New York: Alan R. Liss (pub.) 1978. Pp. 133-135.
-
- 7. Temtamy, S. A.; Shoukry, A. S.; Ghaly, I.; El-Meligy, R.; Boulos,
- S. Y.: The Duane radial dysplasia syndrome: an autosomal dominant
- disorder. Birth Defects Orig. Art. Ser. XI(5): 344-345, 1975.
-
- *FIELD* CS
-
- Limbs:
- Thumb hypoplasia/aplasia;
- Stiff thumb;
- Preaxial polydactyly;
- Radial defects;
- Thenar hypoplasia;
- Syndactyly;
- Clubhand deformity
-
- GU:
- Renal malrotation/ectopia;
- Partial horseshoe kidney;
- Vesicoureteral reflux;
- Bladder diverticula
-
- Eyes:
- Complete coloboma;
- Optic nerve coloboma;
- Ptosis;
- Duane anomaly (126800)
-
- Skin:
- Abnormal dermatoglyphics
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CN
- Iosif W. Lurie - updated: 7/1/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 07/02/1996
- carol: 7/1/1996
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
-
- *RECORD*
- *FIELD* NO
- 102500
- *FIELD* TI
- *102500 ACROOSTEOLYSIS WITH OSTEOPOROSIS AND CHANGES IN SKULL AND MANDIBLE
- CHENEY SYNDROME;;
- HAJDU-CHENEY SYNDROME;;
- ARTHRODENTOOSTEODYSPLASIA
- *FIELD* TX
- Cheney (1965) described this connective tissue disorder in a family
- living in the upper peninsula of Michigan. The mother and 4 children had
- acroosteolysis, multiple wormian bones, and hypoplasia of ramus of
- mandible. Unlike pycnodysostosis (265800), a recessive with
- osteosclerosis, the condition in Cheney's patients included osteoporosis
- with basilar impression as a feature. The mother was 57 and the affected
- children (4 of 6) were 35, 26, 21 and 13 years of age. Dorst and
- McKusick (1969) described a case. Herrmann et al. (1973) exhaustively
- reviewed the previously reported cases and described 1 new case. They
- pointed out that the changes in the terminal phalanges in this condition
- as well as in pycnodysostosis are 'pseudo-osteolysis,' that is, the
- disorder is one of defective development of bone rather than destruction
- of bone already formed. They observed that acroosteolysis, generalized
- osteoporosis and multiple fractures of the skull, spine and digits,
- short stature, persistent cranial sutures, multiple wormian bones, early
- loss of teeth, and joint laxity were features associated in varying
- degrees. The authors suggested the name arthrodentoosteodysplasia and
- the eponym Hajdu-Cheney syndrome for this disorder. The patients show
- bathrocephaly (projection of the occipital area and a deep groove at the
- lambdoidal sutures between the occipital and parietal bones).
- Loose-jointedness, dislocations of the patella, and hernia occur. Some
- have suggested that short stature is a consistent feature; a patient of
- mine (P20775) had height of 173 cm at age 16. In addition to
- micrognathia and narrow high palate, prominent (projecting) ears may be
- a feature. Unusually deep voice has also been noted. Silverman et al.
- (1974) provided useful long-term follow-up on 2 cases. They believed the
- patient reported by Gilula et al. (1976) had a nonfamilial disorder.
- Although literally true in that instance, the disorder may have been
- genetic and may be the same as (or perhaps an allelic form of) the
- Cheney syndrome.
-
- Elias et al. (1978) reported Cheney syndrome in a mother and son, one of
- whom had an enlarged sella turcica associated with normal endocrine
- function. Histologic studies made in an area of active osteolysis in a
- phalanx suggested to the authors 'a neurovascular dysfunction with local
- release of osteolytic mediators.' Matisonn and Ziady (1973) described
- affected father and 2 sons; only the sons were personally examined.
- Udell et al. (1986) found this disorder in a 27-year-old man who for 7
- years had gradually progressive loss of distal phalangeal mass with pain
- in the affected fingers. His mother had similar 'shrinking fingers,'
- which first appeared at about age 50, progressed for 2 years, and then
- became asymptomatic. Udell et al. (1986) were impressed with the
- abundance of mast cells in the affected tissues and suggested that these
- cells might be elaborating a local factor causing or promoting
- osteolysis. They pointed to the osteopenia that occurs with large doses
- of heparin and with systemic mast cell disease (154800). Magnetic
- resonance imaging was reported by Kawamura et al. (1991). Ades et al.
- (1993) described a child with this disorder complicated by basilar
- invagination and hydrocephalus. MRI showed Arnold-Chiari malformation
- and obstruction to cerebrospinal fluid flow at the level of the foramen
- magnum. A ventriculoperitoneal shunt was inserted at the age of 10
- years. Kaler et al. (1990) described a 21-year-old woman with
- Hajdu-Cheney syndrome who had severe mitral regurgitation and mild
- aortic stenosis necessitating mitral valve replacement and aortic
- valvotomy at the age of 14 years. Pathologic examination of the mitral
- valve showed myxomatous degeneration with thickened valve leaflets and
- foci of calcification. At the age of 18, pacemaker implantation was
- necessitated by the development of heart block. At the age of 20,
- balloon aortic valvuloplasty was attempted for worsening aortic
- stenosis, but was unsuccessful because of thick and calcified valve
- leaflets; aortic valve replacement was required. O'Reilly and Shaw
- (1994) gave an extensive description of the radiologic features in a
- 15-year-old girl. From early in life the face was dysmorphic with a
- prominent premaxilla, hypertelorism, and downward sloping eyes with
- narrow palpebral fissures. Joint laxity and hyperextensibility developed
- as the child grew older. Height and weight remained at the third
- percentile for age but head circumference was above the 98th percentile,
- with an enlarged pituitary fossa on skull radiographs. Kyphoscoliosis
- required bracing and eventually spinal fusion. The permanent teeth were
- all lost soon after eruption. Basilar impression with multiple wormian
- bones and osteolysis of the terminal phalanges with overlying soft
- tissue swelling were illustrated.
-
- On the basis of 2 unrelated patients with typical Hajdu-Cheney syndrome
- and cystic kidneys with ultrasonographic changes similar to those of
- autosomal dominant polycystic kidney disease (173900), Kaplan et al.
- (1995) concluded that cystic kidneys are an important component of this
- disorder. Neither patient had a family history of polycystic kidney or
- Hajdu-Cheney syndrome. One of the patients died of complications of the
- latter condition at the age of 16 years.
-
- *FIELD* SA
- Brown et al. (1976); Hajdu and Kauntze (1948); Weleber and Beals (1976)
- *FIELD* RF
- 1. Ades, L. C.; Morris, L. L.; Haan, E. A.: Hydrocephalus in Hajdu-Cheney
- syndrome. (Letter) J. Med. Genet. 30: 175 only, 1993.
-
- 2. Brown, D. M.; Bradford, D. S.; Gorlin, R. J.; Desnick, R. J.; Langer,
- L. O., Jr.; Jowsey, J.; Sauk, J. J., Jr.: The acro-osteolysis syndrome:
- morphologic and biochemical studies. J. Pediat. 88: 573-580, 1976.
-
- 3. Cheney, W. D.: Acro-osteolysis. Am. J. Roentgen. 94: 595-607,
- 1965.
-
- 4. Dorst, J. P.; McKusick, V. A.: Acro-osteolysis (Cheney syndrome).
- Birth Defects Orig. Art. Ser. V(3): 215-217, 1969.
-
- 5. Elias, A. N.; Pinals, R. S.; Anderson, H. C.; Gould, L. V.; Streeten,
- D. H. P.: Hereditary osteodysplasia with acro-osteolysis (the Hajdu-Cheney
- syndrome). Am. J. Med. 65: 627-636, 1978.
-
- 6. Gilula, L. A.; Bliznak, J.; Staple, T. W.: Idiopathic nonfamilial
- acro-osteolysis with cortical defects and mandibular ramus osteolysis.
- Radiology 121: 63-68, 1976.
-
- 7. Hajdu, N.; Kauntze, R.: Cranioskeletal dysplasia. Brit. J. Radiol. 21:
- 42-48, 1948.
-
- 8. Herrmann, J.; Zugibe, F. T.; Gilbert, E. F.; Opitz, J. M.: Arthro-dento-osteodysplasia
- (Hajdu-Cheney syndrome): review of a genetic 'acro-osteolysis' syndrome.
- Z. Kinderheilk. 114: 93-110, 1973.
-
- 9. Kaler, S. G.; Geggel, R. L.; Sadeghi-Nejad, A.: Hajdu-Cheney syndrome
- associated with severe cardiac valvular and conduction disease. Dysmorph.
- Clin. Genet. 4: 43-47, 1990.
-
- 10. Kaplan, P.; Ramos, F.; Zackai, E. H.; Bellah, R. D.; Kaplan, B.
- S.: Cystic kidney disease in Hajdu-Cheney syndrome. Am. J. Med.
- Genet. 56: 25-30, 1995.
-
- 11. Kawamura, J.; Miki, Y.; Yamazaki, S.; Ogawa, M.: Hajdu-Cheney
- syndrome: MR imaging. Neuroradiology 33: 441-442, 1991.
-
- 12. Matisonn, A.; Ziady, F.: Familial acro-osteolysis. S. Afr.
- Med. J. 47: 2060-2063, 1973.
-
- 13. O'Reilly, M. A. R.; Shaw, D. G.: Hajdu-Cheney syndrome. Ann.
- Rheum. Dis. 53: 276-279, 1994.
-
- 14. Silverman, F. N.; Dorst, J. P.; Hajdu, N.: Acro-osteolysis (Hajdu-Cheney
- syndrome). In: Bergsma, D.: Skeletal Dysplasias. Amsterdam: Excerpta
- Medica (pub.) 1974. Pp. 106-123.
-
- 15. Udell, J.; Schumacher, H. R., Jr.; Kaplan, F.; Fallon, M. D.:
- Idiopathic familial acroosteolysis: histomorphometric study of bone
- and literature review of the Hajdu-Cheney syndrome. Arthritis Rheum. 29:
- 1032-1038, 1986.
-
- 16. Weleber, R. G.; Beals, R. K.: Hajdu-Cheney syndrome--report of
- 2 cases and review of literature. J. Pediat. 88: 243-249, 1976.
-
- *FIELD* CS
-
- Limbs:
- Acroosteolysis;
- Terminal phalangeal pseudo-osteolysis;
- Patellar dislocation
-
- Skull:
- Multiple wormian bones;
- Mandibular ramus hypoplasia;
- Osteoporosis with basilar impression;
- Persistent cranial sutures
-
- Skel:
- Generalized osteoporosis;
- Multiple fractures
-
- Growth:
- Short stature
-
- Teeth:
- Early teeth loss
-
- Joints:
- Joint laxity
-
- Abdomen:
- Hernia
-
- Mouth:
- Micrognathia;
- Narrow high palate
-
- Ears:
- Prominent (projecting) ears
-
- Voice:
- Unusually deep voice
-
- Neuro:
- Hydrocephalus
-
- Radiology:
- Bathrocephaly;
- Arnold-Chiari malformation on MRI
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 4/25/1995
- jason: 6/13/1994
- carol: 4/6/1994
- mimadm: 3/11/1994
- carol: 3/20/1993
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 102510
- *FIELD* TI
- *102510 ACROPECTOROVERTEBRAL DYSPLASIA, F-FORM OF
- *FIELD* TX
- Grosse et al. (1969) described 8 persons in 4 generations of a kindred
- (of surname beginning with F) who showed a skeletal dysplasia.
- Male-to-male transmission was observed. The hand malformation was mainly
- abnormal segmentation of the first ray. The broad, short thumbs showed
- incipient duplication of the distal phalanx and were, to a variable
- degree, webbing with the index finger, which deviated radially,
- especially when the webbing was extensive. In some, the web contained an
- extra bone, which seemed to be derived from the thumb phalanges and was
- associated with the formation of a bony bridge between the tip of the
- thumb and a radial projection from the distal end of the first index
- phalanx. In some, the web between the first two digits was complete and
- the two distal phalanges of the index finger were then hypoplastic and
- formed part of a bone 'chain' connecting the tips of the thumb and index
- finger. Capitate and hamate were invariably fused; other carpals were
- sometimes incorporated into the fusion. The toes were also webbed,
- especially the first and second, and malformed. Pectoral and vertebral
- anomalies were sternal deformity and spina bifida occulta at L5 or S1.
- According to Opitz (1982), this family remained a unique observation.
-
- Camera et al. (1995) reported on a father and daughter in a second
- family. Synostoses between capitate and hamate, and between talus and
- navicular, invariable features in the 8 affected members of the family
- reported by Grosse et al. (1969), were found. The hand malformation
- involved principally the first 2 rays. In the father and daughter, the
- short and malformed thumb was webbed with the index finger, which was
- radially deviated with duplication of the middle and distal phalanges.
- In the feet, polydactyly and severe metatarsal and toe anomalies were
- present. The father had a prominent sternum with pectus excavatum,
- whereas the daughter had no sternal deformity. Both of them had a mild
- failure of fusion of posterior arch L5 and/or S1.
-
- *FIELD* RF
- 1. Camera, G.; Camera, A.; Pozzolo, S.; Costa, M.; Mantero, R.: F-syndrome
- (F-form of acro-pectoro-vertebral dysplasia): report on a second family.
- Am. J. Med. Genet. 57: 472-475, 1995.
-
- 2. Grosse, F. R.; Herrmann, J.; Opitz, J. M.: The F-form of acropectorovertebral
- dysplasia: the F-syndrome. Birth Defects Orig. Art. Ser. V(3):
- 48-63, 1969.
-
- 3. Opitz, J. M.: Personal Communication. Helena, Mont. 1982.
-
- *FIELD* CS
-
- Skel:
- Skeletal dysplasia
-
- Limbs:
- Abnormal segmentation of the first ray;
- Broad, short thumbs;
- Incipient distal thumb phalanx duplication;
- Thumb and index finger syndactyly;
- Index finger deviated radially;
- Fused capitate and hamate;
- Syndactyly of toes;
- Malformed toes
-
- Thorax:
- Pectoral anomaly;
- Sternal deformity
-
- Spine:
- Vertebral anomalies;
- Spina bifida occulta at L5 or S1
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 7/16/1995
- warfield: 4/7/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 102520
- *FIELD* TI
- 102520 ACRORENAL SYNDROME
- *FIELD* TX
- Dieker and Opitz (1969) described 3 patients with the association of
- major malformations of the kidneys and limbs, mainly absence deformities
- of digits. Curran and Curran (1972) described a case and pointed out
- that paternal age was sometimes increased (44 years in their case and 57
- years in one of Dieker and Opitz). All cases have been male and
- sporadic, without parental consanguinity. Opitz (1982) pointed out that
- this is not, to use his terminology, a causal entity, but rather a
- nonspecific developmental field defect.
-
- *FIELD* RF
- 1. Curran, A. S.; Curran, J. P.: Associated acral and renal malformations:
- a new syndrome?. Pediatrics 49: 716-725, 1972.
-
- 2. Dieker, H.; Opitz, J. M.: Associated acral and renal malformations.
- Birth Defects Orig. Art. Ser. V(3): 68-77, 1969.
-
- 3. Opitz, J. M.: Personal Communication. Helena, Mont. 4/1982.
-
- *FIELD* CS
-
- GU:
- Renal malformation
-
- Limbs:
- Absent digits
-
- Misc:
- Male, sporadic developmental field defect
-
- Inheritance:
- ? Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- warfield: 4/7/1994
- mimadm: 4/2/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 102525
- *FIELD* TI
- *102525 ACROSOMAL VESICLE PROTEIN-1; ACRV1
- SP-10 PROTEIN
- *FIELD* TX
- The SP-10 protein is a testis-specific, differentiation antigen that
- arises within the acrosomal vesicle during spermatogenesis. Herr et al.
- (1991) used a 634-bp fragment of the SP-10 sequence as a probe on
- Southern blots of EcoRI digested DNA from human-mouse somatic cell
- hybrids. Cosegregation of the ACRV1 gene with human chromosome 11 was
- observed. Use of hybrid cell lines containing translocations of human
- chromosome 11 allowed further refinement of localization to 11p12-q13.
- However, by fluorescence in situ hybridization using cDNA, ribo, and
- genomic versions of probes for SP-10 coupled to analysis of an expanded
- series of somatic cell hybrids, Golden et al. (1993) showed that the
- location of ACRV1 is at the junction between 11q23 and 11q24. Golden et
- al. (1993) emphasized the utility of riboprobes for chromosome
- localization of single-copy genes. Riboprobes are complementary RNA
- (cRNA) probes produced using a phage-encoded RNA polymerase. Golden
- (1994) found them better than cDNA probes when the probe was short.
-
- *FIELD* RF
- 1. Golden, W. L.: Personal Communication. Charlottesville, Va.
- 1/12/1994.
-
- 2. Golden, W. L.; von Kap-herr, C.; Kurth, B.; Wright, R. M.; Flickinger,
- C. J.; Eddy, R.; Shows, T.; Herr, J. C.: Refinement of the localization
- of the gene for human intraacrosomal protein SP-10 (ACRV1) to the
- junction of bands q23-q24 of chromosome 11 by nonisotopic in situ
- hybridization. Genomics 18: 446-449, 1993.
-
- 3. Herr, J. C.; Wright, R. M.; Flickinger, C. J.; Eddy, R. L.; Shows,
- T. B.: Assignment of the gene for human intra-acrosomal protein SP-10
- (ACRV1) to the p12-q13 region of chromosome 11. (Abstract) Cytogenet.
- Cell Genet. 58: 1963 only, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 9/30/1991
-
- *FIELD* ED
- carol: 1/14/1994
- carol: 11/30/1993
- supermim: 3/16/1992
- carol: 2/23/1992
- carol: 9/30/1991
-
- *RECORD*
- *FIELD* NO
- 102530
- *FIELD* TI
- 102530 ACROSOME MALFORMATION OF SPERMATOZOA
- ROUND-HEADED SPERMATOZOA;;
- SPERMATOZOA, ROUND-HEADED
- GLOBOZOOSPERMIA, INCLUDED
- *FIELD* TX
- Vegni-Talluri et al. (1977) observed acrosome malformations of
- spermatids and spermatozoa in the testes of 2 infertile males who were
- investigated by light and electron microscopy. The first visible
- abnormality appeared at early spermatid stages. Defective
- differentiation of the acrosome granule in spermatids appeared to be
- responsible for the malformation of mature spermatozoa. The fact that
- about half the early spermatids lacked the acrosome granule suggested
- that the original cause is genetic and that the gene is expressed in the
- haploid phase. The gene might be X-linked or autosomal. The authors
- referred to comparable abnormalities of the acrosome observed in bulls
- and boars and thought to have a mendelian basis. Complete lack of the
- acrosome during spermiogenesis, resulting in round-headed spermatozoa
- incapable of fertilization, has been observed in man and has been
- thought to have a primary genetic basis. Furthermore, the authors drew
- analogies to abnormalities of spermatozoa related to the T-locus of the
- mouse. Abnormalities of spermiogenesis in mammals were reviewed by
- Bishop (1972). Kullander and Rausing (1975) observed only round-headed
- spermatozoa in 2 infertile brothers and suggested that homozygosity for
- an autosomal gene defect underlies this phenotype. In Friesian bulls, a
- characteristic defect of the acrosome ('knobbed' spermatozoa) associated
- with sterility appears to be autosomal recessive.
-
- Florke-Gerloff et al. (1983) showed that the acrosomal membrane proteins
- are first detectable in early spermatids. (The acrosome is a caplike
- compartment in the apical part of the sperm head. It is a lysosome-like
- organelle derived from the Golgi apparatus. In the fertilization
- process, fusion of the sperm plasma membrane and outer acrosomal
- membrane (OAM) occurs with discharge of the acrosomal endosol.)
- Florke-Gerloff et al. (1983) found that the round-headed spermatozoa of
- an infertile patient with globozoospermia lacked the constituting
- components of the outer acrosomal membrane as well as the intraacrosomal
- acrosin system (see 102480). Nistal et al. (1978) observed 2 infertile
- brothers with round-headed spermatozoa. Florke-Gerloff et al. (1984)
- also found 2 affected brothers and studied their father as well. Whereas
- the brothers, like other reported cases, had all round-headed
- spermatozoa, the father had more than 94% normally shaped sperm. Theirs
- was the first study to quantitate the abnormality; in 9 infertile men
- the proportion of round-headed sperm varied from 14 to 71%. They showed
- that the round-headed spermatozoa lacked both acrosin and OAM, as
- indicated by immunofluorescent and immunoperoxidase staining techniques
- and confirmed by the gelatinolysis test. The normally shaped sperm of 6
- of the 9 men were positive for acrosin and OAM. In the father of the
- affected brothers, only 10% of the normally shaped spermatozoa were
- acrosin positive and only 30% were positive for OAM. Florke-Gerloff et
- al. (1984) suggested that the round-headed spermatozoa syndrome is
- polygenic in its inheritance.
-
- *FIELD* SA
- Donald and Hancock (1953)
- *FIELD* RF
- 1. Bishop, M. W. H.: Genetically determined abnormalities of the
- reproductive system. J. Reprod. Fertil. 15 (suppl.): 51-78, 1972.
-
- 2. Donald, H. P.; Hancock, J. L.: Evidence of gene-controlled sterility
- in bulls. J. Agricult. Sci. 43: 178-181, 1953.
-
- 3. Florke-Gerloff, S.; Topfer-Petersen, E.; Muller-Esterl, W.; Mansouri,
- A.; Schatz, R.; Schirren, C.; Schill, W.; Engel, W.: Biochemical
- and genetic investigation of round-headed spermatozoa in infertile
- men including two brothers and their father. Andrologia 16: 187-202,
- 1984.
-
- 4. Florke-Gerloff, S.; Topfer-Petersen, E.; Muller-Esterl, W.; Schill,
- W.-B.; Engel, W.: Acrosin and the acrosome in human spermatogenesis.
- Hum. Genet. 65: 61-67, 1983.
-
- 5. Kullander, S.; Rausing, A.: On round-headed human spermatozoa.
- Int. J. Fertil. 20: 33-40, 1975.
-
- 6. Nistal, M.; Herruzo, A.; Sanchez-Corral, F.: Teratozoospermia
- absoluta de presentacion familiar. Espermatozoides microcefalos irregulares
- sin acrosoma. Andrologia 10: 234-240, 1978.
-
- 7. Vegni-Talluri, M.; Menchini-Fabris, F.; Renieri, T.: A possible
- haploid effect in acrosome malformations of human spermatozoa. Andrologia 9:
- 315-322, 1977.
-
- *FIELD* CS
-
- GU:
- Infertility
-
- Lab:
- Malformed acrosomes of spermatids and spermatozoa
-
- Inheritance:
- Autosomal dominant vs. X-linked or polygenic
-
- *FIELD* CD
- Victor A. McKusick: 6/16/1986
-
- *FIELD* ED
- carol: 4/6/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- carol: 5/5/1989
-
- *RECORD*
- *FIELD* NO
- 102540
- *FIELD* TI
- *102540 ACTIN, ALPHA, CARDIAC MUSCLE; ACTC
- SMOOTH MUSCLE ACTIN;;
- ALPHA-ACTIN;;
- ACTIN, ALPHA
- *FIELD* TX
- Actin has been identified in many kinds of cells including muscle, where
- it is a major constituent of the thin filament, and platelets. Muscle
- actins from sources as diverse as rabbits and fish are very similar in
- amino acid sequence. Elzinga et al. (1976) examined whether actin in
- different tissues of the same organism are products of the same gene.
- They found that human platelet and human cardiac actins differ by one
- amino acid, viz., threonine and valine, respectively, at position 129.
- Thus they must be determined by different genes. Actins can be separated
- by isoelectric focusing into 3 main groups which show more than 90%
- homology of amino acid sequence. Firtel (1981) referred to the actin of
- smooth muscle, the most acidic form, as alpha type and the two
- cytoplasmic forms as beta and gamma. Beta and gamma actins are involved
- in the cytoskeleton and in internal cell mobility phenomena. The actins
- constitute multiple gene families. There is only a 4% amino acid
- difference in the actins of Physarum and mammals. In mammals, 4
- different muscle actins have been sequenced: from fast muscle, heart,
- aorta, and stomach. These vary only by 4 to 6 amino acids from each
- other, and by about 25 amino acids from the beta and gamma actins. Thus,
- from the protein data, at least 6 actin genes would be expected in
- mammals. Recombinant DNA probes for both actin and myosin of the mouse
- have been made (Weydert et al., 1981). Because actin is a highly
- conserved protein, Engel et al. (1981) could use cloned actin genes from
- Drosophila and from chicken to isolate 12 actin gene fragments from a
- human DNA library. Restriction endonuclease studies of each indicated
- that they are not allelic and are from nonoverlapping regions of the
- genome. In all, 25 to 30 EcoRI fragments homologous to actin genes were
- found in the human genome and no restriction site polymorphism was found
- indicating evolutionary conservatism. Humphries et al. (1981) used
- probes from the mouse to detect actin genes in human DNA and concluded
- that there are about 20 actin genes in the human genome. Three lines of
- evidence supported this number: the rate of hybridization of the mouse
- probe with human DNA; the fact that the probe hybridizes to 17-20 bands
- in Southern blots of restriction enzyme digests of total human DNA;
- restriction enzyme mapping of individual human actin genes indicating at
- least 9 different genes, judged on probability grounds to have been
- picked from a pool of at least 20. Litt and Luty (1989) used PCR to
- amplify a microsatellite hypervariable repeat in the human cardiac actin
- gene. They detected 12 different allelic fragments in 37 unrelated
- individuals, of whom 32 were heterozygous. (Weber and May (1989) also
- found that (GT)n repeats within human loci are highly polymorphic.) In
- vertebrates, 6 actin isoforms are known: 4 muscle types (skeletal,
- cardiac, and 2 smooth muscle types) and 2 nonmuscle types (cytoplasmic
- actins).
-
- Hamada et al. (1982) isolated and characterized the human cardiac actin
- gene. The cardiac and skeletal actin genes showed close similarity,
- suggesting a relatively recent derivation from a common ancestral gene.
- Nucleotide sequences of all exon/intron boundaries agreed with the GT/AG
- rule (GT at the 5-prime and AG at the 3-prime termini of each intron).
- The cardiac actin gene and the skeletal actin gene (102610; on
- chromosome 1) are coexpressed in both skeletal and heart muscle.
- Buckingham et al. (1986) provided a summary of the actin and myosin
- multigene families in mouse and man. Certain inbred mouse lines, e.g.,
- BALB/c, have a mutant cardiac actin locus (Garner et al., 1986). The
- first 3 coding exons and promoter region of the gene are present as a
- duplication immediately upstream from the cardiac actin gene. The
- upstream promoter is active, and partial gene transcripts are generated
- which are correctly spliced for the first 3 coding exons but which
- terminate at cryptic sites in the region between the duplication and the
- gene. Transcriptional activity at the upstream promoter interferes with
- the downstream promoter of the bona fide cardiac actin gene, leading to
- a 5- to 6-fold reduction in cardiac actin mRNA in the hearts of BALB/c
- mice. In this situation there is an accumulation of skeletal actin gene
- transcripts in the adult hearts of these mice, which partially
- compensates for the reduction in cardiac actin transcripts. BALB/c mice
- have a normal life span and their hearts do not undergo hypertrophy.
- Apparently, cardiac and skeletal actins, which differ only by 4 out of
- 375 amino acids, are to some extent interchangeable. Schwartz et al.
- (1986) found that under conditions of aortic stenosis leading to cardiac
- overload and cardiac hypertrophy, skeletal actin gene transcripts are
- found in adult rodent hearts in addition to the cardiac actin gene
- products normally present.
-
- Using a cDNA fragment from an intron of the human cardiac actin gene in
- somatic hybrid cell studies, Shows et al. (1984) showed that the gene is
- coded by the segment 15q11-qter. Crosby et al. (1989) showed that in the
- mouse the cardiac actin gene (Actc-1) is not on chromosome 17 as
- previously reported (Czosnek et al., 1983) but is located on chromosome
- 2. It is closely linked to beta-2-microglobulin as indicated by mapping
- studies using restriction fragment variants in recombinant inbred
- strains. Using a highly polymorphic CA repeat microsatellite within
- intron 4 of the ACTC gene, Kramer et al. (1992) did family linkage
- studies with multiple markers on 15q, thus permitting the gene to be
- placed on the chromosome linkage map. They demonstrated that it lies
- about 0.06 cM proximal to D15S49 which is about 0.05 cM proximal to
- D15S25 which in turn is about 0.07 cM proximal to D15S1; D15S1 is
- tightly linked to the Marfan syndrome and to fibrillin. Thus ACTC may be
- about 0.18 cM proximal to the fibrillin locus and no more distal than
- 15q21.1.
-
- By fluorescence in situ hybridization, Ueyama et al. (1995) assigned the
- ACTC gene to 15q14.
-
- *FIELD* SA
- Gunning et al. (1984)
- *FIELD* RF
- 1. Buckingham, M.; Alonso, S.; Barton, P.; Cohen, A.; Daubas, P.;
- Garner, I.; Robert, B.; Weydert, A.: Actin and myosin multigene families:
- their expression during the formation and maturation of striated muscle. Am.
- J. Med. Genet. 25: 623-634, 1986.
-
- 2. Crosby, J. L.; Phillips, S. J.; Nadeau, J. H.: The cardiac actin
- locus (Actc-1) is not on mouse chromosome 17 but is linked to beta-2-microglobulin
- on chromosome 2. Genomics 5: 19-23, 1989.
-
- 3. Czosnek, H.; Nudel, U.; Mayer, Y.; Barker, P. E.; Pravtcheva, D.
- D.; Ruddle, F. H.; Yaffe, D.: The genes coding for the cardiac muscle
- actin, the skeletal muscle actin and the cytoplasmic beta-actin are
- located on three different mouse chromosomes. EMBO J. 2: 1977-1979,
- 1983.
-
- 4. Elzinga, M.; Maron, B. J.; Adelstein, R. S.: Human heart and platelet
- actins are products of different genes. Science 191: 94-95, 1976.
-
- 5. Engel, J. N.; Gunning, P. W.; Kedes, L.: Isolation and characterization
- of human actin genes. Proc. Nat. Acad. Sci. 78: 4674-4678, 1981.
-
- 6. Firtel, R. A.: Multigene families encoding actin and tubulin. Cell 24:
- 6-7, 1981.
-
- 7. Garner, I.; Minty, A. J.; Alonso, S.; Barton, P. J.; Buckingham,
- M. E.: A 5-prime duplication of the alpha-cardiac actin gene in BALB/c
- mice is associated with abnormal levels of alpha-cardiac and alpha-skeletal
- actin mRNAs in adult cardiac tissue. EMBO J. 5: 2559-2567, 1986.
-
- 8. Gunning, P.; Ponte, P.; Kedes, L.; Eddy, R.; Shows, T.: Chromosomal
- location of the co-expressed human skeletal and cardiac actin genes. Proc.
- Nat. Acad. Sci. 81: 1813-1817, 1984.
-
- 9. Hamada, H.; Petrino, M. G.; Kakunaga, T.: Molecular structure
- and evolutionary origin of human cardiac muscle actin gene. Proc.
- Nat. Acad. Sci. 79: 5901-5905, 1982.
-
- 10. Humphries, S. E.; Whittall, R.; Minty, A.; Buckingham, M.; Williamson,
- R.: There are approximately 20 actin genes in the human genome. Nucleic
- Acids Res. 9: 4895-4908, 1981.
-
- 11. Kramer, P. L.; Luty, J. A.; Litt, M.: Regional localization of
- the gene for cardiac muscle actin (ACTC) on chromosome 15q. Genomics 13:
- 904-905, 1992.
-
- 12. Litt, M.; Luty, J. A.: A hypervariable microsatellite revealed
- by in vitro amplification of a dinucleotide repeat within the cardiac
- muscle actin gene. Am. J. Hum. Genet. 44: 397-401, 1989.
-
- 13. Schwartz, K.; de la Bastie, D.; Bouveret, P.; Oliviero, P.; Alonso,
- S.; Buckingham, M.: Alpha-skeletal muscle actin mRNAs accumulate
- in hypertrophied adult rat hearts. Circulation Res. 59: 551-555,
- 1986.
-
- 14. Shows, T.; Eddy, R. L.; Haley, L.; Byers, M.; Henry, M.; Gunning,
- P.; Ponte, P.; Kedes, L.: The coexpressed genes for human alpha (ACTA)
- and cardiac actin (ACTC) are on chromosomes 1 and 15, respectively.
- (Abstract) Cytogenet. Cell Genet. 37: 583 only, 1984.
-
- 15. Ueyama, H.; Inazawa, J.; Ariyama, T.; Nishino, H.; Ochiai, Y.;
- Ohkubo, I.; Miwa, T.: Reexamination of chromosomal loci of human
- muscle actin genes by fluorescence in situ hybridization. Jpn. J.
- Hum. Genet. 40: 145-148, 1995.
-
- 16. Weber, J. L.; May, P. E.: Abundant class of human DNA polymorphisms
- which can be typed using the polymerase chain reaction. Am. J. Hum.
- Genet. 44: 388-396, 1989.
-
- 17. Weydert, A.; Robert, B.; Alonso, S.; Caravatti, M.; Cohen, A.;
- Daubas, P.; Minty, A.; Buckingham, M.: Multigene families of contractile
- proteins: the actins and myosins. (Abstract) Sixth Int. Cong. Hum.
- Genet., Jerusalem 39 only, 1981.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 11/27/1996
- terry: 6/16/1995
- carol: 11/18/1994
- carol: 10/13/1993
- carol: 8/25/1992
- carol: 6/29/1992
- carol: 3/20/1992
-
- *RECORD*
- *FIELD* NO
- 102545
- *FIELD* TI
- *102545 ACTIN, GAMMA-2, SMOOTH MUSCLE, ENTERIC; ACTG2
- ACTSG;;
- ACTE;;
- ACTIN, ALPHA-3, PREVIOUSLY;;
- ACTA3, PREVIOUSLY
- *FIELD* TX
- Miwa et al. (1991) isolated recombinant phages that carried the human
- smooth muscle (enteric) gamma-actin gene (which they symbolized ACTSG)
- from human genomic DNA libraries. The gene, designated ACTG2, contained
- one 5-prime untranslated exon and 8 coding exons extending for 27 kb;
- the mapping of the gene to chromosome 2 was demonstrated by study of
- rodent-human somatic cell hybrids. Ueyama et al. (1995) isolated genomic
- clones containing the gene (which has also been symbolized ACTA3) and
- mapped the gene to 2p13.1 by fluorescence in situ hybridization. From
- the characterized molecular structures of the 6 human actin isoform
- genes, Miwa et al. (1991) proposed a hypothesis of the evolutionary
- pathway of the actin gene family. Each of the 5 other actin genes maps
- to a separate chromosome. Ueyama et al. (1995) demonstrated that the
- HindIII RFLP in the first intron of the gene is due to the
- presence/absence of a 24-bp sequence harboring a HindIII restriction
- site. A biallelic system was found to have allelic frequencies of 45
- (HindIII-minus):55 (HindIII-Plus).
-
- Szucsik and Lessard (1995) characterized the mouse smooth muscle
- (enteric) gamma-actin gene. It represented the largest isoactin gene
- characterized to that time, measuring over 23,000 bp from the
- transcription start site to the polyadenylation signal. The gene had 9
- exons and encoded a mature actin protein of 374 amino acids.
-
- *FIELD* RF
- 1. Miwa, T.; Manabe, Y.; Kurokawa, K.; Kamada, S.; Kanda, N.; Bruns,
- G.; Ueyama, H.; Kakunaga, T.: Structure, chromosome location, and
- expression of the human smooth muscle (enteric type) gamma-actin gene:
- evolution of six human actin genes. Molec. Cell. Biol. 11: 3296-3306,
- 1991.
-
- 2. Szucsik, J. C.; Lessard, J. L.: Cloning and sequence analysis
- of the mouse smooth muscle gamma-enteric actin gene. Genomics 28:
- 154-162, 1995.
-
- 3. Ueyama, H.; Inazawa, J.; Nishino, H.; Han-Xiang, D.; Ochiai, Y.;
- Ohkubo, I.: Chromosomal mapping of the human smooth muscle actin
- gene (enteric type, ACTA3) to 2p13.1 and molecular nature of the HindIII
- polymorphism. Genomics 25: 720-723, 1995.
-
- *FIELD* CD
- Victor A. McKusick: 7/10/1991
-
- *FIELD* ED
- mark: 8/25/1995
- supermim: 3/16/1992
- carol: 8/22/1991
- carol: 7/10/1991
-
- *RECORD*
- *FIELD* NO
- ^102550
- *FIELD* TI
- ^102550 MOVED TO 102630
- *FIELD* TX
- This entry was incorporated into entry 102630 on 10 April 1997.
-
- *FIELD* CN
- Mark H. Paalman - edited: 4/10/1997
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
- *FIELD* ED
- jenny: 04/15/1997
- jenny: 4/10/1997
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
- *RECORD*
- *FIELD* NO
- 102560
- *FIELD* TI
- *102560 ACTIN, GAMMA-1; ACTG1
- ACTIN, GAMMA; ACTG;;
- CYTOSKELETAL GAMMA-ACTIN;;
- ACTIN, CYTOPLASMIC, 2
- *FIELD* TX
- Microfilaments, which are involved in cell motility, organelle
- transport, cytokinesis, and muscle contraction, are linear polymers of
- actin. In mammalian nonmuscle cells, 2 classes of actin are recognized
- on isoelectric focusing gels: beta and gamma. These 2 isoforms differ by
- 4 amino acid substitutions at the conserved NH2-end of the molecule.
- They are coexpressed in nonmuscle cells. Erba et al. (1986) presented
- the complete sequence of gamma cytoskeletal actin mRNA. Erba et al.
- (1988) cloned and sequenced the human gamma-actin gene and demonstrated
- that it is located on chromosome 17 by Southern analysis of DNA from
- human-mouse somatic cell hybrids. Hybridization of the probe to the
- genome of a human-mouse cell hybrid containing a 17;9 translocation
- indicated that the gene is located in the region 17p11-qter.
-
- Ueyama et al. (1996) mapped the ACTG1 gene to 17q25 and 3 ACTG
- pseudogenes to other chromosomes.
-
- *FIELD* RF
- 1. Erba, H. P.; Eddy, R.; Shows, T.; Kedes, L.; Gunning, P.: Structure,
- chromosome location, and expression of the human gamma-actin gene:
- differential evolution, location, and expression of the cytoskeletal
- beta- and gamma-actin genes. Molec. Cell. Biol. 8: 1775-1789, 1988.
-
- 2. Erba, H. P.; Gunning, P.; Kedes, L.: Nucleotide sequence of the
- human gamma cytoskeletal actin mRNA: anomalous evolution of vertebrate
- non-muscle actin genes. Nucleic Acids Res. 14: 5275-5294, 1986.
-
- 3. Ueyama, H.; Inazawa, J.; Nishino, H.; Ohkubo, I.; Miwa, T.: FISH
- localization of human cytoplasmic actin genes ACTB to 7p22 and ACTG1
- to 17q25 and characterization of related pseudogenes. Cytogenet.
- Cell Genet. 74: 221-224, 1996.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 03/20/1997
- terry: 1/13/1997
- supermim: 3/16/1992
- carol: 7/3/1991
- carol: 3/19/1991
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 6/3/1988
-
- *RECORD*
- *FIELD* NO
- 102565
- *FIELD* TI
- *102565 FILAMIN 2; FLN2
- ACTIN BINDING PROTEIN-280, AUTOSOMAL FORM; ABP-280A;;
- ABPA
- *FIELD* TX
- See 300017. Gariboldi et al. (1994) mapped the FLN2 gene to human
- 7q32-q35 by analysis of somatic cell hybrids containing portions of
- chromosome 7. By using a mapping panel from an interspecific murine
- cross, they mapped the corresponding murine locus to chromosome 6 in a
- region homologous to human chromosome 7.
-
- *FIELD* RF
- 1. Gariboldi, M.; Maestrini, E.; Canzian, F.; Manenti, G.; De Gregorio,
- L.; Rivella, S.; Chatterjee, A.; Herman, G. E.; Archidiacono, N.;
- Antonacci, R.; Pierotti, M. A.; Dragani, T. A.; Toniolo, D.: Comparative
- mapping of the actin-binding protein 280 genes in human and mouse. Genomics 21:
- 428-430, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 7/8/1993
-
- *FIELD* ED
- mark: 04/10/1997
- jason: 6/8/1994
- carol: 4/13/1994
- carol: 8/16/1993
- carol: 7/8/1993
-
- *RECORD*
- *FIELD* NO
- 102570
- *FIELD* TI
- *102570 ACTIN, PLATELET
- *FIELD* TX
- See 102540.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
- *RECORD*
- *FIELD* NO
- 102573
- *FIELD* TI
- *102573 ACTININ, ALPHA-2; ACTN2
- *FIELD* TX
- Alpha-actinin is an actin-binding protein with multiple roles in
- different cell types. In nonmuscle cells, the cytoskeletal isoform is
- found along microfilament bundles and adherens-type junctions, where it
- is involved in binding actin to the membrane (see ACTN1; 102575). In
- contrast, skeletal, cardiac, and smooth muscle isoforms are localized to
- the Z-disc and analogous dense bodies, where they help anchor the
- myofibrillar actin filaments. Beggs et al. (1992) characterized 2 human
- muscle-specific alpha-actinin genes, ACTN2 and ACTN3, and mapped them to
- chromosomes 1 and 11, respectively, using somatic cell hybrids. In situ
- hybridization placed the ACTN2 locus at 1q42-q43. Beggs et al. (1992)
- identified a polymorphic (CA)n repeat within the ACTN2 gene and used it
- to position the ACTN2 gene on the CEPH linkage map of chromosome 1.
-
- *FIELD* SA
- Beggs et al. (1992)
- *FIELD* RF
- 1. Beggs, A. H.; Byers, T. J.; Knoll, J. H. M.; Boyce, F. M.; Bruns,
- G. A. P.; Kunkel, L. M.: Cloning and characterization of two human
- skeletal muscle alpha-actinin genes located on chromosomes 1 and 11.
- J. Biol. Chem. 267: 9281-9288, 1992.
-
- 2. Beggs, A. H.; Phillips, H. A.; Kozman, H.; Mulley, J. C.; Wilton,
- S. D.; Kunkel, L. M.; Laing, N. G.: A (CA)n repeat polymorphism for
- the human skeletal muscle alpha-actinin gene ACTN2 and its localization
- on the linkage map of chromosome 1. Genomics 13: 1314-1315, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 8/14/1992
-
- *FIELD* ED
- carol: 10/13/1992
- carol: 9/9/1992
- carol: 8/14/1992
-
- *RECORD*
- *FIELD* NO
- 102574
- *FIELD* TI
- *102574 ACTININ, ALPHA-3; ACTN3
- *FIELD* TX
- See ACTN2 (102573). Beggs et al. (1992) assigned ACTN3 to human
- chromosome 11 by use of somatic cell hybrids and narrowed the
- localization to 11q13-q14 by fluorescence in situ hybridization.
-
- *FIELD* RF
- 1. Beggs, A. H.; Byers, T. J.; Knoll, J. H. M.; Boyce, F. M.; Bruns,
- G. A. P.; Kunkel, L. M.: Cloning and characterization of two human
- skeletal muscle alpha-actinin genes located on chromosomes 1 and 11.
- J. Biol. Chem. 267: 9281-9288, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 8/14/1992
-
- *FIELD* ED
- carol: 4/7/1993
- carol: 9/9/1992
- carol: 8/14/1992
-
- *RECORD*
- *FIELD* NO
- 102575
- *FIELD* TI
- *102575 ACTININ, ALPHA-1; ACTN1
- *FIELD* TX
- Alpha-actinin was initially isolated from rabbit skeletal muscle as a
- factor that induces the gelation of F-actin and promotes the
- superprecipitation of actomyosin. Subsequently, a number of different
- isoforms were isolated from both muscle and nonmuscle cells and from a
- wide variety of organisms. The native molecule is thought to be a
- homodimer of 97-kD subunits arranged in antiparallel fashion. In
- myofibrillar cells, alpha-actinin constitutes a major component of
- Z-disks in striated muscle and of the functionally analogous dense
- bodies and dense plaques in smooth muscle. In nonmuscle cells, it is
- distributed along microfilament bundles and is thought to mediate their
- attachment to the membrane at adherens-type junctions. Youssoufian et
- al. (1990) cloned and characterized a full-length cDNA encoding the
- human cytoskeletal isoform. The gene encodes 891 amino acids with 96 to
- 98% sequence identity at the amino acid level to chicken nonskeletal
- muscle alpha-actinin. Transient expression in COS cells produced a
- protein of about 104 kD. By analysis of somatic cell hybrids and by in
- situ hybridization, Youssoufian et al. (1990) mapped the gene to
- 14q22-q24. Pulsed-field gel analysis of genomic DNA showed that the
- ACTN1 gene and that for erythroid beta-spectrin (182870) are located in
- the same restriction fragment. This finding is of great interest because
- of the structural homology between spectrin and actinin.
-
- *FIELD* RF
- 1. Youssoufian, H.; McAfee, M.; Kwiatkowski, D. J.: Cloning and chromosomal
- localization of the human cytoskeletal alpha-actinin gene reveals
- linkage to the beta-spectrin gene. Am. J. Hum. Genet. 47: 62-72,
- 1990.
-
- *FIELD* CD
- Victor A. McKusick: 7/12/1990
-
- *FIELD* ED
- mark: 04/10/1997
- carol: 8/14/1992
- supermim: 3/16/1992
- carol: 8/20/1990
- carol: 7/12/1990
-
- *RECORD*
- *FIELD* NO
- 102576
- *FIELD* TI
- *102576 ACTIVIN A RECEPTOR, TYPE I; ACVR1
- *FIELD* TX
- Although activins were discovered by virtue of their capacity to
- stimulate the production of follicle-stimulating hormone (FSH; 136530)
- by the pituitary gland and inhibins were initially characterized as FSH
- inhibitors, activins and inhibins are dimeric proteins that share a
- common subunit. There are 3 activins (A, B, and A-B), comprising
- different combinations of 2 closely related beta subunits
- (beta-A/beta-A; beta-B/beta-B; and beta-A/beta-B, respectively) and 2
- inhibins (A and B), consisting of 1 beta-subunit and an inhibin-specific
- alpha subunit (alpha/beta-A and alpha/beta-B). Activins impinge on a
- much broader spectrum of cells than do inhibins; however, in those
- systems in which both proteins are functional, they have opposing
- biologic effects. Activins are members of a family of polypeptide growth
- factors that includes also the transforming growth factors-beta (190180,
- 190220, 190230), mullerian duct-inhibiting substance, and several bone
- morphogenetic proteins. Mathews and Vale (1991) cloned the activin
- receptor by use of a method that has been used to clone other receptors,
- such as that for erythropoietin. The cloning is based on the ability of
- the receptor to bind a labeled ligand following expression of a cDNA
- library in mammalian cells. The cDNA coded for a protein of 494 amino
- acids comprising a ligand-binding extracellular domain, a single
- membrane-spanning domain, and an intracellular kinase domain with
- predicted serine/threonine specificity.
-
- Two types of activin receptors were identified on the basis of
- affinity-crosslinking studies. The type I receptor has a molecular size
- of 65 kD, while the molecular size of the type II receptor is 85 kD
- (Mathews and Vale, 1991).
-
- *FIELD* RF
- 1. Mathews, L. S.; Vale, W. W.: Expression cloning of an activin
- receptor, a predicted transmembrane serine kinase. Cell 65: 973-982,
- 1991.
-
- *FIELD* CD
- Victor A. McKusick: 8/9/1991
-
- *FIELD* ED
- carol: 3/30/1994
- supermim: 3/16/1992
- carol: 8/30/1991
- carol: 8/9/1991
-
- *RECORD*
- *FIELD* NO
- 102577
- *FIELD* TI
- *102577 ACTIVATOR 1, 37-KILODALTON SUBUNIT
- A1, 37-KD SUBUNIT;;
- REPLICATION FACTOR C, 37-KD SUBUNIT;;
- RFC, 37-KD SUBUNIT;;
- REPLICATION FACTOR C4; RFC4
- *FIELD* TX
- The elongation of primed DNA templates by DNA polymerase delta and DNA
- polymerase epsilon requires the action of 2 accessory proteins,
- proliferating cell nuclear antigen (PCNA; 176740) and activator 1 (A1;
- also called replication factor C). A1 is an enzyme that contains 5
- different subunits of 140, 40, 38, 37, and 36 kD. Chen et al. (1992)
- isolated the gene encoding the 37-kD subunit from HeLa cells. The
- deduced amino acid sequence showed a high degree of homology to the
- 40-kD subunit of A1 but, unlike the 40-kD protein, the 37-kD expressed
- protein did not bind ATP. Other findings suggested that both the 37- and
- 40-kD subunits of A1 are required for the biologic role of A1 and that
- they may function differently in this process.
-
- Okumura et al. (1995) mapped RFC4 to 3q27 by a combination of PCR
- amplification of DNAs from a panel of somatic hybrids and by
- fluorescence in situ hybridization.See replication factor C, subunit 2
- (RFC2; 600404).
-
- *FIELD* RF
- 1. Chen, M.; Pan, Z.-Q.; Hurwitz, J.: Studies of the cloned 37-kDa
- subunit of activator 1 (replication factor C) of HeLa cells. Proc.
- Nat. Acad. Sci. 89: 5211-5215, 1992.
-
- 2. Okumura, K.; Nogami, M.; Taguchi, H.; Dean, F. B.; Chen, M.; Pan,
- Z.-Q.; Hurwitz, J.; Shiratori, A.; Murakami, Y.; Ozawa, K.; Eki, T.
- : Assignment of the 36.5-kDa (RFC5), 37-kDa (RFC4), 38-kDa (RFC3),
- and 40-kDa (RFC2) subunit genes of human replication factor C to chromosome
- bands 12q24.2-q24.3, 3q27, 13q12.3-q13, and 7q11.23. Genomics 25:
- 274-278, 1995.
-
- *FIELD* CD
- Victor A. McKusick: 7/7/1992
-
- *FIELD* ED
- carol: 3/19/1995
- carol: 12/14/1993
- carol: 7/7/1992
-
- *RECORD*
- *FIELD* NO
- 102578
- *FIELD* TI
- *102578 ACUTE PROMYELOCYTIC LEUKEMIA, INDUCER OF; PML
- *FIELD* TX
- In the process of analyzing the retinoic acid receptor alpha (RARA;
- 180240) gene in the t(15;17)(q22;q11.2-q12) translocation specifically
- associated with acute promyelocytic leukemia (APL), de The et al. (1990)
- identified a new gene on chromosome 15 which is involved with the RARA
- gene in the formation of a fusion product. This gene, which they called
- MYL for 'myelocytic leukemia,' was transcribed in the same direction as
- RARA on the translocated allele. They identified a 144-bp region,
- flanked by canonical splice acceptor and donor sequences, that had a
- high probability of being an exon and showed no significant similarity
- to any sequence in a protein data bank, thus suggesting that MYL is a
- previously undescribed gene. In the chimeric gene, the promoter and
- first exon of the RARA gene were replaced by part of the MYL gene. De
- The et al. (1990) established that the translocation chromosome
- generates an MYL/RARA chimeric transcript. The findings strongly
- implicated retinoic acid receptor alpha in leukemogenesis. The
- possibility was raised that the altered retinoic acid receptor behaves
- as a dominant negative mutant that blocks the expression of retinoic
- acid target genes involved in granulocytic differentiation. In a later
- report, de The et al. (1991) changed the name of the gene from MYL to
- PML. They reported, furthermore, that the gene product contains a novel
- zinc finger motif common to several DNA-binding proteins. The PML-RARA
- mRNA encoded a predicted 106-kd chimeric protein containing most of the
- PML sequences fused to a large part of the RARA gene, including its DNA-
- and hormone-binding domains. Goddard et al. (1991) demonstrated that PML
- is a putative zinc finger protein and potential transcription factor
- that is commonly expressed, with at least 3 major transcription
- products. The PML breakpoints are clustered in 2 regions on either side
- of an alternatively spliced exon. Although leukemic cells with
- translocations characteristically expressed only one fusion product,
- both PML/RARA (on the 15q+ derivative chromosome) and RARA/PML (on the
- 17q- derivative) were transcribed. The contribution of PML to the
- oncogenicity of the fusion products was demonstrated by the following:
- no mutations affecting RARA alone were observed in 20 APLs analyzed; 2
- APLs cytogenetically lacking t(15;17) chromosomes were found to have
- rearrangements of both PML and RARA; and PML, but not RARA, was
- molecularly rearranged in a variant APL translocation in which
- chromosome 15 had been translocated to another chromosome with no
- visible involvement of chromosome 17. Tong et al. (1992) found that in
- 20 of 22 patients with a detectable MYL rearrangement the breakpoints
- were clustered within a 4.4-kb segment, which they designated MYL(bcr).
- The 2 remaining patients exhibited a more 5-prime rearrangement at about
- 10-kb upstream of the MYL(bcr) region, indicating the lack of at least
- one MYL gene exon in the resulting MYL-RARA fusion gene. Cleary (1991)
- pointed out that detection of the PML-RARA fusion links a specific
- molecular defect in neoplasia with a characteristic biologic and
- clinical response to pharmacologic therapy. It is a useful marker for
- the diagnosis of APL and for the identification of patients who may
- benefit from retinoid treatment.
-
- PML, the gene involved in the breakpoint on chromosome 15, is a putative
- transcription factor: it contains a cysteine-rich motif that resembles a
- zinc finger DNA-binding domain common to several classes of
- transcriptional factors. Its physiologic role is unknown. Two fusion
- genes, PML-RARA and RARA-PML, are formed as a result of the
- characteristic translocation in acute promyelocytic leukemia.
- Heterogeneity of the chromosome 15 breakpoints accounts for the diverse
- architecture of the PML-RARA mRNAs isolated from different APL patients,
- and alternative splicing of PML exons gives rise to multiple isoforms of
- the PML-RARA mRNAs even within a single patient. Alcalay et al. (1992)
- investigated the organization and expression pattern of the RARA-PML
- gene in a series of APL patients. An RARA-PML transcript was present in
- most, but not all, APL patients. Among 70 patients with APL, Diverio et
- al. (1992) found an abnormality in intron 2 of the RARA gene in all
- cases, with clustering of rearrangements within the 20-kb intronic
- region separating exons 2 and 3. A curious difference was found in the
- location of breakpoints in males and females: breakpoints at the 5-prime
- end of intron 2 of the RARA gene occurred in females and 3-prime
- breakpoints predominated in males.
-
- From their analysis of the phosphoamino acids of the PML protein, Chang
- et al. (1995) concluded that both tyrosine and serine residues are
- phosphorylated. To investigate whether expression of the PML protein is
- cell-cycle related, HeLa cells synchronized at various phases of the
- cell cycle were analyzed by immunofluorescence staining and confocal
- microscopy. They found that PML was expressed at a lower level in S, G2,
- and M phases and at a significantly higher level in G1 phase. Other
- studies showed that PML is a phosphoprotein and is associated with the
- nuclear matrix. Chang et al. (1995) noted that PML shares many
- properties with tumor suppressors, such as RB (180200).
-
- Goddard et al. (1995) cloned the murine Pml gene and determined its
- intron/exon organization. The predicted amino acid sequence of the mouse
- Pml, a ring-finger protein, shows 80% similarity to that of the human
- homolog with greater than 90% similarity in the proposed functional
- domains. Chromosomal localization of the Pml locus by somatic cell
- hybrids and by linkage analysis indicated that the gene maps to a region
- of mouse chromosome 9 with known homology of synteny to the region of
- 15q where PML is located.
-
- Brown et al. (1997) established a transgenic mouse model that documented
- the ability of the chimeric PMLRAR-alpha gene to initiate
- leukemogenesis. The mice developed 2 currently unrelated abnormalities.
- The first was a severe papillomatosis of the skin; the second was a
- disturbance of hematopoiesis that presented as a partial block of
- differentiation in the neutrophil lineage of the transgenic mice and
- then progressed at low frequency to overt APL. The leukemia appeared to
- be a faithful reproduction of the human disease, including a therapeutic
- response to retinoic acid that reflected differentiation of the leukemic
- cells. Both the preleukemic state and the overt leukemia could be
- transplanted into nontransgenic hosts. Brown et al. (1997) commented
- that the model should be useful for exploring the pathogenesis and
- treatment of APL.
-
- *FIELD* RF
- 1. Alcalay, M.; Zangrilli, D.; Fagioli, M.; Pandolfi, P. P.; Mencarelli,
- A.; Lo Coco, F.; Biondi, A.; Grignani, F.; Pelicci, P. G.: Expression
- pattern of the RAR-alpha-PML fusion gene in acute promyelocytic leukemia. Proc.
- Nat. Acad. Sci. 89: 4840-4844, 1992.
-
- 2. Brown, D.; Kogan, S.; Lagasse, E.; Weissman, I.; Alcalay, M.; Pelicci,
- P. G.; Atwater, S.; Bishop, J. M.: A PMLRAR-alpha transgene initiates
- murine acute promyelocytic leukemia. Proc. Nat. Acad. Sci. 94: 2551-2556,
- 1997.
-
- 3. Chang, K.-S.; Fan, Y.-H.; Andreeff, M.; Liu, J.; Mu, Z.-M.: The
- PML gene encodes a phosphoprotein associated with the nuclear matrix. Blood 85:
- 3646-3653, 1995.
-
- 4. Cleary, M. L.: Oncogenic conversion of transcription factors by
- chromosomal translocations. Cell 66: 619-622, 1991.
-
- 5. de The, H.; Chomienne, C.; Lanotte, M.; Degos, L.; Dejean, A.:
- The t(15;17) translocation of acute promyelocytic leukaemia fuses
- the retinoic acid receptor alpha gene to a novel transcribed locus. Nature 347:
- 558-561, 1990.
-
- 6. de The, H.; Lavau, C.; Marchio, A.; Chomienne, C.; Degos, L.; Dejean,
- A.: The PML-RAR-alpha fusion mRNA generated by the t(15;17) translocation
- in acute promyelocytic leukemia encodes a functionally altered RAR. Cell 66:
- 675-684, 1991.
-
- 7. Diverio, D.; Lo Coco, F.; D'Adamo, F.; Biondi, A.; Fagioli, M.;
- Grignani, F.; Rambaldi, A.; Rossi, V.; Avvisati, G.; Petti, M. C.;
- Testi, A. M.; Liso, V.; Specchia, G.; Fioritoni, G.; Recchia, A.;
- Frassoni, F.; Ciolli, S.; Pelicci, P. G.: Identification of DNA rearrangements
- at the retinoic acid receptor-alpha (RAR-alpha) locus in all patients
- with acute promyelocytic leukemia and mapping of APL breakpoints within
- the RAR-alpha second intron. Blood 79: 3331-3336, 1992.
-
- 8. Goddard, A. D.; Borrow, J.; Freemont, P. S.; Solomon, E.: Characterization
- of a zinc finger gene disrupted by the t(15;17) in acute promyelocytic
- leukemia. Science 254: 1371-1374, 1991.
-
- 9. Goddard, A. D.; Yuan, J. Q.; Fairbairn, L.; Dexter, M.; Borrow,
- J.; Kozak, C.; Solomon, E.: Cloning of the murine homolog of the
- leukemia-associated PML gene. Mammalian Genome 6: 732-737, 1995.
-
- 10. Tong, J.-H.; Dong, S.; Geng, J.-P.; Huang, W.; Wang, Z.-Y.; Sun,
- G.-L.; Chen, S.-J.; Chen, Z.; Larsen, C.-J.; Berger, R.: Molecular
- rearrangements of the MYL gene in acute promyelocytic leukemia (APL,
- M3) define a breakpoint cluster region as well as some molecular variants. Oncogene 7:
- 311-316, 1992.
-
- *FIELD* CN
- Victor A. McKusick - updated: 04/21/1997
-
- *FIELD* CD
- Victor A. McKusick: 11/30/1990
-
- *FIELD* ED
- jenny: 04/21/1997
- terry: 4/12/1997
- mark: 11/30/1995
- mark: 10/5/1995
- carol: 8/13/1992
- carol: 6/16/1992
- carol: 5/28/1992
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 102579
- *FIELD* TI
- *102579 ACTIVATOR 1, 140-KILODALTON SUBUNIT
- A1, 140-KD SUBUNIT;;
- REPLICATION FACTOR C, 140-KD SUBUNIT;;
- RFC, 140-KD SUBUNIT;;
- RFC140;;
- RFC1;;
- RECC1
- *FIELD* TX
- Replication factor C is a multisubunit, DNA polymerase accessory protein
- required for the coordinated synthesis of both DNA strands during simian
- virus 40 DNA replication in vitro. It is a DNA-dependent ATPase that
- binds in a structure-specific manner to the 3-prime end of a primer
- hybridized to a template DNA, an activity thought intrinsic to the
- 140-kD component of this multisubunit complex. Bunz et al. (1993)
- isolated and analyzed cDNAs encoding the 140-kD subunit. An open reading
- frame of 3.4 kb was predicted to encode a 1,148-amino acid protein with
- a predicted molecular mass of 130 kD. A putative ATP-binding motif was
- observed that is similar to a motif in several of the smaller subunits
- of RFC and in functionally homologous replication factors of bacterial
- and viral origin. The predicted protein showed similarities to other
- DNA-binding proteins.
-
- Luckow et al. (1994) isolated a full-length mouse cDNA which encodes a
- protein that binds in a sequence-unspecific manner to DNA, is localized
- exclusively in the nucleus, and represented, they concluded, the 140-kD
- subunit of mouse replication factor C. They found that it showed 83%
- identity to the human protein. Luckow et al. (1994) assigned the gene
- for the largest subunit of replication factor C (RFC1) to 4p14-p13 by
- fluorescence in situ hybridization. They mapped the homolog in the mouse
- to chromosome 5. Lossie et al. (1995) likewise mapped this gene, which
- they symbolized Recc1, to human chromosome 4 by human/rodent somatic
- cell hybrid analysis and to mouse chromosome 5 by haplotype analysis of
- an interspecific backcross.
-
- *FIELD* RF
- 1. Bunz, F.; Kobayashi, R.; Stillman, B.: cDNAs encoding the large
- subunit of human replication factor C. Proc. Nat. Acad. Sci. 90:
- 11014-11018, 1993.
-
- 2. Lossie, A. C.; Haugen, B. R.; Wood, W. M.; Camper, S. A.; Gordon,
- D. F.: Chromosomal localization of the large subunit of mouse replication
- factor C in the mouse and human. Mammalian Genome 6: 58-59, 1995.
-
- 3. Luckow, B.; Bunz, F.; Stillman, B.; Lichter, P.; Schutz, G.: Cloning,
- expression, and chromosomal localization of the 140-kilodalton subunit
- of replication factor C from mice and humans. Molec. Cell. Biol. 14:
- 1626-1634, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 12/14/1993
-
- *FIELD* ED
- terry: 4/18/1995
- carol: 2/20/1995
- carol: 12/14/1993
-
- *RECORD*
- *FIELD* NO
- 102581
- *FIELD* TI
- *102581 ACTIVIN A RECEPTOR, TYPE II; ACVR2
- *FIELD* TX
- Two types of activin receptors were identified by affinity-crosslinking
- studies. The type I receptor (ACVR1; 102576) has a molecular weight of
- 65 kD, while the molecular size of the type II receptor is 85 kD
- (Mathews and Vale, 1991). Donaldson et al. (1992) cloned cDNAs encoding
- type II activin receptor of the human. Activin has been suggested to be
- an autocrine/paracrine regulator in the human placenta. This is
- supported by the work of Peng et al. (1993), who demonstrated ACVR2 mRNA
- in human trophoblast cells. They also provided the first evidence of
- expression of the gene in human brain and ovary.
-
- Two different forms of activin receptor type 2 have been found in mouse
- and chick (Feijen et al., 1994). Both forms show tissue-specific and
- temporal-specific differences in the timing of their expression during
- mouse embryogenesis.
-
- *FIELD* RF
- 1. Donaldson, C. J.; Mathews, L. S.; Vale, W. W.: Molecular cloning
- and binding properties of the human type II activin receptor. Biochem.
- Biophys. Res. Commun. 184: 310-316, 1992.
-
- 2. Feijen, A.; Goumans, M. J.; van den Eijnden-van Raaij, A. J.:
- Expression of activin subunits, activin receptors and follistatin
- in postimplantation mouse embryos suggests specific developmental
- functions for different activins. Development 120: 3621-3637, 1994.
-
- 3. Mathews, L. S.; Vale, W. W.: Expression cloning of an activin
- receptor, a predicted transmembrane serine kinase. Cell 65: 973-982,
- 1991.
-
- 4. Peng, C.; Huang, T.-H. J.; Jeung, E.-B.; Donaldson, C. J.; Vale,
- W. W.; Leung, P. C. K.: Expression of the type II activin receptor
- gene in the human placenta. Endocrinology 133: 3046-3049, 1993.
-
- *FIELD* CN
- Moyra Smith - Updated: 05/16/1996
-
- *FIELD* CD
- Victor A. McKusick: 3/30/1994
-
- *FIELD* ED
- carol: 05/16/1996
- carol: 3/30/1994
-
- *RECORD*
- *FIELD* NO
- 102582
- *FIELD* TI
- *102582 SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION 3; STAT3
- ACUTE-PHASE RESPONSE FACTOR; APRF
- *FIELD* TX
- Acute-phase response factor is a latent cytoplasmic transcription factor
- that is rapidly activated in response to interleukin-5 (147850),
- interleukin-6 (147620), epidermal growth factor (131530), leukemia
- inhibitory factor (159540), oncostatin M (165095), interleukin-11
- (147681), and ciliary neurotrophic factor (118945). After activation,
- the 89-kD protein binds to IL6 response elements identified in the
- promoter regions of various IL6-induced plasma-protein and
- intermediate-early genes. Lutticken et al. (1994) demonstrated that the
- above listed cytokines cause tyrosine phosphorylation of the APRF.
- Protein kinases of the JAK family (e.g., 147795) were also rapidly
- tyrosine phosphorylated, and both APRF and JAK1 associated with the
- signal transducer gp130 (162820). Akira et al. (1994) suggested that
- APRF may play a major role in the gp130-mediated signaling pathway. They
- purified APRF and cloned the cDNA. At the amino acid level, APRF
- exhibited 52.5% overall homology with p91, a component of the interferon
- (IFN)-stimulated gene factor-3 complexes. See STAT1 (600555).
-
- Binding of interleukin-5 to its specific receptor activates JAK2
- (147796) which leads to the tyrosine phosphorylation of STAT3 proteins.
- Caldenhoven et al. (1996) reported the cloning of a cDNA encoding a
- variant of the transcription factor STAT3 (named STAT3-beta) that was
- isolated by screening an eosinophil cDNA library. Compared to wildtype
- STAT3, STAT3-beta lacks an internal domain of 50 bp located near the C
- terminus. This splice product is a naturally occurring isoform of STAT3
- and encodes an 80-kD protein. Like STAT3, STAT3-beta is phosphorylated
- on tyrosine and binds to the pIRE from the ICAM1 (147840) promoter after
- IL-5 stimulation. Coexpression of STAT3-beta inhibits the
- transactivation potential of STAT3. These results suggested that
- STAT3-beta functions as a negative regulator of transcription.
-
- The leptin receptor (601007) is found in many tissues in several
- alternatively spliced forms, raising the possibility that leptin exerts
- effects on many tissues including the hypothalamus. The leptin receptor
- is a member of the gp130 family of cytokine receptors that are known to
- stimulate gene transcription via activation of cytosolic STAT proteins.
- In order to identify the sites of leptin action in vivo, Vaisse et al.
- (1996) assayed for activation of STAT proteins in mice treated with
- leptin. The STAT proteins bind to phosphotyrosine residues in the
- cytoplasmic domain of the ligand-activated receptor, where they are
- subsequently phosphorylated. The activated STAT proteins dimerize and
- translocate to the nucleus where they bind DNA and activate
- transcription. The investigators assayed the activation of STAT proteins
- in response to leptin in a variety of mouse tissues known to express
- Obr. Leptin injection activated Stat3 but no other STAT protein in the
- hypothalamus of ob/ob and wildtype mice but not db/db mice, mutants that
- lack an isoform of the leptin receptor. Leptin did not induce STAT
- activation in any of the other tissues tested. The dose-dependent
- activation of STAT3 by leptin was first observed after 15 minutes and
- maximal in 30 minutes. The data indicated to Vaisse et al. (1996) that
- the hypothalamus is a direct target of leptin action and this activation
- is critically dependent on the gp130-like leptin receptor isoform
- missing in db/db mice.
-
- *FIELD* RF
- 1. Akira, S.; Nishio, Y.; Inoue, M.; Wang, X.-J.; Wei, S.; Matsusaka,
- T.; Yoshida, K.; Sudo, T.; Naruto, M.; Kishimoto, T.: Molecular cloning
- of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription
- factor involved in the gp130-mediated signaling pathway. Cell 77:
- 63-71, 1994.
-
- 2. Caldenhoven, E.; van Dijk, T. B.; Solari, R.; Armstrong, J.; Raaijmakers,
- J. A. M.; Lammers, J.-W. J.; Koenderman, L.; de Groot, R. P.: STAT3-beta,
- a splice variant of transcription factor STAT3, is a dominant negative
- regulator of transcription. J. Biol. Chem. 271: 13221-13227, 1996.
-
- 3. Lutticken, C.; Wegenka, U. M.; Yuan, J.; Buschmann, J.; Schindler,
- C.; Ziemiecki, A.; Harpur, A. G.; Wilks, A. F.; Yasukawa, K.; Taga,
- T.; Kishimoto, T.; Barbieri, G.; Pellegrini, S.; Sendtner, M.; Heinrich,
- P. C.; Horn, F.: Association of transcription factor APRF and protein
- kinase Jak1 with the interleukin-6 signal transducer gp130. Science 263:
- 89-92, 1994.
-
- 4. Vaisse, C.; Halaas, J. L.; Horvath, C. M.; Darnell, J. E., Jr.;
- Stoffel, M.; Friedman, J. M.: Leptin activation of Stat3 in the hypothalamus
- of wildtype and ob/ob mice but not in db/db mice. Nature Genet. 14:
- 95-100, 1996.
-
- *FIELD* CN
- Mark H. Paalman - edited: 9/10/1996
-
- *FIELD* CD
- Victor A. McKusick: 7/13/1994
-
- *FIELD* ED
- terry: 12/30/1996
- terry: 12/11/1996
- mark: 9/12/1996
- mark: 9/11/1996
- mark: 9/10/1996
- jason: 7/13/1994
-
- *RECORD*
- *FIELD* NO
- 102590
- *FIELD* TI
- 102590 ACYLASE, COBALT-ACTIVATED
- *FIELD* TX
- By polyacrylamide gel electrophoresis, Ziomek and Szewczuk (1978)
- demonstrated polymorphism of Co(2+)-activated acylase of human liver,
- kidney and small intestine as well as serum from patients with viral
- hepatitis. Family studies were not reported. This enzyme is an
- N-acylamino acid amidohydrolase that cleaves the low-molecular-weight
- carboxylic acids from acylated amino acids. It is distinct from
- aminoacylases 1 and 2 (104620).
-
- *FIELD* RF
- 1. Ziomek, E.; Szewczuk, A.: Polymorphism of the cobalt-activated
- acylase in human tissues. Acta Biochim. Polon. 25: 3-14, 1978.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 102593
- *FIELD* TI
- *102593 ACYLOXYACYL HYDROLASE; AOAH
- *FIELD* TX
- Acyloxyacyl hydrolase (AOAH) is a 2-subunit lipase present in phagocytic
- cells. This enzyme specifically hydrolyzes the secondary acyl chains of
- the lipopolysaccharide found in the walls of gram-negative bacteria.
- Although the physiologic function of AOAH has not been clearly defined,
- its action on lipopolysaccharide (or endotoxin) suggests that it
- modulates the host's inflammatory response to gram-negative bacteria.
- This hypothesis is supported by studies showing that the deacylation of
- lipopolysaccharide by AOAH in vitro greatly reduces its toxicity and
- activity. Hagen et al. (1991) cloned and characterized cDNA for human
- AOAH and showed that its 2 subunits are translated from a single mRNA
- molecule about 2.2 kb long. By fluorescence in situ hybridization,
- Whitmore et al. (1994) mapped the AOAH gene to 7p14-p12.
-
- *FIELD* RF
- 1. Hagen, F. S.; Grant, F. J.; Kuijper, J. L.; Slaughter, C. A.; Moomaw,
- C. R.; Orth, K.; O'Hara, P. J.; Munford, R. S.: Expression and characterization
- of recombinant human acyloxyacyl hydrolase, a leukocyte enzyme that
- deacylates bacterial lipopolysaccharides. Biochemistry 30: 8415-8423,
- 1991.
-
- 2. Whitmore, T. E.; Mathewes, S. L.; O'Hara, P. J.; Durnam, D. M.
- : Chromosomal localization of the acyloxyacyl hydrolase (AOAH) gene
- to 7p14-p12 using fluorescence in situ hybridization. Genomics 21:
- 457-458, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 6/17/1994
-
- *FIELD* ED
- jason: 6/17/1994
-
- *RECORD*
- *FIELD* NO
- 102595
- *FIELD* TI
- *102595 ACYLPHOSPHATASE, MUSCLE; ACYP
- *FIELD* TX
- Acylphosphatase (EC 3.6.1.7) is a hydrolase that specifically catalyzes
- the hydrolysis of the carboxyl-phosphate bond of acylphosphates. It is a
- small (relative molecular mass about 11,000) and stable enzyme that is
- distributed among a wide variety of species and tissues. The enzyme has
- been purified from skeletal muscle of various mammals and birds and the
- primary structures determined. The primary structure is well conserved
- among different species. Liguri et al. (1986) reported the isolation and
- characterization of a human erythrocyte acylphosphatase isoenzyme; see
- 600875. Modesti et al. (1993) constructed a DNA sequence coding for
- human muscle acylphosphatase and studied its expression in E. coli and
- S. cerevisiae.
-
- *FIELD* RF
- 1. Liguri, G.; Camici, G.; Manao, G.; Cappugi, G.; Nassi, P.; Modesti,
- A.; Ramponi, G.: A new acylphosphatase isoenzyme from human erythrocytes:
- purification, characterization, and primary structure. Biochemistry 25:
- 8089-8094, 1986.
-
- 2. Modesti, A.; Raugei, G.; Taddei, N.; Marzocchini, R.; Vecchi, M.;
- Camici, G.; Manao, G.; Ramponi, G.: Chemical synthesis and expression
- of a gene coding for human muscle acylphosphatase. Biochim. Biophys.
- Acta 1216: 369-374, 1993.
-
- *FIELD* CD
- Victor A. McKusick: 3/26/1994
-
- *FIELD* ED
- mark: 10/16/1995
- carol: 3/26/1994
-
- *RECORD*
- *FIELD* NO
- 102600
- *FIELD* TI
- *102600 ADENINE PHOSPHORIBOSYLTRANSFERASE; APRT
- 2,8-@DIHYDROXYADENINE UROLITHIASIS, INCLUDED;;
- DHA-UROLITHIASIS, INCLUDED;;
- APRT, AUTOSOMAL RECESSIVE, INCLUDED
- *FIELD* MN
- Patients with complete deficiency of APRT excrete gravel consisting of
- stones of 2,8-dihydroxyadenine (DHA) in urine, but do not have
- hyperuricemia or gout. Treatment with allopurinol and a low purine diet
- stops stone formation. Homozygotes can be detected by raised urinary
- adenine levels and no detectable red cell APRT (Simmonds et al., 1992).
- In Japanese, partial deficiency of APRT may lead to 2,8-dihydroxyadenine
- urolithiasis (Kamatani et al., 1992), whereas all Caucasian patients
- with 2,8-DHA urolithiasis have been completely deficient. The common
- Japanese mutant allele is known as APRT*J.
-
- Renal biopsy shows changes similar to those of uric acid nephropathy.
- Families carrying the mutant APRT gene need to be aware of it since
- acute renal failure may be the presenting symptom and this may be
- reversible, although some patients progress to chronic renal failure
- requiring dialysis and transplantation. There is a simple test for
- distinguishing uric acid calculi from 2,8-DHA calculi (Maddocks, 1992)
- and even visual examination can distinguish the two: 2,8-DHA stones are
- soft, friable, reddish-brown when wet and grayish when dry (Ward and
- Addison, 1992). The presence of round, brownish urine crystals, even
- without radiolucent kidney stones, should alert the physician to the
- diagnosis.
-
- The APRT gene is located at 16q24 (Fratini et al., 1986). It is about
- 2.6 kb long and contains 5 exons. Its promoter region, like that of
- several other 'housekeeping' genes, lacks the 'TATA' and 'CCAAT' boxes
- but contains 5 GC boxes that are potential binding sites for the Sp1
- transcription factor (Broderick et al., 1987). Mutations include
- basepair deletions, insertions, and substitutions. The estimated gene
- frequency among Japanese is about 1.2% (Kamatani et al., 1992).
-
- *FIELD* ED
- carol: 07/06/1996 joanna: 6/25/1996
-
- *FIELD* CD
- F. Clarke Fraser: 5/9/1996
- *FIELD* TX
- Mutant forms of APRT (EC 2.4.2.7) have been described by Kelley et al.
- (1968) and by Henderson et al. (1969) who found the inheritance to be
- autosomal. (The other purine phosphoribosyltransferase (HGPRT) is
- determined by an X-linked locus and is mutant in the Lesch-Nyhan
- syndrome (308000).) The heat-stable enzyme allele has a frequency of
- about 15% and the heat-labile enzyme allele a frequency of about 85%.
- Kelley et al. (1968) found apparent heterozygosity in 4 persons in 3
- generations of a family. The level of enzyme activity ranged from 21 to
- 37%, requiring some special explanation. That the enzyme is a dimer is
- one possibility. Fox et al. (1973) described a second family with
- partial deficiency of red cell APRT. Delbarre et al. (1974) found
- deficiency of APRT in persons with gout but recognized that purine
- overproduction was not necessarily caused by the APRT deficiency.
- Emmerson et al. (1975) described a family with dominant inheritance of
- APRT deficiency. Although the proband was a female with gout, a
- relationship to the APRT deficiency was considered unproved. The
- partially purified enzyme showed no difference in Michaelis constants,
- heat stability, or electrophoresis.
-
- Debray et al. (1976) observed a child with urolithiasis and complete
- deficiency of APRT. Both parents had partial deficiency. Van Acker et
- al. (1977) described brothers with complete deficiency of APRT. They
- were detected by the fact that one had from birth excreted gravel
- consisting of stones of 2,8-dihydroxyadenine in urine. Neither showed
- hyperuricemia or gout. Treatment with allopurinol and a low purine diet
- stopped stone formation. Homozygotes can be detected by raised urinary
- adenine levels and absence of detectable red cell APRT. Rappaport and
- DeMars (1973) identified clones of cells resistant to 2,6-diaminopurine
- (DAP) in skin fibroblast cultures derived from 13 of 21 normal humans.
- In some of the mutant cultures adenine phosphoribosyltransferase was
- normal. Two mutants from unrelated boys had little or no detectable APRT
- activity. Resistance resulted from reduced ability to convert DAP to its
- toxic ribonucleotide. The authors reasoned that mutant-yielding cultures
- were heterozygous to begin with. If so, DAP resistance has a
- heterozygote frequency as high as 0.2. This contrasts with the very low
- frequency of electrophoretic variants of APRT. There may be other
- mechanisms (mutation at other loci) for DAP-resistance. Azaguanine
- resistance is determined by mutation at the X-linked HGPRT locus.
- Barratt et al. (1979) reported a child of consanguineous Arab parents,
- the third case in which 2,8-dihydroxyadenine stones have been identified
- as the result of complete lack of APRT. Kishi et al. (1984) found only
- 10 reported cases of complete deficiency of APRT, beginning with the
- case of Cartier et al. (1974). Kishi et al. (1984) reported 3 cases in 2
- families. Although APRT deficiency occurred in mononuclear cells and
- polymorphonuclear leukocytes as well as in red cells, no abnormality of
- immunologic or phagocytic function was detected. The sole clinical
- manifestation was urinary calculi composed of 2,8-DHA. In Japanese,
- partial deficiency of APRT leads to 2,8-dihydroxyadenine urolithiasis,
- whereas all Caucasian patients with 2,8-DHA urolithiasis have been
- completely deficient. Fujimori et al. (1985) found that partially
- purified enzyme from Japanese families has a reduced affinity for
- phosphoribosylpyrophosphate (PRPP), as well as increased resistance to
- heat and reduced sensitivity to the stabilizing effect of PRPP. They
- referred to this common Japanese mutant allele as APRT*J. Kamatani et
- al. (1987) examined samples from 19 Japanese families with
- DHA-urolithiasis. In 15 of the 19 families, the patients had only
- partial APRT deficiency. All patients with DHA-urolithiasis were
- homozygotes regardless of whether the deficiency was complete or
- partial. They estimated that about 1% of the Japanese population are
- carriers. Kamatani et al. (1987) described a method for identifying
- heterozygotes for the Japanese allele of APRT. Manyak et al. (1987)
- found DHA-urolithiasis in a 50-year-old white woman. The patient was
- homozygous for APRT deficiency. Glicklich et al. (1988) reported the
- second case of homozygous APRT deficiency from the United States. The
- disorder was recognized 23 years after the patient, a black woman from
- Bermuda, had her initial episode of renal colic, and after
- 2,8-dihydroxyadenine stones had recurred after renal transplant.
- Ishidate et al. (1991) reported father and daughter with
- DHA-urolithiasis. The father and his wife were first cousins; thus, this
- was an example of pseudodominance.
-
- Gault et al. (1981) described 2,8-dihydroxyadenine urolithiasis in a
- white woman who lived in Newfoundland and first developed symptoms of
- urolithiasis at the age of 42. The use of infrared or x-ray diffraction
- analysis of calculi that are positive for uric acid with standard wet
- chemical tests can make the diagnosis. Adults may first present with
- renal failure. Renal biopsy shows changes like those of uric acid
- nephropathy. Maddocks and Al-Safi (1988) used identification of adenine
- in the urine by thin layer chromatography to diagnose APRT deficiency.
- Simmonds et al. (1992) pointed out that patients who are mistakenly
- diagnosed as having uric acid lithiasis will be treated successfully
- with allopurinol despite the incorrect diagnosis. This may be
- responsible for underdiagnosis of the disorder. Families carrying the
- mutant APRT gene need to be aware of it since acute renal failure may be
- the presenting symptom and this may be reversible, though some patients
- progress to chronic renal failure requiring dialysis and
- transplantation. Maddocks (1992) described a simple test for
- distinguishing uric acid calculi from 2,8-DHA calculi. Ward and Addison
- (1992) indicated that even visual examination can distinguish the two:
- 2,8-DHA stones are reddish-brown when wet and grayish when dry; they are
- also very soft and friable. Stones composed mainly of uric acid are very
- rare in children. Laxdal and Jonasson (1988) found 2 children and 2
- adults in 4 unrelated families with 2,8-dihydroxyadenine crystalluria.
- They suggested that the presence of round, brownish urine crystals, even
- without radiolucent kidney stones, should alert the physician to the
- diagnosis. Thirteen heterozygotes were identified by study of the
- families. Laxdal (1992) pointed out that Iceland contributed 8 of the 62
- APRT-deficient type I homozygotes. The 8 cases were from 8 different
- families. Although remote ancestral connections were identified, all 8
- cases were detected by the finding of typical round reddish-brown
- crystals in the urine on light microscopy. The importance of alert
- laboratory technicians in making the diagnosis was emphasized.
-
- By cell hybridization studies, Tischfield and Ruddle (1974) concluded
- that the APRT locus is on chromosome 16. Marimo and Giannelli (1975)
- confirmed this assignment by demonstrating a 1.69-fold increase in
- enzyme level in trisomy 16 cells. The same cells showed no difference in
- the levels of HGPRT, G6PD (305900) or adenosine kinase (102750) from
- controls. Barg et al. (1982) assigned APRT to 16q12-pter. Lavinha et al.
- (1984) assigned APRT and DIA4 (125860) to 16q12-q22 by study of
- rearranged chromosomes 16 in somatic cell hybrids. For APRT,
- Ferguson-Smith and Cox (1984) found a smallest region of overlap (SRO)
- of 16q22.2-q22.3. Castiglione et al. (1985) found no evidence of linkage
- between HP (140100) and HPRT within 12 map units, despite both loci
- having been mapped to band 16q22. Fratini et al. (1986) mapped the APRT
- locus with respect to the HP locus and the fragile site at 16q23.2
- (FRA16D). A subclone of the APRT gene and a cDNA clone of HP were used
- for molecular hybridization to DNA from mouse-human hybrid cell lines
- containing specific chromosome 16 translocations. The APRT subclone was
- used for in situ hybridization to chromosomes expressing FRA16D. APRT
- was found to be distal to HP and FRA16D and was localized at 16q24,
- making the gene order cen--FRA16B--HP--FRA16D--APRT--qter. Broderick et
- al. (1987) found that in species as widely separated in evolution as
- man, mouse, hamster, and E. coli, CpG dinucleotides are conserved at a
- frequency higher than expected on the basis of randomness considering
- the G+C content of the gene. This suggested some importance of this
- sequence to the function of the gene. Although the intron I sequences of
- mouse and man had no apparent homology, both had retained a very high
- CpG content. The APRT gene is about 2.6 kb long and contains 5 exons.
- The promoter region of the human APRT gene, like that of several other
- 'housekeeping' genes, lacks the 'TATA' and 'CCAAT' boxes but contains 5
- GC boxes that are potential binding sites for the Sp1 transcription
- factor. Hidaka et al. (1987) also prepared a complete sequence of the
- APRT gene and found a number of discrepancies from the sequence reported
- by Broderick et al. (1987), all occurring within noncoding regions.
- Hakoda et al. (1990) made the interesting observation that 2-step
- mutations leading to homozygous deficiencies at the somatic cell level,
- as proposed by the Knudson hypothesis of carcinogenesis in
- retinoblastoma (180200) and some other human tumors, occur at other
- autosomal loci. They cloned and enumerated somatic T cells with
- mutations at the APRT locus by taking advantage of the presence of
- heterozygous APRT deficiency and an effective selection procedure for
- homozygosity. They cultured peripheral blood mononuclear cells with
- 2,6-diaminopurine, an APRT-dependent cytotoxin, to search for in vivo
- mutational cells. In all 4 heterozygotes studied, homozygously deficient
- T cells were found, at an average frequency of 1.3 x 10(-4). Among 310
- normal persons, Hakoda et al. (1990) identified only 1 homozygous
- APRT-deficient clone, with a calculated frequency of 5.0 x 10(-9).
- Homozygous cells were found at rather high frequencies in 15 putative
- heterozygotes, as reported by Hakoda et al. (1991). Analysis of genomic
- DNA in 82 resistant clones from 2 of the heterozygotes showed that 64
- (78%) had lost the germinally intact alleles. This approach may prove
- useful for identifying heterozygotes for other enzyme deficiencies.
-
- Kamatani et al. (1992) stated that about 70 Japanese families with
- homozygous APRT deficiency have been reported, whereas the number of
- reported non-Japanese families is about 36. The estimated gene frequency
- among Japanese is about 1.2%.
-
- Terai et al. (1995) detected homozygous APRT deficiency by the finding
- of 2,8-dihydroxyadenine-like spherical crystals in the urinary sediment.
- The molecular diagnosis was established using PCR-SSCP with the
- demonstration of the APRT*J allele (102600.0003).
-
- According to the numerology used by Hidaka et al. (1988), the adenine in
- the initiation codon ATG is counted as nucleotide no. 1 and the
- initiator methionine is counted as amino acid no. 1.
-
- Engle et al. (1996) used targeted homologous recombination in embryonic
- stem cells to produce mice that lack APRT. Mice homozygous for a null
- Aprt allele excreted adenine and DHA crystals in their urine. Renal
- histopathology showed extensive tubular dilation, inflammation,
- necrosis, and fibrosis that varied in severity between different mouse
- backgrounds.
-
- *FIELD* AV
- .0001
- APRT DEFICIENCY
- APRT, PHE173DEL
- In cell line '904,' a lymphoblastoid cell line from a Caucasian patient
- in Belgium, Hidaka et al. (1987) studied the molecular basis of APRT
- deficiency by sequencing both alleles of a patient with complete
- deficiency. In 1 allele, a trinucleotide deletion, TTC at positions 2179
- to 2181 in exon 4, which corresponded to phenylalanine-173 in the
- deduced amino acid sequence, was demonstrated. In the other allele, a
- single nucleotide insertion, a T, was found immediately adjacent to the
- splice site at the 5-prime end of intron 4. This insertion led to
- aberrant splicing, as was demonstrated by the absence of exon 4 in the
- cDNA and by altered RNase mapping analysis of the abnormal mRNA.
- Frameshift led to premature termination at amino acid 110. The enzyme
- activity was less than 1% of normal and the enzyme protein was
- immunologically undetectable.
-
- .0002
- APRT DEFICIENCY
- APRT, IVS4DS INS T
- In the second allele of cell line '904,' Hidaka et al. (1987) found
- insertion of a thymine at the 5-prime end of intron 4 between
- nucleotides 1834 and 1835 resulting in deletion of exon 4 and frameshift
- with premature termination at amino acid 110. The insertion changed the
- IVS4 splice donor site from gtaa to gttaa. In identical twin brothers
- born to nonconsanguineous German parents, Gathof et al. (1991)
- demonstrated that the cause of APRT deficiency was a single base
- insertion, a T, between bases 1831 and 1832 or 1832 and 1833. (In the
- numbering system they used, nucleotide 1831 is the first in intron 4.
- The insertion changed the donor site from gtaa to gttaa.) The insertion
- altered the consensus sequence at the splice donor site between exon 4
- and intron 4, leading to aberrant splicing. They quoted finding of the
- same mutation in 2 other Caucasian patients living in the U.S. and as
- one of 2 alleles in a Belgian patient with compound heterozygosity. This
- is the same mutation as that found by Hidaka et al. (1987).
-
- .0003
- APRT DEFICIENCY, JAPANESE TYPE
- APRT*J
- APRT, MET136THR
- Hidaka et al. (1988) identified a T-to-C substitution in exon 5 at
- position 2069, giving rise to substitution of threonine for methionine
- at position 136 in the Japanese-type APRT deficiency. The enzyme showed
- abnormal kinetics and activity that was less than 10.3% of normal. Six
- other Japanese homozygotes carried the same mutation on at least 1
- allele. In the Japanese type of APRT deficiency, Kamatani et al. (1989)
- took advantage of the fact that the only methionine residue in normal
- APRT (at position 136) has been changed to threonine. By means of
- specific cleavage of the peptide at the methionine residue with cyanogen
- bromide (BrCN), they could distinguish normal from mutant proteins.
- Kamatani et al. (1989) found that 79% of all Japanese patients with this
- disease and more than half of the world's patients have this particular
- mutation. Kamatani et al. (1990) found that 24 of 39 Japanese
- 2,8-dihydroxyadenine urolithiasis patients had only APRT*J alleles. They
- found that normal alleles occur in 4 major haplotypes, whereas all
- APRT*J alleles occurred in only 2. They interpreted this as meaning that
- all APRT*J alleles had a single origin and that this mutant sequence has
- been maintained for a long time, as reflected in the frequency of the
- recombinant alleles. Sahota et al. (1991) described DHA-lithiasis in a
- patient heterozygous for the Japanese mutation. Lithiasis had previously
- been observed only in homozygotes. The polyamine pathway is thought to
- be the major source of endogenous adenine in the human. Whether
- increased polyamine synthesis can lead to increased adenine production,
- enhancer to DHA-lithiasis in an APRT heterozygote, remains to be
- determined. Among 141 defective APRT alleles from 72 different Japanese
- families, Kamatani et al. (1992) found the met136-to-thr mutation in 96
- (68%); 30 (21%) and 10 (7%) had the TGG-to-TGA nonsense mutation at
- codon 98 (102600.0005) and duplication of a 4-bp sequence in exon 3
- (102600.0006), respectively.
-
- .0004
- APRT DEFICIENCY, COMPLETE, ICELANDIC TYPE
- APRT, ASP65VAL
- Chen et al. (1990) analyzed the molecular nature of the mutation in all
- 5 patients with complete APRT deficiency reported from Iceland. The same
- mutation, an A-to-T transversion at position 1350, was identified in all
- of the patients (the A of the ATG start codon was designated number 1).
- The substitution led to the replacement of aspartic acid (GAC) by valine
- (GTC) at amino acid 65 in exon 3. In all 5 patients the mutation was
- homozygous. Common ancestors could be identified for only 2 of the
- cases.
-
- .0005
- APRT DEFICIENCY DUE TO TYPE I ALLELE
- APRT, TRP98TER
- Mimori et al. (1991) analyzed 7 APRT*Q0 (null) alleles from 4 unrelated
- Japanese subjects (3 homozygotes and a heterozygote). In all 7, they
- found a G-to-A transition at nucleotide position 1453, which changed
- tryptophan-98 to a stop codon. There was also a C-to-T transition at
- 1456, which did not alter alanine-99. The G-to-A change at 1453 resulted
- in the elimination of a PflMI site in the APRT gene.
-
- .0006
- APRT DEFICIENCY
- APRT, 4-BP DUP, EX3
- Among 141 defective APRT alleles from 72 different Japanese families,
- Kamatani et al. (1992) found that 10 (7%) had duplication of a CCGA
- sequence in exon 3. Duplication resulted in an APRT*Q0 (null) allele.
- Two other alleles, APRT*J (102600.0003) and trp98-to-ter (102600.0005),
- accounted for 68% and 21%, respectively. The different alleles with the
- same mutation had the same haplotype, except for APRT*J. Evidence for a
- crossover or a gene conversion event within the APRT gene was observed
- in an APRT*J mutant allele.
-
- .0007
- APRT DEFICIENCY
- APRT, LEU110PRO
- Sahota et al. (1994) described 2 sisters from Newfoundland who carried a
- leucine-to-proline missense transition at codon position 110 (nucleotide
- position 1759). One of the sisters exhibited 2,8-dihyroxyadenine
- urolithiasis, whereas the other was disease-free. Restriction mapping
- and DNA sequence data were compatible with both sisters being homozygous
- for the mutation, although hemizygosity could not be ruled out.
-
- *FIELD* SA
- Doppler et al. (1981); Fox et al. (1977); Hidaka et al. (1987); Hirsch-Kauffmann
- and Doppler (1981); Johnson et al. (1977); Kamatani et al. (1990);
- Kamatani et al. (1987); Lester et al. (1980); Nesterova et al. (1987);
- Simmonds (1979); Simon and Taylor (1983); Takeuchi et al. (1985);
- Wilson et al. (1986)
- *FIELD* RF
- 1. Barg, R.; Barton, P.; Caine, A.; Clements, R. L.; Ferguson-Smith,
- M. A.; Malcolm, S.; Morrison, N.; Murphy, C. S.: Regional localization
- of the human alpha-globin gene to the short arm of chromosome 16 (16p12-pter)
- using both somatic cell hybrids and in situ hybridization. Cytogenet.
- Cell Genet. 32: 252-253, 1982.
-
- 2. Barratt, T. M.; Simmonds, H. A.; Cameron, J. S.; Potter, C. F.;
- Rose, G. A.; Arkell, D. G.; Williams, D. I.: Complete deficiency
- of adenine phosphoribosyltransferase: a third case presenting as renal
- stones in a young child. Arch. Dis. Child. 54: 25-31, 1979.
-
- 3. Broderick, T. P.; Schaff, D. A.; Bertino, A. M.; Dush, M. K.; Tischfield,
- J. A.; Stambrook, P. J.: Comparative anatomy of the human APRT gene
- and enzyme: nucleotide sequence divergence and conservation of a nonrandom
- CpG dinucleotide arrangement. Proc. Nat. Acad. Sci. 84: 3349-3353,
- 1987.
-
- 4. Cartier, P.; Hamet, M.; Hamburger, J.: Une nouvelle maladie metabolique:
- le deficit complet en adenine phosphoribosyltransferase avec lithiase
- de 2,8-dihydroxyadenine. C. R. Seances Acad. Sci. 279: 883-886,
- 1974.
-
- 5. Castiglione, C. M.; Kidd, J. R.; Tischfield, J. A.; Stambrook,
- P. J.; Murphy, P. D.; Sparkes, R. A.; Kidd, K. K.: Polymorphism and
- linkage of APRT.(Abstract) Cytogenet. Cell Genet. 40: 601 only,
- 1985.
-
- 6. Chen, J.; Sahota, A.; Laxdal, T.; Stambrook, P. J.; Tischfield,
- J. A.: Demonstration of a common mutation at the adenine phosphoribosyltransferase
- (APRT) locus in the Icelandic population.(Abstract) Am. J. Hum. Genet. 47
- (suppl.): A152 only, 1990.
-
- 7. Debray, H.; Cartier, P.; Temstet, A.; Cendron, J.: Child's urinary
- lithiasis revealing a complete deficit in adenine phosphoribosyl transferase.
- Pediat. Res. 10: 762-766, 1976.
-
- 8. Delbarre, F.; Aucher, C.; Amor, B.; de Gery, A.; Cartier, P.; Hamet,
- M.: Gout with adenine phosphoribosyltransferase deficiency. Biomedicine 21:
- 82-85, 1974.
-
- 9. Doppler, W.; Hirsch-Kauffmann, M.; Schabel, F.; Schweiger, M.:
- Characterization of the biochemical basis of a complete deficiency
- of the adenine phosphoribosyl transferase (APRT). Hum. Genet. 57:
- 404-410, 1981.
-
- 10. Emmerson, B. T.; Gordon, R. B.; Thompson, L.: Adenine phosphoribosyltransferase
- deficiency: its inheritance and occurrence in a female with gout and
- renal disease. Aust. New Zeal. J. Med. 5: 440-446, 1975.
-
- 11. Engle, S. J.; Stockelman, M. G.; Chen, J.; Boivin, G.; Yum, M.-N.;
- Davies, P. M.; Ying, M. Y.; Sahota, A.; Simmonds, H. A.; Stambrook,
- P. J.; Tischfield, J. A.: Adenine phosphoribosyltransferase-deficient
- mice develop 2,8-dihydroxyadenine nephrolithiasis. Proc. Nat. Acad.
- Sci. 93: 5307-5312, 1996.
-
- 12. Ferguson-Smith, M. A.; Cox, D. R.: Report of the committee on
- the genetic constitution of chromosomes 13, 14, 15, 16 and 17. Cytogenet.
- Cell Genet. 37: 127-154, 1984.
-
- 13. Fox, I. H.; Lacroix, S.; Planet, G.; Moore, M.: Partial deficiency
- of adenine phosphoribosyltransferase in man. Medicine 56: 515-526,
- 1977.
-
- 14. Fox, I. H.; Meade, J. C.; Kelley, W. N.: Adenine phosphoribosyltransferase
- deficiency in man: report of a second family. Am. J. Med. 55: 614-619,
- 1973.
-
- 15. Fratini, A.; Simmers, R. N.; Callen, D. F.; Hyland, V. J.; Tischfield,
- J. A.; Stambrook, P. J.; Sutherland, G. R.: A new location for the
- human adenine phosphoribosyltransferase gene (APRT) distal to the
- haptoglobin (HP) and fra(16)(q23) (FRA16D) loci. Cytogenet. Cell
- Genet. 43: 10-13, 1986.
-
- 16. Fujimori, S.; Akaoka, I.; Sakamoto, K.; Yamanaka, H.; Nishioka,
- K.; Kamatani, N.: Common characteristics of mutant adenine phosphoribosyltransferases
- from four separate Japanese families with 2,8-dihydroxyadenine urolithiasis
- associated with partial enzyme deficiencies. Hum. Genet. 71: 171-176,
- 1985.
-
- 17. Gathof, B. S.; Sahota, A.; Gresser, U.; Chen, J.; Stambrook, P.
- J.; Tischfield, J. A.; Zollner, N.: Identification of a splice mutation
- at the adenine phosphoribosyltransferase locus in a German family.
- Klin. Wschr. 69: 1152-1155, 1991.
-
- 18. Gault, M. H.; Simmonds, H. A.; Snedden, W.; Dow, D.; Churchill,
- D. N.; Penney, H.: Urolithiasis due to 2,8-dihydroxyadenine in an
- adult. New Eng. J. Med. 305: 1570-1572, 1981.
-
- 19. Glicklich, D.; Gruber, H. E.; Matas, A. J.; Tellis, V. A.; Karwa,
- G.; Finley, K.; Salem, C.; Soberman, R.; Seegmiller, J. E.: 2,8-Dihydroxyadenine
- urolithiasis: report of a case first diagnosed after renal transplant.
- Quart. J. Med. (N.S.) 69: 785-793, 1988.
-
- 20. Hakoda, M.; Nishioka, K.; Kamatani, N.: Homozygous deficiency
- at autosomal locus APRT in human somatic cells in vivo induced by
- two different mechanisms. Cancer Res. 50: 1738-1741, 1990.
-
- 21. Hakoda, M.; Yamanaka, H.; Kamatani, N.; Kamatani, N.: Diagnosis
- of heterozygous states for adenine phosphoribosyltransferase deficiency
- based on detection of in vivo somatic mutants in blood T cells: application
- to screening of heterozygotes. Am. J. Hum. Genet. 48: 552-562,
- 1991.
-
- 22. Henderson, J. F.; Kelley, W. N.; Rosenbloom, F. M.; Seegmiller,
- J. E.: Inheritance of purine phosphoribosyltransferases in man. Am.
- J. Hum. Genet. 21: 61-70, 1969.
-
- 23. Hidaka, Y.; Palella, T. D.; O'Toole, T. E.; Tarle, S. A.; Kelley,
- W. N.: Human adenine phosphoribosyltransferase: identification of
- allelic mutations at the nucleotide level as a cause of complete deficiency
- of the enzyme. J. Clin. Invest. 80: 1409-1415, 1987.
-
- 24. Hidaka, Y.; Tarle, S. A.; Fujimori, S.; Kamatani, N.; Kelley,
- W. N.; Palella, T. D.: Human adenine phosphoribosyltransferase deficiency:
- demonstration of a single mutant allele common to the Japanese. J.
- Clin. Invest. 81: 945-950, 1988.
-
- 25. Hidaka, Y.; Tarle, S. A.; O'Toole, T. E.; Kelley, W. N.; Palella,
- T. D.: Nucleotide sequence of the human APRT gene. Nucleic Acids
- Res. 15: 9086, 1987.
-
- 26. Hirsch-Kauffmann, M.; Doppler, W.: Biochemical studies on a patient
- with complete APRT-deficiency.(Abstract) Sixth Int. Cong. Hum. Genet.,
- Jerusalem 96 only, 1981.
-
- 27. Ishidate, T.; Igarashi, S.; Kamatani, N.: Pseudodominant transmission
- of an autosomal recessive disease, adenine phosphoribosyltransferase
- deficiency. J. Pediat. 118: 90-91, 1991.
-
- 28. Johnson, L. A.; Gordon, R. B.; Emmerson, B. T.: Adenine phosphoribosyltransferase:
- a simple spectrophotometric assay and the incidence of mutation in
- the normal population. Biochem. Genet. 15: 265-272, 1977.
-
- 29. Kamatani, N.; Hakoda, M.; Otsuka, S.; Yoshikawa, H.; Kashiwazaki,
- S.: Only three mutations account for almost all defective alleles
- causing adenine phosphoribosyltransferase deficiency in Japanese patients.
- J. Clin. Invest. 90: 130-135, 1992.
-
- 30. Kamatani, N.; Kuroshima, S.; Hakoda, M.; Palella, T. D.; Hidaka,
- Y.: Crossovers within a short DNA sequence indicate a long evolutionary
- history of the APRT*J mutation. Hum. Genet. 85: 600-604, 1990.
-
- 31. Kamatani, N.; Kuroshima, S.; Terai, C.; Hidaka, Y.; Palella, T.
- D.; Nishioka, K.: Detection of an amino acid substitution in the
- mutant enzyme for a special type of adenine phosphoribosyltransferase
- (APRT) deficiency by sequence-specific protein cleavage. Am. J.
- Hum. Genet. 45: 325-331, 1989.
-
- 32. Kamatani, N.; Kuroshima, S.; Terai, C.; Kawai, K.; Mikanagi, K.;
- Nishioka, K.: Selection of human cells having two different types
- of mutations in individual cells (genetic/artificial mutants): application
- to the diagnosis of the heterozygous state for a type of adenine phosphoribosyltransferase
- deficiency. Hum. Genet. 76: 148-152, 1987.
-
- 33. Kamatani, N.; Kuroshima, S.; Yamanaka, H.; Nakashe, S.; Take,
- H.; Hakoda, M.: Identification of a compound heterozygote for adenine
- phosphoribosyltransferase deficiency (APRT*J/APRT*Q0) leading to 2,8-dihydroxyadenine
- urolithiasis. Hum. Genet. 85: 500-504, 1990.
-
- 34. Kamatani, N.; Terai, C.; Kuroshima, S.; Nishioka, K.; Mikanagi,
- K.: Genetic and clinical studies on 19 families with adenine phosphoribosyltransferase
- deficiencies. Hum. Genet. 75: 163-168, 1987.
-
- 35. Kelley, W. N.; Levy, R. I.; Rosenbloom, F. M.; Henderson, J. F.;
- Seegmiller, J. E.: Adenine phosphoribosyltransferase deficiency:
- a previously undescribed genetic defect in man. J. Clin. Invest. 47:
- 2281-2289, 1968.
-
- 36. Kishi, T.; Kidani, K.; Komazawa, Y.; Sakura, N.; Matsuura, R.;
- Kobayashi, M.; Tanabe, A.; Hyodo, S.; Kittaka, E.; Sakano, T.; Tanaka,
- Y.; Kobayashi, Y.; Nakamoto, T.; Nakatsu, H.; Moriyama, H.; Hayashi,
- M.; Nihira, H.; Usui, T.: Complete deficiency of adenine phosphoribosyltransferase:
- a report of three cases and immunologic and phagocytic investigations.
- Pediat. Res. 18: 30-34, 1984.
-
- 37. Lavinha, J.; Morrison, N.; Glasgow, L.; Ferguson-Smith, M. A.
- : Further evidence for the regional localization of human APRT and
- DIA4 on chromosome 16.(Abstract) Cytogenet. Cell Genet. 37: 517
- only, 1984.
-
- 38. Laxdal, T.: 2,8-Dihydroxyadenine crystalluria vs urolithiasis.(Letter) Lancet 340:
- 184 only, 1992.
-
- 39. Laxdal, T.; Jonasson, T. A.: Adenine phosphoribosyltransferase
- deficiency in Iceland. Acta Med. Scand. 224: 621-626, 1988.
-
- 40. Lester, S. C.; LeVan, S. K.; Steglich, C.; DeMars, R.: Expression
- of human genes of adenine phosphoribosyltransferase and hypoxanthine-guanine
- phosphoribosyltransferase after genetic transformation of mouse cells
- with purified human DNA. Somat. Cell Genet. 6: 241-259, 1980.
-
- 41. Maddocks, J. L.: 2,8-Dihydroxyadenine urolithiasis.(Letter) Lancet 339:
- 1296 only, 1992.
-
- 42. Maddocks, J. L.; Al-Safi, S. A.: Adenine phosphoribosyltransferase
- deficiency: a simple diagnostic test. Clin. Sci. 75: 217-220, 1988.
-
- 43. Manyak, M. J.; Frensilli, F. J.; Miller, H. C.: 2,8-Dihydroxyadenine
- urolithiasis: report of an adult case in the United States. J. Urol. 137:
- 312-314, 1987.
-
- 44. Marimo, B.; Giannelli, F.: Gene dosage effect in human trisomy
- 16. Nature 256: 204-206, 1975.
-
- 45. Mimori, A.; Hidaka, Y.; Wu, V. C.; Tarle, S. A.; Kamatani, N.;
- Kelley, W. N.; Pallela, T. D.: A mutant allele common to the type
- I adenine phosphoribosyltransferase deficiency in Japanese subjects.
- Am. J. Hum. Genet. 48: 103-107, 1991.
-
- 46. Nesterova, T. B.; Borodin, P. M.; Zakian, S. M.; Serov, O. L.
- : Assignment of the gene for adenine phosphoribosyltransferase on
- the genetic map of mouse chromosome 8. Biochem. Genet. 25: 563-568,
- 1987.
-
- 47. Rappaport, H.; DeMars, R.: Diaminopurine-resistant mutants of
- cultured, diploid human fibroblasts. Genetics 75: 335-345, 1973.
-
- 48. Sahota, A.; Chen, J.; Behzadian, M. A.; Ravindra, R.; Takeuchi,
- H.; Stambrook, P. J.; Tischfield, J. A.: 2,8-Dihydroxyadenine lithiasis
- in a Japanese patient heterozygous at the adenine phosphoribosyltransferase
- locus. Am. J. Hum. Genet. 48: 983-989, 1991.
-
- 49. Sahota, A.; Chen, J.; Boyadijev, S. A.; Gault, M. H.; Tischfield,
- J. A.: Missense mutation in the adenine phosphoribosyltransferase
- gene causing 2,8-dihydroxyadenine urolithiasis. Hum. Molec. Genet. 3:
- 817-818, 1994.
-
- 50. Simmonds, H. A.: 2,8-Dihydroxyadeninuria--or when is a uric acid
- stone not a uric acid stone?. Clin. Nephrol. 12: 195-197, 1979.
-
- 51. Simmonds, H. A.; Van Acker, K. J.; Sahota, A. S.: 2,8-Dihydroxyadenine
- urolithiasis.(Letter) Lancet 339: 1295-1296, 1992.
-
- 52. Simon, A. E.; Taylor, M. W.: High-frequency mutation at the adenine
- phosphoribosyltransferase locus in Chinese hamster ovary cells due
- to deletion of the gene. Proc. Nat. Acad. Sci. 80: 810-814, 1983.
-
- 53. Takeuchi, F.; Matsuta, K.; Miyamoto, T.; Enomoto, S.; Fujimori,
- S.; Akaoka, I.; Kamatani, N.; Nishioka, K.: Rapid method for the
- diagnosis of partial adenine phosphoribosyltransferase deficiencies
- causing 2,8-dihydroxyadenine urolithiasis. Hum. Genet. 71: 167-170,
- 1985.
-
- 54. Terai, C.; Hakoda, M.; Yamanaka, H.; Kamatani, N.; Okai, M.; Takahashi,
- F.; Kashiwazaki, S.: Adenine phosphoribosyltransferase deficiency
- identified by urinary sediment analysis: cellular and molecular confirmation. Clin.
- Genet. 48: 246-250, 1995.
-
- 55. Tischfield, J. A.; Ruddle, F. H.: Assignment of the gene for
- adenine phosphoribosyltransferase to human chromosome 16 by mouse-human
- somatic cell hybridization. Proc. Nat. Acad. Sci. 71: 45-49, 1974.
-
- 56. Van Acker, K. J.; Simmonds, H. A.; Potter, C.; Cameron, J. S.
- : Complete deficiency of adenine phosphoribosyltransferase: report
- of a family. New Eng. J. Med. 297: 127-132, 1977.
-
- 57. Ward, I. D.; Addison, G. M.: 2,8-Dihydroxyadenine urolithiasis.
- (Letter) Lancet 339: 1296, 1992.
-
- 58. Wilson, J. M.; O'Toole, T. E.; Argos, P.; Shewach, D. S.; Daddona,
- P. E.; Kelley, W. N.: Human adenine phosphoribosyltransferase: complete
- amino acid sequence of the erythrocyte enzyme. J. Biol. Chem. 261:
- 13677-13683, 1986.
-
- *FIELD* CS
-
- GU:
- Urolithiasis;
- Renal failure
-
- Lab:
- APRT deficiency;
- 2,8-dihydroxyadenine urinary stones;
- Round, brownish urine crystals
-
- Inheritance:
- Autosomal dominant (16q22.2-q22.3), with homozygosity or compound
- heterozygosity in complete deficiency
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 07/06/1996
- mark: 6/24/1996
- terry: 6/12/1996
- carol: 5/18/1996
- mark: 1/17/1996
- pfoster: 11/29/1994
- mimadm: 4/14/1994
- warfield: 4/6/1994
- carol: 7/9/1993
- carol: 2/17/1993
- carol: 10/28/1992
-
- *RECORD*
- *FIELD* NO
- 102610
- *FIELD* TI
- *102610 ACTIN, ALPHA, SKELETAL MUSCLE 1; ACTA1
- ASMA
- *FIELD* TX
- By use of a cDNA probe in somatic cell hybrids, Hanauer et al. (1984)
- assigned the gene for the alpha chain of skeletal muscle actin to
- chromosome 1. Actin sequences were found at high stringency also at
- 2p23-qter and 3pter-q21. Under conditions of low or medium stringency,
- actin sequences were demonstrated on the X (p11-p12) and Y chromosomes.
- Using a cDNA copy of the 3-prime untranslated region of the human
- skeletal alpha actin gene, Shows et al. (1984) mapped the gene to
- 1p12-1qter. This gene and that for cardiac alpha-actin (102540) are
- coexpressed in both human skeletal muscle and heart. Coexpression is not
- a function of linkage; the loci are on separate chromosomes: 1p21-qter
- and 15q11-qter, respectively (Gunning et al., 1984). Akkari et al.
- (1994) narrowed the assignment of the ACTA1 gene to 1q42 by fluorescence
- in situ hybridization. Also by fluorescence in situ hybridization,
- Ueyama et al. (1995) mapped the gene to 1q42.1. Using a panel of somatic
- cell hybrids, Alonso et al. (1993) confirmed the localization of the
- ACTA1 gene on human chromosome 1. On the basis of analysis of
- mouse/hamster somatic cell hybrids segregating mouse chromosomes,
- Czosnek et al. (1982) concluded that the skeletal actin gene is located
- on mouse chromosome 3. However, Alonso et al. (1993) found by PCR
- analysis of a microsatellite in an interspecific backcross that the
- gene, symbolized Actsk-1, is closely linked to tyrosine aminotransferase
- and adenine phosphoribosyltransferase on mouse chromosome 8. The Actsk-1
- gene is situated between Tat and Aprt; the human homologs TAT (276600)
- and APRT (102600) are on human chromosome 16. Abonia et al. (1993)
- likewise mapped the Actsk-1 gene to mouse chromosome 8 by segregation of
- RFLVs in 2 interspecific backcross sets and in 4 recombinant inbred (RI)
- mouse sets.
-
- Actin makes up 10 to 20% of cellular protein and has vital roles in cell
- integrity, structure, and motility. It is highly conserved throughout
- evolution. Its function depends on the balance between monomeric
- (globular) G-actin (42 kD) and filamentous F-actin, a linear polymer of
- G-actin subunits. Among the cytosolic actin-binding proteins, 3 appear
- to be of primary importance in limiting polymerization: profilin
- (176590, 176610), thymosin beta-4 (188395), and gelsolin (GSN; 137350).
- The existence of intracellular actin-binding proteins allows the
- concentration of G-actin to be maintained substantially above the
- threshold at which polymerization and the formation of filaments would
- normally occur. When released into the extracellular space, actin, which
- otherwise is known to have a pathologic effect, is bound by gelsolin and
- by the Gc protein (GC; 139200). This is the so-called extracellular
- actin-scavenger system (Lee and Galbraith, 1992).
-
- *FIELD* RF
- 1. Abonia, J. P.; Abel, K. J.; Eddy, R. L.; Elliott, R. W.; Chapman,
- V. M.; Shows, T. B.; Gross, K. W.: Linkage of Agt and Actsk-1 to
- distal mouse chromosome 8 loci: a new conserved linkage. Mammalian
- Genome 4: 25-32, 1993.
-
- 2. Akkari, P. A.; Eyre, H. J.; Wilton, S. D.; Callen, D. F.; Lane,
- S. A.; Meredith, C.; Kedes, L.; Laing, N. G.: Assignment of the human
- skeletal muscle alpha actin gene (ACTA1) to 1q42 by fluorescence in
- situ hybridisation. Cytogenet. Cell Genet. 65: 265-267, 1994.
-
- 3. Alonso, S.; Montagutelli, X.; Simon-Chazottes, D.; Guenet, J.-L.;
- Buckingham, M.: Re-localization of Actsk-1 to mouse chromosome 8,
- a new region of homology with human chromosome 1. Mammalian Genome 4:
- 15-20, 1993.
-
- 4. Czosnek, H.; Nudel, U.; Shani, M.; Barker, P. E.; Pravtcheva, D.
- D.; Ruddle, F. H.; Yaffe, D.: The genes coding for the muscle contractile
- proteins, myosin heavy chain, myosin light chain 2, and skeletal muscle
- actin are located on three different mouse chromosomes. EMBO J. 1:
- 1299-1305, 1982.
-
- 5. Gunning, P.; Ponte, P.; Kedes, L.; Eddy, R.; Shows, T.: Chromosomal
- location of the co-expressed human skeletal and cardiac actin genes. Proc.
- Nat. Acad. Sci. 81: 1813-1817, 1984.
-
- 6. Hanauer, A.; Heilig, R.; Levin, M.; Moisan, J. P.; Grzeschik, K.
- H.; Mandel, J. L.: The actin gene family in man: assignment of the
- gene for skeletal muscle alpha-actin to chromosome 1, and presence
- of actin sequences on autosomes 2 and 3, and on the X and Y chromosomes.
- (Abstract) Cytogenet. Cell Genet. 37: 487-488, 1984.
-
- 7. Lee, W. M.; Galbraith, R. M.: The extracellular actin-scavenger
- system and actin toxicity. New Eng. J. Med. 326: 1335-1341, 1992.
-
- 8. Shows, T.; Eddy, R. L.; Haley, L.; Byers, M.; Henry, M.; Gunning,
- P.; Ponte, P.; Kedes, L.: The coexpressed genes for human alpha (ACTA)
- and cardiac actin (ACTC) are on chromosomes 1 and 15, respectively.
- (Abstract) Cytogenet. Cell Genet. 37: 583 only, 1984.
-
- 9. Ueyama, H.; Inazawa, J.; Ariyama, T.; Nishino, H.; Ochiai, Y.;
- Ohkubo, I.; Miwa, T.: Reexamination of chromosomal loci of human
- muscle actin genes by fluorescence in situ hybridization. Jpn. J.
- Hum. Genet. 40: 145-148, 1995.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 03/20/1997
- terry: 6/16/1995
- carol: 5/27/1994
- carol: 2/3/1993
- carol: 5/28/1992
- supermim: 3/16/1992
- carol: 7/3/1991
-
- *RECORD*
- *FIELD* NO
- 102620
- *FIELD* TI
- *102620 ACTIN, ALPHA, SMOOTH MUSCLE, AORTIC; ACTSA
- ACTIN, ALPHA-2, SMOOTH MUSCLE, AORTA; ACTA2;;
- ACTIN, VASCULAR SMOOTH MUSCLE
- *FIELD* TX
- Six different actin isoforms have been identified in vertebrates by
- amino acid sequencing: skeletal muscle, cardiac muscle, 2 smooth muscle
- (enteric and aortic), and 2 cytoplasmic (beta and gamma) (Vandekerckhove
- and Weber, 1979). Their amino acid sequences are very similar and well
- conserved in evolution; e.g., skeletal and cardiac actins differ by only
- 4 amino acids, and skeletal muscle and cytoplasmic beta-actins differ by
- only 25 amino acids out of a total of 374. Ueyama et al. (1984) isolated
- and characterized the human aortic smooth muscle actin gene. It was
- found to contain 2 more introns than do skeletal and cardiac muscle
- actin genes: between codons 84 and 85 and 121 and 122. The gene also has
- a transition point mutation in position 309, substituting thymine for
- cytosine. Ueyama et al. (1990) assigned the ACTSA gene to chromosome 10
- by Southern blot analysis of DNAs from 18 rodent-human somatic cell
- hybrids. Regional mapping by in situ hybridization localized the gene to
- 10q22-q24. By fluorescence in situ hybridization, Ueyama et al. (1995)
- localized the ACTSA gene to 10q23.3.
-
- *FIELD* RF
- 1. Ueyama, H.; Bruns, G.; Kanda, N.: Assignment of the vascular smooth
- muscle actin gene ACTSA to human chromosome 10. Jpn. J. Hum. Genet. 35:
- 145-150, 1990.
-
- 2. Ueyama, H.; Hamada, H.; Battula, N.; Kakunaga, T.: Structure of
- a human smooth muscle actin gene (aortic type) with a unique intron
- site. Molec. Cell. Biol. 4: 1073-1078, 1984.
-
- 3. Ueyama, H.; Inazawa, J.; Ariyama, T.; Nishino, H.; Ochiai, Y.;
- Ohkubo, I.; Miwa, T.: Reexamination of chromosomal loci of human
- muscle actin genes by fluorescence in situ hybridization. Jpn. J.
- Hum. Genet. 40: 145-148, 1995.
-
- 4. Vandekerckhove, J.; Weber, K.: The complete amino acid sequence
- of actins from bovine aorta, bovine heart, bovine fast skeletal muscle,
- and rabbit slow skeletal muscle. Differentiation 14: 123-133, 1979.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 6/16/1995
- supermim: 3/16/1992
- carol: 2/27/1992
- carol: 7/3/1991
- carol: 3/19/1991
- carol: 9/27/1990
-
- *RECORD*
- *FIELD* NO
- 102630
- *FIELD* TI
- *102630 ACTIN, BETA; ACTB
- BETA-ACTIN
- *FIELD* TX
- From studies of the amino acid sequence of cytoplasmic and muscle
- actins, Vandekerckhove and Weber (1978) concluded that mammalian
- cytoplasmic actins are the products of 2 different genes and differ by
- many amino acids from muscle actin. In a neoplastic cell line resulting
- from treatment of cultured human diploid fibroblasts with a chemical
- mutagen, Leavitt et al. (1982) observed a mutant form of beta actin.
- Toyama and Toyama (1984) isolated and characterized lines of KB cells
- resistant to cytochalasin B. They found that one resistant line had an
- alteration in beta-actin. Such cells bound less cytochalasin B than did
- parental KB cells. The authors suggested that the primary site of action
- of cytochalasin B on cell motility processes is beta-actin.
-
- There are 6 known actin proteins in mammalian cells: 2 sarcomeric muscle
- actins (alpha-skeletal and alpha-cardiac), 2 smooth muscle actins (alpha
- and gamma), and 2 nonmuscle, cytoskeletal actins (beta and gamma) (Kedes
- et al., 1985). The genes of 3 of these have been mapped: beta-actin on
- chromosome 7, alpha-skeletal actin (102610) on chromosome 1, and
- alpha-cardiac actin (102540) on chromosome 15. Ng et al. (1985) assigned
- the ACTB gene to 7pter-q22 by Southern blot analysis of DNA from somatic
- cell hybrids. Habets et al. (1992) generated hybrids that harbor only
- specific regions of human chromosome 7 and assigned the ACTB locus to
- 7p15-p12.
-
- Ueyama et al. (1996) used fluorescence in situ hybridization to map ACTB
- to 7p22. By PCR of somatic cell hybrid DNAs, they mapped 4 ACTB
- pseudogenes to other chromosomes.
-
- - PSEUDOGENES
-
- Ng et al. (1985, 1985) showed that there are about 20 pseudogenes widely
- distributed in the genome. ACTBP1 is on Xq13-q22; ACTBP2, on chromosome
- 5; ACTBP3, on chromosome 18; ACTBP4, on chromosome 5 and ACTBP5, on
- 7q22-7qter. All have been mapped in somatic cell hybrids by use of DNA
- clones.
-
- *FIELD* SA
- Erba et al. (1988); Nakajima-Iijima et al. (1985)
- *FIELD* RF
- 1. Erba, H. P.; Eddy, R.; Shows, T.; Kedes, L.; Gunning, P.: Structure,
- chromosome location, and expression of the human gamma-actin gene:
- differential evolution, location, and expression of the cytoskeletal
- beta- and gamma-actin genes. Molec. Cell. Biol. 8: 1775-1789, 1988.
-
- 2. Habets, G. G. M.; van der Kammen, R. A.; Willemsen, V.; Balemans,
- M.; Wiegant, J.; Collard, J. G.: Sublocalization of an invasion-inducing
- locus and other genes on human chromosome 7. Cytogenet. Cell Genet. 60:
- 200-205, 1992.
-
- 3. Kedes, L.; Ng, S.-Y.; Lin, C.-S.; Gunning, P.; Eddy, R.; Shows,
- T.; Leavitt, J.: The human beta-actin multigene family. Trans. Assoc.
- Am. Phys. 98: 42-46, 1985.
-
- 4. Leavitt, J.; Bushar, G.; Kakunaga, T.; Hamada, H.; Hirakawa, T.;
- Goldman, D.; Merril, C.: Variations in expression of mutant beta-actin
- accompanying incremental increases in human fibroblast tumorigenicity. Cell 28:
- 259-268, 1982.
-
- 5. Nakajima-Iijima, S.; Hamada, H.; Reddy, P.; Kakunaga, T.: Molecular
- structure of the human cytoplasmic beta-actin gene; interspecies homology
- of sequences in the introns. Proc. Nat. Acad. Sci. 82: 6133-6137,
- 1985.
-
- 6. Ng, S.-Y.; Gunning, P.; Eddy, R.; Ponte, P.; Leavitt, J.; Kedes,
- L.; Shows, T.: Chromosome 7 assignment of the human beta-actin functional
- gene (ACTB) and the chromosomal dispersion of pseudogenes. (Abstract) Cytogenet.
- Cell Genet. 40: 712 only, 1985.
-
- 7. Ng, S.-Y.; Gunning, P.; Eddy, R.; Ponte, P.; Leavitt, J.; Shows,
- T.; Kedes, L.: Evolution of the functional human beta-actin gene
- and its multi-pseudogene family: conservation of the noncoding regions
- and chromosomal dispersion of pseudogenes. Molec. Cell. Biol. 5:
- 2720-2732, 1985.
-
- 8. Toyama, S.; Toyama, S.: A variant form of beta-actin in a mutant
- of KB cells resistant to cytochalasin B. Cell 37: 609-614, 1984.
-
- 9. Ueyama, H.; Inazawa, J.; Nishino, H.; Ohkubo, I.; Miwa, T.: FISH
- localization of human cytoplasmic actin genes ACTB to 7p22 and ACTG1
- to 17q25 and characterization of related pseudogenes. Cytogenet.
- Cell Genet. 74: 221-224, 1996.
-
- 10. Vandekerckhove, J.; Weber, K.: Mammalian cytoplasmic actins are
- the products of at least two genes and differ in primary structure
- in at least 25 identified positions from skeletal muscle actins. Proc.
- Nat. Acad. Sci. 75: 1106-1110, 1978.
-
- *FIELD* CN
- Mark H. Paalman - edited: 4/18/1997
- Mark H. Paalman - edited: 4/10/1997
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 04/18/1997
- mark: 4/18/1997
- jenny: 4/10/1997
- terry: 1/13/1997
- carol: 7/1/1993
- supermim: 3/16/1992
- carol: 2/29/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- carol: 5/18/1988
-
- *RECORD*
- *FIELD* NO
- ^102640
- *FIELD* TI
- ^102640 MOVED TO 102630
- *FIELD* TX
- This entry was incorporated into entry 102630 on 18 April 1997.
-
- *FIELD* CN
- Mark H. Paalman - edited: 04/18/1997
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
- *FIELD* ED
- mark: 04/18/1997
- supermim: 3/16/1992
- carol: 3/3/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
- *RECORD*
- *FIELD* NO
- 102642
- *FIELD* TI
- *102642 STEROL O-ACYLTRANSFERASE; SOAT
- ACYL-CoA:CHOLESTEROL ACYLTRANSFERASE; ACACT;;
- STEROL ACYLTRANSFERASE
- *FIELD* TX
- Accumulation of cholesterol esters as cytoplasmic lipid droplets within
- macrophages and smooth muscle cells is a characteristic feature of the
- early stages of atherosclerotic plaques. Intracellularly, an essential
- element in forming cholesterol ester from cholesterol is the enzyme
- acyl-coenzyme A:cholesterol acyltransferase (ACACT; EC 2.3.1.26). ACACT
- is a membrane protein located in the endoplasmic reticulum. Cadigan et
- al. (1988) isolated a cell line lacking ACACT activity from mutagenized
- Chinese hamster ovary cells. By DNA-mediated gene transfer into
- ACACT-deficient cells, Cadigan et al. (1989) obtained transfectant cells
- stably expressing human ACACT activity. Using genomic DNAs of these
- transfectant cells as starting materials, Chang et al. (1993) cloned a
- human macrophage cDNA encoding ACACT. The cDNA contained a single open
- reading frame of approximately 1.7 kb. Protein homology analysis of this
- ORF indicated that it represents a structural gene for ACACT.
-
- By fluorescence in situ hybridization and by Southern blot analysis of
- human/hamster somatic cell hybrid panels, Chang et al. (1994) mapped the
- ACACT gene to 1q25.
-
- Unesterified sterol modulates the function of eukaryotic membranes. In
- human cells, sterol is esterified to a storage form by acyl-coenzyme A
- (CoA):cholesterol acyltransferase. Yang et al. (1996) identified 2 genes
- designated ARE1 and ARE2 by them that encode related enzymes in yeast.
- The yeast enzymes are 49% identical to each other and exhibit 23%
- identity and 49% similarity to human sterol O-acyltransferase. A
- deletion of ARE2 reduced the sterol ester levels to approximately 25% of
- normal levels, whereas disruption of ARE1 did not affect sterol ester
- biosynthesis. Deletion of both genes resulted in a viable cell with
- undetectable esterified sterol. With the use of a consensus sequence to
- the yeast and human genes, an additional member of the SOAT gene family
- was identified in humans; see 601311.
-
- Meiner et al. (1996) noted that ACAT activity is found in many tissues,
- including macrophages, adrenal glands, and liver. In macrophages, ACAT
- is thought to participate in foam cell formation and thereby to
- contribute to the development of atherosclerotic lesions. Meiner et al.
- (1996) disrupted the homologous gene (Acact) in mice, which resulted in
- decreased cholesterol esterification in Acact-deficient fibroblasts and
- adrenal membranes and markedly reduced cholesterol ester levels in
- adrenal glands and peritoneal macrophages. In contrast, the livers of
- Acact-deficient mice contained substantial amounts of cholesterol esters
- and exhibited no reduction in cholesterol esterification activity. These
- tissue-specific reductions in cholesterol esterification provided
- evidence that in mammals this process involves more than 1 form of
- esterification enzyme.
-
- Nomenclature: The preferred symbol for this gene is SOAT, for steryl
- O-acyltransferase. Chang et al. (1993) and Yang et al. (1996) used the
- abbreviation ACAT for the enzyme; this, however, has been used for
- another enzyme with ketothiolase activity (203750). Literature symbols
- used for this gene include ACACT and STAT (not to be confused with a
- family of signal transducer/transcription activator genes; see 600555).
-
- *FIELD* RF
- 1. Cadigan, K. M.; Chang, C. C. Y.; Chang, T.-Y.: Isolation of Chinese
- hamster ovary cell lines expressing human acyl-coenzyme A/cholesterol
- acyltransferase activity. J. Cell Biol. 108: 2201-2210, 1989.
-
- 2. Cadigan, K. M.; Heider, J. G.; Chang, T.-Y.: Isolation and characterization
- of Chinese hamster ovary cell mutants deficient in acyl-coenzyme A:cholesterol
- acyltransferase activity. J. Biol. Chem. 263: 274-282, 1988.
-
- 3. Chang, C. C. Y.; Huh, H. Y.; Cadigan, K. M.; Chang, T. Y.: Molecular
- cloning and functional expression of human acyl-coenzyme A:cholesterol
- acyltransferase cDNA in mutant Chinese hamster ovary cells. J. Biol.
- Chem. 268: 20747-20755, 1993.
-
- 4. Chang, C. C. Y.; Noll, W. W.; Nutile-McMenemy, N.; Lindsay, E.
- A.; Baldini, A.; Chang, W.; Chang, T. Y.: Localization of acyl coenzyme
- A:cholesterol acyltransferase gene to human chromosome 1q25. Somat.
- Cell Molec. Genet. 20: 71-74, 1994.
-
- 5. Meiner, V. L.; Cases, S.; Myers, H. M.; Sande, E. R.; Bellosta,
- S.; Schambelan, M.; Pitas, R. E.; McGuire, J.; Herz, J.; Farese, R.
- V., Jr.: Disruption of the acyl-CoA:cholesterol acyltransferase gene
- in mice: evidence suggesting multiple cholesterol esterification enzymes
- in mammals. Proc. Nat. Acad. Sci. 93: 14041-14046, 1996.
-
- 6. Yang, H.; Bard, M.; Bruner, D. A.; Gleeson, A.; Deckelbaum, R.
- J.; Aljinovic, G.; Pohl, T. M.; Rothstein, R.; Sturley, S. L.: Sterol
- esterification in yeast: a two-gene process. Science 272: 1353-1356,
- 1996.
-
- *FIELD* CD
- Victor A. McKusick: 11/10/1993
-
- *FIELD* ED
- terry: 01/23/1997
- mark: 1/18/1997
- terry: 1/10/1997
- mark: 6/17/1996
- terry: 6/17/1996
- terry: 6/13/1996
- mark: 3/8/1996
- carol: 10/10/1994
- terry: 8/25/1994
- carol: 11/12/1993
- carol: 11/10/1993
-
- *RECORD*
- *FIELD* NO
- 102645
- *FIELD* TI
- *102645 ACYLPEPTIDE HYDROLASE; APH
- N-ACYLAMINOACYLPEPTIDE HYDROLASE; APEH
- *FIELD* TX
- Harper and Saunders (1981) mapped a probe called lambda-H3 to chromosome
- 1 by in situ hybridization. This was subsequently called D1S1. Further
- studies by Carritt et al. (1986) and Goode et al. (1986) indicated that
- this single copy sequence actually originated from chromosome 3 and that
- several homologous sequences were located on chromosome 1. The locus on
- chromosome 3 was designated DNF15S2 and the locus on chromosome 1 was
- designated DNF15S1. The DNF15S2 locus was shown to have a high rate of
- allele loss in both small cell lung cancer and renal cell carcinoma.
- Naylor et al. (1989) showed that the DNF15S2 locus is located at 3p21
- and that it is transcribed in normal lung and in small cell lung cancer.
- They presented the sequence of the gene. They pointed out that the
- activity of aminoacylase-1, which is encoded by the ACY1 gene located at
- 3p21 (104620), was lacking in the same small cell lung cancer cell line
- that lacked DNF15S1. Jones et al. (1991) pointed out an 87% identity
- between the cDNA sequence that encodes acylpeptide hydrolase from
- porcine liver (Mitta et al., 1989) and the cDNA transcribed from DNF15S2
- (Naylor et al., 1989). Acylpeptide hydrolase (EC 3.4.19.1) catalyzes the
- hydrolysis of the terminal acetylated amino acid preferentially from
- small acetylated peptides. The acetylamino acid formed by acylpeptide
- hydrolase is further processed to acetate and a free amino acid by an
- aminoacylase. The substrates for the acylpeptide hydrolase and the
- acylase behave in a reciprocal manner since acylpeptide hydrolase binds
- but does not process acetylamino acids and the acylase binds
- acetylpeptides but does not hydrolyze them; however, the 2 enzymes share
- the same specificity for the acyl group. All of these findings indicate
- common functional features in the protein structures of the 2 enzymes,
- which are encoded by the same region of human chromosome 3, namely,
- 3p21. Jones et al. (1991) suggested that there may be a relationship
- between the expression of these 2 enzymes and acetylated peptide growth
- factors in some carcinomas. The locus on 3p21, formerly called DNF15S2
- and now symbolized APH, is known to have 2 polymorphic sites, both
- detectable with HindIII (Carritt et al., 1986; Goode et al., 1986).
- (This locus was labeled DNF15S2 by HGM9 in Paris in 1987, D3F15S2E by
- HGM10 in New Haven in 1989, and D3F15S2 by HGM10.5 in Oxford in 1990.)
-
- A polymorphic locus, D3S94, previously localized to 3pter-p14.2 (Kiousis
- et al., 1989), contains 2 CpG islands and sequences conserved in the
- hamster and mouse. Ginzinger et al. (1992) isolated cDNAs homologous to
- the conserved fragments and found 96% sequence similarity to a cDNA
- derived from the DNF15S2 locus. Furthermore, the sequence of cDNAs
- derived from both the rat and pig acylpeptide hydrolase showed a high
- degree of sequence similarity to cDNAs derived from D3S94 and DNF15S2,
- suggesting that they are all the same locus. The locus in question was
- mapped to 3p21.3 by fluorescence in situ hybridization (FISH). ACY1 and
- APH map to slightly different regions of 3p, 3p21.1 and 3p21.3,
- respectively. Using pulsed field gel electrophoresis, Boldog et al.
- (1989) showed that the DNF15S2 locus is not linked to D3S2; since D3S2
- is within the same 2.5-Mb region as ACY1, it is likely that ACY1 and APH
- are not closely linked physically. The homologous gene is located on
- mouse chromosome 9 and rat chromosome 8 in a region highly homologous to
- human chromosome 3 (Pausova et al., 1994).
-
- *FIELD* RF
- 1. Boldog, F.; Erlandsson, R.; Klein, G.; Sumegi, J.: Long-range
- restriction enzyme maps of DNF15S2, D3S2 and c-raf1 loci on the short
- arm of human chromosome 3. Cancer Genet. Cytogenet. 42: 295-306,
- 1989.
-
- 2. Carritt, B.; Welch, H. M.; Parry-Jones, N. J.: Sequences homologous
- to the human D1S1 locus present on human chromosome 3. Am. J. Hum.
- Genet. 38: 428-436, 1986.
-
- 3. Ginzinger, D. G.; Shridhar, V.; Baldini, A.; Taggart, R. T.; Miller,
- O. J.; Smith, D. I.: The human loci DNF15S2 and D3S94 have a high
- degree of sequence similarity to acyl-peptide hydrolase and are located
- at 3p21.3. Am. J. Hum. Genet. 50: 826-833, 1992.
-
- 4. Goode, M. E.; vanTuinen, P.; Ledbetter, D. H.; Daiger, S. P.:
- The anonymous polymorphic DNA clone D1S1, previously mapped to human
- chromosome 1p36 by in situ hybridization, is from chromosome 3 and
- is duplicated on chromosome 1. Am. J. Hum. Genet. 38: 437-446,
- 1986.
-
- 5. Harper, M. E.; Saunders, G. E.: Localization of single copy DNA
- sequences on G-banded human chromosomes by in situ hybridization.
- Chromosoma 83: 431-439, 1981.
-
- 6. Jones, W. M.; Scaloni, A.; Bossa, F.; Popowicz, A. M.; Schneewind,
- O.; Manning, J. M.: Genetic relationship between acylpeptide hydrolase
- and acylase, two hydrolytic enzymes with similar binding but different
- catalytic specificities. Proc. Nat. Acad. Sci. 88: 2194-2198, 1991.
-
- 7. Kiousis, S.; Drabkin, H.; Smith, D. I.: Isolation and mapping
- of a polymorphic DNA sequence (cA476) on chromosome 3 (D3S94). Nucleic
- Acids Res. 17: 5876 only, 1989.
-
- 8. Mitta, M.; Asada, K.; Uchimura, Y.; Kimizuka, F.; Kato, I.; Sakiyama,
- F.; Tsunasawa, S.: The primary structure of porcine liver acylamino
- acid-releasing enzyme deduced from cDNA sequences. J. Biochem. 106:
- 548-551, 1989.
-
- 9. Naylor, S. L.; Marshall, A.; Hensel, C.; Martinez, P. F.; Holley,
- B.; Sakaguchi, A. Y.: The DNF15S2 locus at 3p21 is transcribed in
- normal lung and small cell lung cancer. Genomics 4: 355-361, 1989.
-
- 10. Pausova, Z.; Bourdon, J.; Clayton, D.; Mattei, M.-G.; Seldin,
- M. F.; Janicic, N.; Riviere, M.; Szpirer, J.; Levan, G.; Szpirer,
- C.; Goltzman, D.; Hendy, G. N.: Cloning of a parathyroid hormone/parathyroid
- hormone-related peptide receptor (PTHR) cDNA from a rat osteosarcoma
- (UMR 106) cell line: chromosomal assignment of the gene in the human,
- mouse, and rat genomes. Genomics 20: 20-26, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 3/25/1991
-
- *FIELD* ED
- carol: 4/5/1994
- carol: 4/6/1993
- carol: 10/13/1992
- supermim: 7/28/1992
-
- *RECORD*
- *FIELD* NO
- 102650
- *FIELD* TI
- 102650 ADACTYLIA, UNILATERAL
- TERMINAL TRANSVERSE DEFECTS OF HAND, UNILATERAL
- *FIELD* TX
- Graham et al. (1986) described adult female twins with unilateral
- terminal transverse defects affecting the left hand in one and the right
- hand in the other. The latter woman had a daughter with a unilateral
- transverse defect affecting the left hand. The hand anomaly was
- characterized by absence of the terminal portions of digits 2 to 5 with
- a mildly hypoplastic thumb. Tiny nail remnants were evident on the
- digital stumps. No soft tissue syndactyly was present. The other hand
- and both feet were clinically and radiologically normal in each of the 3
- persons. No other similar families were found in the literature.
-
- *FIELD* RF
- 1. Graham, J. M., Jr.; Brown, F. E.; Struckmeyer, C. L.; Hallowell,
- C.: Dominantly inherited unilateral terminal transverse defects of
- the hand (adactylia) in twin sisters and one daughter. Pediatrics 78:
- 103-106, 1986.
-
- *FIELD* CS
-
- Limbs:
- Unilateral terminal transverse hand defect;
- Absent terminal portions of digits 2 to 5;
- Mildly hypoplastic thumb
-
- Nails:
- Tiny nail remnants on digital stumps
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 9/8/1988
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 9/13/1988
- root: 9/8/1988
-
- *RECORD*
- *FIELD* NO
- 102660
- *FIELD* TI
- 102660 ADAMANTINOMA OF LONG BONES
- *FIELD* TX
- Adamantinoma of the long bones is a rare, low-grade malignant neoplasm
- of unknown histogenesis, which affects mainly the tibia of young adults
- (Keeney et al., 1989). Sozzi et al. (1990) demonstrated a translocation
- t(7;13)(q32;q14) in a lung metastasis from an adamantinoma of the tibia
- in a boy who showed the same translocation constitutionally (in normal
- fibroblasts and lymphoid cells). The identical translocation was found
- in his normal father. The breakpoint in chromosome 13 was in the same
- region as that in retinoblastoma (180200). The level of esterase D was
- normal in the patient and his parents.
-
- *FIELD* RF
- 1. Keeney, G. L.; Unni, K. K.; Beabout, J. W.; Pritchard, D. J.:
- Adamantinoma of long bones: a clinicopathologic study of 85 cases.
- Cancer 64: 730-737, 1989.
-
- 2. Sozzi, G.; Miozzo, M.; Di Palma, S.; Minelli, A.; Calderone, C.;
- Danesino, C.; Pastorino, U.; Pierotti, M. A.; Della Porta, G.: Involvement
- of the region 13q14 in a patient with adamantinoma of the long bones.
- Hum. Genet. 85: 513-515, 1990.
-
- *FIELD* CS
-
- Oncology:
- Adamantinoma of long bones
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 11/21/1990
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 11/21/1990
-
- *RECORD*
- *FIELD* NO
- 102670
- *FIELD* TI
- *102670 ADDRESSIN, MUCOSAL
- MUCOSAL ADDRESSIN CELL ADHESION MOLECULE-1;;
- MAdCAM-1; MACAM1
- *FIELD* TX
- Tissue-specific homing of lymphocytes is regulated by interactions with
- the endothelium of specialized venules, such as the high endothelial
- venules (HEV) in lymph nodes and mucosal lymphoid tissues. The mucosal
- vascular addressin, a 58-66K glycoprotein adhesion receptor for
- lymphocytes, is selectively expressed on HEV of the mucosal lymphoid
- organ and on lamina propria venules and helps direct lymphocyte traffic
- to these mucosal tissues. Briskin et al. (1993) isolated a cDNA that, on
- transfection into COS cells, encoded immunoreactive addressin that
- specifically bound a mucosal HEV-binding T-cell lymphoma. The predicted
- amino acid sequence defined the mucosal addressin as a novel
- immunoglobulin family member with 2 amino-terminal domains that
- displayed strong homology to previously described vascular adhesion
- receptors for leukocytes: ICAM1 (147840) and VCAM1 (192225). The
- membrane proximal domain was found to be homologous to the third domain
- of another mucosa-associated member of the immunoglobulin family,
- namely, IgA1.
-
- *FIELD* RF
- 1. Briskin, M. J.; McEvoy, L. M.; Butcher, E. C.: MAdCAM-1 has homology
- to immunoglobulin and mucin-like adhesion receptors and to IgA1. Nature 363:
- 461-464, 1993.
-
- *FIELD* CD
- Victor A. McKusick: 6/22/1993
-
- *FIELD* ED
- carol: 6/22/1993
-
- *RECORD*
- *FIELD* NO
- 102680
- *FIELD* TI
- *102680 ADDUCIN, ALPHA SUBUNIT; ADDA
- ADDUCIN-1; ADD1
- *FIELD* TX
- Adducin is a cell-membrane skeletal protein that was first purified from
- human erythrocytes by Gardner and Bennett (1986) and subsequently
- isolated from bovine brain membranes. Isoforms of this protein have been
- detected in lung, kidney, testes, and liver. Erythrocyte adducin is a
- 200-kD heterodimer protein present at about 30,000 copies per cell. It
- binds with high affinity to Ca(2+)/calmodulin and is a substrate for
- protein kinases A and C. Joshi and Bennett (1990) investigated the
- structure and function of the separate domains of the protein. Adducin
- is a heterodimeric protein. The related subunits, alpha and beta
- (102681), are produced from distinct genes but share a similar
- structure, with a protease-resistant N-terminal region and a
- protease-sensitive, hydrophilic C-terminal region. Joshi et al. (1991)
- isolated reticulocyte cDNAs for alpha- and beta-adducin and, by somatic
- cell hybrid analysis, provisionally assigned the ADDA gene to chromosome
- 4 and the ADDB gene to chromosome 2. Both alpha-adducin and beta-adducin
- show alternative splicing; thus, there may be several different
- heterodimeric or homodimeric forms of adducin, each with a different
- functional specificity. Adducin is thought to promote assembly of
- spectrin-actin complexes in the formation of the membrane cytoskeleton
- (the name comes from the Latin adducere, meaning 'to bring together').
- At least in brain, alpha-adducin is encoded by alternatively spliced
- mRNAs. See Gilligan and Bennett (1993) for a review of adducin and the
- other components of the junctional complex of the cell membrane
- skeleton.
-
- Using the technique of exon amplification to isolate genes from the
- 4p16.3 region where Huntington disease (HD; 143100) appears to be
- located, Taylor et al. (1992) identified exons corresponding to the
- alpha subunit of adducin. The alpha-adducin gene (ADDA) maps immediately
- telomeric to D4S95, in a region likely to contain the HD defect, and
- therefore is a candidate gene for Huntington disease. (Buckler et al.
- (1991) described a vector system that allows selection and amplification
- of exons from genomic DNA, a method referred to as 'exon trapping.')
- Goldberg et al. (1992) reported the isolation and cloning of cDNA for
- the human brain alpha-adducin gene which they found to be located within
- 20 kb of D4S95, a marker showing strong linkage disequilibrium with HD.
- Ankyrin and adducin appear to have different functions in the membrane
- skeleton but both play a role in the interaction with spectrin and the
- maintenance of normal membrane integrity. Studies of red cells,
- fibroblasts, lymphocytes and neurons in HD patients pointed to a
- possible generalized disturbance in membrane structure and function in
- this disorder (review by Hayden, 1981). The functional consequences of
- defects in the adducin gene are unknown. However, mice deficient in
- ankyrin have, in addition to hemolytic anemia, significant neurologic
- dysfunction associated with Purkinje cell degeneration in the cerebellum
- and the development of a late-onset neurologic syndrome characterized by
- persistent tremor and gait disturbance (Peters et al., 1991). Goldberg
- et al. (1992) identified a 4-kb alpha-adducin transcript that was
- abundantly expressed in the caudate nucleus, the site of major neuronal
- loss in HD. No sequence alterations specific to HD were discovered in
- sequencing the brain alpha-adducin cDNA from 2 HD patients and an
- age-matched control. Brain cDNA from both patients and control showed 2
- alternately spliced brain exons not previously described in erythrocyte
- cDNA. Further assessment of the role of this gene in the pathogenesis of
- HD was considered warranted.
-
- Bianchi et al. (1994) showed that 1 point mutation in each of the 2
- genes coding for adducin is associated with blood pressure level in the
- Milan strain of hypertensive rats. The hypertensive and normal rats
- differed, respectively, by the amino acids tyrosine and phenylalanine at
- position 316 of the alpha subunit; at the beta-adducin locus, the
- hypertensive strain was always homozygous for arginine at position 529,
- while the normal strain showed either arginine or glutamine in that
- position. The arg/gln heterozygotes showed lower blood pressure than any
- of the homozygotes. In vitro phosphorylation studies suggested that both
- of these amino acid substitutions occurred within protein kinase
- recognition sites. Analysis of an F2 generation demonstrated that Y
- (tyrosine) alleles segregated with a significant increment in blood
- pressure. This effect was modulated by the presence of the R (arginine)
- allele of the beta subunit. Taken together, these findings strongly
- supported a role for adducin polymorphisms in causing variation of blood
- pressure in the Milan strain of rats. In the rat, the beta- and
- alpha-adducin genes were said to be located on chromosomes 4 and 14,
- respectively, according to unpublished data.
-
- Nasir et al. (1994) used an interspecific backcross to map the mouse
- homolog of human alpha-adducin (Add1) to mouse chromosome 5, within the
- region of conserved synteny with the short arm of human chromosome 4.
- Grosson et al. (1994) also mapped the murine homolog to mouse chromosome
- 5 in a continuous linkage group that included the Huntington disease
- homolog.
-
- *FIELD* RF
- 1. Bianchi, G.; Tripodi, G.; Casari, G.; Salardi, S.; Barber, B. R.;
- Garcia, R.; Leoni, P.; Torielli, L.; Cusi, D.; Ferrandi, M.; Pinna,
- L. A.; Baralle, F. E.; Ferrari, P.: Two point mutations within the
- adducin genes are involved in blood pressure variation. Proc. Nat.
- Acad. Sci. 91: 3999-4003, 1994.
-
- 2. Buckler, A. J.; Chang, D. D.; Graw, S. L.; Brook, J. D.; Haber,
- D. A.; Sharp, P. A.; Housman, D. E.: Exon amplification: a strategy
- to isolate mammalian genes based on RNA splicing. Proc. Nat. Acad.
- Sci. 88: 4005-4009, 1991.
-
- 3. Gardner, K.; Bennett, V.: A new erythrocyte membrane-associated
- protein with calmodulin binding activity: identification and purification.
- J. Biol. Chem. 261: 1339-1348, 1986.
-
- 4. Gilligan, D. M.; Bennett, V.: The junctional complex of the membrane
- skeleton. Seminars Hemat. 30: 74-83, 1993.
-
- 5. Goldberg, Y. P.; Lin, B.-Y.; Andrew, S. E.; Nasir, J.; Graham,
- R.; Glaves, M. L.; Hutchinson, G.; Theilmann, J.; Ginzinger, D. G.;
- Schappert, K.; Clarke, L.; Rommens, J. M.; Hayden, M. R.: Cloning
- and mapping of the alpha-adducin gene close to D4S95 and assessment
- of its relationship to Huntington disease. Hum. Molec. Genet. 1:
- 669-675, 1992.
-
- 6. Grosson, C. L. S.; MacDonald, M. E.; Duyao, M. P.; Ambrose, C.
- M.; Roffler-Tarlov, S.; Gusella, J. F.: Synteny conservation of the
- Huntington's disease gene and surrounding loci on mouse chromosome
- 5. Mammalian Genome 5: 424-428, 1994.
-
- 7. Hayden, M. R.: Huntington's Chorea. New York: Springer-Verlag
- (pub.) 1981.
-
- 8. Joshi, R.; Bennett, V.: Mapping the domain structure of human
- erythrocyte adducin. J. Biol. Chem. 265: 13130-13136, 1990.
-
- 9. Joshi, R.; Gilligan, D. M.; Otto, E.; McLaughlin, T.; Bennett,
- V.: Primary structure and domain organization of human alpha and
- beta adducin. J. Cell Biol. 115: 665-675, 1991.
-
- 10. Nasir, J.; Lin, B.; Bucan, M.; Koizumi, T.; Nadeau, J. H.; Hayden,
- M. R.: The murine homologues of the Huntington disease gene (Hdh)
- and the alpha-adducin gene (Add1) map to mouse chromosome 5 within
- a region of conserved synteny with human chromosome 4p16.3. Genomics 22:
- 198-201, 1994.
-
- 11. Peters, L. L.; Birkenmeier, C. S.; Bronson, R. T.; White, R. A.;
- Lux, S. E.; Otto, E.; Bennett, V.; Higgins, A.; Barker, J. E.: Purkinje
- cell degeneration associated with erythroid ankyrin deficiency in
- nb/nb mice. J. Cell Biol. 114: 1233-1241, 1991.
-
- 12. Taylor, S. A. M.; Snell, R. G.; Buckler, A.; Ambrose, C.; Duyao,
- M.; Church, D.; Lin, C. S.; Altherr, M.; Bates, G. P.; Groot, N.;
- Barnes, G.; Shaw, D. J.; Lehrach, H.; Wasmuth, J. J.; Harper, P. S.;
- Housman, D. E.; MacDonald, M. E.; Gusella, J. F.: Cloning of the
- alpha-adducin gene from the Huntington's disease candidate region
- of chromosome 4 by exon amplification. Nature Genet. 2: 223-227,
- 1992.
-
- *FIELD* CD
- Victor A. McKusick: 12/9/1991
-
- *FIELD* ED
- terry: 8/26/1994
- jason: 7/19/1994
- carol: 6/1/1994
- carol: 3/20/1993
- carol: 2/18/1993
- carol: 2/2/1993
-
- *RECORD*
- *FIELD* NO
- 102681
- *FIELD* TI
- *102681 ADDUCIN 2; ADD2
- ADDUCIN, BETA SUBUNIT; ADDB
- *FIELD* TX
- See adducin, alpha subunit (102680). Adducin is a heterodimeric
- calmodulin (114180)-binding protein of the cell-membrane skeleton, which
- is thought to play a role in assembly of the spectrin-actin lattice that
- underlies the plasma membrane (see also 182860 and 102560). Missense
- mutations in both the alpha and beta ADD genes that alter amino acids
- that are normally phosphorylated have been associated with the
- regulation of blood pressure in the Milan Hypertensive Strain (MHS) of
- rats (Bianchi et al., 1994).
-
- Joshi et al. (1991) determined the sequence of cDNAs encoding both the
- alpha and beta human adducins. The 726-amino acid predicted beta subunit
- is 49% identical to the alpha adducin sequence. Tisminetzky et al.
- (1995) determined the genomic organization of the human beta adducin
- gene and showed that it consists of 13 exons spanning approximately 50
- kb. The authors showed that alternative splicing results in the
- production of several different transcripts.
-
- By somatic cell hybrid analysis, Joshi et al. (1991) found that the
- alpha and beta subunits are encoded by separate genes, the alpha gene
- being located on 4p16.3 and the ADDB gene (symbol = ADD2) being located
- on chromosome 2. Gilligan et al. (1995) mapped ADD2 to 2p14-p13 by
- fluorescence in situ hybridization. White et al. (1995) mapped the mouse
- Add2 gene to chromosome 6 by haplotype analysis in interspecific
- backcross mice. Mapping of the human gene to chromosome 2 was confirmed
- by study of somatic cell hybrid panels by Southern blotting. The gene
- was further localized to 2pter-p11.2 by study of somatic cell hybrids
- containing portions of chromosome 2. Tisminetzky et al. (1995)
- regionally mapped ADD2 to 2p15-cen by in situ hybridization.
-
- *FIELD* RF
- 1. Bianchi, G.; Tripodi, G.; Casari, G.; Salardi, S.; Barber, B. R.;
- Garcia, R.; Leoni, P.; Torielli, L.; Cusi, D.; Ferrandi, M.; Pinna,
- L. A.; Baralle, F. E.; Ferrari, P.: Two point mutations within the
- adducin genes are involved in blood pressure variation. Proc. Nat.
- Acad. Sci. 91: 3999-4003, 1994.
-
- 2. Gilligan, D. M.; Lieman, J.; Bennett, V.: Assignment of the human
- beta-adducin gene (ADD2) to 2p13-p14 by in situ hybridization. Genomics 28:
- 610-612, 1995.
-
- 3. Joshi, R.; Gilligan, D. M.; Otto, E.; McLaughlin, T.; Bennett,
- V.: Primary structure and domain organization of human alpha and
- beta adducin. J. Cell Biol. 115: 665-675, 1991.
-
- 4. Tisminetzky, S.; Devescovi, G.; Tripodi, G.; Muro, A.; Bianchi,
- G.; Colombi, M.; Moro, L.; Barlati, S.; Tuteja, R.; Baralle, F. E.
- : Genomic organisation and chromosomal localisation of the gene encoding
- human beta adducin. Gene 167: 313-316, 1995.
-
- 5. White, R. A.; Angeloni, S. V.; Pasztor, L. M.: Chromosomal localization
- of the beta-adducin gene to mouse chromosome 6 and human chromosome
- 2. Mammalian Genome 6: 741-743, 1995.
-
- *FIELD* CN
- Alan F. Scott - updated: 5/13/1996
- Alan F. Scott - updated: 9/27/1995
-
- *FIELD* CD
- Victor A. McKusick: 11/23/1992
-
- *FIELD* ED
- terry: 05/13/1996
- mark: 5/13/1996
- terry: 4/17/1996
- mark: 4/1/1996
- mark: 1/21/1996
- mark: 11/30/1995
- carol: 4/8/1994
- carol: 1/4/1993
- carol: 11/23/1992
-
- *RECORD*
- *FIELD* NO
- 102699
- *FIELD* TI
- *102699 ADENO-ASSOCIATED VIRUS INTEGRATION SITE 1; AAVS1
- *FIELD* TX
- Kotin et al. (1990) isolated cellular sequences flanking integrated
- copies of the adeno-associated virus (AAV) genome from a latently
- infected clonal human cell line and used them to probe genomic blots
- derived from an additional 21 independently derived clones of human
- cells latently infected with AAV. In genomic blots of uninfected human
- cell lines and of primary human tissue, each flanking-sequence probe
- hybridized to unique bands. Kotin et al. (1990) concluded that the AAV
- genome preferentially integrates into a specific region of the cellular
- genome. By somatic cell hybrid mapping, they determined that the
- integration site is unique to chromosome 19. The human parvovirus AAV is
- unique among eukaryotic DNA viruses in its ability to integrate site
- specifically. By means of in situ hybridization, Kotin et al. (1991)
- mapped the integration site to 19q13-qter.
-
- Samulski et al. (1991) mapped the AAVS1 gene to 19q13.4-qter by in situ
- hybridization of AAV DNA to chromosomes from latently infected cells.
- The findings suggested that this nonpathogenic parvovirus establishes
- viral latency by integrating its DNA specifically into 1 chromosomal
- region. Such specific integration was considered unique among the
- eukaryotic DNA viruses. The incorporation of site-specific integration
- into AAV vector schemes should make this vector system attractive for
- human gene therapy strategies.
-
- By analysis of the proviral junctions, Kotin et al. (1992) determined
- that integration of the AAV DNA occurred via a nonhomologous
- recombination pathway. Direct repeats at a much greater than random
- occurrence were found distributed nonuniformly throughout the AAVS1
- sequence.
-
- *FIELD* RF
- 1. Kotin, R. M.; Linden, R. M.; Berns, K. I.: Characterization of
- a preferred site on human chromosome 19q for integration of adeno-associated
- virus DNA by non-homologous recombination. EMBO J. 11: 5071-5078,
- 1992.
-
- 2. Kotin, R. M.; Menninger, J. C.; Ward, D. C.; Berns, K. I.: Mapping
- and direct visualization of a region-specific viral DNA integration
- site on chromosome 19q13-qter. Genomics 10: 831-834, 1991.
-
- 3. Kotin, R. M.; Siniscalco, M.; Samulski, R. J.; Zhu, X. D.; Hunter,
- L.; Laughlin, C. A.; McLaughlin, S.; Muzyczka, N.; Rocchi, M.; Berns,
- K. I.: Site-specific integration by adeno-associated virus. Proc.
- Nat. Acad. Sci. 87: 2211-2215, 1990.
-
- 4. Samulski, R. J.; Zhu, X.; Xiao, X.; Brook, J. D.; Housman, D. E.;
- Epstein, N.; Hunter, L. A.: Targeted integration of adeno-associated
- virus (AAV) into human chromosome 19. EMBO J. 10: 3941-3950, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 9/9/1990
-
- *FIELD* ED
- carol: 2/4/1993
- carol: 1/15/1993
- supermim: 3/16/1992
- carol: 6/4/1991
- carol: 2/19/1991
- carol: 2/15/1991
-
- *RECORD*
- *FIELD* NO
- 102700
- *FIELD* TI
- *102700 ADENOSINE DEAMINASE; ADA
- ADENOSINE AMINOHYDROLASE
- SEVERE COMBINED IMMUNODEFICIENCY DUE TO ADA DEFICIENCY, INCLUDED;;
- SCID DUE TO ADA DEFICIENCY, INCLUDED;;
- ADA-SCID, INCLUDED
- *FIELD* MN
- ADA deficiency is the cause of one form of severe combined
- immunodeficiency disease (SCID), in which there is dysfunction of both B
- and T lymphocytes with impaired cellular immunity and decreased
- production of immunoglobulins. ADA deficiency accounts for about
- one-half of cases of autosomal recessive SCID. In 85 to 90% of cases the
- disorder is severe with skeletal lesions. In the remainder the disorder
- is milder with progressive manifestations, mainly involving cellular
- immunity, beginning after age 2 years or even in adulthood (Shovlin et
- al., 1993).
-
- The ADA gene is located on 20q12-q13.11 (Rothschild et al., 1993). The
- complete sequence and structure of the gene is known (Wiginton et al.,
- 1986). A variety of mutant alleles have been identified, including
- basepair substitutions (Hirschhorn et al., 1990) and deletions (Berkvens
- et al., 1990). These mutations and compound heterozygosity account for
- much of the variation in expression. Somatic mosaicism may be the basis
- for delayed presentation and unusual course of ADA deficiency in some
- cases (Hirschhorn et al., 1994). Striking disparity in clinical
- phenotype of sibs may also result from differences in efficiency of
- splicing (Arredondo-Vega et al., 1994). See 102710 and 102720 for
- descriptions of adenosine deaminase complexing proteins, coded by loci
- on chromosomes 6 and 2, respectively, which may be involved in some
- cases.
-
- There are 3 genetically determined isozymes of erythrocyte adenosine
- deaminase: ADA 1, ADA 2-1 and ADA 2. The ADA 2 allozyme is a more basic
- electrophoretic variant that is codominantly inherited with the usual
- ADA 1 allozyme (Hirschhorn et al., 1994). The variant has been found in
- all populations studied and results in only minimally reduced enzyme
- activity in erythrocytes. The frequency of the ADA2 allele was estimated
- at 0.06 in Europeans, 0.04 in Blacks, and 0.11 in Asiatic Indians
- (Spencer et al., 1968). An overrepresention of West Indian ancestry and
- the finding of multiple new mutations suggest that partial ADA
- deficiency may have had a selective advantage.
-
- Most lymphocyte ADA is of the same electrophoretic type as red cell ADA.
- Bone marrow or fetal liver has been used for transplantation purposes.
- Blood transfusion can result in graft-versus-host disease due to donor
- lymphocytes. However, use of packed erythrocytes, subjected to freezing
- and irradiation to eliminate lymphocytes, has been effective therapy
- (Markert et al., 1987). Successful use of polyethylene glycol
- (PEG)-modified bovine intestinal ADA administered intramuscularly has
- been reported (Hershfield et al., 1987). Gene therapy trials are in
- progress.
-
- *FIELD* ED
- carol: 07/23/1996 marlene: 7/23/1996 joanna: 7/11/1996
-
- *FIELD* CD
- F. Clarke Fraser: 5/9/1996
- *FIELD* TX
- By means of a new and specific method, Spencer et al. (1968)
- demonstrated isozymes of erythrocyte adenosine deaminase (adenosine
- aminohydrolase; EC 3.5.4.4) and showed that there are 3 genetically
- determined phenotypes: ADA 1, ADA 2-1 and ADA 2. The frequency of the
- ADA 2 allele was estimated at 0.06 in Europeans, 0.04 in Blacks, and
- 0.11 in Asiatic Indians. Data on gene frequencies of allelic variants
- were tabulated by Roychoudhury and Nei (1988).
-
- Wiginton et al. (1986) reported the complete sequence and structure of
- the gene for human ADA. By study of mouse-man somatic cell hybrids,
- Creagan et al. (1973) and Tischfield et al. (1974) showed that the locus
- for ADA is on chromosome 20. Gene dosage studies of adenosine deaminase
- and inosine triphosphatase provided corroboration of partial trisomy 20
- diagnosed cytogenetically (Rudd et al., 1979). Valerio et al. (1984)
- used an ADA cDNA probe in Southern hybridizations with DNA from a hybrid
- cell panel to assign the gene to chromosome 20. Mohandas et al. (1984)
- reported that the genes for ADA and SAHH are on separate parts of 20q,
- separated by 20q13.1. Nielsen et al. (1986) studied ADA in a case of
- partial trisomy 20q resulting from a familial t(3;20) translocation.
- Gene dosage studies seemed to exclude the ADA gene from the distal part
- of 20q (20q13.1-qter). By dosage effect in a patient with deletion of
- 20q, Petersen et al. (1987) assigned the ADA locus to 20q13.11. By means
- of in situ hybridization to high resolution spreads of somatic and
- pachytene chromosomes, Jhanwar et al. (1989) localized the ADA gene to
- 20q12-q13.11. Rothschild et al. (1993) identified and mapped new
- dinucleotide repeat polymorphisms associated with the ADA locus. These
- increased the PIC of the ADA locus to 0.89.
-
- Adenosine deaminase shows not only polymorphism but also deficiency. ADA
- deficiency is the cause of one form of severe combined immunodeficiency
- disease (SCID), in which there is dysfunction of both B and T
- lymphocytes with impaired cellular immunity and decreased production of
- immunoglobulins. Multiple forms of SCID exist; see Swiss type of
- agammaglobulinemia (202500, 300400), nucleoside phosphorylase (164050),
- and transcobalamin II deficiency (275350). ADA deficiency accounts for
- about one-half of cases of autosomal recessive SCID. In 85 to 90% of
- cases the disorder is severe with skeletal lesions. In the remainder the
- disorder is milder with progressive manifestations, mainly involving
- cellular immunity, beginning after age 2 years. Bony changes in patients
- with ADA-deficient SCID suggest that ADA may be the defect in at least
- some cases of reported 'achondroplasia and Swiss-type
- agammaglobulinemia' (200900). Note also that cartilage-hair hypoplasia
- (250250) involves a defect in cellular immunity in association with
- skeletal changes. Giblett et al. (1972) described 2 girls in separate
- families with impaired cellular immunity and absent red cell adenosine
- deaminase. One child, aged 22 months, showed recurrent respiratory
- infections, candidiasis, and marked lymphopenia from birth. The other,
- aged 3.5 years, was allegedly normal in the first 2 years of life. Mild
- upper respiratory infections began at age 24 months and progressed to
- severe pulmonary insufficiency and hepatosplenomegaly by age 30 months.
- The parents of the first child were related and the second child had a
- sister who died in consequence of a major immunologic defect (Hong et
- al., 1970). The finding that both pairs of parents had an intermediate
- level of red cell ADA supports recessive inheritance. Possibly a
- different allele is present in the 2 families because in the first
- family the parents showed about a 50% level of ADA whereas it was about
- two-thirds normal in the second pair. Hirschhorn et al. (1980) pointed
- to the neurologic abnormalities that had been reported in 2 of 23
- ADA-deficient patients and reported a third who showed improvement of
- these features with enzyme replacement by red cell infusion. Bortin and
- Rimm (1977) reported on the characteristics and results of treatment in
- 69 patients with SCID; in 25 patients tested, deficiency of ADA was
- found in 4 (16%). In surveying 18 cases of SCID that survived bone
- marrow transplantation, Kenny and Hitzig (1979) found that 3 had ADA
- deficiency. Mitchell et al. (1978) found that deoxyadenosine and
- deoxyguanosine are particularly toxic to T cells but not to B cells.
- Addition of deoxycytidine or dipyridamole prevented deoxyribonucleoside
- toxicity. See 102710 and 102720 for descriptions of adenosine deaminase
- complexing proteins, coded by loci on chromosomes 6 and 2, respectively.
- Are some cases of SCID due to deficiency of ADCP rather than of the
- enzyme itself? Koch and Shows (1980) showed that ADA deficiency in SCID
- segregates with chromosome 20 alone in interspecific somatic cell
- hybrids, suggesting that a structural gene mutation at the ADA locus is
- the primary cause of ADA-deficient SCID. Boss et al. (1981) concluded
- that ecto-5-prime-nucleotidase deficiency is secondary to the primary
- defect of ADA. Herbschleb-Voogt et al. (1983) demonstrated
- CRM-negativity in a patient with ADA-deficiency SCID. Wiginton et al.
- (1983) cloned cDNA sequences of human ADA. Two B-lymphoblast lines from
- cases of hereditary ADA deficiency contained unstable ADA protein but
- had 3 to 4 times the normal level of ADA mRNA. ADA and
- S-adenosylhomocysteine hydrolase (SAHH; 180960) have related metabolic
- functions. In SCID due to ADA deficiency, red cells also show very low
- levels (less than 2% of controls) of SAHH. The latter finding has been
- attributed to a suicide-like inactivation of SAHH by
- 2-prime-deoxyadenine. SAHH is also coded by a gene on chromosome 20.
-
- Shovlin et al. (1993) described an adult form of ADA deficiency in 2
- sisters who presented with chronic chest disease and recurrent
- bacterial, viral, and fungal infections together with laboratory
- phenotypes similar to those of advanced HIV disease, including severe
- CD4 lymphopenia. Both were HIV negative. These were the oldest patients
- ever described with a new diagnosis of primary ADA deficiency. One
- woman, aged 34 years, had had asthma and recurrent chest infections from
- childhood. Records revealed lymphopenia from age 20 years. She had
- widespread viral warts, recurrent oral and vaginal candidosis, and had
- had 2 episodes of dermatomal zoster. The sister, aged 35 years, was well
- until age 17 when she developed idiopathic thrombocytopenic purpura
- necessitating splenectomy, azathioprine for 7 years, and prednisolone
- until the time of report. By age 20 she had asthma, recurrent chest
- infections, vaginal and oral candidosis, widespread viral warts, and
- recurrent dermatomal zoster. Records showed lymphopenia from age 17.
- Both sisters had clinical and radiologic evidence of extensive lung
- damage. Shovlin et al. (1994) demonstrated that the sisters were
- compound heterozygotes: in the paternal allele, there was a deletion
- resulting from homologous recombination between 2 Alu elements; this
- allele predicted a null phenotype. In the mutant allele inherited from
- the mother, a C-to-T transition in a CpG dinucleotide changed the codon
- for arginine-211, which lies in a conserved sequence close to the active
- site, to that for cysteine. This mutation had previously been observed
- in a child thought to have partial ADA deficiency by Hirschhorn et al.
- (1990); see 102700.0014. Shovlin et al. (1994) suggested that immune
- function in children with partial ADA deficiency may deteriorate with
- time.
-
- The enzyme defect in ADA deficiency is expressed in all cells, and
- therefore the substrates for the enzyme, adenosine and
- 2-prime-deoxyadenosine, accumulate in cells of all types.
- Immunodeficiency is the consequence of the particular sensitivity of
- immature lymphoid cells to the toxic effects of these 2 substrates. In
- addition, some patients have neurologic abnormalities that may be due to
- ADA deficiency (Hershfield and Mitchell, 1995). Unlike humans, mice that
- express no adenosine deaminase die perinatally of severe hepatocellular
- degeneration (Migchielsen et al., 1995; Wakamiya et al., 1995).
- Bollinger et al. (1996) described a human neonate with ADA deficiency
- and prolonged hyperbilirubinemia with hepatitis that resolved after the
- institution of adenonsine deaminase replacement therapy. Percutaneous
- liver biopsy showed early giant-cell transformation, with enlarged foamy
- hepatocytes and portal and lobular eosinophilic infiltrates. The patient
- was a compound heterozygote for the gly74-to-val mutation (102700.0025)
- and the ala329-to-val mutation (102700.0006).
-
- In studies of 4 unrelated patients with 'partial' ADA deficiency,
- Hirschhorn et al. (1983) found in 3 of them evidence of a different
- mutation at the structural locus: 1) an acidic, low activity,
- heat-labile mutation; 2) a basic, somewhat higher activity, heat-labile
- mutation; and 3) a relatively normal activity, heat-labile mutation. In
- the fourth patient, there was no compelling evidence for a mutation at
- the structural locus for ADA and a mutation at a regulatory locus could
- not be excluded. These children lacked ADA in red cells but retained
- variable amounts of activity in lymphoid cells; none had significant
- immunologic deficiency. Since at least 2 of the partially deficient
- families were black and a third came from the Mediterranean basin,
- Hirschhorn et al. (1983) were tempted to speculate that a partial ADA
- gene might confer some advantage against intraerythrocytic parasites
- such as malaria. Hirschhorn and Ellenbogen (1986) found 5 different
- mutations in 5 unrelated new patients. Of the 5, 3 were shown to be
- genetic compounds by the presence of 2 electrophoretically
- distinguishable allozymes or by family studies that demonstrated a
- 'null' allele in addition to an electrophoretically abnormal enzyme. A
- seemingly increased West Indian ethnic representation strengthened the
- speculation that partial ADA deficiency may have a selective advantage,
- perhaps because many intraerythrocytic parasites such as those of
- malaria and babesiosis require exogenous purines derived from the host.
- Hart et al. (1986) reported an example of partial adenosine deaminase
- deficiency of the general type previously reported by Hirschhorn et al.
- (1979), Daddona et al. (1983), and Hirschhorn and Ellenbogen (1986),
- among others. Their proband, a Bantu-speaking Xhosa man, proved to be a
- genetic compound. The previous case observed in South Africa had been a
- Kalahari San ('Bushman') reported by Jenkins et al. (1976). Akeson et
- al. (1987) reported an ADA-deficient patient who was a genetic compound;
- one allele caused an amino acid change of alanine to valine
- (102700.0006) and the other a change from arginine to histidine
- (102700.0004). In a second cell line from an ADA-deficient patient, one
- allele was found to cause an alanine to valine substitution whereas the
- other allele was found to produce an mRNA in which exon 4 had been
- spliced out (102700.0007). Several of the ADA cDNA clones extended
- 5-prime of the major initiation start site, indicating multiple start
- sites for ADA transcription. Furthermore, analysis of ADA cDNAs from
- different cell lines detected aberrant RNA species that either included
- intron 7 or excluded exon 7. This was interpreted as indicating aberrant
- splicing of pre-mRNAs, unrelated to the mutations that cause ADA
- deficiency. Tzall et al. (1989) identified and/or characterized at least
- 9 RFLPs at the ADA locus and studied these in 17 patients with complete
- deficiency and in 10 patients with partial deficiency. Genetic compounds
- were identified among both types of patients, but there was, as
- expected, a decreased incidence of heterozygosity. Two additional
- haplotypes not found in the normal population were identified in
- homozygous form in patients. Akeson et al. (1989) reviewed substitutions
- found in ADA in cases of ADA deficiency. Out of the 7 different
- mutations found in the 14 chromosomes of 7 consecutively ascertained
- patients in the New York State newborn screening program, 6 were found
- by Hirschhorn et al. (1990) to have mutations involving CpG
- dinucleotides. Six of the 7 children either came from a limited area in
- the Caribbean or shared a black ethnic background, suggesting that a
- single mutation might have been derived from a common progenitor through
- a founder effect. The fact that multiple new mutations were found
- suggests that partial ADA deficiency may have had a selective advantage.
-
- Most lymphocyte ADA is of the same electrophoretic type as red cell ADA.
- Bone marrow or fetal liver has been used for transplantation purposes.
- Blood transfusion can result in graft-versus-host disease due to donor
- lymphocytes. However, use of packed erythrocytes, subjected to freezing
- and irradiation to eliminate lymphocytes, has been effective therapy.
- The infused normal red cells are in equilibrium with freely diffusing
- adenosine. The ADA they contain lowers the level of adenosine in the
- plasma. The lymphocyte count rises and responsiveness to mixed
- lymphocyte culture and phytohemagglutinin returns. Retransfusion is
- necessary every few weeks (Hirschhorn, 1976). Markert et al. (1987)
- evaluated response to therapy in ADA deficiency and in purine nucleoside
- phosphorylase deficiency. Hershfield et al. (1987) reported successful
- use of polyethylene glycol-modified ADA (PEG-ADA) administered
- intramuscularly. Covalent attachment of polyethylene glycol appears to
- block access to sites on the surface of the protein, inhibiting
- clearance from the circulation, attack by degraded enzymes and binding
- of antibodies, and processing by antigen-presenting cells required for
- generation of an immune response. Hershfield et al. (1987) used
- PEG-modified bovine intestinal ADA in 2 children with SCID due to ADA
- deficiency. They found that the modified enzyme was rapidly absorbed
- after intramuscular injection and had a half-life in plasma of 48 to 72
- hours. Weekly doses could maintain plasma ADA activity at 2 to 3 times
- the level of red cell ADA in normal subjects. The principal biochemical
- consequences of the deficiency were almost completely reversed. In red
- cells, adenosine nucleotides increased and the toxic deoxyadenosine
- nucleotides decreased to less than 0.5% of total adenine nucleotides.
- The activity of S-adenosylhomocysteine hydrolase, which is inactivated
- by deoxyadenosine, increased to normal in red cells and nucleated marrow
- cells. Neither toxic effects nor hypersensitivity reactions were
- observed. In vitro tests of cellular immune function of each patient
- showed marked improvement, together with an increase in T lymphocytes.
- This approach might be useful in other inherited metabolic diseases in
- which accumulated metabolites equilibrate with plasma. Gaucher disease,
- Fabry disease, nucleoside phosphorylase deficiency, and some disorders
- of amino acid and urea cycle metabolism in which accumulated metabolites
- equilibrate with plasma are candidates for this therapeutic approach.
- Levy et al. (1988) reported a child who did not develop trouble from her
- ADA deficiency until age 3 years. Treatment with PEG-modified ADA was
- effective. Hershfield (1995) summarized the results of treatment with
- PEG-ADA. This treatment is indicated for patients who lack an
- HLA-identical bone marrow donor but are at too high a risk for
- HLA-haploidentical marrow transplantation. Treatment almost completely
- corrects metabolic abnormalities, allowing the recovery of a variable
- degree of immune function that in most cases has been sufficient to
- protect against opportunistic infections. Mortality with PEG-ADA is
- lower than that with haploidentical bone marrow transplantation.
- Hershfield (1995) noted, however, that the cost per patient of PEG-ADA
- is 'very high,' approximately $100,000 yearly for an infant and 2 to 3
- times this in older patients.
-
- Santisteban et al. (1993) examined the genetic basis for ADA deficiency
- in 7 patients with late/delayed onset of immunodeficiency, which they
- characterized as an underdiagnosed and relatively unstudied condition.
- Deoxyadenosine-mediated metabolic abnormalities were less severe than in
- the usual, early-onset disorder. Six patients were compound
- heterozygotes; 7 of 10 mutations found were novel. Tissue-specific
- variation in splicing efficiency may ameliorate disease severity in
- patients with splicing mutations, of which 3 were found.
-
- Hirschhorn et al. (1994) found that somatic mosaicism was the basis for
- delayed presentation and unusual course of ADA deficiency in a currently
- healthy young adult who had received no therapy. He was diagnosed at age
- 2.5 years because of life-threatening pneumonia, recurrent infections,
- failure of normal growth, and lymphopenia, but retained significant
- cellular immune function. A fibroblast cell line and a B-cell line,
- established at the time of diagnosis, lacked ADA activity and were
- heteroallelic for a splice-donor-site mutation in IVS1 and a missense
- mutation, arg101-to-gln (102700.0003). All clones isolated from the
- B-cell mRNA carried the missense mutation, indicating that the allele
- with the splice site mutation produced unstable mRNA. In striking
- contrast, a B-cell line established at age 16 expressed 50% of normal
- ADA; 50% of ADA mRNA had normal sequence, and 50% had the missense
- mutation. Genomic DNA contained the missense mutation but not the splice
- site mutation. In vivo somatic mosaicism was demonstrated in genomic DNA
- from peripheral blood cells obtained at 16 years of age, in that less
- than half the DNA carried the splice-site mutation (P less than 0.002,
- vs original B-cell line). Consistent with the mosaicism, erythrocyte
- content of the toxic metabolite deoxyATP was only minimally elevated.
- Somatic mosaicism could have arisen by somatic mutation or by reversion
- at the site of mutation. Selection in vivo for ADA normal hematopoietic
- cells may have played a role in the return to normal health, in the
- absence of therapy.
-
- Abbott et al. (1986) presented evidence that 'wasted' (wst) in mice is
- caused by a mutation in the structural gene for ADA. As occurs in humans
- with ADA deficiency, wasted mice are immunodeficient, develop neurologic
- abnormalities, and die soon after weaning. This animal model may be
- useful in studies of gene therapy. Using a retroviral vector for human
- ADA, Ferrari et al. (1991) transduced peripheral blood lymphocytes from
- patients affected by ADA-negative SCID and injected them into
- immunodeficient mice. Longterm survival of vector-transduced human cells
- was demonstrated in recipient animals. Expression of vector-derived ADA
- restored immune functions, as indicated by the presence of human
- immunoglobulin and antigen-specific T cells in reconstituted animals.
- The experiments demonstrated that gene transfer is necessary and
- sufficient for development of specific immune functions in vivo and has
- therapeutic potential.
-
- Bordignon et al. (1995) used 2 different retroviral vectors to transfer
- the human ADA minigene ex vivo into bone marrow cells and peripheral
- blood lymphocytes from 2 patients undergoing exogenous enzyme
- replacement therapy. After 2 years of treatment, longterm survival of T
- and B lymphocytes, marrow cells, and granulocytes expressing the
- transferred ADA gene was demonstrated and resulted in normalization of
- the immune repertoire and restoration of cellular and humeral immunity.
- After discontinuation of treatment, T lymphocytes, derived from
- transduced peripheral blood lymphocytes, were progressively replaced by
- marrow-derived T cells in both patients. These results indicated
- successful gene transfer into long lasting progenitor cells, producing a
- functional multilineage progeny. Blaese et al. (1995) reported results
- of a clinical trial which started in 1990 using retroviral-mediated
- transfer of the ADA gene into the T cells of 2 children with
- ADA-deficient SCID. Patient 1 was begun on gene therapy on 14 September
- 1990 and received a total of 11 infusions. Patient 2 began gene therapy
- on 31 January 1991 and received a total of 12 infusions. The number of
- blood T lymphocytes normalized as did many cellular and humeral immune
- responses. Gene treatment ended after 2 years, but integrated vector and
- ADA gene expression in T cells persisted. Blaese et al. (1995) concluded
- that although many components remained to be perfected, gene therapy was
- a safe and effective addition the treatment for some patients with this
- form of SCID.
-
- Hirschhorn et al. (1996) described an unusual instance of somatic
- mosaicism due to in vivo reversion to normal of an inherited mutation in
- the ADA gene. In the proband ADA activity was not detectable in
- erythroctyes at age 5, but concentrations of deoxy-ATP in RBCs and
- deoxyadenosine in urine were only minimally elevated, as compared to
- concentrations found in patients with early onset ADA(-) SCID. Both
- parents exhibited approximately 50% of the normal erythrocyte ADA as did
- 2 young adult healthy sibs. Enzyme activity in lymphocytes was
- diminished to approximately 15% of normal in the proband and 20-25% of
- normal (within the heterozygote range) in both parents. Lymphoid cell
- lines established from the proband and both parents also exhibited
- markedly diminished ADA. The considerable residual enzyme activity in
- nonerythroid cells and low concentrations of metabolites were similar to
- findings in 'partially' ADA-deficient children ascertained by population
- screening who had remained healthy during the first year of life
- (Hirschhorn et al., 1990). By contrast, the death in infancy due to
- immunodeficiency of a prior sib and the abnormal immunologic findings in
- the proband during the first years of life were more consistent with
- complete ADA deficiency. Hirschhorn et al. (1996) provided an
- explanation by molecular analysis of the family. The father was
- heterozygous for a splice site mutation at the invariant G of the
- 5-prime donor site in IVS5 of the ADA gene leading to deletion of the
- 116-bp sequence contained in exon 5 (102700.0026). The mother was a
- mosaic of normal lymphocytes and lymphocytes containing a G-to-A
- transition at nt 467, predicting an arg156-to-his substitution (a
- deleterious mutation previously reported by Santisteban et al. (1993) in
- ADA-deficient immunodeficient patients); in 13/15 authenticated B cell
- lines and in 17% of single alleles cloned from blood DNA, the maternally
- transmitted deleterious mutation was absent in the proband, despite
- retention of a maternal 'private' ADA polymorphism linked to the
- mutation. Hirschhorn et al. (1996) speculated that these cells had a
- strong selective advantage, thus accounting for the mild phenotype
- compared to the brother.
-
- *FIELD* AV
- .0001
- ADA DEFICIENCY
- ADA, LYS80ARG
- In cell line GM2471, Valerio et al. (1986) found 2 point mutations in
- the ADA gene of a patient with severe combined immunodeficiency: a
- change from lys to arg at position 80 and a change from leu to arg at
- position 304 (102700.0005). Studies with expression clones mutagenized
- in vitro showed that the mutation at position 304 was responsible for
- ADA inactivation. This resulted from a T-to-G mutation at nucleotide
- 1006. This was the change on only 1 of the chromosomes in the cell line
- studied; the patient was a genetic compound. (The GM numbers relate to
- individuals from whom cell lines were derived for deposit in the human
- genetic mutant cell repository at the Coriell Institute in Camden, New
- Jersey.)
-
- .0002
- ADA DEFICIENCY
- ADA, ARG101TRP
- Akeson et al. (1988) summarized the point mutations identified in ADA
- deficiency cases. They came from 5 different patients, each of whom
- proved to be a compound heterozygote. GM2606 was found to have change of
- arg101 to trp resulting from a change of CGG to TGG as well as
- substitution of his for arg211 (102700.0004) as a result of change of
- CGT to CAT (Akeson et al., 1988). Arredondo-Vega et al. (1990) studied T
- cells from the patient from whom the ADA-deficient B-cell line GM2606
- had been established. They found that the arg101-to-trp mutation can be
- expressed selectively in IL2-dependent T cells as a stable, active
- enzyme. Cultured T cells from other patients with the arg211his mutation
- did not express significant ADA activity, while some B-cell lines from a
- patient with an arg101-to-gln mutation had been found to express normal
- ADA activity. Arredondo-Vega et al. (1990) speculated that arg101 may be
- at a site that determines degradation of ADA by a protease that is under
- negative control by IL2 in T cells, and is variably expressed in B
- cells.
-
- .0003
- ADA DEFICIENCY
- ADA, ARG101GLN
- In cell line GM1715 from an immunodeficient patient, Bonthron et al.
- (1985) found a point mutation in codon 101 (CGG to CAG) of ADA; this
- change predicts an amino acid change from arginine to glutamine. The
- mutation was apparently responsible for loss of function in the gene
- because the predicted primary structure of the enzyme was otherwise
- entirely normal. The demonstration of 2 different mutations in codon 101
- leading to ADA deficiency indicates that this amino acid position is
- critical for stability and/or activity of the enzyme protein. In GM2756,
- Akeson et al. (1987) demonstrated 2 different mutant alleles: one was
- arg101 to gln (as in GM1715); the other was ala329 to val (102700.0006).
-
- .0004
- ADA DEFICIENCY
- ADA, ARG211HIS
- Akeson et al. (1988) found this change in cell line GM2606 and Akeson et
- al. (1987) found it in cell line GM2756.
-
- .0005
- ADA DEFICIENCY
- ADA, LEU304ARG
- In cell line GM2471 from a genetic compound, Valerio et al. (1986)
- demonstrated 2 point mutations: lys80 to arg and leu304 to arg. The
- latter resulted from a T-to-G mutation in nucleotide 1006 and was shown
- to cause ADA inactivation in studies with expression clones mutagenized
- in vitro.
-
- .0006
- ADA DEFICIENCY
- ADA, ALA329VAL
- In cell line GM2756, Akeson et al. (1987) demonstrated 2 different
- mutant alleles: one was arg101 to gln (102700.0003); the other was
- ala329 to val. Cell line GM2825A was found to have a substitution of
- valine for alanine-329 resulting from a C-to-T transition at base 1081.
- Markert et al. (1989) also identified a point mutation at position 1081
- of the adenosine deaminase cDNA, causing an alanine-to-valine
- substitution at position 329 of the protein sequence. Because the
- mutation created a new BalI restriction site, Southern analysis was used
- to screen for the frequency of this mutation. It was found in 7 of 22
- alleles with known or suspected point mutations and was associated with
- 3 distinct ADA haplotypes. Hirschhorn et al. (1992) found that 5
- missense mutations accounted for one-third of 45 'ADA-negative'
- chromosomes studied. The ala329-to-val mutation was the most frequent,
- being found in 4 persons heterozygous for the mutation and 1 person
- homozygous for it.
-
- .0007
- ADA DEFICIENCY
- ADA, ALA39VAL
- Akeson et al. (1987, 1988) found that cell line GM2825A was a genetic
- compound. One allele had an ala39-to-val change (102700.0006); the other
- allele had a point mutation from A to G in the 3-prime splice site of
- intron 3, resulting in elimination of exon 4 from the mature mRNA.
-
- .0008
- ADA DEFICIENCY
- ADA, 3.25KB DEL, ALU-RELATED
- Berkvens et al. (1987) found a 3.2-kb deletion spanning the ADA promoter
- and the first exon in an infant with ADA deficiency. The parents were
- consanguineous, and the infant was homozygous for the deletion. Markert
- et al. (1987) reported an apparent deletion mutation in a patient with
- ADA deficiency and SCID who had a major structural alteration in the
- 5-prime end of the ADA gene. The patient had no ADA enzyme activity in
- his lymphocytes, no detectable ADA mRNA by Northern RNA analysis, and a
- deletion in the region of the first exon of the ADA gene by Southern DNA
- analysis. Markert et al. (1988) defined the precise boundaries of the
- deletion and the mechanism of the defect, namely, homologous
- recombination between 2 repetitive DNA sequences of the Alu family,
- resulting in a deletion of the ADA promoter and first exon. By direct
- sequencing of in vitro amplified DNA, Berkvens et al. (1990) showed that
- the 3,250-bp deletion in their patient was due to recombination within
- the left arms of 2 direct AluI repeats. They pointed out that the
- mutation was identical to that in the unrelated patient reported by
- Markert et al. (1988). Neither the pedigree of the Belgian family nor a
- comparison of haplotype data suggested a relationship between the
- American and Belgian patients.
-
- .0009
- ADA DEFICIENCY
- ADA, PRO297GLN
- In a partially ADA-deficient child from Santo Domingo, Hirschhorn et al.
- (1989) demonstrated a C-to-A transversion that resulted in the
- replacement of a proline by a glutamine residue at codon 297. Since this
- mutation generated a new recognition site in exon 10 of genomic DNA for
- the enzyme AluI, Hirschhorn et al. (1989) could use Southern blot
- analysis to establish that this child was homozygous for the mutation
- and that the same mutation was present in another patient. The point
- mutation resulted in heat-lability of the enzyme.
-
- .0010
- ADA DEFICIENCY
- ADA, ARG76TRP
- In cell lines GM5816, GM6200 and GM7103, Hirschhorn et al. (1990) found
- a C-to-T transition at nucleotide 226 resulting in a change of
- arginine-76 to tryptophan.
-
- .0011
- ADA DEFICIENCY
- ADA, ARG149GLN
- In cell line GM6143A, Hirschhorn et al. (1990) found a substitution of
- glutamine for arginine at amino acid 149 resulting from a G-to-A
- transition at nucleotide 446.
-
- .0012
- ADA DEFICIENCY
- ADA, PRO274LEU
- In cell line GM5816, Hirschhorn et al. (1990) found a substitution of
- leucine for proline-274 resulting from a C-to-T transition at nucleotide
- 821.
-
- .0013
- ADA DEFICIENCY
- ADA, LEU107PRO
- In GM7103 and GM4396, both cell lines from compound heterozygous
- patients, Hirschhorn et al. (1990) found a substitution of proline for
- leucine at amino acid 107 resulting from a T-to-C transition in
- nucleotide 320 in exon 4.
-
- .0014
- ADA DEFICIENCY
- ADA, ARG211CYS
- In cell line GM4396, from a compound heterozygous patient, Hirschhorn et
- al. (1990) found substitution of cysteine for arginine at amino acid 211
- resulting from a C-to-T transition of nucleotide 631.
-
- .0015
- ADA DEFICIENCY
- ADA, ALA215THR
- In cell line GM2294, Hirschhorn et al. (1990) found homozygosity for a
- G-to-A transition of nucleotide 643 in exon 7 resulting in a change of
- alanine215-to-threonine.
-
- .0016
- ADA DEFICIENCY
- ADA, GLY216ARG
- In a patient with very severe combined immunodeficiency, Hirschhorn et
- al. (1991) identified a transition of G-646 to A at a CG dinucleotide,
- predicting a glycine-to-arginine substitution at codon 216 of the ADA
- protein. The patient was homozygous, the offspring of consanguineous
- Amish parents from eastern Pennsylvania. Onset of symptoms was at 3 days
- of age with respiratory distress from pneumonia unresponsive to
- antibiotics. Of 9 patients, this one had the highest concentration of
- the toxic metabolite deoxyATP and a relatively poor immunologic response
- during the initial 2 years of therapy with polyethylene glycol-adenosine
- deaminase. Heterozygosity for the same mutation was found in 2 of 21
- additional patients with ADA-SCID.
-
- .0017
- ADA DEFICIENCY
- ADA, A-G, 3-PRIME IVS3, EX4DEL
- See 102700.0007.
-
- .0018
- ADA DEFICIENCY
- ADA, ARG156CYS
- In 2 patients with SCID who were unusual for reportedly responding to
- the limited form of enzyme therapy provided by repeated partial exchange
- transfusions (Polmar et al., 1976; Dyminski et al., 1979), Hirschhorn
- (1992) found two new missense mutations, arg156-to-cys and ser291-to-leu
- (102700.0019). The first of these was found in cell line GM2471 and
- represented a CGC-to-TGC transition at codon 156.
-
- .0019
- ADA DEFICIENCY
- ADA, SER291LEU
- See 102700.0018. Hirschhorn (1992) found the S291L mutation in cell line
- GM4258.
-
- .0020
- COMBINED IMMUNODEFICIENCY DISEASE, LATE/DELAYED ONSET
- ADA, IVS10AS, G-A, -34
- In a patient with late-onset combined immunodeficiency in whom the
- diagnosis of ADA deficiency was first made at the age of 15 years,
- Santisteban et al. (1993) found homozygosity for a single base change in
- intron 10 which activated a cryptic splice acceptor, resulting in a
- protein with 100 extra amino acids. The G(-34) was changed to A, thereby
- converting a GG dinucleotide to AG, and creating a new splice acceptor
- site with all the cis-acting elements of a functional 3-prime splice
- junction. Besides introducing 9 new codons after leu325, use of the
- cryptic splice site shifted the reading frame to include 268 bp of the
- normal 3-prime noncoding region before a new TGA stop codon was
- generated 16 bp from the polyA addition signal. The mutant protein was
- predicted to consist of 463 residues.
-
- .0021
- ADENOSINE DEAMINASE 2 ALLOZYME
- ADA*2
- ADA, ASP8ASN
- Hirschhorn et al. (1994) determined the molecular basis for the common
- electrophoretic variant of ADA, the ADA2 allozyme, which is a more basic
- electrophoretic variant that is codominantly inherited with the usual
- ADA1 allozyme. The variant has been found in all populations studied and
- results in only minimally reduced enzyme activity in erythrocytes. The
- gene frequency of the ADA2 allozyme is estimated as 0.06 in Western
- populations, lower among individuals of African descent, and higher in
- Southeast Asian populations. Hirschhorn et al. (1994) found that the
- ADA*2 allele contains a G-to-A transition at nucleotide 22 (counting
- from the ATG initiator methionine) that results in substitution of
- asparagine for aspartic acid at codon 8. Introduction of the nucleotide
- substitution into an ADA1 cDNA and transfection into monkey kidney (COS)
- cells confirmed that the mutation resulted in expression of an enzyme
- that comigrated with a naturally occurring ADA2 allozyme. The nucleotide
- substitution was found on at least 2 different genetic backgrounds, 1 of
- Ashkenazi Jewish ancestry and 1 in a large Mormon pedigree from Utah,
- suggesting independent recurrence of the mutation. Consistent with
- independent recurrence, the G-to-A transition was located in a CpG
- dinucleotide of the type subject to a high frequency of mutation.
- Hirschhorn et al. (1994) also found a probable intragenic crossover in
- the very large first intron that is rich in repetitive DNA sequences.
-
- .0022
- ADA DEFICIENCY
- ADA, IVS2DS, G-A, +1
- Arredondo-Vega et al. (1994) characterized the mutations responsible for
- ADA deficiency in sibs with striking disparity in clinical phenotype.
- Residual ADA activity was detectable in the cultured T cells,
- fibroblasts, and B lymphoblasts of 1 sib but not in the cells of the
- other. ADA mRNA was undetectable by Northern analysis in the cells of
- both patients. Both sibs were found to be compound heterozygotes for the
- following novel splicing defects: (1) a G-to-A substitution at the +1
- position of the 5-prime splice site of IVS2, and (2) a complex 17-bp
- rearrangement of the 3-prime splice site of IVS8, which inserted a run
- of 7 purines into the polypyrimidine tract and altered the reading frame
- of exon 9 (102700.0023). PCR-amplified ADA cDNA clones with premature
- translation stop codons arising from aberrant pre-mRNA splicing were
- identified, which were consistent with these mutations. However, some
- cDNA clones from T cells of both patients and from fibroblasts and
- EBV-transformed B cells of the first patient were normally spliced at
- both the exon 2/3 and 8/9 junctions. A normal coding sequence was
- documented for clones from both sibs. Findings were interpreted as
- indicating that a low level of normal pre-mRNA splicing may occur
- despite mutation of the invariant first nucleotide of the 5-prime splice
- donor sequence and that differences in efficiency of such splicing may
- account for the difference in residual ADA activity, immune dysfunction,
- and clinical severity in the 2 sibs. These 2 sisters were reported by
- Umetsu et al. (1994). The second-born child presented first with serious
- infections and failure to thrive at age 4 months; the diagnosis of SCID
- and ADA deficiency was made at age 9 months when the child was
- hospitalized for Pseudomonas sepsis and Pneumocystis pneumonia. Her
- healthy 39-month-old sister was then tested and found to be ADA
- deficient. She had an unremarkable history, including normal development
- (weight in 97th percentile) and uncomplicated varicella zoster at age 6
- months. Although she was lymphopenic, antibody production, delayed
- hypersensitivity, and in vitro T-cell function were intact. She became
- more lymphopenic over a period of 6 to 7 months and developed persistent
- upper respiratory infections. Along with her sister, she was then
- treated by enzyme replacement with polyethylene glycol (PEG)-ADA.
-
- .0023
- ADA DEFICIENCY
- ADA, IVS8AS, 7BP INS
- See 102700.0022 and Arredondo-Vega et al. (1994).
-
- .0024
- ADA DEFICIENCY
- ADA, IVS1DS, G-C, +1
- Hirschhorn et al. (1994) found that fibroblast and B-cell lines
- established at the time of diagnosis of ADA deficiency (GM2445 and
- GM1715) were heteroallelic for a newly identified splice-site mutation
- (+1 GT-to-CT transversion) at the donor splice site in IVS1 and for a
- previously described arg101-to-gln missense mutation in exon 4
- (102700.0003). As described earlier, by the time the patient was 16
- years of age, the mutation had disappeared from the B cells but not from
- the fibroblasts and the patient had undergone spontaneous recovery from
- ADA deficiency.
-
- .0025
- ADA DEFICIENCY
- ADA, GLY74VAL
- In a newborn with hepatic dysfunction as a complication of ADA
- deficiency, Bollinger et al. (1996) found compound heterozygosity for
- the ala329-to-val (102700.0006) mutation and a change of codon 74 from
- GGC (gly) to GTC (val).
-
- .0026
- ADA DEFICIENCY
- ADA, IVS5DS, G-A, +1, 116BP DEL, EX5 DEL
- Hirschhorn et al. (1996) identified compound heterozygosity for this
- splice site mutation, which resulted in deletion of the 116-bp sequence
- contained in exon 5 of the ADA gene. The other allele of the patient
- carried a G-to-A transition at nucleotide 467, predicting an
- arg156-to-his substitution, a previously reported deleterious mutation
- found in ADA SCID patients (Santisteban et al., 1993). Hirschhorn et al.
- (1996) found that this mutation had undergone reversion in a certain
- proportion of cells, leading to a relatively mild phenotype.
-
- *FIELD* SA
- Adrian et al. (1984); Adrian et al. (1984); Aitken and Ferguson-Smith
- (1978); Aitken et al. (1980); Chen et al. (1978); Chen et al. (1979);
- Chen et al. (1974); Cohen et al. (1978); Cook et al. (1970); Daddona
- and Kelley (1979); Detter et al. (1970); Dissing and Knudsen (1972);
- Dissing and Knudsen (1969); Hershfield and Kredich (1978); Hirschhorn
- et al. (1974); Hirschhorn et al. (1979); Hirschhorn et al. (1994);
- Honig et al. (1984); Hopkinson et al. (1969); Hutton et al. (1981);
- Kaitila et al. (1976); Kellems et al. (1985); Kredich and Martin (1977);
- Markert et al. (1987); Meuwissen et al. (1975); Orkin et al. (1983);
- Palmer et al. (1987); Parkman et al. (1975); Ratech et al. (1985);
- Ritter et al. (1971); Rubinstein et al. (1979); Schmalstieg et al.
- (1983); Schrader et al. (1978); Scott et al. (1974); Tariverdian and
- Ritter (1969); Valerio et al. (1985); Valerio et al. (1984); Valerio
- et al. (1984); Van der Weyden and Kelley (1974); Weitkamp (1971);
- Weitkamp (1972); Wiginton et al. (1984); Wiginton and Hutton (1982);
- Yokoyama et al. (1979); Yount et al. (1974); Ziegler et al. (1980);
- Ziegler et al. (1981)
- *FIELD* RF
- 1. Abbott, C. M.; Skidmore, C. J.; Searle, A. G.; Peters, J.: Deficiency
- of adenosine deaminase in the wasted mouse. Proc. Nat. Acad. Sci. 83:
- 693-695, 1986.
-
- 2. Adrian, G. S.; Wiginton, D. A.; Hutton, J. J.: Characterization
- of normal and mutant adenosine deaminase messenger RNAs by translation
- and hybridization to a cDNA probe. Hum. Genet. 68: 169-172, 1984.
-
- 3. Adrian, G. S.; Wiginton, D. A.; Hutton, J. J.: Structure of adenosine
- deaminase mRNAs from normal and adenosine deaminase-deficient human
- cell lines. Molec. Cell. Biol. 4: 1712-1717, 1984.
-
- 4. Aitken, D. A.; Ferguson-Smith, M. A.: Investigation of the intrachromosomal
- position of the ADA locus on chromosome 20 by gene dosage studies. Cytogenet.
- Cell Genet. 22: 514-517, 1978.
-
- 5. Aitken, D. A.; Kleijer, W. J.; Niermeijer, M. F.; Herbschleb-Voogt,
- E.; Galjaard, H.: Prenatal detection of a probable heterozygote for
- ADA deficiency and severe combined immunodeficiency disease using
- a microradioassay. Clin. Genet. 17: 293-298, 1980.
-
- 6. Akeson, A. L.; Wiginton, D. A.; Dusing, M. R.; States, J. C.; Hutton,
- J. J.: Mutant human adenosine deaminase alleles and their expression
- by transfection into fibroblasts. J. Biol. Chem. 263: 16291-16296,
- 1988.
-
- 7. Akeson, A. L.; Wiginton, D. A.; Hutton, J. J.: Normal and mutant
- human adenosine deaminase genes. J. Cell. Biochem. 39: 217-228,
- 1989.
-
- 8. Akeson, A. L.; Wiginton, D. A.; States, J. C.; Perme, C. M.; Dusing,
- M. R.; Hutton, J. J.: Mutations in the human adenosine deaminase
- gene that affect protein structure and RNA splicing. Proc. Nat. Acad.
- Sci. 84: 5947-5951, 1987.
-
- 9. Arredondo-Vega, F. X.; Kurtzberg, J.; Chaffee, S.; Santisteban,
- I.; Reisner, E.; Povey, M. S.; Hershfield, M. S.: Paradoxical expression
- of adenosine deaminase in T cells cultured from a patient with adenosine
- deaminase deficiency and combined immunodeficiency. J. Clin. Invest. 86:
- 444-452, 1990.
-
- 10. Arredondo-Vega, F. X.; Santisteban, I.; Kelly, S.; Schlossman,
- C. M.; Umetsu, D. T.; Hershfield, M. S.: Correct splicing despite
- mutation of the invariant first nucleotide of a 5-prime splice site:
- a possible basis for disparate clinical phenotypes in siblings with
- adenosine deaminase deficiency. Am. J. Hum. Genet. 54: 820-830,
- 1994.
-
- 11. Berkvens, T. M.; Gerritsen, E. J. A.; Oldenburg, M.; Breukel,
- C.; Wijnen, J. T.; van Ormondt, H.; Vossen, J. M.; van der Eb, A.
- J.; Meera Khan, P.: Severe combined immune deficiency due to a homozygous
- 3.2-kb deletion spanning the promoter and first exon of the adenosine
- deaminase gene. Nucleic Acids Res. 15: 9365-9378, 1987.
-
- 12. Berkvens, T. M.; van Ormondt, H.; Gerritsen, E. J. A.; Meera Khan,
- P.; van der Eb, A. J.: Identical 3250-bp deletion between two AluI
- repeats in the ADA genes of unrelated ADA-SCID patients. Genomics 7:
- 486-490, 1990.
-
- 13. Blaese, R. M.; Culver, K. W.; Miller, A. D.; Carter, C. S.; Fleisher,
- T.; Clerici, M.; Shearer, G.; Chang, L., Chiang, Y.; Tolstoshev, P.;
- Greenblatt, J. J.; Rosenberg, S. A.; Klein, H.; Berger, M.; Mullen,
- C. A.; Ramsey, W. J.; Muul, L.; Morgan, R. A.; Anderson, W. F.: T
- lymphocyte-directed gene therapy for ADA-SCID: initial trial results
- after 4 years. Science 270: 475-480, 1995.
-
- 14. Bollinger, M. E.; Arredondo-Vega, F. X.; Santisteban, I.; Schwarz,
- K.; Hershfield, M. S.; Lederman, H. M.: Hepatic dysfunction as a
- complication of adenosine deaminase deficiency. New Eng. J. Med. 334:
- 1367-1371, 1996.
-
- 15. Bonthron, D. T.; Markham, A. F.; Ginsburg, D.; Orkin, S. H.:
- Identification of a point mutation in the adenosine deaminase gene
- responsible for immunodeficiency. J. Clin. Invest. 76: 894-897,
- 1985.
-
- 16. Bordignon, C.; Notarangelo, L. D.; Nobili, N.; Ferrari, G.; Casorati,
- G.; Panina, P.; Mazzolari, E.; Maggioni, D.; Rossi, C.; Servida, P.;
- Ugazio, A. G.; Mavilio, F.: Gene therapy in peripheral blood lymphocytes
- and bone marrow for ADA: immunodeficient patients. Science 270:
- 470-475, 1995.
-
- 17. Bortin, M. M.; Rimm, A. A. (eds.): Severe combined immunodeficiency
- disease: characterization of the disease and results of transplantation. J.A.M.A. 238:
- 591-600, 1977.
-
- 18. Boss, G. R.; Thompson, L. F.; O'Connor, R. D.; Ziering, R. W.;
- Seegmiller, J. E.: Ecto-5-prime-nucleotidase deficiency: association
- with adenosine deaminase deficiency and nonassociation with deoxyadenosine
- toxicity. Clin. Immun. Immunopath. 19: 1-7, 1981.
-
- 19. Chen, S.-H.; Ochs, H. D.; Scott, C. R.: Adenosine deaminase deficiency:
- disappearance of adenine deoxynucleotides from a patient's erythrocytes
- after successful marrow transplantation. J. Clin. Invest. 62: 1386-1389,
- 1978.
-
- 20. Chen, S.-H.; Ochs, H. D.; Scott, C. R.; Giblett, E. R.: Adenosine
- deaminase and nucleoside phosphorylase activity in patients with immunodeficiency
- syndromes. Clin. Immun. Immunopath. 13: 156-160, 1979.
-
- 21. Chen, S.-H.; Scott, C. R.; Giblett, E. R.: Adenosine deaminase:
- demonstration of a 'silent' gene associated with combined immunodeficiency
- disease. Am. J. Hum. Genet. 26: 103-107, 1974.
-
- 22. Cohen, A.; Hirschhorn, R.; Horowitz, S. D.; Rubinstein, A.; Polmar,
- S. H.; Hong, R.; Martin, D. W., Jr.: Deoxyadenosine triphosphate
- as a potentially toxic metabolite in adenosine deaminase deficiency. Proc.
- Nat. Acad. Sci. 75: 472-476, 1978.
-
- 23. Cook, P. J. L.; Hopkinson, D. A.; Robson, E. B.: The linkage
- relationships of adenosine deaminase. Ann. Hum. Genet. 34: 187-188,
- 1970.
-
- 24. Creagan, R. P.; Tischfield, J. A.; Nichols, E. A.; Ruddle, F.
- H.: Autosomal assignment of the gene for the form of adenosine deaminase
- which is deficient in patients with combined immunodeficiency syndrome.
- (Letter) Lancet II: 1449, 1973.
-
- 25. Daddona, P. E.; Kelley, W. N.: Human adenosine deaminase: stoichiometry
- of the adenosine deaminase-binding protein complex. Biochim. Biophys.
- Acta 580: 302-311, 1979.
-
- 26. Daddona, P. E.; Mitchell, B. S.; Meuwissen, H. J.; Davidson, B.
- L.; Wilson, J. M.; Koller, C. A.: Adenosine deaminase deficiency
- with normal immune function: an acidic enzyme mutation. J. Clin.
- Invest. 72: 483-492, 1983.
-
- 27. Detter, J. C.; Stamatoyannopoulos, G.; Giblett, E. R.; Motulsky,
- A. G.: Adenosine deaminase: racial distribution and report of a new
- phenotype. J. Med. Genet. 7: 356-357, 1970.
-
- 28. Dissing, J.; Knudsen, B.: Adenosine-deaminase deficiency and
- combined immunodeficiency syndrome. (Letter) Lancet II: 1316, 1972.
-
- 29. Dissing, J.; Knudsen, J. B.: A new red cell adenosine deaminase
- phenotype in man. Hum. Hered. 19: 375-377, 1969.
-
- 30. Dyminski, J. W.; Daoud, A.; Lampkin, B. C.; Limouze, S.; Donofrio,
- J.; Coleman, M. S.; Hutton, J. J.: Immunological and biochemical
- profiles in response to transfusion therapy in adenosine deaminase-deficient
- patient with severe combined immunodeficiency disease. Clin. Immun.
- Immunopath. 14: 307-326, 1979.
-
- 31. Ferrari, G.; Rossini, S.; Giavazzi, R.; Maggioni, D.; Nobili,
- N.; Soldati, M.; Ungers, G.; Mavilio, F.; Gilboa, E.; Bordignon, C.
- : An in vivo model of somatic cell gene therapy for human severe combined
- immunodeficiency. Science 251: 1363-1366, 1991.
-
- 32. Giblett, E. R.; Anderson, J. E.; Cohen, F.; Pollara, B.; Meuwissen,
- H. J.: Adenosine-deaminase deficiency in two patients with severely
- impaired cellular immunity. Lancet I: 1067-1069, 1972.
-
- 33. Hart, S. L.; Lane, A. B.; Jenkins, T.: Partial adenosine deaminase
- deficiency: another family from southern Africa. Hum. Genet. 74:
- 307-312, 1986.
-
- 34. Herbschleb-Voogt, E.; Scholten, J.-W.; Meera Khan, P.: Basic
- defect in the expression of adenosine deaminase in ADA SCID disease.
- II. Deficiency of ADA-CRM detected in heterozygote human-Chinese hamster
- cell hybrids. Hum. Genet. 63: 121-125, 1983.
-
- 35. Hershfield, M. S.: PEG-ADA: an alternative to haploidentical
- bone marrow transplantation and an adjunct to gene therapy for adenosine
- deaminase deficiency. Hum. Mutat. 5: 107-112, 1995.
-
- 36. Hershfield, M. S.; Buckley, R. H.; Greenberg, M. L.; Melton, A.
- L.; Schiff, R.; Hatem, C.; Kurtzberg, J.; Markert, M. L.; Kobayashi,
- R. H.; Kobayashi, A. L.; Abuchowski, A.: Treatment of adenosine deaminase
- deficiency with polyethylene glycol-modified adenosine deaminase. New
- Eng. J. Med. 316: 589-596, 1987.
-
- 37. Hershfield, M. S.; Kredich, N. M.: S-adenosylhomocysteine hydrolase
- is an adenosine-binding protein: a target for adenosine toxicity. Science 202:
- 757-760, 1978.
-
- 38. Hershfield, M. S.; Mitchell, B. S.: Immunodeficiency diseases
- caused by adenosine deaminase deficiency and purine nucleoside phosphorylase
- deficiency.In: Scriver, C. R.; Beaudet, A. L.; Sly, W. S.; Valle,
- D. (eds.): The Metabolic and Molecular Bases of Inherited Disease.
- Vol. 2. New York: McGraw-Hill (7th ed.): 1995. Pp. 1725-1768.
-
- 39. Hirschhorn, R.: Personal Communication. New York, N.Y. 1976.
-
- 40. Hirschhorn, R.: Identification of two new missense mutations
- (R156C and S291L) in two ADA(-) SCID patients unusual for response
- to therapy with partial exchange transfusions. Hum. Mutat. 1: 166-168,
- 1992.
-
- 41. Hirschhorn, R.; Chakravarti, V.; Puck, J.; Douglas, S. D.: Homozygosity
- for a newly identified missense mutation in a patient with very severe
- combined immunodeficiency due to adenosine deaminase deficiency (ADA-SCID). Am.
- J. Hum. Genet. 49: 878-885, 1991.
-
- 42. Hirschhorn, R.; Ellenbogen, A.: Genetic heterogeneity in adenosine
- deaminase (ADA) deficiency: five different mutations in five new patients
- with partial ADA deficiency. Am. J. Hum. Genet. 38: 13-25, 1986.
-
- 43. Hirschhorn, R.; Ellenbogen, A.; Tzall, S.: Five missense mutations
- at the adenosine deaminase locus (ADA) detected by altered restriction
- fragments and their frequency in ADA-patients with severe combined
- immunodeficiency (ADA-SCID). Am. J. Med. Genet. 42: 201-207, 1992.
-
- 44. Hirschhorn, R.; Levytska, V.; Parkman, R.: A mutant form of adenosine
- deaminase in severe combined immunodeficiency. (Abstract) J. Clin.
- Invest. 53: 33A, 1974.
-
- 45. Hirschhorn, R.; Martiniuk, F.; Roegner-Maniscalco, V.; Ellenbogen,
- A.; Perignon, J.-L.; Jenkins, T.: Genetic heterogeneity in partial
- adenosine deaminase deficiency. J. Clin. Invest. 71: 1887-1892,
- 1983.
-
- 46. Hirschhorn, R.; Papageorgiou, P. S.; Kesarwala, H. H.; Taft, L.
- T.: Amelioration of neurologic abnormalities after 'enzyme replacement'
- in adenosine deaminase deficiency. New Eng. J. Med. 303: 377-380,
- 1980.
-
- 47. Hirschhorn, R.; Roegner, V.; Jenkins, T.; Seaman, C.; Piomelli,
- S.; Borkowsky, W.: Erythrocyte adenosine deaminase deficiency without
- immunodeficiency: evidence for an unstable mutant enzyme. J. Clin.
- Invest. 64: 1130-1139, 1979.
-
- 48. Hirschhorn, R.; Tzall, S.; Ellenbogen, A.: Hot spot mutations
- in adenosine deaminase deficiency. Proc. Nat. Acad. Sci. 87: 6171-6175,
- 1990.
-
- 49. Hirschhorn, R.; Tzall, S.; Ellenbogen, A.; Orkin, S. H.: Identification
- of a point mutation resulting in a heat-labile adenosine deaminase
- (ADA) in two unrelated children with partial ADA deficiency. J. Clin.
- Invest. 83: 497-501, 1989.
-
- 50. Hirschhorn, R.; Vawter, G. F.; Kirkpatrick, J. A., Jr.; Rosen,
- F. S.: Adenosine deaminase deficiency: frequency and comparative
- pathology in autosomally recessive severe combined immunodeficiency. Clin.
- Immun. Immunopath. 14: 107-120, 1979.
-
- 51. Hirschhorn, R.; Yang, D. R.; Israni, A.: An asp8-to-asn substitution
- results in the adenosine deaminase (ADA) genetic polymorphism (ADA
- 2 allozyme): occurrence on different chromosomal backgrounds and apparent
- intragenic crossover. Ann. Hum. Genet. 58: 1-9, 1994.
-
- 52. Hirschhorn, R.; Yang, D. R.; Israni, A.; Huie, M. L.; Ownby, D.
- R.: Somatic mosaicism for a newly identified splice-site mutation
- in a patient with adenosine deaminase-deficient immunodeficiency and
- spontaneous clinical recovery. Am. J. Hum. Genet. 55: 59-68, 1994.
-
- 53. Hirschhorn, R.; Yang, D. R.; Puck, J. M.; Huie, M. L.; Jiang,
- C.-K.; Kurlandsky, L. E.: Spontaneous in vivo reversion to normal
- of an inherited mutation in a patient with adenosine deaminase deficiency. Nature
- Genet. 13: 290-295, 1996.
-
- 54. Hong, R.; Galti, R.; Rathbun, J. C.; Good, R. A.: Thymic hypoplasia
- and thyroid dysfunction. New Eng. J. Med. 282: 470-474, 1970.
-
- 55. Honig, J.; Martiniuk, F.; D'Eustachio, P.; Zamfirescu, C.; Desnick,
- R.; Hirschhorn, K.; Hirschhorn, L. R.; Hirschhorn, R.: Confirmation
- of the regional localization of the genes for human acid alpha-glucosidase
- (GAA) and adenosine deaminase (ADA) by somatic cell hybridization. Ann.
- Hum. Genet. 48: 49-56, 1984.
-
- 56. Hopkinson, D. A.; Cook, P. J. L.; Harris, H.: Further data on
- the adenosine deaminase (ADA) polymorphism and a report of a new phenotype. Ann.
- Hum. Genet. 32: 361-368, 1969.
-
- 57. Hutton, J. J.; Wiginton, D. A.; Coleman, M. S.; Fuller, S. A.;
- Limouze, S.; Lampkin, B. C.: Biochemical and functional abnormalities
- in lymphocytes from an adenosine deaminase-deficient patient during
- enzyme replacement therapy. J. Clin. Invest. 68: 413-421, 1981.
-
- 58. Jenkins, T.; Rabson, A. R.; Nurse, G. T.; Lane, A. B.; Hopkinson,
- D. A.: Deficiency of adenosine deaminase not associated with severe
- combined immunodeficiency. J. Pediat. 89: 732-736, 1976.
-
- 59. Jhanwar, S. C.; Berkvens, T. M.; Breukel, C.; van Ormondt, H.;
- van der Eb, A. J.; Meera Khan, P.: Localization of human adenosine
- deaminase (ADA) gene sequences to the q12-q13.11 region of chromosome
- 20 by in situ hybridization. Cytogenet. Cell Genet. 50: 168-171,
- 1989.
-
- 60. Kaitila, I.; Rimoin, D. L.; Cederbaum, S. D.; Stiehm, E. R.; Lechman,
- R. S.: Chondroosseous histopathology in adenosine deaminase deficient
- combined immunodeficiency disease. Birth Defects Orig. Art. Ser. XII(6):
- 115-121, 1976.
-
- 61. Kellems, R. E.; Yeung, C.-Y.; Ingolia, D. E.: Adenosine deaminase
- deficiency and severe combined immunodeficiencies. Trends Genet. 1:
- 278-283, 1985.
-
- 62. Kenny, A. B.; Hitzig, W. H.: Bone marrow transplantation for
- severe combined immunodeficiency disease: reported from 1968-1977. Europ.
- J. Pediat. 131: 155-176, 1979.
-
- 63. Koch, G.; Shows, T. B.: Somatic cell genetics of adenosine deaminase
- expression and severe combined immunodeficiency disease in humans. Proc.
- Nat. Acad. Sci. 77: 4211-4215, 1980.
-
- 64. Kredich, N. M.; Martin, D. W., Jr.: Role of 5-adenosylhomocysteine
- in adenosine-mediated toxicity in cultured mouse T-lymphoma cells. Cell 12:
- 931-938, 1977.
-
- 65. Levy, Y.; Hershfield, M. S.; Fernandez-Mejia, C.; Polmar, S. H.;
- Scudiery, D.; Berger, M.; Sorensen, R. U.: Adenosine deaminase deficiency
- with late onset of recurrent infections: response to treatment with
- polyethylene glycol-modified adenosine deaminase. J. Pediat. 113:
- 312-317, 1988.
-
- 66. Markert, M. L.; Hershfield, M. S.; Schiff, R. I.; Buckley, R.
- H.: Adenosine deaminase and purine nucleoside phosphorylase deficiencies:
- evaluation of therapeutic interventions in eight patients. J. Clin.
- Immun. 7: 389-399, 1987.
-
- 67. Markert, M. L.; Hershfield, M. S.; Wiginton, D. A.; States, J.
- C.; Ward, F. E.; Bigner, S. H.; Buckley, R. H.; Kaufman, R. E.; Hutton,
- J. J.: Identification of a deletion in the adenosine deaminase gene
- in a child with severe combined immunodeficiency. J. Immun. 138:
- 3203-3206, 1987.
-
- 68. Markert, M. L.; Hutton, J. J.; Wiginton, D. A.; States, J. C.;
- Kaufman, R. E.: Adenosine deaminase (ADA) deficiency due to deletion
- of the ADA gene promoter and first exon by homologous recombination
- between two Alu elements. J. Clin. Invest. 81: 1323-1327, 1988.
-
- 69. Markert, M. L.; Norby-Slycord, C.; Ward, F. E.: A high proportion
- of ADA point mutations associated with a specific alanine-to-valine
- substitution. Am. J. Hum. Genet. 45: 354-361, 1989.
-
- 70. Meuwissen, H. J.; Pollara, B.; Pickering, R. J.: Combined immunodeficiency
- disease associated with adenosine deaminase deficiency (report on
- a workshop held in Albany, New York, October 1, 1973). J. Pediat. 86:
- 169-181, 1975.
-
- 71. Migchielsen, A. A. J.; Breuer, M. L.; van Roon, M. A.; te Riele,
- H.; Zurcher, C.; Ossendorp, F.; Toutain, S.; Hershfield, M. S.; Berns,
- A.; Valerio, D.: Adenosine-deaminase-deficient mice die perinatally
- and exhibit liver-cell degeneration, atelectasis and small intestinal
- cell death. Nature Genet. 10: 279-287, 1995.
-
- 72. Mitchell, B. S.; Mejias, E.; Daddona, P. E.; Kelley, W. N.: Purinogenic
- immunodeficiency disease: selective toxicity of deoxyribonucleosides
- for T-cells. Proc. Nat. Acad. Sci. 75: 5011-5014, 1978.
-
- 73. Mohandas, T.; Sparkes, R. S.; Suh, E. J.; Hershfield, M. S.:
- Regional localization of the human genes for S-adenosylhomocysteine
- hydrolase (cen-q131) and adenosine deaminase (q131-qter) on chromosome
- 20. Hum. Genet. 66: 292-295, 1984.
-
- 74. Nielsen, K. B.; Tommerup, N.; Jespersen, B.; Nygaard, P.; Kleif,
- L.: Segregation of a t(3;20) translocation through three generations
- resulting in unbalanced karyotypes in six persons. J. Med. Genet. 23:
- 446-451, 1986.
-
- 75. Orkin, S. H.; Daddona, P. E.; Shewach, D. S.; Markham, A. F.;
- Bruns, G. A.; Goff, S. C.; Kelley, W. N.: Molecular cloning of human
- adenosine deaminase gene sequences. J. Biol. Chem. 258: 12753-12756,
- 1983.
-
- 76. Palmer, T. D.; Hock, R. A.; Osborne, W. R. A.; Miller, A. D.:
- Efficient retrovirus-mediated transfer and expression of a human adenosine
- deaminase gene in diploid skin fibroblasts from an adenosine deaminase-deficient
- human. Proc. Nat. Acad. Sci. 84: 1055-1059, 1987.
-
- 77. Parkman, R.; Gelfand, E. W.; Rosen, F. S.; Sanderson, A.; Hirschhorn,
- R.: Severe combined immunodeficiency and adenosine deaminase deficiency. New
- Eng. J. Med. 292: 714-719, 1975.
-
- 78. Petersen, M. B.; Tranebjaerg, L.; Tommerup, N.; Nygaard, P.; Edwards,
- H.: New assignment of the adenosine deaminase gene locus to chromosome
- 20q13.11 by study of a patient with interstitial deletion 20q. J.
- Med. Genet. 24: 93-96, 1987.
-
- 79. Polmar, S. H.; Stern, R. C.; Schwartz, A. L.; Wetzler, E. M.;
- Chase, P. A.; Hirschhorn, R.: Enzyme replacement therapy for adenosine
- deaminase deficiency and severe combined immunodeficiency. New Eng.
- J. Med. 295: 1337-1343, 1976.
-
- 80. Ratech, H.; Greco, M. A.; Gallo, G.; Rimoin, D. L.; Kamino, H.;
- Hirschhorn, R.: Pathologic findings in adenosine-deaminase-deficient
- severe combined immunodeficiency. I. Kidney, adrenal, and chondro-osseous
- tissue alterations. Am. J. Path. 120: 157-169, 1985.
-
- 81. Ritter, H.; Wendt, G. G.; Tariverdian, G.; Zelch, J.; Rube, M.;
- Kirchberg, G.: Genetics and linkage analysis of adenosine deaminase. Humangenetik 14:
- 69-71, 1971.
-
- 82. Rothschild, C. B.; Akots, G.; Hayworth, R.; Pettenati, M. J.;
- Rao, P. N.; Wood, P.; Stolz, F.-M.; Hansmann, I.; Serino, K.; Keith,
- T. P.; Fajans, S. S.; Bowden, D. W.: A genetic map of chromosome
- 20q12-q13.1: multiple highly polymorphic microsatellite and RFLP markers
- linked to the maturity-onset diabetes of the young (MODY) locus. Am.
- J. Hum. Genet. 52: 110-123, 1993.
-
- 83. Roychoudhury, A. K.; Nei, M.: Human Polymorphic Genes: World
- Distribution. New York: Oxford Univ. Press (pub.) 1988.
-
- 84. Rubinstein, A.; Hirschhorn, R.; Sicklick, M.; Murphy, R. A.:
- In vivo and in vitro effects of thymosin and adenosine deaminase on
- adenosine-deaminase-deficient lymphocytes. New Eng. J. Med. 300:
- 387-392, 1979.
-
- 85. Rudd, N. L.; Bain, H. W.; Giblett, E.; Chen, S.-H.; Worton, R.
- G.: Partial trisomy 20 confirmed by gene dosage studies. Am. J.
- Med. Genet. 4: 357-364, 1979.
-
- 86. Santisteban, I.; Arredondo-Vega, F. X.; Kelly, S.; Mary, A.; Fischer,
- A.; Hummell, D. S.; Lawton, A.; Sorensen, R. U.; Stiehm, E. R.; Uribe,
- L.; Weinberg, K.; Hershfield, M. S.: Novel splicing, missense, and
- deletion mutations in seven adenosine deaminase-deficient patients
- with late/delayed onset of combined immunodeficiency disease: contribution
- of genotype to phenotype. J. Clin. Invest. 92: 2291-2302, 1993.
-
- 87. Schmalstieg, F. C.; Mills, G. C.; Tsuda, H.; Goldman, A. S.:
- Severe combined immunodeficiency in a child with a healthy adenosine
- deaminase deficient mother. Pediat. Res. 17: 935-940, 1983.
-
- 88. Schrader, W. P.; Pollara, B.; Meuwissen, H. J.: Characterization
- of the residual adenosine deaminating activity in the spleen of a
- patient with combined immunodeficiency disease and adenosine deaminase
- deficiency. Proc. Nat. Acad. Sci. 75: 446-450, 1978.
-
- 89. Scott, C. R.; Chen, S.-H.; Giblett, E. R.: Deletion of the carrier
- state in combined immunodeficiency disease associated with deaminase
- deficiency. J. Clin. Invest. 53: 1194-1196, 1974.
-
- 90. Shovlin, C. L.; Hughes, J. M. B.; Simmonds, H. A.; Fairbanks,
- L.; Deacock, S.; Lechler, R.; Roberts, I.; Webster, A. D. B.: Adult
- presentation of adenosine deaminase deficiency. (Letter) Lancet 341:
- 1471, 1993.
-
- 91. Shovlin, C. L.; Simmonds, H. A.; Fairbanks, L. D.; Deacock, S.
- J.; Hughes, J. M. B.; Lechler, R. I.; Webster, A. D. B.; Sun, X.-M.;
- Webb, J. C.; Soutar, A. K.: Adult onset immunodeficiency caused by
- inherited adenosine deaminase deficiency. J. Immun. 153: 2331-2339,
- 1994.
-
- 92. Spencer, N.; Hopkinson, D. A.; Harris, H.: Adenosine deaminase
- polymorphism in man. Ann. Hum. Genet. 32: 9-14, 1968.
-
- 93. Tariverdian, G.; Ritter, H.: Adenosine deaminase polymorphism
- (EC 3.5.4.4): formal genetics and linkage relations. Humangenetik 7:
- 176-178, 1969.
-
- 94. Tischfield, J. A.; Creagan, R. P.; Nichols, E. A.; Ruddle, F.
- H.: Assignment of a gene for adenosine deaminase to human chromosome
- 20. Hum. Hered. 24: 1-11, 1974.
-
- 95. Tzall, S.; Ellenbogen, A.; Eng, F.; Hirschhorn, R.: Identification
- and characterization of nine RFLPs at the adenosine deaminase (ADA)
- locus. Am. J. Hum. Genet. 44: 864-875, 1989.
-
- 96. Umetsu, D. T.; Schlossman, C. M.; Ochs, H. D.; Hershfield, M.
- S.: Heterogeneity of phenotype in two siblings with adenosine deaminase
- deficiency. J. Allergy Clin. Immunol. 93: 543-550, 1994.
-
- 97. Valerio, D.; Dekker, B. M. M.; Duyvesteyn, M. G. C.; van der Voorn,
- L.; Berkvens, T. M.; van Ormondt, H.; van der Eb, A. J.: One adenosine
- deaminase allele in a patient with severe combined immunodeficiency
- contains a point mutation abolishing enzyme activity. EMBO J. 5:
- 113-119, 1986.
-
- 98. Valerio, D.; Duyvesteyn, M. G. C.; Dekker, B. M. M.; Weeda, G.;
- Berkvens, T. M.; van der Voorn, L.; van Ormondt, H.; van der Eb, A.
- J.: Adenosine deaminase: characterization and expression of a gene
- with a remarkable promoter. EMBO J. 4: 437-443, 1985.
-
- 99. Valerio, D.; Duyvesteyn, M. G. C.; Meera Khan, P.; Pearson, P.
- L.; Geurts van Kessel, A.; van Ormondt, H.: Direct assignment of
- ADA gene to chromosome 20. (Abstract) Cytogenet. Cell Genet. 37:
- 599, 1984.
-
- 100. Valerio, D.; Duyvesteyn, M. G. C.; van Ormondt, H.; Meera Khan,
- P.; van der Eb, A. J.: Adenosine deaminase (ADA) deficiency in cells
- derived from humans with severe combined immunodeficiency is due to
- an aberration of the ADA protein. Nucleic Acids Res. 12: 1015-1024,
- 1984.
-
- 101. Valerio, D.; McIvor, R. S.; Williams, S. R.; Duyvesteyn, M. G.
- C.; van Ormondt, H.; van der Eb, A. J.; Martin, D. W., Jr.: Cloning
- of human adenosine deaminase cDNA and expression in mouse cells. Gene 31:
- 147-153, 1984.
-
- 102. Van der Weyden, M. B.; Kelley, W. N.: Adenosine deaminase deficiency
- in severe combined immunodeficiency: evidence for a posttranslational
- defect. (Abstract) J. Clin. Invest. 53: 81A-82A, 1974.
-
- 103. Wakamiya, M.; Blackburn, M. R.; Jurecic, R.; McArthur, M. J.;
- Geske, R. S.; Cartwright, Jr., J.; Mitani, K.; Vaishnav, S.; Belmont,
- J. W.; Kellems, R. E.; Finegold, M. J.; Montgomery, Jr., C. A.; Bradley,
- A.; Caskey, C. T.: Disruption of the adenosine deaminase gene causes
- hepatocellular impairment and perinatal lethality in mice. Proc.
- Nat. Acad. Sci. 92: 3673-3677, 1995.
-
- 104. Weitkamp, L. R.: Further data on the genetic linkage relations
- of the adenosine deaminase locus. Hum. Hered. 21: 351-356, 1971.
-
- 105. Weitkamp, L. R.: Genetic linkage relationships of the ADA and
- 6-PGD loci in 'Humangenetik.' (Letter) Humangenetik 15: 359-360,
- 1972.
-
- 106. Wiginton, D. A.; Adrian, G. S.; Friedman, R. L.; Suttle, D. P.;
- Hutton, J. J.: Cloning of cDNA sequences of human adenosine deaminase. Proc.
- Nat. Acad. Sci. 80: 7481-7485, 1983.
-
- 107. Wiginton, D. A.; Adrian, G. S.; Hutton, J. J.: Sequence of human
- adenosine deaminase cDNA including the coding region and a small intron. Nucleic
- Acids Res. 12: 2439-2446, 1984.
-
- 108. Wiginton, D. A.; Hutton, J. J.: Immunoreactive protein in adenosine
- deaminase deficient human lymphoblast cell lines. J. Biol. Chem. 257:
- 3211-3217, 1982.
-
- 109. Wiginton, D. A.; Kaplan, D. J.; States, J. C.; Akeson, A. L.;
- Perme, C. M.; Bilyk, I. J.; Vaughn, A. J.; Lattier, D. L.; Hutton,
- J. J.: Complete sequence and structure of the gene for human adenosine
- deaminase. Biochemistry 25: 8234-8244, 1986.
-
- 110. Yokoyama, S.; Hayashi, T.; Yoshimura, Y.; Irimada, K.; Saito,
- T.; Akiba, T.; Tsuchiya, S.: Severe combined immunodeficiency disease
- with adenosine deaminase deficiency. Tohoku J. Exp. Med. 129: 197-202,
- 1979.
-
- 111. Yount, J.; Nichols, P.; Ochs, H. D.; Hammar, S. P.; Scott, C.
- R.; Chen, S.-H.; Giblett, E. R.; Wedgwood, R. J.: Absence of erythrocyte
- adenosine deaminase associated with severe combined immunodeficiency. J.
- Pediat. 84: 173-177, 1974.
-
- 112. Ziegler, J. B.; Lee, C. H.; Van Der Weyden, M. B.; Bagnara, A.
- S.; Beveridge, J.: Severe combined immunodeficiency and adenosine
- deaminase deficiency: failure of enzyme replacement therapy. Arch.
- Dis. Child. 55: 452-457, 1980.
-
- 113. Ziegler, J. B.; Van Der Weyden, M. B.; Lee, C. H.; Daniel, A.
- : Prenatal diagnosis for adenosine deaminase deficiency. J. Med.
- Genet. 18: 154-156, 1981.
-
- *FIELD* CS
-
- Immunology:
- Severe combined immunodeficiency disease
-
- Skel:
- Skeletal dysplasia
-
- Head:
- Normocephaly
-
- Facies:
- Normal
-
- Heme:
- B-cell deficiency;
- T-cell deficiency;
- CD4 lymphopenia;
- Idiopathic thrombocytopenic purpura
-
- Pulm:
- Recurrent respiratory infections;
- Asthma
-
- GI:
- Hepatosplenomegaly
-
- Misc:
- Late onset CID with allelic variant .0020;
- Recurrent bacterial, viral, and fungal infections
-
- Lab:
- Adenosine deaminase deficiency
-
- Inheritance:
- Autosomal dominant (20q13.11);
- the deficiency syndrome is an autosomal recessive disorder
-
- *FIELD* CN
- Iosif W. Lurie - updated: 09/26/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 09/26/1996
- carol: 7/23/1996
- carol: 6/29/1996
- mark: 6/27/1996
- terry: 6/25/1996
- terry: 6/6/1996
- terry: 6/4/1996
- carol: 5/18/1996
- mark: 12/12/1995
- terry: 12/5/1995
- carol: 11/10/1994
- terry: 8/30/1994
- jason: 7/26/1994
- warfield: 4/7/1994
- pfoster: 3/25/1994
- mimadm: 3/13/1994
-
- *RECORD*
- *FIELD* NO
- 102710
- *FIELD* TI
- 102710 ADENOSINE DEAMINASE COMPLEXING PROTEIN-1; ADCP1
- *FIELD* TX
- ADA occurs in a small molecular form (MW 33,000) called red cell ADA
- (102700) and in a large molecular form (MW 200,000) called
- tissue-specific ADA. The five ADA tissue enzymes consist of one or more
- molecules of red cell ADA and one molecule of adenosine deaminase
- complexing protein (also known as a conversion factor). Koch and Shows
- (1978) concluded that one tissue enzyme, ADA-d, is dependent upon at
- least two genes--the chromosome 20 gene for ADA and a gene on chromosome
- 6 which determines an ADA-complexing protein (ADCP1). Herbschleb-Voogt
- et al. (1979) and Koch and Shows (1979) concluded that expression of
- ADA-d is dependent on another gene, ADCP2 (102720), located on
- chromosome 2. The assignment of an ADCP gene to chromosome 6 might be
- considered 'in limbo' (Shows, 1982).
-
- *FIELD* SA
- Daddona and Kelley (1979); Koch and Shows (1980)
- *FIELD* RF
- 1. Daddona, P. E.; Kelley, W. N.: Human adenosine deaminase: stoichiometry
- of the adenosine deaminase-binding protein complex. Biochim. Biophys.
- Acta 580: 302-311, 1979.
-
- 2. Herbschleb-Voogt, E.; Grzeschik, K.-H.; de Wit, J.; Pearson, P.
- L.; Meera Khan, P.: Assignment of a structural gene for adenosine
- deaminase complexing protein (ADCP) to human chromosome 2 in interspecific
- somatic cell hybrids. (Abstract) Cytogenet. Cell Genet. 25: 163
- only, 1979.
-
- 3. Koch, G.; Shows, T. B.: A gene on human chromosome 6 functions
- in assembly of tissue-specific adenosine deaminase isozymes. Proc.
- Nat. Acad. Sci. 75: 3876-3880, 1978.
-
- 4. Koch, G.; Shows, T. B.: Somatic cell genetics of adenosine deaminase
- expression and severe combined immune deficiency disease in man. Proc.
- Nat. Acad. Sci. 77: 4211-4215, 1980.
-
- 5. Koch, G. A.; Shows, T. B.: Genes on human chromosomes 2 and 6
- are required for expression of the adenosine deaminase complexing
- protein (ADCP) in human-mouse somatic cell hybrids. (Abstract) Cytogenet.
- Cell Genet. 25: 174 only, 1979.
-
- 6. Shows, T. B.: Personal Communication. Buffalo, N. Y. 5/5/1982.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- warfield: 4/7/1994
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 3/30/1988
-
- *RECORD*
- *FIELD* NO
- 102720
- *FIELD* TI
- *102720 ADENOSINE DEAMINASE COMPLEXING PROTEIN-2; ADCP2
- T-CELL ACTIVATION ANTIGEN CD26; CD26;;
- DIPEPTIDYLPEPTIDASE IV; DPP4;;
- DIPEPTIDYLPEPTIDASE, INTESTINAL
- *FIELD* TX
- Koch and Shows (1979, 1980) concluded that at least 3 genes are involved
- in the expression of adenosine deaminase complexing protein: ADA
- (102700) on chromosome 20, ADCP1 (102710) on chromosome 6, and ADCP2 on
- chromosome 2. On the other hand, from studies in mouse-man and
- hamster-man hybrid cells, Herbschleb-Voogt et al. (1981) concluded that
- a gene or genes on human chromosome 2 determine the expression of ADCP
- and that neither chromosome 6 nor any other chromosome of man carries
- genes involved in the formation of ADCP. Van Cong et al. (1981)
- concluded that the gene for ADCP on chromosome 2 is located between MDH1
- (154200) and IDH1 (147700), i.e., in the segment 2p23-q32. Could one
- form of adenosine deaminase deficiency (leading to severe combined
- immunodeficiency) represent, in fact, deficiency of the complexing
- protein?
-
- Data presented by Kameoka et al. (1993) and partial amino acid sequence
- data presented by Morrison et al. (1993) indicated that the ADA binding
- protein is identical to CD26, a T-cell activation molecule and a 110-kD
- glycoprotein that is present also on epithelial cells of various tissues
- including the liver, kidney, and intestine. Kameoka et al. (1993) listed
- the reasons for thinking that ADA on the T-cell surface is regulated
- during the process of T-cell activation, that CD26 may be involved in
- regulating the extracellular concentration of ADA, and that some cases
- of SCID may be related to mutation in this gene.
-
- The CD4 antigen (186940) is essential for binding human immunodeficiency
- virus (HIV) particles, but is not sufficient for efficient viral entry
- and infection. Callebaut et al. (1993) demonstrated that a cofactor
- necessary for efficient function is CD26. Coexpression of human CD4 and
- CD26 in murine NIH 3T3 cells rendered them permissive to infection by
- HIV. They suggested the possibility of developing specific inhibitors
- that would block the function of CD26 and thus be useful as effective
- therapeutic agents in AIDS patients.
-
- Dipeptidylpeptidase IV (DPP4; EC 3.4.14.5) is identical to ADA
- complexing protein-2 and to the T-cell activation antigen CD26. DPP4 is
- a serine exopeptidase that cleaves X-proline dipeptides from the
- N-terminus of polypeptides. It is an intrinsic membrane glycoprotein
- anchored into the cell membrane by its N-terminal end. High levels of
- the enzyme are found in the brush-border membranes of the kidney
- proximal tubule and of the small intestine, but several other tissues
- also express the enzyme. The enzyme is present in the fetal colon but
- disappears at birth. It is ectopically expressed in some human colon
- adenocarcinomas and human colon cancer cell lines. From such a colon
- cancer cell line, Darmoul et al. (1990) isolated a cDNA probe for
- intestinal dipeptidylpeptidase IV and, by Southern analysis of somatic
- cell hybrids, assigned the gene to chromosome 2. This assignment was
- confirmed by Mathew et al. (1994), who sublocalized the DPP4 gene to
- 2q23 by fluorescence in situ hybridization. Misumi et al. (1992)
- isolated and sequenced the cDNA coding for DPP4. The nucleotide sequence
- (3,465 bp) of the cDNA contained an open reading frame encoding a
- polypeptide comprising 766 amino acids, 1 residue less than those of the
- rat protein. The predicted amino acid sequence exhibited 84.9% identity
- to that of the rat enzyme.
-
- Abbott et al. (1994) demonstrated that CD26 spans approximately 70 kb
- and contains 26 exons, ranging in size from 45 bp to 1.4 kb. The
- nucleotides that encode the serine recognition site (G-W-S-Y-G) are
- split between 2 exons. This clearly distinguishes the genomic
- organization of the prolyl oligopeptidase family from that of the
- classic serine protease family. CD26 encodes 2 messages sized at about
- 4.2 and 2.8 kb. These are both expressed at high levels in the placenta
- and kidney and at moderate levels in the lung and liver. Only the 4.2 kb
- mRNA was expressed at low levels in skeletal muscle, heart, brain, and
- pancreas. By fluorescence in situ hybridization, Abbott et al. (1994)
- mapped the gene to 2q24.3.
-
- *FIELD* RF
- 1. Abbott, C. A.; Baker, E.; Sutherland, G. R.; McCaughan, G. W.:
- Genomic organization, exact localization, and tissue expression of
- the human CD26 (dipeptidyl peptidase IV) gene. Immunogenetics 40:
- 331-338, 1994.
-
- 2. Callebaut, C.; Krust, B.; Jacotot, E.; Hovanessian, A. G.: T cell
- activation antigen, CD26, as a cofactor for entry of HIV in CD4+ cells.
- Science 262: 2045-2050, 1993.
-
- 3. Darmoul, D.; Lacasa, M.; Chantret, I.; Swallow, D. M.; Trugnan,
- G.: Isolation of a cDNA probe for the human intestinal dipeptidylpeptidase
- IV and assignment of the gene locus DPP4 to chromosome 2. Ann. Hum.
- Genet. 54: 191-197, 1990.
-
- 4. Herbschleb-Voogt, E.; Grzeschik, K.-H.; Pearson, P. L.; Meera Khan,
- P.: Assignment of adenosine deaminase complexing protein (ADCP) gene(s)
- to human chromosome 2 in rodent-human somatic cell hybrids. Hum.
- Genet. 59: 317-323, 1981.
-
- 5. Kameoka, J.; Tanaka, T.; Nojima, Y.; Schlossman, S. F.; Morimoto,
- C.: Direct association of adenosine deaminase with a T cell activation
- antigen, CD26. Science 261: 466-469, 1993.
-
- 6. Koch, G.; Shows, T. B.: Somatic cell genetics of adenosine deaminase
- expression and severe combined immune deficiency disease in man. Proc.
- Nat. Acad. Sci. 77: 4211-4215, 1980.
-
- 7. Koch, G. A.; Shows, T. B.: Genes on human chromosomes 2 and 6
- are required for expression of the adenosine deaminase complexing
- protein (ADCP) in human-mouse somatic cell hybrids. (Abstract) Cytogenet.
- Cell Genet. 25: 174, 1979.
-
- 8. Mathew, S.; Morrison, M. E.; Murty, V. V. V. S.; Houghton, A. N.;
- Chaganti, R. S. K.: Assignment of the DPP4 gene encoding adenosine
- deaminase binding protein (CD26/dipeptidylpeptidase IV) to 2q23. Genomics 22:
- 211-212, 1994.
-
- 9. Misumi, Y.; Hayashi, Y.; Arakawa, F.; Ikehara, Y.: Molecular cloning
- and sequence analysis of human dipeptidyl peptidase IV, a serine proteinase
- on the cell surface. Biochim. Biophys. Acta 1131: 333-336, 1992.
-
- 10. Morrison, M. E.; Vijayasaradhi, S.; Engelstein, D.; Albino, A.
- P.; Houghton, A. N.: A marker for neoplastic progression of human
- melanocytes is a cell surface ectopeptidase. J. Exp. Med. 117:
- 1135-1143, 1993.
-
- 11. Van Cong, N.; Weil, D.; Gross, M.-S.; Foubert, C.; Jami, J.; Frezal,
- J.: Controle genetique et epigenetique de l'expression de l'adenosine
- deaminase. Analyse des cellules humaines et hybrides homme-rongeur.
- Ann. Genet. 24: 141-147, 1981.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 05/18/1996
- carol: 1/19/1995
- carol: 12/22/1993
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 102730
- *FIELD* TI
- 102730 ADENOSINE DEAMINASE, ELEVATED, HEMOLYTIC ANEMIA DUE TO
- *FIELD* TX
- In addition to the polymorphism of red cell ADA (EC 3.5.4.4.) and the
- deficiency state of the enzyme leading to immunodeficiency (102700),
- elevated red cell ADA (with decreased ATP) has been reported, first by
- Valentine et al. (1977) in Los Angeles and later by Miwa et al. (1978)
- and Fujii et al. (1980) in Japan and by Perignon et al. (1982) in
- France. The proband in the case reported by Miwa et al. (1978) was a
- 38-year-old Japanese male with compensated hemolytic anemia. His red
- cells showed moderate stomatocytosis and his red cell ADA activity was
- 40 times normal. The mother showed a 4-fold increase in red cell ADA;
- the father's enzyme levels were normal. In lymphocytes ADA levels were
- nearly normal. Valentine's patient also showed stomatocytosis. In his
- family 12 affected persons in 3 generations showed ADA levels of 45 to
- 70 times the normal and no one showed intermediate levels as in the
- mother of Miwa's family. Serum uric acid levels were mildly elevated.
- This mutation probably involves a regulatory gene at a locus separate
- from the structural locus for ADA carried on chromosome 20. In the
- 10-year-old affected male with severe hemolytic disease reported by
- Perignon et al. (1982), the level of ADA was about 85 times the normal.
- Evidence was presented that the excessive ADA activity in red cells was
- due to an abnormal amount of a catalytically and immunologically normal
- enzyme. Novelli et al. (1986) found a 4-fold increase in red cell ADA in
- a 16-month-old Libyan infant without hemolytic anemia but with mild
- anisopoikilocytosis. The parents, who were related as first cousins, and
- a healthy brother had normal red cell ADA levels. Glader et al. (1983)
- suggested that elevated ADA activity is a feature of Blackfan-Diamond
- anemia (205900).
-
- Chottiner et al. (1987) studied the family originally described by
- Valentine et al. (1977). They verified that red cell ADA-specific
- activity was 70 to 100 times the normal levels. Western blots
- demonstrated a corresponding increase in red cell ADA-specific
- immunoreactive protein. Analysis of genomic DNA showed no evidence for
- amplification or major structural changes in the ADA gene. ADA-specific
- mRNA from proband reticulocytes was comparable in size and amount to
- mRNA from control reticulocytes. This finding excluded increased
- transcription of the gene or increased stability of red cell ADA mRNA.
- On the other hand, Chottiner et al. (1987) found evidence of
- posttranslational abnormality. In vitro translation and
- immunoprecipitation experiments consistently showed a band of about
- 42,000 molecular weight synthesized from proband reticulocyte mRNA but
- not control mRNA. These data strongly suggested that red cell ADA
- overabundance in this disorder was due to an abnormality intrinsic to
- reticulocyte ADA mRNA that results in its increased translation. There
- have been several examples of mutations that affect the translational
- efficiency of specific mRNAs, usually mutations in the 5-prime noncoding
- region. The reason for the tissue specificity of the abnormality was not
- clear. The in vitro translation experiments made the possibility of a
- transacting factor coded by a separate locus less likely.
-
- In the form of severe combined immunodeficiency with deficiency of ADA,
- structural changes such as point mutations have been identified in the
- ADA gene on chromosome 20 and the deficiency is found in all tissues. In
- the disorder of ADA excess, only the erythroid elements show the
- abnormality and the ADA molecule is structurally normal by all the usual
- criteria, including electrophoretic migration, kinetics for various
- substrates and inhibitors, heat stability, specific activity, pH
- optimum, immunologic reactivity, amino acid composition, and peptide
- patterns. The defect is transmitted as an autosomal dominant. The
- mutation is presumably in a gene separate from the structural gene for
- ADA. The study of these families with DNA markers located in the region
- of the ADA gene on 20q might prove conclusively that the determinant is
- at a site remote from the ADA gene. Such experiments were performed by
- Chen et al. (1993), who, to determine whether increased ADA mRNA is due
- to a cis-acting or a trans-acting mutation, took advantage of a highly
- polymorphic TAAA repeat located at the tail end of an Alu repeat
- approximately 1.1 kb upstream of the ADA gene. Using PCR to amplify this
- region, they identified 5 different alleles in 19 members of an affected
- family. All 11 affected individuals had an ADA allele with 12 TAAA
- repeats, whereas none of the 8 normal individuals did. They concluded
- that this disorder results from a cis-acting mutation in the vicinity of
- the ADA gene. Chen and Mitchell (1994) examined reporter gene activity
- using constructs containing 10.6 kb of 5-prime flanking sequence and
- 12.3 kb of the first intron of the ADA gene from normal and mutant
- alleles. No differences in chloramphenicol acetyltransferase (CAT)
- activity were found in transient transfection experiments using
- erythroleukemia cell lines. Furthermore, transgenic mice containing the
- ADA constructs showed CAT activities in erythrocytes and bone marrow
- that did not differ between the normal and mutant alleles. Results were
- interpreted as indicating that the mutation responsible for ADA
- overexpression is unlikely to reside in the 5-prime and promoter regions
- or in the regulatory regions of the first intron.
-
- *FIELD* RF
- 1. Chen, E. H.; Mitchell, B. S.: Hereditary overexpression of adenosine
- deaminase in erythrocytes: studies in erythroid cell lines and transgenic
- mice. Blood 84: 2346-2353, 1994.
-
- 2. Chen, E. H.; Tartaglia, A. P.; Mitchell, B. S.: Hereditary overexpression
- of adenosine deaminase in erythrocytes: evidence for a cis-acting
- mutation. Am. J. Hum. Genet. 53: 889-893, 1993.
-
- 3. Chottiner, E. C.; Cloft, H. J.; Tartaglia, A. P.; Mitchell, B.
- S.: Elevated adenosine deaminase activity and hereditary hemolytic
- anemia: evidence for abnormal translational control of protein synthesis.
- J. Clin. Invest. 79: 1001-1005, 1987.
-
- 4. Fujii, H.; Miwa, S.; Suzuki, K.: Purification and properties of
- adenosine deaminase in normal and hereditary hemolytic anemia with
- increased red cell activity. Hemoglobin 4: 693-705, 1980.
-
- 5. Glader, B. E.; Backer, K.; Diamond, L. K.: Elevated erythrocyte
- adenosine deaminase activity in congenital hypoplastic anemia. New
- Eng. J. Med. 309: 1486-1490, 1983.
-
- 6. Miwa, S.; Fujii, H.; Matsumoto, N.; Nakatsuji, T.; Oda, S.; Asano,
- H.; Asano, S.; Miura, Y.: A case of red-cell adenosine deaminase
- over-production associated with hereditary hemolytic anemia found
- in Japan. Am. J. Hemat. 5: 107-115, 1978.
-
- 7. Novelli, G.; Stocchi, V.; Giannotti, A.; Magnani, M.; Dallapiccola,
- B.: Increased erythrocyte adenosine deaminase activity without haemolytic
- anaemia. Hum. Hered. 36: 37-40, 1986.
-
- 8. Perignon, J.-L.; Hamet, M.; Buc, H. A.; Cartier, P. H.; Derycke,
- M.: Biochemical study of a case of hemolytic anemia with increased
- (85-fold) red cell adenosine deaminase. Clin. Chim. Acta 124: 205-212,
- 1982.
-
- 9. Valentine, W. N.; Paglia, D. E.; Tartaglia, A. P.; Gilsanz, F.
- : Hereditary hemolytic anemia with increased red cell adenosine deaminase
- (45- to 70-fold) and decreased adenosine triphosphate. Science 195:
- 783-785, 1977.
-
- *FIELD* CS
-
- Heme:
- Hemolytic anemia;
- Red cell stomatocytosis;
- Anisopoikilocytosis
-
- Lab:
- Elevated red cell ADA;
- Decreased ATP;
- Serum uric acid mildly elevated
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/16/1986
-
- *FIELD* ED
- terry: 12/20/1994
- carol: 4/6/1994
- mimadm: 3/11/1994
- carol: 10/12/1993
- carol: 10/7/1993
- carol: 3/31/1992
-
- *RECORD*
- *FIELD* NO
- 102750
- *FIELD* TI
- *102750 ADENOSINE KINASE; ADK
- *FIELD* TX
- Adenosine kinase (ATP:adenosine 5-prime-phosphotransferase; EC 2.7.1.20)
- is an abundant enzyme in mammalian tissues that catalyzes the transfer
- of the gamma-phosphate from ATP to adenosine, thereby serving as a
- potentially important regulator of concentrations of both extracellular
- adenosine and intracellular adenine nucleotides. Adenosine has
- widespread effects on the cardiovascular, nervous, respiratory, and
- immune systems and inhibitors of ADK could play an important
- pharmacological role in increasing intravascular adenosine
- concentrations and acting as antiinflammatory agents. Spychala et al.
- (1996) obtained full-length cDNA clones encoding catalytically active
- ADK from lymphocyte, placental, and liver cDNA libraries. On Northern
- blots of all tissues examined, they identified mRNA species of 1.3 and
- 1.8 kb, attributable to alternative polyadenylation sites at the 3-prime
- end of the gene. The encoded protein consisted of 345 amino acids with a
- calculated molecular size of 38.7 kD and without any sequence
- similarities to other well-characterized mammalian nucleoside kinases.
- In contrast, 2 regions were identified with significant sequence
- identity to microbial ribokinase and fructokinases and a bacterial
- inosine/guanosine kinase. Thus, ADK is a structurally distinct mammalian
- nucleoside kinase that appears to be akin to sugar kinases of microbial
- origin.
-
- The structural gene for this enzyme was tentatively assigned to
- chromosome 10 by somatic cell hybrid studies (Klobutcher et al., 1976).
- By the principle of gene dosage, Francke and Thompson (1979) concluded
- by exclusion that ADK must be in the region 10q11-10q24. In a case of
- trisomy 10p, Snyder et al. (1984) found normal levels of ADK.
-
- *FIELD* SA
- Chan et al. (1978)
- *FIELD* RF
- 1. Chan, T.-S.; Cregan, R. P.; Reardon, M. P.: Adenosine kinase as
- a new selective marker in somatic cell genetics: isolation of adenosine
- kinase-deficient mouse cell lines and human-mouse hybrid cell lines
- containing adenosine kinase. Somat. Cell Genet. 4: 1-12, 1978.
-
- 2. Francke, U.; Thompson, L.: Regional mapping, by exclusion, of
- adenosine kinase (ADK) on human chromosome 10 using the gene dosage
- approach. (Abstract) Cytogenet. Cell Genet. 25: 156, 1979.
-
- 3. Klobutcher, L. A.; Nichols, E. A.; Kucherlapati, R. S.; Ruddle,
- F. H.: Assignment of the gene for human adenosine kinase to chromosome
- 10 using a somatic cell hybrid clone panel. Cytogenet. Cell Genet. 16:
- 171-174, 1976.
-
- 4. Snyder, F. F.; Lin, C. C.; Rudd, N. L.; Shearer, J. E.; Heikkila,
- E. M.; Hoo, J. J.: A de novo case of trisomy 10p: gene dosage studies
- of hexokinase, inorganic pyrophosphatase and adenosine kinase. Hum.
- Genet. 67: 187-189, 1984.
-
- 5. Spychala, J.; Datta, N. S.; Takabayashi, K.; Datta, M.; Fox, I.;
- Gribbin, T.; Mitchell, B.: Cloning of human adenosine kinase cDNA:
- sequenced similarity to microbial ribokinases and fructokinases, Proc.
- Nat. Acad. Sci. 93: 1232-1237, 1996.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 02/26/1996
- mark: 2/20/1996
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 102770
- *FIELD* TI
- *102770 ADENOSINE MONOPHOSPHATE DEAMINASE-1; AMPD1
- AMP DEAMINASE
- MYOADENYLATE DEAMINASE DEFICIENCY, MYOPATHY DUE TO, INCLUDED;;
- MAD DEFICIENCY, INCLUDED;;
- MADA DEFICIENCY, INCLUDED
- *FIELD* TX
- Morton et al. (1989) used in situ hybridization and somatic cell hybrid
- analysis to demonstrate that the AMPD1 gene maps to human chromosome 1.
- Moseley et al. (1990) demonstrated that the murine equivalent is located
- close to Ampd-2 on distal mouse chromosome 3. Sabina et al. (1990)
- stated that tissue-specific isoforms of AMP deaminase are produced by
- differential expression of 2 genes as well as by alternative splicing of
- the primary transcript of 1 of these genes. The gene is approximately 20
- kb long with 16 exons ranging in size from 101 to 220 nucleotides, with
- the exception of exon 2, which comprises only 12 nucleotides. Intron
- size ranges from 159 bp for intron 14 to several kilobases. By in situ
- hybridization and analysis of human-mouse somatic cell hybrids, Sabina
- et al. (1990) localized the AMPD1 gene to 1p21-p13.
-
- Morisaki et al. (1990) found that AMPD1 is expressed at high levels in
- skeletal muscle of the adult rat, whereas AMPD2 is the predominant gene
- expressed in nonmuscle tissues and smooth muscle of the adult rat and is
- also the predominant gene expressed in embryonic muscle and
- undifferentiated myoblasts. Both genes are expressed in cardiac muscle
- of the adult rat. The peptides encoded by these 2 genes have distinct
- immunologic properties. The conservation of nucleotide sequence and
- exon/intron boundaries in these 2 genes, as well as their close linkage,
- suggests that they arose by duplication of a common primordial gene.
-
- Myoadenylate deaminase (MADA; EC 3.5.4.6) catalyzes the deamination of
- AMP to IMP in skeletal muscle and plays an important role in the purine
- nucleotide cycle. Deficiency of the muscle-specific myoadenylate
- deaminase is apparently a common cause of exercise-induced myopathy and
- probably the most common cause of metabolic myopathy in the human. It is
- the experience of most large centers that 1 to 2% of all muscle biopsies
- submitted for pathologic examination are deficient in AMP deaminase
- enzyme activity. Fishbein et al. (1978) found deficiency of MADA in 5
- unrelated white males with muscle weakness and/or postexertional
- cramping. Adenosine deaminase and creatine phosphokinase were normal in
- muscle. MADA is 10 times higher in skeletal muscle than in any other
- tissue. Increase in plasma ammonia (relative to lactate) after the
- exercise of sponge-squeezing may be low in this disorder, and this may
- be a useful clinical test. The authors suggested that this may be a
- common form of myopathy of the nonprogressive, 'limp infant' and benign
- congenital hypotonia type. Red cell adenylate deaminase was normal,
- suggesting that it is under different genetic control from that of
- muscle. This accords with evidence that myoadenylate deaminase is
- antigenically unique to muscle and that the isozyme from red cells has
- distinctive kinetic properties. No instances of multiple affected sibs
- have been encountered but since muscle biopsy was relied on by Fishbein
- et al. (1979) for diagnosis this may mean little. Family study using the
- ammonia-lactate ratio in the ischemic forearm exercise test would be of
- interest. Fishbein et al. (1979) had one instance of a mother with an
- intermediate value in the test. Sabina et al. (1980) reported studies of
- a 35-year-old woman which indicated that depletion of the ATP pool of
- muscle and slow repletion are responsible for the symptoms. The chief
- complaint, often dating from childhood, is muscle weakness or cramping
- after exercise. Fatigue after exertion is prolonged. Valen et al. (1987)
- found decreased purine release after exercise in MADA-deficient patients
- compared with that in normal subjects and pointed out that this finding
- increases the specificity of the forearm ischemic exercise test. Using
- the standardized ischemic forearm test, Sinkeler et al. (1988) studied
- 36 relatives of 9 unrelated MAD-deficient patients. Eight new cases of
- myoadenylate deaminase deficiency were detected, 5 of which were
- confirmed histochemically and biochemically. Obligate heterozygotes
- showed a normal ammonia production and MAD staining, but the mean
- activity of the enzyme was significantly less than in controls. Only 2
- of the 8 newly found MAD-deficient persons complained of exertional
- myalgia.
-
- Normally, AMP deaminase is about 95% inhibited by guanosine triphosphate
- (GTP) and may be the limiting step in adenine nucleotide catabolism. Van
- den Berghe and Hers (1980) studied the liver from a man with familial
- primary gout and found defective inhibition of AMP deaminase by GTP. The
- authors had suggested that a genetically determined reduction in
- sensitivity of AMP deaminase to inhibition might be a basis for primary
- gout. Morisaki et al. (1993) presented a study that provided the
- possible molecular explanation for the fact that this AMPD1 mutation so
- rarely causes significant symptoms. Alternative splicing eliminates exon
- 2 in 0.6-2% of AMPD1 mRNA transcripts in adult skeletal muscle.
- Expression studies documented that AMPD1 mRNA, which has exon 2 deleted,
- encodes a functional AMPD peptide. Variations in splicing patterns may
- contribute to the variability in clinical symptoms.
-
- *FIELD* AV
- .0001
- AMPD DEFICIENCY
- AMPD1, GLN12TER, PRO48LEU
- The index case in the family studied by Morisaki et al. (1992) was an
- 18-year-old German female, who first noted calf pain at 4 years of age,
- usually related to exercise. Because of persistence of these symptoms
- and weakness of the upper arms, muscle biopsy was performed,
- demonstrating absence of AMPD activity with normal phosphorylase and
- phosphofructokinase activities. In this patient and 10 other unrelated
- individuals with AMPD deficiency, Morisaki et al. (1992) demonstrated
- homozygosity for a C-to-T transition at nucleotide 34 (codon 12 in exon
- 2) and at nucleotide 143 (codon 48 in exon 3). The C-to-T transition
- resulted in a nonsense mutation predicting a severely truncated AMPD
- peptide (gln12-to-ter). Consistent with this prediction, no
- immunoreactive AMPD1 peptide was detectable in skeletal muscle of these
- patients. The mutation at nucleotide 143 resulted in a change of
- proline-48 to leucine. The mutant allele was found in 12% of Caucasians
- and 19% of African-Americans, whereas none of 106 Japanese subjects
- surveyed had this mutant allele. The frequency of the mutant allele
- would account for the 2% reported incidence of AMPD deficiency in muscle
- biopsies. The restricted distribution and high frequency of this doubly
- mutated allele suggested that it arose in a remote ancestor of
- individuals of western European descent.
-
- *FIELD* SA
- Fishbein (1985); Fishbein et al. (1984); Kar and Pearson (1981);
- Kelemen et al. (1983); Kelemen et al. (1982); Lecky (1983); Sabina
- et al. (1984); Shumate (1983); Shumate et al. (1980)
- *FIELD* RF
- 1. Fishbein, W. N.: Myoadenylate deaminase deficiency: inherited
- and acquired forms. Biochem. Med. 33: 158-169, 1985.
-
- 2. Fishbein, W. N.; Armbrustmacher, V. W.; Griffin, J. L.: Myo-adenylate
- deaminase deficiency: a new disease of muscle. Science 200: 545-548,
- 1978.
-
- 3. Fishbein, W. N.; Armbrustmacher, V. W.; Griffin, J. L.; Davis,
- J. I.; Foster, W. D.: Levels of adenylate deaminase, adenylate kinase,
- and creatine kinase in frozen human muscle biopsy specimens relative
- to type1/type2 fiber distribution: evidence for a carrier state of
- myoadenylate deaminase deficiency. Ann. Neurol. 15: 271-277, 1984.
-
- 4. Fishbein, W. N.; Griffin, J. L.; Nagarajan, K.; Winkert, J. W.;
- Armbrustmacher, V. W.: Immunologic uniqueness of muscle adenylate
- deaminase (mAD) and genetic transmission of the deficiency state.
- (Abstract) Clin. Res. 27: 274A only, 1979.
-
- 5. Kar, N. C.; Pearson, C. M.: Muscle adenylate deaminase deficiency:
- report of six new cases. Arch. Neurol. 38: 279-281, 1981.
-
- 6. Kelemen, J.; Bradley, W. G.; DiMauro, S.: Reply to J. B. Shumate.
- (Letter) Neurology 33: 1534 only, 1983.
-
- 7. Kelemen, J.; Rice, D. R.; Bradley, W. G.; Munsat, T. L.; DiMauro,
- S.; Hogan, E. L.: Familial myoadenylate deaminase deficiency and
- exertional myalgia. Neurology 32: 857-863, 1982.
-
- 8. Lecky, B. R. F.: Failure of D-ribose in myoadenylate deaminase
- deficiency. (Letter) Lancet I: 193 only, 1983.
-
- 9. Morisaki, H.; Morisaki, T.; Newby, L. K.; Holmes, E. W.: Alternative
- splicing: a mechanism for phenotypic rescue of a common inherited
- defect. J. Clin. Invest. 91: 2275-2280, 1993.
-
- 10. Morisaki, T.; Gross, M.; Morisaki, H.; Pongratz, D.; Zollner,
- N.; Holmes, E. W.: Molecular basis of AMP deaminase deficiency in
- skeletal muscle. Proc. Nat. Acad. Sci. 89: 6457-6461, 1992.
-
- 11. Morisaki, T.; Sabina, R. L.; Holmes, E. W.: Adenylate deaminase:
- a multigene family in humans and rats. J. Biol. Chem. 265: 11482-11486,
- 1990.
-
- 12. Morton, C. C.; Eddy, R. L.; Shows, T. B.; Clark, P. R. H.; Sabina,
- R. L.; Holmes, E. W.: Human AMP deaminase-1 gene (AMPD1) is mapped
- to chromosome 1. (Abstract) Cytogenet. Cell Genet. 51: 1048-1049,
- 1989.
-
- 13. Moseley, W. S.; Morisaki, T.; Sabina, R. L.; Holmes, E. W.; Seldin,
- M. F.: Ampd-2 maps to distal mouse chromosome 3 in linkage with Ampd-1.
- Genomics 6: 572-574, 1990.
-
- 14. Sabina, R. L.; Morisaki, T.; Clarke, P.; Eddy, R.; Shows, T. B.;
- Morton, C. C.; Holmes, E. W.: Characterization of the human and rat
- myoadenylate deaminase genes. J. Biol. Chem. 265: 9423-9433, 1990.
-
- 15. Sabina, R. L.; Swain, J. L.; Olanow, C. W.; Bradley, W. G.; Fishbein,
- W. N.; DiMauro, S.; Holmes, E. W.: Myoadenylate deaminase deficiency:
- functional and metabolic abnormalities associated with disruption
- of the purine nucleotide cycle. J. Clin. Invest. 73: 720-730, 1984.
-
- 16. Sabina, R. L.; Swain, J. L.; Patten, B. M.; Ashizawa, T.; O'Brien,
- W. E.; Holmes, E. W.: Disruption of the purine nucleotide cycle:
- a potential explanation for muscle dysfunction in myoadenylate deaminase
- deficiency. J. Clin. Invest. 66: 1419-1423, 1980.
-
- 17. Shumate, J. B.: Myoadenylate deaminase deficiency--a nonfamilial,
- nondisease?. (Letter) Neurology 33: 1533-1534, 1983.
-
- 18. Shumate, J. B.; Kaiser, K. K.; Carroll, J. E.; Brooke, M. H.:
- Adenylate deaminase deficiency in a hypotonic infant. J. Pediat. 96:
- 885-887, 1980.
-
- 19. Sinkeler, S. P. T.; Joosten, E. M. G.; Wevers, R. A.; Oei, T.
- L.; Jacobs, A. E. M.; Veerkamp, J. H.; Hamel, B. C. J.: Myoadenylate
- deaminase deficiency: a clinical, genetic, and biochemical study in
- nine families. Muscle Nerve 11: 312-317, 1988.
-
- 20. Valen, P. A.; Nakayama, D. A.; Veum, J.; Sulaiman, A. R.; Wortmann,
- R. L.: Myoadenylate deaminase deficiency and forearm ischemic exercise
- testing. Arthritis Rheum. 30: 661-668, 1987.
-
- 21. van den Berghe, G.; Hers, H. G.: Abnormal AMP deaminase in primary
- gout. (Letter) Lancet II: 1090 only, 1980.
-
- *FIELD* CS
-
- Muscle:
- Exercise-induced myopathy;
- Postexertional muscle weakness or cramping;
- Prolonged fatigue after exertion
-
- Neuro:
- Limp infant;
- Benign congenital hypotonia
-
- Lab:
- Muscle-specific myoadenylate deaminase deficiency;
- Normal muscle adenosine deaminase and creatine phosphokinase;
- Low increase in plasma ammonia (relative to lactate) after sponge-squeezing
- exercise;
- Decreased purine release after exercise
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- carol: 6/4/1993
- carol: 8/28/1992
- carol: 8/19/1992
- supermim: 3/16/1992
- carol: 11/12/1990
-
- *RECORD*
- *FIELD* NO
- 102771
- *FIELD* TI
- *102771 ADENOSINE MONOPHOSPHATE DEAMINASE-2; AMPD2
- *FIELD* TX
- Southern blotting demonstrated that distinct restriction fragments in
- the rat and human genome hybridized to AMPD1 (102770) and AMPD2 cDNAs.
- Indirect evidence suggests that the 2 genes are linked: L6 myoblasts
- resistant to coformycin coamplified both genes while expressing only
- AMPD2. Moseley et al. (1990) demonstrated further that Ampd-1 and Ampd-2
- are closely linked on distal mouse chromosome 3. Mapping of the human
- AMPD2 gene had not been achieved, but it is presumably located in the
- same region (1p21-p13) as AMPD1. Eddy et al. (1993) indeed demonstrated
- that the AMPD2 gene is localized to 1p by studies of human/mouse somatic
- cell hybrids. They indicated that AMPD1 encodes isoform M (muscle) and
- AMPD2 isoform L (liver). AMPD3 encodes the erythrocytic form (102772).
-
- Bausch-Jurken et al. (1992) isolated cDNA clones for human AMPD2 from
- T-lymphoblast and placental lambda-gt11 libraries using a previously
- cloned rat partial AMPD2 cDNA as the probe. By screening of a human
- spleen cDNA library and by use of PCR techniques, Yamada et al. (1992)
- determined the nucleotide sequence of AMPD2 cDNA. The 3.7-kb cDNA
- contained an open reading frame of 2,301 bp that encodes 767 amino acids
- to form an 89-kD protein.
-
- *FIELD* RF
- 1. Bausch-Jurken, M. T.; Mahnke-Zizelman, D. K.; Morisaki, T.; Sabina,
- R. L.: Molecular cloning of AMP deaminase isoform L. J. Biol. Chem. 267:
- 22407-22413, 1992.
-
- 2. Eddy, R. L.; Mahne-Zizelman, D. K.; Bausch-Jurken, M. T.; Sabina,
- R. L.; Shows, T. B.: Distribution of the AMP deaminase multigene
- family within the human genome: assignment of the AMPD2 to chromosome
- 1p21-p34 and AMPD3 to chromosome 11p13-pter. (Abstract) Human Genome
- Mapping Workshop 93 24, 1993.
-
- 3. Moseley, W. S.; Morisaki, T.; Sabina, R. L.; Holmes, E. W.; Seldin,
- M. F.: Ampd-2 maps to distal mouse chromosome 3 in linkage with Ampd-1.
- Genomics 6: 572-574, 1990.
-
- 4. Yamada, Y.; Goto, H.; Ogasawara, N.: Cloning and nucleotide sequence
- of the cDNA encoding human erythrocyte-specific AMP deaminase. Biochim.
- Biophys. Acta 1171: 125-128, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 3/1/1990
-
- *FIELD* ED
- joanna: 02/05/1996
- mimadm: 3/11/1994
- carol: 12/6/1993
- carol: 1/28/1993
- carol: 1/4/1993
- supermim: 3/16/1992
- carol: 7/6/1990
-
- *RECORD*
- *FIELD* NO
- 102772
- *FIELD* TI
- *102772 ADENOSINE MONOPHOSPHATE DEAMINASE-3; AMPD3
- ERYTHROCYTE AMP DEAMINASE DEFICIENCY, INCLUDED
- *FIELD* TX
- AMP deaminase (EC 3.5.4.6) is a highly regulated purine nucleotide
- catabolic and interconverting enzyme. Multiple isoforms have been
- identified. An inherited defect in AMPD1 results in deficiency of
- isoform M (muscle) and associated exercise-induced myopathy (102770).
- The AMPD2 gene (102771) encodes the L (liver) isoform. The AMPD3 gene
- encodes 2 erythrocytic isoforms, E1 and E2. An inherited defect in AMPD3
- results in combined deficiency of these isoforms. Whereas the AMPD1 and
- AMPD2 genes both are situated in the 1p21-p13 region of chromosome 1,
- Eddy et al. (1993) demonstrated that the AMPD3 gene is located on
- chromosome 11 in the region pter-p13.
-
- Ogasawara et al. (1987) observed 6 related individuals with complete
- deficiency of erythrocyte AMP deaminase. All were healthy and had no
- hematologic disorders. The deficiency was limited to isozyme E, which is
- the red cell type. The deficiency was inherited as an autosomal
- recessive trait as demonstrated by the fact that both parents had
- partial deficiency in each case in which this could be studied and all
- children of completely deficient individuals were partially deficient.
- The frequency of the mutant gene was surprisingly high; heterozygotes
- had a frequency of about 1 in 30 in Japan, Seoul, and Taipei. The ATP
- level was approximately 50% higher in AMP-deficient red cells compared
- to the level in the control cells. Degradation of adenine nucleotide was
- slower in the deficient erythrocytes than in the control erythrocytes.
- Yamada et al. (1994) stated that AMPD3 deficiency had been found in
- Europe and that the frequency in northern Poland was almost the same as
- that in east Asia.
-
- *FIELD* AV
- .0001
- AMP DEAMINASE DEFICIENCY OF ERYTHROCYTE
- AMPD3, ARG573CYS
- Yamada et al. (1994) identified a C-to-T transition in the AMPD3 gene,
- resulting in an amino acid change of arg to cys at codon 573. Two
- individuals with complete deficiency were homozygous and 2 with partial
- deficiency were heterozygous. The missense mutation resulted in a
- catalytically inactive enzyme.
-
- *FIELD* RF
- 1. Eddy, R. L.; Mahne-Zizelman, D. K.; Bausch-Jurken, M. T.; Sabina,
- R. L.; Shows, T. B.: Distribution of the AMP deaminase multigene
- family within the human genome: assignment of the AMPD2 to chromosome
- 1p21-p34 and AMPD3 to chromosome 11p13-pter. (Abstract) Human Genome
- Mapping Workshop 93 24 only, 1993.
-
- 2. Ogasawara, N.; Goto, H.; Yamada, Y.; Nishigaki, I.; Itoh, T.; Hasegawa,
- I.; Park, K. S.: Deficiency of AMP deaminase in erythrocytes. Hum.
- Genet. 75: 15-18, 1987.
-
- 3. Yamada, Y.; Goto, H.; Ogasawara, N.: A point mutation responsible
- for human erythrocyte AMP deaminase deficiency. Hum. Molec. Genet. 3:
- 331-334, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 12/6/1993
-
- *FIELD* ED
- carol: 4/13/1994
- carol: 12/6/1993
-
- *RECORD*
- *FIELD* NO
- 102775
- *FIELD* TI
- *102775 ADENOSINE A1 RECEPTOR; ADORA1; RDC7
- *FIELD* TX
- Diverse physiologic effects of adenosine were recognized as early as the
- 1920s (Drury and Szent-Gyorgyi, 1929; Berne, 1963). Once released,
- adenosine activates adenosine receptors, which in turn regulate a
- diverse set of physiologic functions including cardiac rate and
- contractility, smooth muscle tone, sedation, release of
- neurotransmitters, platelet function, lipolysis, renal function, and
- white blood cell function. Stiles (1992) reviewed the structure and
- function of adenosine receptors important in the mediation of these
- multiple effects. Also see adenosine A2 receptor (ADORA2; 102776).
- Libert et al. (1991) obtained cDNA clones for 4 new receptors of the
- G-protein-coupled receptor family by selective amplification of cloning
- from thyroid cDNA and termed them RDC1, RDC4, RDC7, and RDC8. RDC7 and
- RDC8 were identified as A1 and A2 adenosine receptors, respectively. By
- in situ hybridization, Libert et al. (1991) assigned the RDC7 gene to
- 22q11.2-q13.1.
-
- Using fluorescence in situ hybridization, Townsend-Nicholson et al.
- (1995) demonstrated that, in fact, the ADORA1 gene is located on 1q32.1.
-
- *FIELD* RF
- 1. Berne, R. M.: Cardiac nucleotides in hypoxia: possible role in
- regulation of coronary blood flow. Am. J. Physiol. 204: 317-322,
- 1963.
-
- 2. Drury, A. N.; Szent-Gyorgyi, A.: The physiological activity of
- adenine compounds with especial reference to their action upon the
- mammalian heart. J. Physiol. 68: 213-237, 1929.
-
- 3. Libert, F.; Passage, E.; Parmentier, M.; Simons, M.-J.; Vassart,
- G.; Mattei, M.-G.: Chromosomal mapping of A1 and A2 adenosine receptors,
- VIP receptor, and a new subtype of serotonin receptor. Genomics 11:
- 225-227, 1991.
-
- 4. Stiles, G. L.: Adenosine receptors. J. Biol. Chem. 267: 6451-6454,
- 1992.
-
- 5. Townsend-Nicholson, A.; Baker, E.; Schofield, P. R.; Sutherland,
- G. R.: Localization of the adenosine A1 receptor subtype gene (ADORA1)
- to chromosome 1q32.1. Genomics 26: 423-425, 1995.
-
- *FIELD* CD
- Victor A. McKusick: 9/9/1991
-
- *FIELD* ED
- terry: 4/18/1995
- carol: 6/22/1992
- carol: 6/19/1992
- supermim: 3/16/1992
- carol: 9/9/1991
-
- *RECORD*
- *FIELD* NO
- 102776
- *FIELD* TI
- *102776 ADENOSINE A2 RECEPTOR; ADORA2A
- ADORA2;;
- RDC8
- *FIELD* TX
- See 102775. By in situ hybridization, Libert et al. (1991) assigned the
- RDC8 gene to 11q11-q13. Szepetowski et al. (1993) used
- amplification-based mapping of the 11q13 region to demonstrate that the
- ADORA2 gene is located in that band proximal to BCL1 (151400). It was
- found to be in the coamplification group closest to BCL1 in 11q13 along
- with PPP1A (176875) and GST3 (138370). Physical mapping by hybridization
- of the same probes to DNA fragments generated by rare-cutting
- restriction endonucleases and separated by pulsed field gel
- electrophoresis confirmed the findings. MacCollin et al. (1994)
- suggested that the assignment to chromosome 11 was in error; they
- localized the gene to chromosome 22 both by analysis of cosmid clones
- from a human chromosome 22 library and by Southern hybridization with a
- comprehensive somatic cell hybrid panel. It may be that they were
- dealing with a different gene. Libert et al. (1991) and Szepetowski et
- al. (1993) were clearly mapping the same locus since they used precisely
- the same RDC8 probe. Although the probe used by MacCollin et al. (1994)
- was reportedly very similar in sequence, it must in fact have come from
- a different locus (Gusella, 1994; Gaudray, 1994).
-
- By fluorescence in situ hybridization and PCR analysis of human/hamster
- hybrid cell panels, Le et al. (1996) demonstrated that the ADORA2A gene
- is located on 22q11.2. This was in contrast to previous reports
- (subsequently retracted) which mapped the gene to 11q11-q13; see erratum
- for Libert et al. (1991).
-
- *FIELD* RF
- 1. Gaudray, P.: Personal Communication. Nice, France 6/1/1994.
-
- 2. Gusella, J. F.: Personal Communication. Boston, Mass. 4/17/1994.
-
- 3. Le, F.; Townsend-Nicholson, A.; Baker, E.; Sutherland, G. R.; Schofield,
- P. R.: Characterization and chromosomal localization of the human
- A2a adenosine receptor gene: ADORA2A. Biochem. Biophys. Res. Commun. 223:
- 461-467, 1996.
-
- 4. Libert, F.; Passage, E.; Parmentier, M.; Simons, M.-J.; Vassart,
- G.; Mattei, M.-G.: Chromosomal mapping of A1 and A2 adenosine receptors,
- VIP receptor, and a new subtype of serotonin receptor. Genomics 11:
- 225-227, 1991. Note: Erratum: Genomics 23:305 only, 1994.
-
- 5. MacCollin, M.; Peterfreund, R.; MacDonald, M.; Fink, J. S.; Gusella,
- J.: Mapping of a human A2a adenosine receptor (ADORA2) to chromosome
- 22. Genomics 20: 332-333, 1994.
-
- 6. Szepetowski, P.; Perucca-Lostanlen, D.; Gaudray, P.: Mapping genes
- according to their amplification status in tumor cells: contribution
- to the map of 11q13. Genomics 16: 745-750, 1993.
-
- *FIELD* CD
- Victor A. McKusick: 9/9/1991
-
- *FIELD* ED
- jamie: 12/04/1996
- terry: 11/8/1996
- carol: 9/28/1994
- carol: 6/24/1993
- carol: 3/2/1993
- supermim: 3/16/1992
- carol: 2/27/1992
- carol: 9/9/1991
-
- *RECORD*
- *FIELD* NO
- 102777
- *FIELD* TI
- *102777 ADENOSINE A2B RECEPTOR-LIKE
- ADORA2B-LIKE;;
- ADORA2L
- *FIELD* TX
- The nucleoside adenosine acts through cell surface receptors to
- influence a wide variety of physiologic processes. Based on
- pharmacologic and functional properties, adenosine receptors have been
- divided into 2 main types: A1 adenosine receptors, which inhibit
- adenylyl cyclase, and A2 adenosine receptors which stimulate adenylyl
- cyclase. A2 adenosine receptors are further divided into A2a and A2b
- subtypes based on pharmacologic criteria. Rivkees and Reppert (1992)
- characterized the pharmacologic properties of a cDNA clone for A2b
- adenosine receptor in stably transfected CHO cells by examining cAMP
- responses to drug treatments. Libert et al. (1991), who used the gene
- symbol ADORA2L, mapped the gene to 10q25.3-q26.3 by in situ
- hybridization.
-
- *FIELD* RF
- 1. Libert, F.; Passage, E.; Parmentier, M.; Simons, M.-J.; Vassart,
- G.; Mattei, M.-G.: Chromosomal mapping of A1 and A2 adenosine receptors,
- VIP receptor, and a new subtype of serotonin receptor. Genomics 11:
- 225-227, 1991.
-
- 2. Rivkees, S. A.; Reppert, S. M.: RFL9 encodes an A2b adenosine
- receptor. Molec. Endocr. 6: 1598-1604, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 1/12/1993
-
- *FIELD* ED
- carol: 3/9/1995
- carol: 3/2/1993
- carol: 1/12/1993
-
- *RECORD*
- *FIELD* NO
- 102800
- *FIELD* TI
- *102800 ADENOSINE TRIPHOSPHATASE DEFICIENCY, ANEMIA DUE TO
- *FIELD* TX
- In 2 kindreds Harvald et al. (1964) observed nonspherocytic hemolytic
- anemia due to deficiency of ATP-ase. At least 2 generations were
- affected in each family and father-son transmission was noted. Hanel et
- al. (1971) restudied the families and concluded that the trait is an
- irregular dominant. Probably a minority of the heterozygotes have
- hemolytic anemia.
-
- *FIELD* SA
- Paglia et al. (1970)
- *FIELD* RF
- 1. Hanel, H. K.; Cohn, J.; Harvald, B.: Adenosine-triphosphatase
- deficiency in a family with non-spherocytic haemolytic anaemia. Hum.
- Hered. 21: 313-319, 1971.
-
- 2. Harvald, B.; Hanel, K. H.; Squires, R.; Trap-Jensen, J.: Adenosine-triphosphatase
- deficiency in patients with non-spherocytic haemolytic anaemia. Lancet II:
- 18-19, 1964.
-
- 3. Paglia, D. E.; Valentine, W. N.; Tartaglia, A. P.; Konrad, P. N.
- : Adenine nucleotide reductions associated with a dominantly transmitted
- form of nonspherocytic hemolytic anemia. (Abstract) Blood 36: 837
- only, 1970.
-
- *FIELD* CS
-
- Heme:
- Infrequent nonspherocytic hemolytic anemia
-
- Lab:
- ATP-ase deficiency
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- supermim: 2/28/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 102900
- *FIELD* TI
- 102900 ADENOSINE TRIPHOSPHATE, ELEVATED, OF ERYTHROCYTES
- PYRUVATE KINASE HYPERACTIVITY
- *FIELD* TX
- Brewer (1965) in the United States and Zurcher et al. (1965) in Holland
- described high erythrocyte adenosine triphosphate as a dominantly
- inherited trait. 'High red cell ATP syndrome' may be a heterogeneous
- category. For example, pyrimidine-5-prime-nucleotidase deficiency
- (266120) hemolytic anemia shows this feature. Max-Audit et al. (1980)
- described a family in which 4 persons had polycythemia and pyruvate
- kinase hyperactivity. They showed low 2,3-diphosphoglycerate (2,3-DPG)
- and high adenosine triphosphate (ATP) levels. The PK electrophoretic
- patterns in these persons were abnormal by the presence of several
- additional bands.
-
- *FIELD* SA
- Loos et al. (1967)
- *FIELD* RF
- 1. Brewer, G. J.: A new inherited abnormality of human erythrocyte--elevated
- erythrocyte adenosine triphosphate. Biochem. Biophys. Res. Commun. 18:
- 430-434, 1965.
-
- 2. Loos, J. A.; Prins, H. K.; Zurcher, C.: Elevated ATP levels in
- human erythrocytes. In: Beutler, E.: Hereditary Disorders of Erythrocyte
- Metabolism. New York: Grune and Stratton (pub.) 1967.
-
- 3. Max-Audit, I.; Rosa, R.; Marie, J.: Pyruvate kinase hyperactivity
- genetically determined: metabolic consequences and molecular characterization.
- Blood 56: 902-909, 1980.
-
- 4. Zurcher, C.; Loos, J. A.; Prins, H. K.: Hereditary high ATP content
- of human erythrocytes. Folia Haemat. 83: 366-376, 1965.
-
- *FIELD* CS
-
- Heme:
- Polycythemia
-
- Lab:
- High erythrocyte adenosine triphosphate;
- Pyruvate kinase hyperactivity;
- Low 2,3-diphosphoglycerate (2,3-DPG);
- Additional PK electrophoretic bands
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 102910
- *FIELD* TI
- *102910 ADENOSINE TRIPHOSPHATE SYNTHASE, MITOCHONDRIAL, BETA SUBUNIT; ATP5B;
- ATPSB; ATPMB
- *FIELD* TX
- The beta subunit of mitochondrial ATP synthase is encoded by a nuclear
- gene and assembled with the other subunits encoded by both mitochondrial
- and nuclear genes. The enzyme catalyzes ATP formation, using the energy
- of proton flux through the inner membrane during oxidative
- phosphorylation. Two subunits are encoded by a mitochondrial gene and
- the others by a nuclear gene. The numbers of mitochondria per cell vary
- greatly depending on the developmental stage, cell activity, and type of
- tissue. The molecular mechanism for coordinating the 2 genetic systems
- is unknown. Ohta et al. (1988) cloned cDNA of the human beta subunit.
- The gene contains 10 exons, with the first exon corresponding to the
- noncoding region and most of the presequence which targets this protein
- to the mitochondria. Neckelmann et al. (1989) sequenced the human ATP
- synthase beta subunit gene and demonstrated that it is preferentially
- expressed in heart and skeletal muscle. The gene was found to have 10
- exons encoding a leader peptide of 49 amino acids and a mature protein
- of 480 amino acids. Kudoh et al. (1989) assigned the ATPMB locus to the
- p13-qter region of human chromosome 12 by analysis of human-mouse
- somatic cell hybrid DNA and by use of flow-sorted chromosomes. They
- assigned 2 related sequences, ATPMBL1 and ATPMBL2, to chromosome 2 and
- 17, respectively.
-
- *FIELD* SA
- Neckelmann et al. (1989)
- *FIELD* RF
- 1. Kudoh, J.; Minoshima, S.; Fukuyama, R.; Maekawa, M.; Neckelmann,
- N.; Wallace, D. C.; Shimizu, Y.; Shimizu, N.: Assignment of ATP synthase
- beta subunit (ATPMB) gene to the p13-qter region of human chromosome
- 12. (Abstract) Cytogenet. Cell Genet. 51: 1026 only, 1989.
-
- 2. Neckelmann, N.; Warner, C. K.; Chung, A.; Kudoh, J.; Minoshima,
- S.; Fukuyama, R.; Maekawa, M.; Shimizu, Y.; Shimizu, N.; Liu, J. D.;
- Wallace, D. C.: The human ATP synthase beta subunit gene: sequence
- analysis, chromosome assignment, and differential expression. Genomics 5:
- 829-843, 1989.
-
- 3. Neckelmann, N. S.; Chung, A. B.; Warner, C. K.; Hodge, J. A.; Wallace,
- D. C.: The human ATP synthase beta subunit gene has been sequenced
- and shown to be preferentially expressed in heart and skeletal muscle.
- (Abstract) Cytogenet. Cell Genet. 51: 1051 only, 1989.
-
- 4. Ohta, S.; Tomura, H.; Matsuda, K.; Kagawa, Y.: Gene structure
- of the human mitochondrial adenosine triphosphate synthase beta subunit.
- J. Biol. Chem. 263: 11257-11262, 1988.
-
- *FIELD* CD
- Victor A. McKusick: 10/10/1988
-
- *FIELD* ED
- jason: 7/29/1994
- supermim: 3/16/1992
- carol: 2/7/1991
- supermim: 3/20/1990
- carol: 12/14/1989
- ddp: 10/27/1989
-
- *RECORD*
- *FIELD* NO
- 102920
- *FIELD* TI
- *102920 ADENOVIRUS-12 CHROMOSOME MODIFICATION SITE-1p; A12M2
- *FIELD* TX
- Steffensen et al. (1976) found a second adenovirus 12 gap in chromosome
- 1, at 1p36. It has been considered that this site may correspond to that
- of adenylate kinase-2 (103020); however, AK2 appears to be at 1p34.
- McDougall (1979) identified 2 sites on 1p: 1p32 and 1p36.
-
- *FIELD* RF
- 1. McDougall, J. K.: The interactions of adenovirus with host cell
- gene loci. (Abstract) Cytogenet. Cell Genet. 25: 183 only, 1979.
-
- 2. Steffensen, D. M.; Szabo, P.; McDougall, J. K.: Adenovirus 12
- uncoiler regions of human chromosome 1 in relation to the 5S rRNA
- genes. Exp. Cell Res. 100: 436-439, 1976.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
-
- *RECORD*
- *FIELD* NO
- 102930
- *FIELD* TI
- *102930 ADENOVIRUS-12 CHROMOSOME MODIFICATION SITE-1q1; A12M1
- *FIELD* TX
- A site on the long arm of chromosome 1 is altered by exposure of cells
- in vitro to adenovirus 12 (HGM2, Rotterdam, July, 1974). See McDougall
- (1971). Steffensen et al. (1976) concluded that this uncoiler region is
- at 1q42 and that 5S rRNA genes are located immediately distal to it at
- 1q42-1q43. This order is the reverse of that presented tentatively at
- the Rotterdam Gene Mapping Conference. This site may be identical to
- that of guanylate kinase (139270). McDougall (1979) identified 2 sites
- on 1q: 1q21 and 1q42.
-
- *FIELD* RF
- 1. McDougall, J. K.: Adenovirus induced chromosome aberrations in
- human cells. J. Gen. Virol. 12: 43-51, 1971.
-
- 2. McDougall, J. K.: The interactions of adenovirus with host cell
- gene loci. (Abstract) Cytogenet. Cell Genet. 25: 183 only, 1979.
-
- 3. Steffensen, D. M.; Szabo, P.; McDougall, J. K.: Adenovirus 12
- uncoiler regions of human chromosome 1 in relation to the 5S rRNA
- genes. Exp. Cell Res. 100: 436-439, 1976.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- davew: 7/20/1994
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 4/28/1988
-
- *RECORD*
- *FIELD* NO
- 102940
- *FIELD* TI
- *102940 ADENOVIRUS-12 CHROMOSOME MODIFICATION SITE-1q2; A12M3
- *FIELD* TX
- This is the site at 1q21. See 102930.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
- marie: 1/7/1987
- *RECORD*
- *FIELD* NO
- 102970
- *FIELD* TI
- *102970 ADENOVIRUS-12 CHROMOSOME MODIFICATION SITE-17; A12M4
- *FIELD* TX
- Adenovirus 12 produces an uncoiled segment in the long arm of chromosome
- 17. This is associated with elevated thymidine kinase (TK) activity. The
- TK locus (188300) is in the same region of 17q as that which shows the
- morphologic change. Lindgren et al. (1985) pointed out that the 3 major
- adenovirus-12 modification sites are the location of small nuclear RNA
- genes: U1 genes (180680) are at 1p36, class 1 U1 pseudogenes are at
- 1q21, and U2 snRNA genes (180690) are at 17q21-17q22. On this basis,
- they suggested that snRNA genes are the major targets of viral
- chromosome modification.
-
- *FIELD* SA
- McDougall (1971); McDougall et al. (1973)
- *FIELD* RF
- 1. Lindgren, V.; Ares, M., Jr.; Weiner, A. M.; Francke, U.: Human
- genes for U2 small nuclear RNA map to a major adenovirus 12 modification
- site on chromosome 17. Nature 314: 115-116, 1985.
-
- 2. McDougall, J. K.: Adenovirus induced chromosome aberrations in
- human cells. J. Gen. Virol. 12: 43-51, 1971.
-
- 3. McDougall, J. K.; Kucherlapati, R. S.; Ruddle, F. H.: Localization
- and induction of the human thymidine kinase gene by adenovirus 12.
- Nature N.B. 245: 172-175, 1973.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 6/1/1988
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 102980
- *FIELD* TI
- *102980 ADENYLATE CYCLASE ACTIVATING POLYPEPTIDE 1
- ADCYAP1;;
- PITUITARY ADENYLATE CYCLASE ACTIVATING POLYPEPTIDE; PACAP
- *FIELD* TX
- Pituitary adenylate cyclase activating polypeptide (PACAP) is a novel
- bioactive peptide that was originally isolated from ovine hypothalamus
- on the basis of its ability to stimulate adenylate cyclase in rat
- anterior pituitary cell cultures. The amino-terminal amino acid sequence
- of PACAP showed 68% identity with vasoactive intestinal peptide (VIP;
- 192320) and more limited similarity with growth hormone releasing
- hormone (GHRH; 139190). Hosoya et al. (1992) isolated the human PACAP
- gene and by comparison with a human PACAP cDNA determined its
- exon/intron organization. On the basis of DNA isolated from a mouse A9
- microcell hybrid clone containing a single human chromosome, the PACAP
- gene was assigned to chromosome 18; it was regionalized to 18p11 by in
- situ hybridization. Perez-Jurado and Francke (1993) described a
- dinucleotide repeat polymorphism in the 3-prime untranslated region of
- the PACAP gene.
-
- *FIELD* RF
- 1. Hosoya, M.; Kimura, C.; Ogi, K.; Ohkubo, S.; Miyamoto, Y.; Kugoh,
- H.; Shimizu, M.; Onda, H.; Oshimura, M.; Arimura, A.; Fujino, M.:
- Structure of the human pituitary adenylate cyclase activating polypeptide
- (PACAP) gene. Biochim. Biophys. Acta 1129: 199-206, 1992.
-
- 2. Perez-Jurado, L. A.; Francke, U.: Dinucleotide repeat polymorphism
- at the human pituitary adenylate cyclase activating polypeptide (PACAP)
- gene. Hum. Molec. Genet. 2: 827 only, 1993.
-
- *FIELD* CD
- Victor A. McKusick: 7/8/1993
-
- *FIELD* ED
- carol: 8/31/1993
- carol: 7/8/1993
-
- *RECORD*
- *FIELD* NO
- 102981
- *FIELD* TI
- *102981 ADENYLATE CYCLASE ACTIVATING POLYPEPTIDE 1, RECEPTOR FOR; ADCYAP1R1
- PITUITARY ADENYLATE CYCLASE ACTIVATING POLYPEPTIDE RECEPTOR, TYPE;;
- I;;
- PACAP RECEPTOR, TYPE I
- *FIELD* TX
- Pituitary adenylate cyclase activating polypeptide (PACAP; 102980) is a
- hormone that was originally isolated from sheep hypothalamus on the
- basis of its ability to stimulate adenylate cyclase in rat anterior
- pituitary cell cultures (Arimura, 1992). PACAP is present not only in
- the central nervous system but also in peripheral tissues, including
- gastrointestinal tract, adrenal gland, and testis. Its actions include
- the stimulation of secretion of growth hormone, ACTH, catecholamines,
- and insulin, as well as other hormones. In addition, it appears to
- function as a neuromodulator/neurotransmitter in the central and
- peripheral nervous systems. The diverse biologic actions of PACAP are
- mediated by receptors that are positively coupled to adenylate cyclase
- by G(s-alpha). Three different receptors for PACAP have been identified,
- each of which contains 7 transmembrane segments and shares significant
- homology with members of the glucagon/secretin receptor family. The type
- 1 receptor, which is found in the hypothalamus, brain stem, pituitary,
- adrenal gland, pancreas, and testes, has a high affinity only for PACAP
- (Ogi et al., 1993). The type 2 receptor is found in the brain. The
- adrenal gland has a high affinity for both PACAP and for vasoactive
- intestinal peptide (VIP; 192320).
-
- By PCR analysis of genomic DNA from a human/rodent somatic cell hybrid
- mapping panel, Stoffel et al. (1994) mapped the human type 1 PACAP
- receptor gene, symbolized ADCYAP1R1, to chromosome 7. The assignment was
- confirmed and the gene localized to 7p14 by fluorescence in situ
- hybridization. Brabet et al. (1996) likewise mapped this gene to
- 7p15-p14 by fluorescence in situ hybridization.
-
- *FIELD* RF
- 1. Arimura, A.: Pituitary adenylate cyclase activating polypeptide
- (PACAP): discovery and current status of research. Regul. Pept. 37:
- 287-303, 1992.
-
- 2. Brabet, P.; Diriong, S.; Journot, L.; Bockaert, J.; Taviaux, S.
- : Localization of the human pituitary adenylate cyclase-activating
- polypeptide receptor (PACAP-1-R) gene to 7p15-p14 by fluorescence
- in situ hybridization. Genomics 38: 100-102, 1996.
-
- 3. Ogi, K.; Miyamoto, Y.; Masuda, Y.; Habata, Y.; Hosoya, M.; Ohtaki,
- T.; Masuo, Y.; Onda, H.; Fujino, M.: Molecular cloning and functional
- expression of a cDNA encoding a human pituitary adenylate cyclase
- activating polypeptide receptor. Biochem. Biophys. Res. Commun. 196:
- 1511-1521, 1993.
-
- 4. Stoffel, M.; Espinosa, R., III; Trabb, J. B.; Le Beau, M. M.; Bell,
- G. I.: Human type I pituitary adenylate cyclase activating polypeptide
- receptor (ADCYAP1R): localization to chromosome band 7p14 and integration
- into the cytogenetic, physical, and genetic map of chromosome 7. Genomics 23:
- 697-699, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 4/20/1995
-
- *FIELD* ED
- terry: 12/11/1996
- carol: 4/20/1995
-
- *RECORD*
- *FIELD* NO
- 102990
- *FIELD* TI
- 102990 ADENYLATE KINASE, MUSCLE, DEFICIENCY OF
- *FIELD* TX
- Schmitt et al. (1974) studied biopsied skeletal muscle from the father,
- mother, brother and sister of 2 children (sex not given) who had died of
- malignant hyperpyrexia (muscle rigidity, hyperthermia, tachycardia,
- hyperventilation, myoglobinuria and renal failure) after halothane
- anesthesia (see 145600). Deficiency of muscle adenylate kinase (AK) was
- found in the mother and sister. Adenylate kinase, also known as
- myokinase, is a phosphotransferase that catalyzes the reversible
- conversion of 2 molecules of ADP to 1 of ATP plus 1 of AMP. Because red
- cell adenylate was normal, the authors concluded that muscle and red
- cell (103000) AK are under separate genetic control.
-
- *FIELD* RF
- 1. Schmitt, J.; Schmidt, K.; Ritter, H.: Hereditary malignant hyperpyrexia
- associated with muscle adenylate kinase deficiency. Humangenetik 24:
- 253-357, 1974.
-
- *FIELD* CS
-
- Misc:
- Malignant hyperpyrexia after halothane anesthesia
-
- Muscle:
- Muscle rigidity
-
- Metabolic:
- Hyperthermia
-
- Cardiac:
- Tachycardia
-
- Resp:
- Hyperventilation
-
- GU:
- Renal failure
-
- Lab:
- Myoglobinuria;
- Muscle adenylate kinase (AK or myokinase) deficiency;
- Normal red cell adenylate kinase
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 103000
- *FIELD* TI
- *103000 ADENYLATE KINASE-1; AK1
- ADENYLATE KINASE, SOLUBLE
- ADENYLATE KINASE DEFICIENCY, INCLUDED
- *FIELD* TX
- Adenylate kinase is present in red cells as well as in muscle (see
- 102990). Fildes and Harris (1966) found electrophoretic variation in red
- cells and defined 3 phenotypes, designated AK1, AK2-1 and AK2. All of
- the 141 children of two AK1 parents (62 such matings) were also AK1.
- Among the 136 children of AK1 by AK2-1 matings, 72 were AK1 and 64
- AK2-1. AK1 and AK2 persons were thought to be homozygotes for a
- two-allele system and AK2-1 persons heterozygotes. The frequency of the
- rarer AK2 allele was about 0.05 in the English and about 1 in 400
- persons would be expected to be homozygous for this allele. Survey and
- family data were consistent. Singer and Brock (1971) identified a
- probably silent allele at the AK locus. Matsuura et al. (1989) cloned
- the AK1 gene and determined its structure. The gene is 12 kb long and
- has 7 exons.
-
- Rapley et al. (1967) concluded that the AK locus is linked to the ABO
- (110300) locus with a recombination value of about 0.20. Schleutermann
- et al. (1969) found that the nail-patella syndrome locus (161200) and
- the AK locus are closely linked. No recombination was found in 53
- opportunities. Fenger and Sorensen (1975) found a 1.33 to 1 ratio for
- the female to male recombination fractions between ABO and AK, but the
- difference between the recombination fractions was not significantly
- different from zero. All published data combined showed the most likely
- recombination fraction to be about 14%. Westerveld et al. (1976) found
- evidence that the AK locus assigned to chromosome 9 is the AK1 locus, or
- so-called red cell AK. Cook et al. (1978) collated evidence that ABO-AK1
- lie in band 9q34. They could exclude MNSs, GPT and Gc from chromosome 9.
- Cavalli-Sforza et al. (1979) presented evidence for linkage of
- transcobalamin II and adenylate kinase (lod score 1.78 at theta 0.139).
- This was not subsequently confirmed. AK1 is proximal to the break in the
- Philadelphia chromosome rearrangement (Geurts van Kessel et al., 1982).
- On the basis of a chromosome 9 aberration, an inverted paracentric
- insertion, inv ins(9)(q22.1q34.3q34.1), Allderdice et al. (1986)
- concluded that AK1 is located in 9q34.1-q34.3. Since AK1 is in 9q34 and
- is proximal to the breakpoint that creates the Philadelphia chromosome
- in chronic myeloid leukemia, located in band 9q34.1, AK1 and probably
- the linked ABO locus may be in the proximal part of 9q34.1. In a patient
- with deletion 9q32-qter secondary to a balanced maternal translocation,
- Zuffardi et al. (1989) found normal levels of adenylate kinase.
- Comparing this to previously published data, the authors concluded that
- the AK1 locus may be situated in 9q32.
-
- In 2 offspring of second-cousin Arab parents, Szeinberg et al. (1969)
- found marked AK deficiency with intermediate levels in the presumed
- heterozygotes. Severe anemia was present in both. Presumably this
- mutation is at the same locus as that which controls the polymorphism of
- AK. In the study of a black family, Beutler et al. (1982) found that
- despite barely detectable levels of adenylate kinase activity, probably
- representing guanylate kinase, red cells are able to maintain their
- adenine nucleotide levels and to circulate normally. They concluded that
- previously reported cases of AK deficiency represent a chance
- association of hemolysis with the enzyme deficiency, and not a
- cause-and-effect relationship. In the family reported by Boivin et al.
- (1971), the proband had psychomotor retardation and moderate congenital
- hemolytic anemia with markedly diminished red cell AK activity. The
- parents had half-normal AK activity. Autosomal recessive inheritance was
- proposed. Another family, Japanese, was reported by Miwa et al. (1983).
- The proband, a 10-year-old girl, had normal physical and mental
- development, mild to moderate hemolytic anemia from the neonatal period,
- and hepatosplenomegaly. Red cell AK activity was 44% of normal.
- Puzzlingly, the proband's mother, younger sister and maternal
- grandfather showed a half-normal enzyme activity. Lachant et al. (1991)
- reported a fifth family with AK deficiency associated with hemolytic
- anemia. In none of the families had a cause-and-effect relationship to
- AK deficiency been established. Lachant et al. (1991) suggested that
- defects occur in multiple phosphotransferases in AK-deficient red blood
- cells and that these other defects produce deleterious lesions that
- promote the shortened red cell survival. Toren et al. (1994) described a
- family in which 6 children showed AK deficiency; in 3 of them, G6PD
- deficiency was found in combination with AK deficiency. Although
- heterozygotes were asymptomatic, homozygotes had congenital chronic
- nonspherocytic hemolytic anemia with hemoglobin levels of 8-9 g/dl.
- Patients also deficient in G6PD suffered from a more severe hemolytic
- anemia with hemoglobin levels around 6 g/dl. The AK-deficient children
- were also mentally retarded. Splenectomy performed in 5 of the 6
- children resulted in complete remission of the hemolytic process.
-
- Data on gene frequencies of allelic variants were tabulated by
- Roychoudhury and Nei (1988).
-
- *FIELD* AV
- .0001
- ADENYLATE KINASE DEFICIENCY, HEMOLYTIC ANEMIA DUE TO
- AK1, ARG128TRP
- In a patient with hemolytic anemia, Matsuura et al. (1989) demonstrated
- a transition (C-to-T) in exon 6 which resulted in an arg-to-trp
- (CGG-to-TGG) substitution at the 128th residue of AK1. Mutant chicken
- AK1, produced by introducing an arg-to-trp substitution at the same
- position by oligodeoxynucleotide-directed mutagenesis, showed reduced
- catalytic activity as well as decreased solubility when expressed in E.
- coli.
-
- *FIELD* SA
- Boivin et al. (1970); Bowman et al. (1967); Brock (1970); Ferguson-Smith
- et al. (1976); Mohandas et al. (1979); Povey et al. (1976); Seger
- et al. (1978); Szeinberg et al. (1969); Weitkamp et al. (1969)
- *FIELD* RF
- 1. Allderdice, P. W.; Kaita, H.; Lewis, M.; McAlpine, P. J.; Wong,
- P.; Anderson, J.; Giblett, E. R.: Segregation of marker loci in families
- with an inherited paracentric insertion of chromosome 9. Am. J.
- Hum. Genet. 39: 612-617, 1986.
-
- 2. Beutler, E.; Carson, D. A.; Dannawi, H.; Forman, L.; Kuhl, W.;
- West, C.; Westwood, B.: Red cell adenylate kinase deficiency: another
- non-disease?. (Abstract) Blood 60: 33A only, 1982.
-
- 3. Boivin, P.; Galand, C.; Hakim, J.; Simony, D.; Seligman, M.: Deficit
- congenital en adenylate-kinase erythrocytaire. (Letter) Presse Med. 78:
- 1443 only, 1970.
-
- 4. Boivin, P.; Galand, C.; Hakim, J.; Simony, D.; Seligman, M.: Une
- nouvelle erythroenzymopathie: anemie hemolytique congenitale non spherocytaire
- et deficit hereditaire en adenylate-kinase erythrocytaire. Presse
- Med. 79: 215-218, 1971.
-
- 5. Bowman, J. E.; Frischer, H.; Ajmar, F.; Carson, P. E.; Gower, M.
- K.: Population, family and biochemical investigation of human adenylate
- kinase polymorphism. Nature 214: 1156-1158, 1967.
-
- 6. Brock, D. J. H.: Evidence against a common subunit in adenylate
- kinase and pyruvate kinase. Humangenetik 10: 30-34, 1970.
-
- 7. Cavalli-Sforza, L. L.; King, M. C.; Go, R. C. P.; Namboodiri, K.
- K.; Lynch, H. T.; Wong, L.; Kaplan, E. B.; Elston, R. C.: Possible
- linkage between transcobalamin II (TC II) and adenylate kinase (AK).
- (Abstract) Cytogenet. Cell Genet. 25: 140-141, 1979.
-
- 8. Cook, P. J. L.; Robson, E. B.; Buckton, K. E.; Slaughter, C. A.;
- Gray, J. E.; Blank, C. E.; James, F. E.; Ridler, M. A. C.; Insley,
- J.; Hulten, M.: Segregation of ABO, AK(1) and ACONs in families with
- abnormalities of chromosome 9. Ann. Hum. Genet. 41: 365-377, 1978.
-
- 9. Fenger, K.; Sorensen, S. A.: Evaluation of a possible sex difference
- in recombination for the ABO-AK linkage. Am. J. Hum. Genet. 27:
- 784-788, 1975.
-
- 10. Ferguson-Smith, M. A.; Aitken, D. A.; Turleau, C.; de Grouchy,
- J.: Localisation of the human ABO: Np-1: AK-1 linkage group by regional
- assignment of AK-1 to 9q34. Hum. Genet. 34: 35-43, 1976.
-
- 11. Fildes, R. A.; Harris, H.: Genetically determined variation of
- adenylate kinase in man. Nature 209: 261-262, 1966.
-
- 12. Geurts van Kessel, A. H. M.; Hagemeijer, A.; Westerveld, A.; Meera
- Khan, P.; de Groot, P. G.; Pearson, P. L.: Characterization of chromosomal
- abnormalities in chronic myeloid leukemia using somatic cell hybrids.
- (Abstract) Cytogenet. Cell Genet. 32: 280 only, 1982.
-
- 13. Lachant, N. A.; Zerez, C. R.; Barredo, J.; Lee, D. W.; Savely,
- S. M.; Tanaka, K. R.: Hereditary erythrocyte adenylate kinase deficiency:
- a defect of multiple phosphotransferases?. Blood 77: 2774-2784,
- 1991.
-
- 14. Matsuura, S.; Igarashi, M.; Tanizawa, Y.; Yamada, M.; Kishi, F.;
- Kajii, T.; Fujii, H.; Miwa, S.; Sakurai, M.; Nakazawa, A.: Human
- adenylate kinase deficiency associated with hemolytic anemia: a single
- base substitution affecting solubility and catalytic activity of the
- cytosolic adenylate kinase. J. Biol. Chem. 264: 10148-10155, 1989.
-
- 15. Miwa, S.; Fujii, H.; Tani, K.; Takahashi, K.; Takizawa, T.; Igarashi,
- T.: Red cell adenylate kinase deficiency associated with hereditary
- nonspherocytic hemolytic anemia: clinical and biochemical studies.
- Am. J. Hemat. 14: 325-333, 1983.
-
- 16. Mohandas, T.; Sparkes, R. S.; Sparkes, M. C.; Shulkin, J. D.;
- Toomey, K. E.; Funderburk, S. J.: Regional localization of human
- gene loci on chromosome 9: studies of somatic cell hybrids containing
- human translocations. Am. J. Hum. Genet. 31: 586-600, 1979.
-
- 17. Povey, S.; Slaughter, C. A.; Wilson, D. E.; Gormley, I. P.; Buckton,
- K. E.; Perry, P.; Bobrow, M.: Evidence for the assignment of loci
- AK 1, AK 3 and ACON to chromosome 9 in man. Ann. Hum. Genet. 39:
- 413-422, 1976.
-
- 18. Rapley, S.; Robson, E. B.; Harris, H.; Smith, S. M.: Data on
- the incidence, segregation and linkage relations of the adenylate
- kinase (AK) polymorphism. Ann. Hum. Genet. 31: 237-242, 1967.
-
- 19. Roychoudhury, A. K.; Nei, M.: Human Polymorphic Genes: World
- Distribution. New York: Oxford Univ. Press (pub.) 1988.
-
- 20. Schleutermann, D. A.; Bias, W. B.; Murdoch, J. L.; McKusick, V.
- A.: Linkage of the loci for the nail-patella syndrome and adenylate
- kinase. Am. J. Hum. Genet. 21: 606-630, 1969.
-
- 21. Seger, J.; Tchen, P.; Feingold, N.; Grenand, F.; Bois, E.: Homozygosity
- of adenylate kinase allele 3: two cases. Hum. Genet. 43: 337-339,
- 1978.
-
- 22. Singer, J. D.; Brock, D. J.: Half-normal adenylate kinase activity
- in three generations. Ann. Hum. Genet. 35: 109-114, 1971.
-
- 23. Szeinberg, A.; Gavendo, S.; Cahane, D.: Erythrocyte adenylate-kinase
- deficiency. (Letter) Lancet I: 315-316, 1969.
-
- 24. Szeinberg, A.; Kahana, D.; Gavendo, S.; Zaidman, J.; Ben-Ezzer,
- J.: Hereditary deficiency of adenylate kinase in red blood cells.
- Acta Haemat. 42: 111-126, 1969.
-
- 25. Toren, A.; Brok-Simoni, F.; Ben-Bassat, I.; Holtzman, F.; Mandel,
- M.; Neumann, Y.; Ramot, B.; Rechavi, G.; Kende, G.: Congenital haemolytic
- anaemia associated with adenylate kinase deficiency. Brit. J. Haemat. 87:
- 376-380, 1994.
-
- 26. Weitkamp, L. R.; Sing, C. F.; Shreffler, D. C.; Guttormsen, S.
- A.: The genetic linkage relations of adenylate kinase: further data
- on the ABO-AK linkage group. Am. J. Hum. Genet. 21: 600-605, 1969.
-
- 27. Westerveld, A.; Jongsma, A. P. M.; Meera Khan, P.; Van Someren,
- H.; Bootsma, D.: Assignment of the AK(1): Np: AKO linkage group to
- human chromosome 9. Proc. Nat. Acad. Sci. 73: 895-899, 1976.
-
- 28. Zuffardi, O.; Caiulo, A.; Maraschio, P.; Tupler, R.; Bianchi,
- E.; Amisano, P.; Beluffi, G.; Moratti, R.; Liguri, G.: Regional assignment
- of the loci for adenylate kinase to 9q32 and for alpha(1)-acid glycoprotein
- to 9q31-q32: a locus for Goltz syndrome in region 9q32-qter?. Hum.
- Genet. 82: 17-19, 1989.
-
- *FIELD* CS
-
- Heme:
- Hemolytic anemia
-
- Lab:
- Red cell adenylate kinase deficiency
-
- Inheritance:
- Autosomal dominant;
- anemia recessive
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 8/30/1994
- mimadm: 3/11/1994
- carol: 5/12/1993
- supermim: 3/16/1992
- carol: 1/27/1992
- carol: 10/3/1991
-
- *RECORD*
- *FIELD* NO
- 103020
- *FIELD* TI
- *103020 ADENYLATE KINASE-2; AK2
- ADENYLATE KINASE, MITOCHONDRIAL
- *FIELD* TX
- The existence of a second adenylate kinase (EC 2.7.4.3) locus linked to
- PGM1 and peptidase C, i.e., on chromosome 1, was suggested by cell
- hybridization studies by Van Cong et al. (1972). The Goss-Harris method
- of mapping combines features of recombinational study in families and
- synteny tests in hybrid cells. As applied to chromosome 1, the method
- shows that AK2 and UMPK are distal to PGM1 and that the order of the
- loci is PGM1: UMPK: (AK2, alpha-FUC): ENO1 (Goss and Harris, 1977).
- Carritt et al. (1982) presented evidence that AK2 is in 1p34.
-
- *FIELD* SA
- Bruns and Regina (1977)
- *FIELD* RF
- 1. Bruns, G. A. P.; Regina, V. M.: Adenylate kinase-2, a mitochondrial
- enzyme. Biochem. Genet. 15: 477-486, 1977.
-
- 2. Carritt, B.; King, J.; Welch, H. M.: Gene order and localization
- of enzyme loci on the short arm of chromosome 1. Ann. Hum. Genet. 46:
- 329-335, 1982.
-
- 3. Goss, S. J.; Harris, H.: Gene transfer by means of cell fusion.
- II. The mapping of 8 loci on human chromosome 1 by statistical analysis
- of gene assortment in somatic cell hybrids. J. Cell Sci. 25: 39-57,
- 1977.
-
- 4. Van Cong, N.; Billardon, C.; Rebourcet, R.; Kaouel, C. L.-B.; Picard,
- J. Y.; Weil, D.; Frezal, J.: The existence of a second adenylate
- kinase locus linked to PGM-1 and peptidase-C. Ann. Genet. 15: 213-218,
- 1972.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
- marie: 1/7/1987
-
- *RECORD*
- *FIELD* NO
- 103030
- *FIELD* TI
- *103030 ADENYLATE KINASE-3; AK3
- ADENYLATE KINASE, MITOCHONDRIAL
- *FIELD* TX
- The adenylate kinases are a family of structurally and functionally
- related enzymes that catalyze a similar reaction, MgNTP + AMP = MgNDP +
- ADP (N = A or G). The AK enzymes are important for maintenance of
- homeostasis of the adenine and guanine nucleotide pools. AK1 (103000) is
- a cytosolic enzyme for which ATP is the substrate. AK2 (103020)
- catalyzes the same reaction as AK1, but it is localized in the
- mitochondrial intermembrane space. AK3 is present in the mitochondrial
- matrix and prefers GTP over ATP as the substrate. Wilson et al. (1976)
- pointed out that AK3 is nucleosidetriphosphate-adenylate kinase. In the
- course of their efforts to identify the gene causing neurofibromatosis
- (NF1; 162200), Viskochil et al. (1990) found a gene first designated
- HB15, which Xu et al. (1992) subsequently concluded is probably a
- processed pseudogene of AK3. It is intronless and contains a
- polyadenylate tract, but retains coding potential because the open
- reading frame was not impaired by any observed base substitutions. One
- presumed processed pseudogene of AK3 is located within an intron of the
- NF1 gene. Xu et al. (1992) also characterized cDNA clones for the
- authentic AK3.
-
- By study of somatic cell hybrids, Povey et al. (1976) assigned AK3 to
- chromosome 9. The SRO (smallest region of overlap) for AK3 was estimated
- to be 9p24-p13 (Robson and Meera Khan, 1982).
-
- By interspecific backcross linkage analysis, Pilz et al. (1995) mapped
- the Ak3 gene to mouse chromosome 4.
-
- *FIELD* SA
- Cook et al. (1976); Mohandas et al. (1979); Steinbach and Benz (1983)
- *FIELD* RF
- 1. Cook, P. J. L.; Buckton, K. E.; Spowart, G.: Family studies on
- chromosome 9. Cytogenet. Cell Genet. 16: 284-288, 1976.
-
- 2. Mohandas, T.; Sparkes, R. S.; Sparkes, M. C.; Shulkin, J. D.; Toomey,
- K. E.; Funderburk, S. J.: Regional localization of human gene loci
- on chromosome 9: studies of somatic cell hybrids containing human
- translocation. Am. J. Hum. Genet. 31: 586-600, 1979.
-
- 3. Pilz, A.; Woodward, K.; Povey, S.; Abbott, C.: Comparative mapping
- of 50 human chromosome 9 loci in the laboratory mouse. Genomics 25:
- 139-149, 1995.
-
- 4. Povey, S.; Slaughter, C. A.; Wilson, D. E.; Gormley, I. P.; Buckton,
- K. E.; Perry, P.; Bobrow, M.: Evidence for the assignment of the
- loci AK 1, AK 3 and ACON to chromosome 9 in man. Ann. Hum. Genet. 39:
- 413-422, 1976.
-
- 5. Robson, E. B.; Meera Khan, P.: Report of the committee on the
- genetic constitution of chromosomes 7, 8, and 9. Cytogenet. Cell
- Genet. 32: 144-152, 1982.
-
- 6. Steinbach, P.; Benz, R.: Demonstration of gene dosage effects
- for AK3 and GALT in fibroblasts from a fetus with 9p trisomy. Hum.
- Genet. 63: 290-291, 1983.
-
- 7. Viskochil, D.; Buchberg, A. M.; Xu, G.; Cawthon, R. M.; Stevens,
- J.; Wolff, R. K.; Culver, M.; Carey, J. C.; Copeland, N. G.; Jenkins,
- N. A.; White, R.; O'Connell, P.: Deletions and a translocation interrupt
- a cloned gene at the neurofibromatosis type 1 locus. Cell 62: 187-192,
- 1990.
-
- 8. Wilson, D. E., Jr.; Povey, S.; Harris, H.: Adenylate kinases in
- man: evidence for a third locus. Ann. Hum. Genet. 39: 305-313,
- 1976.
-
- 9. Xu, G.; O'Connell, P.; Stevens, J.; White, R.: Characterization
- of human adenylate kinase 3 (AK3) cDNA and mapping of the AK3 pseudogene
- to an intron of the NF1 gene. Genomics 13: 537-542, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 2/7/1995
- jason: 6/28/1994
- carol: 8/11/1992
- carol: 6/29/1992
- supermim: 3/16/1992
- carol: 2/29/1992
-
- *RECORD*
- *FIELD* NO
- 103050
- *FIELD* TI
- *103050 ADENYLOSUCCINATE LYASE; ADSL
- ADENYLOSUCCINASE
- ADENYLOSUCCINASE DEFICIENCY, INCLUDED;;
- SUCCINYLPURINEMIC AUTISM, INCLUDED
- *FIELD* TX
- Van Keuren et al. (1986, 1987) used the strategy of somatic cell
- hybridization of human cells with Chinese hamster ovary (CHO-K1) mutants
- deficient in specific steps of the purine biosynthesis pathway to map
- the human gene correcting deficiency of the enzyme adenylosuccinase (EC
- 4.3.2.2). This CHO-K1 mutant has been designated ade(-)I.
- Adenylosuccinase carries out two independent but similar steps of purine
- biosynthesis: the removal of a fumarate from succinylaminoimidazole
- carboxamide (SAICA) ribotide to give aminoimidazole carboxamide ribotide
- and removal of fumarate from adenylosuccinate to give AMP. These are the
- ninth and the thirteenth steps of adenylate biosynthesis. Ade(-)I cells
- require exogenous adenine for growth. Cell hybrids made by fusing
- ade(-)I in human cell lines were selected for purine prototrophy in
- adenine-free medium. Human chromosome 22 was found to be required for
- growth without adenine. Assignment of the gene for adenylosuccinase to
- chromosome 22 was confirmed by Southern blot analysis with a DNA probe
- that had been isolated from a human fetal brain library and previously
- mapped to chromosome 22. By Southern blotting techniques using somatic
- cell hybrids, Budarf et al. (1991) demonstrated that ADSL maps to
- 22q13.1, distal to the Ewing sarcoma breakpoint (133450). Using both a
- somatic cell hybrid mapping panel and fluorescence in situ
- hybridization, Fon et al. (1993) localized the ADSL gene to
- 22q13.1-q13.2.
-
- Homozygosity for mutations in the adenylosuccinase gene results in a
- clinical disorder called succinylpurinemic autism. In 3 children with
- severe psychomotor delay and autism, Jaeken and Van den Berghe (1984)
- found succinyladenosine and succinylaminoimidazole carboxamide ribotide
- in the body fluids. Concentrations of both compounds were about 100
- micromol/l in CSF, between 5 and 10 micromol/l in plasma, and in the
- millimol/l range in urine. Normally these compounds are not found in
- blood and CSF but may be detected in trace amounts in urine. The
- compounds are dephosphorylated derivatives of the intracellular
- metabolites adenylosuccinate and succinylaminoimidazole carboxamide
- ribotide, the 2 substrates of adenylosuccinase (adenylosuccinate lyase).
- This enzyme is involved in both de novo synthesis of purines and
- formation of adenosine monophosphate from inosine monophosphate. Assays
- of the enzyme in 1 patient showed marked reduction of activity in liver
- and absence of activity in the kidney. Two of the 3 affected children
- were brother and sister, offspring of related Moroccan parents. (At one
- point the authors stated that the parents were related; at another they
- stated that the boy's 'grandparents were first cousins.' Does this mean
- that the parents were second cousins?) The authors suggested that
- adenylosuccinase deficiency is a specific autosomal recessive cause of
- autism. (Stone et al. (1992) demonstrated a point mutation in the ADSL
- gene in the 2 Moroccan sibs; see 103050.0001.) Jaeken et al. (1988)
- presented clinical and biochemical data on 8 children with
- adenylosuccinase deficiency. Seven of the 8 children showed severe
- psychomotor retardation. Epilepsy was documented in 5, autistic features
- in 3, and growth retardation associated with muscular wasting in a
- brother and sister. One female patient was strikingly less retarded
- mentally and had only mild psychomotor retardation. In this patient the
- ratio of the 2 metabolites in body fluids was quite different from that
- in the severely retarded patients, showing an approximately 5-fold
- excess of succinyladenosine. In addition, adenylosuccinase activity in
- fibroblasts was only about 6% of normal, whereas it was about 40% of
- normal in 6 severely retarded patients. At least 2 of the patients from
- separate families were the offspring of consanguineous parents. Maddocks
- and Reed (1989) described a seemingly sensitive and specific test for
- succinyladenosine in the urine. Jaeken et al. (1992) described a patient
- with an intermediate severity. Chemical findings in the patient
- supported the impression that there is an inverse relationship between
- the degree of clinical involvement and the excess of succinyladenosine
- over SAICA riboside. Jaeken et al. (1992) concluded that SAICA riboside
- may be the offending compound that interferes with neurofunction and
- that succinyladenosine may protect against its effects. For purposes of
- screening, they suggested that a modified Bratton-Marshall test,
- originally designed as an assay for sulfonamides, is the most practical
- method, provided the patients are not receiving sulfonamides.
-
- Wong and O'Brien (1995) found that the cDNA of human and mouse ADSL has
- 94 and 87% identity at the amino acid and nucleotide levels,
- respectively. (Adenylosuccinate lyase catalyzes 2 similar reactions in
- the de novo purine biosynthetic pathway, both of which are cleavages
- that produce fumarate as one of the products.) The gene in the mouse is
- about 27 kb and contains 13 exons. Comparison of the exon/intron
- structure of this gene with the argininosuccinate lyase gene (ASL;
- 207900) did not suggest gene duplication or exon shuffling as a
- mechanism of evolution in the fumarate gene family.
-
- *FIELD* AV
- .0001
- SUCCINYLPURINEMIC AUTISM
- ADSL, SER413PRO
- In the 2 Moroccan sibs originally reported by Jaeken and Van den Berghe
- (1984), Stone et al. (1992) demonstrated a ser413-to-pro substitution
- that led to structural instability of the mutant enzyme.
-
- *FIELD* RF
- 1. Budarf, M. L.; Emanuel, B. S.; Collins, J.; Fibison, W.; Barshop,
- B. A.: Isolation and regional localization of the human adenylosuccinate
- lyase gene. (Abstract) Cytogenet. Cell Genet. 58: 2046 only, 1991.
-
- 2. Fon, E. A.; Demczuk, S.; Delattre, O.; Thomas, G.; Rouleau, G.
- A.: Mapping of the human adenylosuccinate lyase (ADSL) gene to chromosome
- 22q13.1-q13.2. Cytogenet. Cell Genet. 64: 201-203, 1993.
-
- 3. Jaeken, J.; Van den Bergh, F.; Vincent, M. F.; Casaer, P.; Van
- den Berghe, G.: Adenylosuccinase deficiency: a newly recognized variant. J.
- Inherit. Metab. Dis. 15: 416-418, 1992.
-
- 4. Jaeken, J.; Van den Berghe, G.: An infantile autistic syndrome
- characterised by the presence of succinylpurines in body fluids. Lancet II:
- 1058-1061, 1984.
-
- 5. Jaeken, J.; Wadman, S. K.; Duran, M.; van Sprang, F. J.; Beemer,
- F. A.; Holl, R. A.; Theunissen, P. M.; de Cock, P.; van den Bergh,
- F.; Vincent, M. F.; van den Berghe, G.: Adenylosuccinase deficiency:
- an inborn error of purine nucleotide synthesis. Europ. J. Pediat. 148:
- 126-131, 1988.
-
- 6. Maddocks, J.; Reed, T.: Urine test for adenylosuccinase deficiency
- in autistic children. (Letter) Lancet I: 158-159, 1989.
-
- 7. Stone, R. L.; Aimi, J.; Barshop, B. A.; Jaeken, J.; Van den Berghe,
- G.; Zalkin, H.; Dixon, J. E.: A mutation in adenylosuccinate lyase
- associated with mental retardation and autistic features. Nature
- Genet. 1: 59-63, 1992.
-
- 8. Van Keuren, M. L.; Hart, I.; Kao, F.-T.; Neve, R. L.; Bruns, G.
- A. P.; Kurnit, D. M.; Patterson, D.: Human chromosome 22 corrects
- the defect in the CHO mutant (Ade-I) lacking adenylosuccinase activity.
- (Abstract) Am. J. Hum. Genet. 39: A172 only, 1986.
-
- 9. Van Keuren, M. L.; Hart, I. M.; Kao, F.-T.; Neve, R. L.; Bruns,
- G. A. P.; Kurnit, D. M.; Patterson, D.: A somatic cell hybrid with
- a single human chromosome 22 corrects the defect in the CHO mutant
- (Ade-I) lacking adenylosuccinase activity. Cytogenet. Cell Genet. 44:
- 142-147, 1987.
-
- 10. Wong, L.-J. C.; O'Brien, W. E.: Characterization of the cDNA
- and the gene encoding murine adenylosuccinate lyase. Genomics 28:
- 341-343, 1995.
-
- *FIELD* CS
-
- Neuro:
- Autism;
- Severe psychomotor delay;
- Seizures
-
- Growth:
- Growth retardation
-
- Muscle:
- Muscular wasting
-
- Lab:
- High succinyladenosine and succinylaminoimidazole carboxamide ribotide
- in body fluids;
- Adenylosuccinase deficiency
-
- Inheritance:
- Autosomal recessive (22q13.1)
-
- *FIELD* CD
- Victor A. McKusick: 12/15/1986
-
- *FIELD* ED
- terry: 02/11/1997
- mark: 8/25/1995
- mimadm: 3/11/1994
- carol: 11/3/1993
- carol: 3/25/1993
- carol: 11/5/1992
- carol: 9/29/1992
-
- *RECORD*
- *FIELD* NO
- 103060
- *FIELD* TI
- *103060 ADENYLOSUCCINATE SYNTHETASE; ADSS
- Ade(-)H, COMPLEMENT OF; ADEH
- *FIELD* TX
- Somatic cell hybrids between human cells and Chinese hamster ovary cells
- deficient in specific steps in the purine biosynthetic pathway permitted
- mapping of human genes correcting the defects. The ade(-)H mutant is
- missing the enzyme adenylosuccinate synthetase (IMP:L-aspartate ligase;
- EC 6.3.4.4.), which carries out the first of a 2-step sequence in the
- biosynthesis of AMP from IMP. Thus, ade(-)H cells require exogenous
- adenine for growth. Lai et al. (1989) found that in somatic cell hybrids
- human chromosome 1 corrected the defect so that the hybrid cell
- containing chromosome 1 grew without adenine. Lai et al. (1991) reported
- that analysis of a human/CHO translocation chromosome that arose in 1 of
- the hybrids suggested that the gene correcting the defect lies in the
- region 1cen-q12. (See their Figure 1 for a useful diagram of the purine
- biosynthesis pathway and the purine nucleotide cycle pathway, together
- with the location of the genes for the enzymes when known.) AMP
- deaminase, which converts AMP back to IMP, is coded by a gene, perhaps 2
- genes, in region 1p21-p13; see 102770.
-
- From a human liver library, Powell et al. (1992) isolated a cDNA that
- encoded a protein of 455 amino acids. Alignment with the sequence of the
- ADSS gene in mouse, Dictyostelium discoideum, and E. coli pointed to
- invariant residues that are likely to be important for structure and/or
- catalysis. The human ADSS sequence also showed some similarity to
- argininosuccinate synthetase, which catalyzes a chemically similar
- reaction.
-
- *FIELD* RF
- 1. Lai, L.; Hart, I.; Patterson, D.: Human chromosome 1 corrects
- the defect in the CHO mutant (Ade-H) deficient in a branch point enzyme
- in purine de novo biosynthesis. (Abstract) Cytogenet. Cell Genet. 51:
- 1028 only, 1989.
-
- 2. Lai, L.-W.; Hart, I. M.; Patterson, D.: A gene correcting the
- defect in the CHO mutant Ade(-)H, deficient in a branch point enzyme
- (adenylosuccinate synthetase) of de novo purine biosynthesis, is located
- on the long arm of chromosome 1. Genomics 9: 322-328, 1991.
-
- 3. Powell, S. M.; Zalkin, H.; Dixon, J. E.: Cloning and characterization
- of the cDNA encoding human adenylosuccinate synthetase. FEBS Lett. 303:
- 4-10, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 6/1/1989
-
- *FIELD* ED
- carol: 8/17/1992
- supermim: 3/16/1992
- carol: 2/5/1992
- carol: 1/15/1991
- supermim: 3/20/1990
- ddp: 10/27/1989
-
- *RECORD*
- *FIELD* NO
- 103070
- *FIELD* TI
- *103070 ADENYLYL CYCLASE, BRAIN, TYPE I
- ADENYLATE CYCLASE 8; ADCY8;;
- ADENYLATE CYCLASE 3, FORMERLY; ADCY3, FORMERLY
- *FIELD* TX
- Adenylyl cyclase (EC 4.6.1.1) catalyzes the transformation of ATP into
- cyclic AMP. The enzymatic activity is under the control of several
- hormones, and different polypeptides participate in the transduction of
- the signal from the receptor to the catalytic moiety. Stimulatory or
- inhibitory receptors (Rs and Ri) interact with G proteins (Gs and Gi)
- that exhibit GTPase activity and they modulate the activity of the
- catalytic subunit of the adenylyl cyclase. Parma et al. (1991) cloned a
- cDNA corresponding to human brain adenylyl cyclase, symbolized by them
- as HBAC1. By in situ hybridization to metaphase chromosomal spreads
- using the human brain cDNA probe, Stengel et al. (1992) showed that the
- gene is located on 8q24.2. A highly homologous gene, ADCY2 (103071), was
- assigned to 5p15.3 by the same method.
-
- *FIELD* RF
- 1. Parma, J.; Stengel, D.; Gannage, M.-H.; Poyard, M.; Barouki, R.;
- Hanoune, J.: Sequence of a human brain adenylyl cyclase partial cDNA:
- evidence for a consensus cyclase domain. Biochem. Biophys. Res.
- Commun. 179: 455-462, 1991.
-
- 2. Stengel, D.; Parma, J.; Gannage, M.-H.; Roeckel, N.; Mattei, M.-G.;
- Barouki, R.; Hanoune, J.: Different chromosomal localization of two
- adenylyl cyclase genes expressed in human brain. Hum. Genet. 90:
- 126-130, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 12/4/1992
-
- *FIELD* ED
- carol: 9/19/1994
- carol: 5/27/1993
- carol: 5/26/1993
- carol: 1/12/1993
- carol: 12/30/1992
- carol: 12/4/1992
-
- *RECORD*
- *FIELD* NO
- 103071
- *FIELD* TI
- *103071 ADENYLYL CYCLASE, BRAIN, TYPE II
- ADENYLATE CYCLASE 2; ADCY2
- *FIELD* TX
- Stengel et al. (1992) identified a brain cDNA corresponding to a gene
- that encodes a human brain adenylyl cyclase, which they symbolized
- HBAC2. The amino acid sequence of ADCY2 displayed significant homology
- with ADCY8 (103070) in the highly conserved adenylyl cyclase domain (250
- amino acids) found in the 3-prime cytoplasmic portion of all mammalian
- adenylyl cyclases. However, outside this domain, the homology was
- extremely low. By in situ hybridization to metaphase chromosomal spreads
- using a human brain cDNA probe, they demonstrated that the ADCY2 gene
- maps to 5p15.3. There was no cross-reactivity with the site on 8q24.2
- where ADCY8 was found to map. Using Southern blot analysis of somatic
- cell hybrid DNAs, Gaudin et al. (1994) likewise mapped type II adenylyl
- cyclase to chromosome 5. Furthermore, they determined the chromosomal
- location of 4 other isoforms: type III on chromosome 2, type IV on
- chromosome 14, type V on chromosome 3, and type VI on chromosome 12. By
- fluorescence in situ hybridization, Edelhoff et al. (1995) mapped the
- mouse homolog to chromosome 13 in the C1 region.
-
- *FIELD* RF
- 1. Edelhoff, S.; Villacres, E. C.; Storm, D. R.; Disteche, C. M.:
- Mapping of adenylyl cyclase genes type I, II, III, IV, V, and VI in
- mouse. Mammalian Genome 6: 111-113, 1995.
-
- 2. Gaudin, C.; Homcy, C. J.; Ishikawa, Y.: Mammalian adenylyl cyclase
- family members are randomly located on different chromosomes. Hum.
- Genet. 94: 527-529, 1994.
-
- 3. Stengel, D.; Parma, J.; Gannage, M.-H.; Roeckel, N.; Mattei, M.-G.;
- Barouki, R.; Hanoune, J.: Different chromosomal localization of two
- adenylyl cyclase genes expressed in human brain. Hum. Genet. 90:
- 126-130, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 12/4/1992
-
- *FIELD* ED
- mark: 4/10/1995
- terry: 1/9/1995
- carol: 9/19/1994
- carol: 5/27/1993
- carol: 1/12/1993
- carol: 12/4/1992
-
- *RECORD*
- *FIELD* NO
- 103072
- *FIELD* TI
- *103072 ADENYLYL CYCLASE, FETAL BRAIN, TYPE I
- ADENYLATE CYCLASE 1; ADCY1
- *FIELD* TX
- The neural-specific, calmodulin-sensitive adenylyl cyclase (type I),
- which was first cloned from bovine brain, has been implicated in
- learning and memory. Villacres et al. (1993) cloned the gene for human
- fetal brain type I adenylyl cyclase and showed by in situ hybridization
- that the gene lies in the region 7p13-p12. See 103070 and 103071 for
- genes encoding other forms of brain adenylyl cyclase. Gaudin et al.
- (1994) likewise mapped the ADCY1 gene to chromosome 7 by Southern blot
- analysis of somatic cell hybrid DNAs. By fluorescence in situ
- hybridization, Edelhoff et al. (1995) mapped the mouse homolog to
- chromosome 11 in the A2 region.
-
- *FIELD* RF
- 1. Edelhoff, S.; Villacres, E. C.; Storm, D. R.; Disteche, C. M.:
- Mapping of adenylyl cyclase genes type I, II, III, IV, V, and VI in
- mouse. Mammalian Genome 6: 111-113, 1995.
-
- 2. Gaudin, C.; Homcy, C. J.; Ishikawa, Y.: Mammalian adenylyl cyclase
- family members are randomly located on different chromosomes. Hum.
- Genet. 94: 527-529, 1994.
-
- 3. Villacres, E. C.; Xia, Z.; Bookbinder, L. H.; Edelhoff, S.; Disteche,
- C. M.; Storm, D. R.: Cloning, chromosomal mapping, and expression
- of human fetal brain type I adenylyl cyclase. Genomics 16: 473-478,
- 1993.
-
- *FIELD* CD
- Victor A. McKusick: 5/26/1993
-
- *FIELD* ED
- mark: 4/10/1995
- carol: 1/9/1995
- carol: 5/27/1993
- carol: 5/26/1993
-
- *RECORD*
- *FIELD* NO
- 103100
- *FIELD* TI
- 103100 ADIE SYNDROME
- *FIELD* TX
- This is a stationary, harmless disorder characterized by tonic,
- sluggishly reacting pupil and hypoactive or absent tendon reflexes. De
- Rudolf (1936) described it in mother and daughter, McKinney and Frocht
- (1940) in father and son, and Mylius (1938) in sibs. The pupil (Laties
- and Scheie, 1965) is excessively sensitive to mecholyl (methacholine).
- In familial dysautonomia, a recessive (q.v.), the pupil is also
- mecholyl-sensitive and tendon reflexes are absent. It would be of
- interest to determine whether the reflexes return with parenteral
- administration of mecholyl as occurs in dysautonomia. An autopsied case
- was reported by Harriman and Garland (1968), who found neuronal
- degeneration in the ciliary ganglion. Selective degeneration of neurons
- in dorsal root ganglia may have been the basis for areflexia. Miyasaki
- et al. (1988) concluded from electrophysiologic studies carried out in
- 11 patients with Adie syndrome that the hyporeflexia in this condition
- is due to the loss of large spindle afferents or the reduced
- effectiveness of their monosynaptic connections to motoneurons.
-
- *FIELD* SA
- Adie (1932)
- *FIELD* RF
- 1. Adie, W. J.: Tonic pupils and absent tendon reflexes: a benign
- disorder sui generis: its complete and incomplete forms. Brain 55:
- 98-113, 1932.
-
- 2. De Rudolf, G.: Tonic pupils with absent tendon reflexes in mother
- and daughter. J. Neurol. Neurosurg. Psychiat. 16: 367-368, 1936.
-
- 3. Harriman, D. G. F.; Garland, H.: The pathology of Adie's syndrome.
- Brain 91: 401-418, 1968.
-
- 4. Laties, A. M.; Scheie, H. G.: Adie's syndrome: duration of methacholine
- sensitivity. Arch. Ophthal. 74: 458-459, 1965.
-
- 5. McKinney, J. M.; Frocht, M.: Adie's syndrome: a non-luetic disease
- simulating tabes dorsalis. Am. J. Med. Sci. 199: 546-555, 1940.
-
- 6. Miyasaki, J. M.; Ashby, P.; Sharpe, J. A.; Fletcher, W. A.: On
- the cause of hyporeflexia in the Holmes-Adie syndrome. Neurology 38:
- 262-265, 1988.
-
- 7. Mylius, (NI): Ueber familiaeres Vorkommen der Pupillotonie. Klin.
- Mbl. Augenheilk. 101: 598-599, 1938.
-
- *FIELD* CS
-
- Eyes:
- Sluggish pupillary response;
- Mecholyl-sensitive pupil
-
- Neuro:
- Hyporeflexia
-
- Misc:
- Stationary, harmless disorder
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- warfield: 4/1/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 6/7/1988
-
- *RECORD*
- *FIELD* NO
- 103180
- *FIELD* TI
- *103180 ADP-RIBOSYLATION FACTOR-1; ARF1
- *FIELD* TX
- ADP-ribosylation factors (ARFs), small guanine nucleotide-binding
- proteins that enhance the enzymatic activities of cholera toxin,
- constitute 1 family of the RAS superfamily. Monomeric guanine
- nucleotide-binding proteins of the RAS superfamily function in a variety
- of cellular processes including signaling, growth, immunity, and protein
- transport. ARFs are essential and ubiquitous in eukaryotes, being
- involved in vesicular transport and functioning as an activator of
- phospholipase D. The functions of ARF proteins in membrane traffic and
- organelle integrity are intimately tied to its reversible association
- with membranes and specific interactions with membrane phospholipids. A
- common feature of these functions is their regulation by the binding and
- hydrolysis of GTP. Amor et al. (1994) described the 3-dimensional
- structure of full-length human ARF1 in its GDP-bound nonmyristoylated
- form.
-
- Bobak et al. (1989) cloned 2 ARF cDNAs, ARF1 and ARF3 (103190), from a
- human cerebellum library. Based on deduced amino acid sequences and
- patterns of hybridization of cDNA and oligonucleotide probes with
- mammalian brain poly(a)+ RNA, human ARF1 is the homolog of bovine ARF1.
- Human ARF3, however, appeared to represent a newly identified, third
- type of ARF, which differs from bovine ARF1 and bovine ARF2. Peng et al.
- (1989) also reported cloning of ADP-ribosylation factor.
-
- Lee et al. (1992) found that the human ARF-1 is identical to its bovine
- counterpart, has a distinctive pattern of tissue and developmental
- expression, and is encoded by an mRNA of approximately 1.9 kb. With 4
- introns, the human ARF1 gene spans approximately 16.5 kb. Exon 1 (46 bp)
- contains only untranslated sequence. The 5-prime-flanking region has a
- high GC content but no TATA or CAAT box, as found in housekeeping genes.
- The authors stated that the 2 human class I ARF genes, ARF1 and ARF3,
- have similar exon/intron organizations and use GC-rich promoters.
-
- Hirai et al. (1996) obtained an expressed sequence tag (EST) containing
- the ARF1 gene and used fluorescence in situ hybridization to assign ARF1
- to 1q42.
-
- *FIELD* RF
- 1. Amor, J. C.; Harrison, D. H.; Kahn, R. A.; Ringe, D.: Structure
- of the human ADP-ribosylation factor 1 complexed with GDP. Nature 372:
- 704-708, 1994.
-
- 2. Bobak, D. A.; Nightingale, M. S.; Murtagh, J. J.; Price, S. R.;
- Moss, J.; Vaughan, M.: Molecular cloning, characterization, and expression
- of human ADP-ribosylation factors: two guanine nucleotide-dependent
- activators of cholera toxin. Proc. Nat. Acad. Sci. 86: 6101-6105,
- 1989.
-
- 3. Hirai, M.; Kusuda, J.; Hashimoto, K.: Assignment of human ADP
- ribosylation factor (ARF) genes ARF1 and ARF3 to chromosomes 1q42
- and 12q13, respectively. Genomics 34: 263-265, 1996.
-
- 4. Lee, C.-M.; Haun, R. S.; Tsai, S.-C.; Moss, J.; Vaughan, M.: Characterization
- of the human gene encoding ADP-ribosylation factor 1, a guanine nucleotide-binding
- activator of cholera toxin. J. Biol. Chem. 267: 9028-9034, 1992.
-
- 5. Peng, Z. G.; Calvert, I.; Clark, J.; Helman, L.; Kahn, R.; Kung,
- H. F.: Molecular cloning, sequence analysis and mRNA expression of
- human ADP-ribosylation factor. Biofactors 2: 45-49, 1989.
-
- *FIELD* CN
- Lori M. Kelman - updated: 8/22/1996
-
- *FIELD* CD
- Victor A. McKusick: 9/26/1989
-
- *FIELD* ED
- joanna: 04/10/1997
- mark: 8/22/1996
- terry: 8/22/1996
- mark: 8/21/1996
- mark: 1/5/1996
- terry: 1/3/1996
- terry: 1/6/1995
- carol: 9/23/1994
- supermim: 3/16/1992
- carol: 7/5/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 103188
- *FIELD* TI
- *103188 ADP-RIBOSYLATION FACTOR-5; ARF5
- *FIELD* TX
- ADP-ribosylation factors (ARFs) are guanine nucleotide-binding proteins,
- approximately 20 kD in size, that serve as GTP-dependent allosteric
- activators of cholera toxin ADP-ribosyltransferase activity. To the 4
- species of mammalian ARF, termed ARF1-4, previously identified by
- cloning, Tsuchiya et al. (1991) added new ARF-like genes, ARF5 and 6
- (600464), encoding proteins of 180 and 175 amino acids, respectively.
- Both proteins contain consensus sequences believed to be involved in
- guanine nucleotide binding and GTP hydrolysis. ARF5 was more similar in
- deduced amino acid sequence to ARF4, which also has 180 amino acids.
-
- *FIELD* RF
- 1. Tsuchiya, M.; Price, S. R.; Tsai, S.-C.; Moss, J.; Vaughan, M.
- : Molecular identification of ADP-ribosylation factor mRNAs and their
- expression in mammalian cells. J. Biol. Chem. 266: 2772-2777, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 6/17/1994
-
- *FIELD* ED
- mark: 3/23/1995
- jason: 6/17/1994
-
- *RECORD*
- *FIELD* NO
- 103190
- *FIELD* TI
- *103190 ADP-RIBOSYLATION FACTOR-3; ARF3
- *FIELD* TX
- See 103180.
-
- *FIELD* CD
- Victor A. McKusick: 9/26/1989
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 10/9/1989
- root: 9/26/1989
- *RECORD*
- *FIELD* NO
- 103195
- *FIELD* TI
- *103195 ADIPOSE DIFFERENTIATION-RELATED PROTEIN; ADRP
- *FIELD* TX
- Adipose differentiation-related protein is a novel 50-kD
- membrane-associated protein whose mRNA levels are induced rapidly and
- maximally after triggering adipocyte differentiation. Eisinger and
- Serrero (1993) isolated and characterized the mouse gene, which spans 14
- kb and contains 8 exons and 7 introns. It maps to mouse chromosome 4.
-
- *FIELD* RF
- 1. Eisinger, D. P.; Serrero, G.: Structure of the gene encoding mouse
- adipose differentiation-related protein (ADRP). Genomics 16: 638-644,
- 1993.
-
- *FIELD* CD
- Victor A. McKusick: 6/24/1993
-
- *FIELD* ED
- carol: 1/14/1994
- carol: 6/24/1993
-
- *RECORD*
- *FIELD* NO
- 103200
- *FIELD* TI
- 103200 ADIPOSIS DOLOROSA
- DERCUM DISEASE
- *FIELD* TX
- This disorder, which was first described by Dercum (1892), is
- characterized by painful subcutaneous lipomas in a background of
- obesity. It is about 5 times more frequent in females than in males.
- Onset of symptoms is generally in middle age. The fatty tumors are most
- often located on the trunk and limbs with sparing of the face and hands.
- Severe asthenia has been emphasized as a feature by some (Wohl and
- Pastor, 1938). Lynch and Harlan (1963) observed the disease in 4 members
- of 3 generations of 1 family and in 2, possibly 4, persons in 2
- generations of a second family.
-
- *FIELD* SA
- Cantu et al. (1973)
- *FIELD* RF
- 1. Cantu, J. M.; Ruiz-Barquin, E.; Jimenez, M.; Castillo, L.; Macotela-Ruiz,
- E.: Autosomal dominant inheritance in adiposis dolorosa (Dercum's
- disease). Humangenetik 18: 89-91, 1973.
-
- 2. Dercum, F. X.: Three cases of a hitherto unclassified affection
- resembling in its grosser aspects obesity, but associated with special
- nervous symptoms: adiposis dolorosa. Am. J. Med. Sci. 104: 521-535,
- 1892.
-
- 3. Lynch, H. T.; Harlan, W. L.: Hereditary factors in adiposis dolorosa
- (Dercum's disease). Am. J. Hum. Genet. 15: 184-190, 1963.
-
- 4. Wohl, M. G.; Pastor, N.: Adipositas dolorosa (Dercum's disease).
- J.A.M.A. 110: 1261-1264, 1938.
-
- *FIELD* CS
-
- Skin:
- Painful trunk and limb subcutaneous lipomas
-
- Growth:
- Obesity
-
- Misc:
- Female to male ratio 5:1;
- Middle age onset
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 10/8/1991
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 103220
- *FIELD* TI
- *103220 ADENINE NUCLEOTIDE TRANSLOCATOR 1; ANT1
- ADP/ATP TRANSLOCATOR OF SKELETAL MUSCLE;;
- ANT;;
- ADP/ATP TRANSLOCASE 1
- *FIELD* TX
- The ADP/ATP translocator, or adenine nucleotide translocator (ANT), is
- the most abundant mitochondrial protein. In its functional state, it is
- a homodimer of 30-kD subunits embedded asymmetrically in the inner
- mitochondrial membrane. The dimer forms a gated pore through which ADP
- is moved from the matrix into the cytoplasm. Neckelmann et al. (1987)
- characterized a 1,400-nucleotide cDNA for human skeletal muscle ANT.
- They compared the sequence with that of the human fibroblast ANT cognate
- as reported by Battini et al. (1987). This showed that the 2 distinct
- ANTs diverged about 275 million years ago. The skeletal muscle ANT is
- expressed in heart, kidney, liver, skeletal muscle, and HeLa cells. The
- rate of evolution of the skeletal muscle ANT is 10 to 12 times slower
- than that of the mitochondrial Ox/Phos genes. Mitochondrial energy
- production varies greatly among human tissues. Because the ANT
- determines the rate of ADP/ATP flux between the mitochondrion and the
- cytosol, it is a logical candidate for regulator of cellular dependence
- on oxidative energy metabolism. Li et al. (1989) reported on the cloning
- and differential expression of the human ANT1 locus. The gene is 5.8 kb
- long and contains 4 exons and 3 introns. The mRNA is 1.4 kb and most
- abundant in heart and skeletal muscle, but barely detectable in liver,
- kidney, or brain. A second full-length ANT cDNA, ANT2 (300150), derived
- from fibroblasts is present in all of the above-mentioned tissues at
- relatively constant levels. A third cDNA, ANT3 (300151), has been cloned
- from human liver (Houldsworth and Attardi, 1988). ANT1, ANT2 and ANT3
- are approximately 90% homologous at the amino acid level.
-
- Minoshima et al. (1989) used hybridization to flow-sorted human
- chromosomes and Southern blot hybridization to mouse/human somatic cell
- hybrids to demonstrate that the ANT1 gene localizes to human chromosome
- 4. See Li et al. (1989). Fan et al. (1992) regionalized the ANT1 gene to
- 4q35 by fluorescence in situ hybridization. Haraguchi et al. (1993)
- mapped the ANT1 gene to 4q35-qter using somatic cell hybrids containing
- various deletions of chromosome 4. The regional location was further
- refined through family studies using ANT1 intron and promoter nucleotide
- polymorphisms recognized by 3 different restriction endonucleases.
- Family studies suggested that ANT1 is located centromeric to D4S139
- which in turn is centromeric to the locus for facioscapulohumeral
- muscular dystrophy (FSHD; 158900). Wijmenga et al. (1993) likewise
- mapped the ANT1 gene to 4q35 to a site proximal to the FSHD gene.
- Studies using a polymorphic CA-repeat 5 kb upstream of the ANT1 gene as
- a marker in FSHD and CEPH families suggested that the ANT1 gene is
- centromeric to FSHD and is separated from it by several markers,
- including the factor XI gene (264900).
-
- Mills et al. (1996) demonstrated that the murine homolog Ant1 is located
- on chromosome 8 by studies of an interspecific cross. The gene had been
- previously localized to chromosome 8 by PCR of a somatic cell hybrid
- mapping panel with primers from the cDNA sequence. Only a single
- recombination event in 227 chromosomes was observed between Ant1 and the
- plasma kalikrein gene Klk3 (229000) which in the human maps to 4q35 as
- does also ANT1.
-
- Bakker et al. (1993) described an 8-year-old boy who was first
- investigated at the age of 3.5 years because of shortness of breath and
- rapid fatigue. Lactate levels in serum and cerebrospinal fluid were
- greatly elevated, and histochemical and electron-microscopic examination
- of skeletal muscle suggested a mitochondrial myopathy. Great clinical
- improvement was observed with the administration of vitamin E.
-
- *FIELD* SA
- Bakker et al. (1993); Li et al. (1989)
- *FIELD* RF
- 1. Bakker, H. D.; Scholte, H. R.; Van den Bogert, C.; Jeneson, J.
- A. L.; Ruitenbeek, W.; Wanders, R. J. A.; Abeling, N. G. G. M.; van
- Gennip, A. H.: Adenine nucleotide translocator deficiency in muscle:
- potential therapeutic value of vitamin E. J. Inherit. Metab. Dis. 16:
- 548-552, 1993.
-
- 2. Bakker, H. D.; Scholte, H. R.; Van den Bogert, C.; Ruitenbeek,
- W.; Jeneson, J. A. L.; Wanders, R. J. A.; Abeling, N. G. G. M.; Dorland,
- B.; Sengers, R. C. A.; van Gennip, A. H.: Deficiency of the adenine
- nucleotide translocator in muscle of a patient with myopathy and lactic
- acidosis: a new mitochondrial defect. Pediat. Res. 33: 412-417,
- 1993.
-
- 3. Battini, R.; Ferrari, S.; Kaczmarek, L.; Calabretta, B.; Chen,
- S.; Baserga, R.: Molecular cloning of a cDNA for a human ADP/ATP
- carrier which is growth-regulated. J. Biol. Chem. 262: 4355-4359,
- 1987.
-
- 4. Fan, Y.-S.; Yang, H.-M.; Lin, C. C.: Assignment of the human muscle
- adenine nucleotide translocator gene (ANT1) to 4q35 by fluorescence
- in situ hybridization. Cytogenet. Cell Genet. 60: 29-30, 1992.
-
- 5. Haraguchi, Y.; Chung, A. B.; Torroni, A.; Stepien, G.; Shoffner,
- J. M.; Wasmuth, J. J.; Costigan, D. A.; Polak, M.; Altherr, M. R.;
- Winokur, S. T.; Wallace, D. C.: Genetic mapping of human heart-skeletal
- muscle adenine nucleotide translocator and its relationship to the
- facioscapulohumeral muscular dystrophy locus. Genomics 16: 479-485,
- 1993.
-
- 6. Houldsworth, J.; Attardi, G.: Two distinct genes for ADP/ATP translocase
- are expressed at the mRNA level in adult human liver. Proc. Nat.
- Acad. Sci. 85: 377-381, 1988.
-
- 7. Li, K.; Warner, C. K.; Hodge, J. A.; Minoshima, S.; Kudoh, J.;
- Fukuyama, R.; Maekawa, M.; Shimizu, Y.; Shimizu, N.; Wallace, D. C.
- : A human muscle adenine nucleotide translocator gene has four exons,
- is located on chromosome 4, and is differentially expressed. J. Biol.
- Chem. 264: 13998-14004, 1989.
-
- 8. Li, K.; Warner, C. K.; Hodge, J. A.; Wallace, D. C.: Cloning and
- tissue-differential expression of human heart-skeletal muscle adenine
- nucleotide translocator gene. (Abstract) Cytogenet. Cell Genet. 51:
- 1032-1033, 1989.
-
- 9. Mills, K. A.; Ellison, J. W.; Mathews, K. D.: The Ant1 gene maps
- near Klk3 on proximal mouse chromosome 8. Mammalian Genome 7: 707
- only, 1996.
-
- 10. Minoshima, S.; Kudoh, J.; Fukuyama, R.; Maekawa, M.; Shimizu,
- Y.; Li, K.; Wallace, D. C.; Shimizu, N.: Mapping of the human muscle
- adenine nucleotide translocator gene (ANT1) to chromosome 4. (Abstract) Cytogenet.
- Cell Genet. 51: 1044-1045, 1989.
-
- 11. Neckelmann, N.; Li, K.; Wade, R. P.; Shuster, R.; Wallace, D.
- C.: cDNA sequence of a human skeletal muscle ADP/ATP translocator:
- lack of a leader peptide, divergence from a fibroblast translocator
- cDNA, and coevolution with mitochondrial DNA genes. Proc. Nat. Acad.
- Sci. 84: 7580-7584, 1987.
-
- 12. Wijmenga, C.; Winokur, S. T.; Padberg, G. W.; Skraastad, M. I.;
- Altherr, M. R.; Wasmuth, J. J.; Murray, J. C.; Hofker, M. H.; Frants,
- R. R.: The human skeletal muscle adenine nucleotide translocator
- gene maps to chromosome 4q35 in the region of the facioscapulohumeral
- muscular dystrophy locus. Hum. Genet. 92: 198-203, 1993.
-
- *FIELD* CD
- Victor A. McKusick: 12/3/1987
-
- *FIELD* ED
- mark: 10/26/1996
- terry: 10/17/1996
- carol: 5/10/1994
- carol: 10/26/1993
- carol: 9/13/1993
- carol: 5/26/1993
- carol: 4/7/1993
- carol: 1/26/1993
-
- *RECORD*
- *FIELD* NO
- 103230
- *FIELD* TI
- 103230 ADRENOCORTICAL HYPOFUNCTION, CHRONIC PRIMARY CONGENITAL
- ADDISON DISEASE, CONGENITAL
- *FIELD* TX
- Chuandi et al. (1985) reported a Chinese kindred in which persons in 3
- generations, and by implication at least 1 person in a fourth earlier
- generation, had chronic adrenal insufficiency. This was manifest by
- hyperpigmentation, hypernatriuria, hypokaliuria, and decreased plasma
- total cortisol and urine free cortisol; PTC, UFC and 17-OHCS did not
- respond to ACTH stimulation. Eleven affected persons in 5 sibships were
- identified, including several instances of male-to-male transmission.
-
- *FIELD* RF
- 1. Chuandi, L.; Junqing, C.; Ruohua, S.; Ruqian, Z.; Guilin, Y.; Wei,
- L.; Wenying, Y.; Qing, Z.; Guirong, L.; Heling, L.; Shiqin, D.: Addison's
- disease of autosomal dominant inheritance: a report of 11 cases in
- one family. Kexue Tongbao 30: 981-984, 1985.
-
- *FIELD* CS
-
- Endocrine:
- Chronic adrenal insufficiency
-
- Skin:
- Hyperpigmentation
-
- Lab:
- Hypernatriuria;
- Hypokaliuria;
- Decreased plasma total cortisol;
- Decreased urine free cortisol;
- No response of PTC, UFC and 17-OHCS to ACTH stimulation
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- root: 1/11/1988
-
- *RECORD*
- *FIELD* NO
- 103260
- *FIELD* TI
- *103260 ADRENODOXIN; ADX
- FERREDOXIN 1, INCLUDED;;
- FDX1, INCLUDED
- *FIELD* TX
- Ferredoxin is a small, acidic, iron-sulfur protein that functions as an
- electron transport intermediate for mitochondrial cytochromes P450
- involved in steroid, vitamin D, and bile acid metabolism. Electrons are
- transferred from NADPH through a flavin-containing protein (ferredoxin
- oxidoreductase) and ferredoxin to the terminal cytochrome P450 for
- oxidation/reduction reactions. Mitochondrial P450s and their ferredoxin
- are found mainly in the steroidogenic tissues, including adrenal, ovary,
- testis, and placenta (Jefcoate et al., 1986). Small amounts of them are
- also found in the liver and kidney for bile acid and vitamin D
- synthesis. Because of its relative abundance, the adrenal ferredoxin,
- designated adrenodoxin, has been characterized in the most detail. It is
- synthesized as a precursor in which 60 amino acids of the signal peptide
- are later cleaved upon transport into the mitochondrial inner matrix to
- form a mature protein of 124 amino acids (Okamura et al., 1985). In
- almost all human tissues, Morel et al. (1987, 1988) found ADX mRNA in 3
- sizes: 1.1, 1.4, and 1.65 kb. Cloning and sequencing of 3 ADX cDNAs
- showed that the mRNAs of various sizes resulted from alternate
- polyadenylation sites yielding 3-prime untranslated regions of 229, 530,
- and 790 bp, respectively. The 540-bp coding region and the 5-prime
- untranslated region were identical in all cases. By means of Southern
- blot analysis of DNA from somatic cell hybrids using stringent
- conditions of hybridization, 2 chromosomal sites were identified for the
- ADX gene: chromosomes 11 and 20. One sequence was suspected to represent
- a processed, intronless pseudogene. Because of the restriction pattern,
- Morel et al. (1987) suggested that the sequence on chromosome 20 is a
- pseudogene. Chang et al. (1988) found that the ADX gene spans more than
- 20 kb and contains 4 exons and 3 introns. The first exon encodes the
- 60-amino acid signal peptide, which directs transport of the protein
- into the inner mitochondrial matrix. The mature peptide of 124 amino
- acids is encoded by the other 3 exons. The third exon encodes the
- portion of the protein containing the ion-sulfur center and a domain
- that binds other components of the electron transport chain.
-
- By analysis of somatic cell hybrids, Morel et al. (1988) and Chang et
- al. (1990) assigned the ADX gene to 11q13-qter. Chang et al. (1990)
- identified pseudogenes on both chromosome 20 and chromosome 21. The
- pseudogenes lacked introns and contained numerous mutations, including
- an insertion, deletion, and substitution, which rendered them inactive.
- They concluded that there are 2 expressed genes, but only 1 gene product
- and that both expressed genes are located on chromosome 11. Human
- adrenodoxin and placental ferredoxin cDNAs share an identical sequence,
- suggesting that they are the same (Mittal et al., 1988). Chashchin et
- al. (1986) found that adrenodoxin is identical in sequence to liver
- ferredoxin (hepatoredoxin). Renal ferredoxin (renodoxin) has similar
- optic, renal, and immunochemical properties to adrenodoxin, although
- Maruya et al. (1983) suggested that the 2 have minor differences.
- Because they identified only 1 protein sequence, Chang et al. (1990)
- suggested that there is no need to designate ferredoxin according to the
- tissue origin. By in situ hybridization, Sparkes et al. (1991) refined
- the assignment of ADX to 11q22 and demonstrated pseudogenes on
- 20q11-q12.
-
- *FIELD* SA
- Picado-Leonard et al. (1988)
- *FIELD* RF
- 1. Chang, C.-Y.; Wu, D.-A.; Lai, C.-C.; Miller, W. L.; Chung, B.-C.
- : Cloning and structure of the human adrenodoxin gene. DNA 7: 609-615,
- 1988.
-
- 2. Chang, C.-Y.; Wu, D.-A.; Mohandas, T. K.; Chung, B.-C.: Structure,
- sequence, chromosomal location, and evolution of the human ferredoxin
- gene family. DNA Cell Biol. 9: 205-212, 1990.
-
- 3. Chashchin, V. L.; Lapko, V. N.; Adamovich, T. B.; Kirillova, N.
- M.; Lapko, A. G.; Akhrem, A. A.: The primary structure of hepatoredoxin
- from bovine liver mitochondria. Bioorg. Khim. 12: 1286-1289, 1986.
-
- 4. Jefcoate, C. R.; McNamara, B. C.; DiBartolomeis, M. J.: Control
- of steroid synthesis in adrenal fasciculata cells. Endocr. Res. 12:
- 314-350, 1986.
-
- 5. Maruya, N.; Hiwatashi, A.; Ichikawa, Y.; Yamano, T.: Purification
- and characterization of renal ferredoxin from bovine renal mitochondria.
- J. Biochem. 93: 1239-1247, 1983.
-
- 6. Mittal, S.; Zhu, Y. Z.; Vickery, L. E.: Molecular cloning and
- sequence analysis of human placental ferredoxin. Arch. Biochem.
- Biophys. 264: 383-391, 1988.
-
- 7. Morel, Y.; Picado-Leonard, J.; Mohandas, T. K.; Miller, W. L.:
- Two highly homologous genes for adrenodoxin lie on human chromosomes
- 11 and 20. (Abstract) Am. J. Hum. Genet. 41: A178 only, 1987.
-
- 8. Morel, Y.; Picado-Leonard, J.; Wu, D.-A.; Chang, C.-Y.; Mohandas,
- T. K.; Chung, B.-C.; Miller, W. L.: Assignment of the functional
- gene for human adrenodoxin to chromosome 11q13-qter and of adrenodoxin
- pseudogenes to chromosome 20cen-q13.1. Am. J. Hum. Genet. 43: 52-59,
- 1988.
-
- 9. Okamura, T.; John, M. E.; Zuber, M. X.; Simpson, E. R.; Waterman,
- M. R.: Molecular cloning and amino acid sequence of the precursor
- form of bovine adrenodoxin: evidence for a previously unidentified
- COOH-terminal peptide. Proc. Nat. Acad. Sci. 82: 5705-5709, 1985.
-
- 10. Picado-Leonard, J.; Voutilainen, R.; Kao, L.-C.; Chung, B.-C.;
- Strauss, J. F., III; Miller, W. L.: Human adrenodoxin: cloning of
- three cDNAs and cycloheximide enhancement in JEG-3 cells. J. Biol.
- Chem. 263: 3240-3244, 1988.
-
- 11. Sparkes, R. S.; Klisak, I.; Miller, W. L.: Regional mapping of
- genes encoding human steroidogenic enzymes: P450scc to 15q23-q24;
- adrenodoxin to 11q22; adrenodoxin reductase to 17q24-q25; and P450c17
- to 10q24-q25. DNA Cell Biol. 10: 359-365, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 10/22/1987
-
- *FIELD* ED
- mimadm: 4/14/1994
- carol: 10/15/1993
- carol: 10/27/1992
- carol: 10/26/1992
- supermim: 3/16/1992
- carol: 2/29/1992
-
- *RECORD*
- *FIELD* NO
- 103270
- *FIELD* TI
- *103270 ADRENODOXIN REDUCTASE; ADXR
- FERREDOXIN:NADP(+) REDUCTASE; FDXR
- *FIELD* TX
- Adrenodoxin reductase (ferredoxin:NADP(+) oxidoreductase; EC 1.18.1.2)
- is a mitochondrial flavoprotein that receives electrons from NADPH, thus
- initiating the electron-transport chain serving mitochondrial
- cytochromes P450. Solish et al. (1988) cloned and sequenced 2 human ADXR
- cDNAs that differed by the presence of 6 additional codons in the middle
- of 1 clone. The sequence in this region of the clones indicated that
- these 6 extra codons rose by alternative splicing of the pre-mRNA.
- Southern blot analysis indicated that the human genome contains only 1
- ADXR gene. Lin et al. (1990) found that the ADXR gene is 12 kb long and
- consists of 12 exons. The first exon encodes the first 26 of the 32
- amino acids of the signal peptide, and the second exon encodes the
- remainder of the signal peptide and the apparent FAD binding site. The
- remaining 10 exons are clustered in a region of only 4.3 kb, separated
- from the first 2 exons by a large intron of about 5.6 kb. Lin et al.
- (1990) also found 2 forms of mRNA, which differed by the absence or
- presence of 18 bases in the middle of the sequence; these arise through
- alternative splicing at the 5-prime end of exon 7. By analysis of DNA
- from a panel of mouse-human somatic cell hybrids, Solish et al. (1988)
- localized the gene to 17cen-q25. By in situ hybridization, Sparkes et
- al. (1991) refined the assignment to 17q24-q25.
-
- *FIELD* RF
- 1. Lin, D.; Shi, Y.; Miller, W. L.: Cloning and sequence of the human
- adrenodoxin reductase gene. Proc. Nat. Acad. Sci. 87: 8516-8520,
- 1990.
-
- 2. Solish, S. B.; Picado-Leonard, J.; Morel, Y.; Kuhn, R. W.; Mohandas,
- T. K.; Hanukoglu, I.; Miller, W. L.: Human adrenodoxin reductase:
- two mRNAs encoded by a single gene on chromosome 17cen-q25 are expressed
- in steroidogenic tissues. Proc. Nat. Acad. Sci. 85: 7104-7108,
- 1988.
-
- 3. Sparkes, R. S.; Klisak, I.; Miller, W. L.: Regional mapping of
- genes encoding human steroidogenic enzymes: P450scc to 15q23-q24;
- adrenodoxin to 11q22; adrenodoxin reductase to 17q24-q25; and P450c17
- to 10q24-q25. DNA Cell Biol. 10: 359-365, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 10/12/1988
-
- *FIELD* ED
- carol: 10/26/1992
- supermim: 3/16/1992
- carol: 8/19/1991
- carol: 12/3/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 103275
- *FIELD* TI
- *103275 ADRENOMEDULLIN; AM
- ADM
- *FIELD* TX
- Adrenomedullin, a hypotensive peptide found in human pheochromocytoma,
- consists of 52 amino acids, has 1 intramolecular disulfide bond, and
- shows a slight homology with the calcitonin gene-related peptide (CGRP;
- 114130). It may function as a hormone in circulation control because it
- is found in blood in a considerable concentration. Kitamura et al.
- (1993) constructed a cDNA library of pheochromocytoma and isolated
- therefrom a cDNA clone encoding an adrenomedullin precursor. The
- precursor, called preproadrenomedullin, is 185 amino acids long. By
- RNA-blot analysis, human adrenomedullin mRNA was found to be highly
- expressed in several tissues, including adrenal medulla, cardiac
- ventricle, lung, and kidney, as well as pheochromocytoma. Ishimitsu et
- al. (1994) found that the genomic AM DNA consists of 4 exons and 3
- introns, with the 5-prime flanking region containing TATA, CAAT, and GC
- boxes. There are also multiple binding sites for activator protein-2
- (AP2TF; 107580) and a cAMP-regulated enhancer element. Southern blot
- analyses of human/hamster somatic hybrid cell lines demonstrated that
- the AM gene is represented by a single locus on chromosome 11.
-
- Richards et al. (1996) reviewed information accumulated on
- adrenomedullin since its original description by Kitamura et al. (1993).
-
- *FIELD* RF
- 1. Ishimitsu, T.; Kojima, M.; Kangawa, K.; Hino, J.; Matsuoka, H.;
- Kitamura, K.; Eto, T.; Matsuo, H.: Genomic structure of human adrenomedullin
- gene. Biochem. Biophys. Res. Commun. 203: 631-639, 1994.
-
- 2. Kitamura, K.; Sakata, J.; Kangawa, K.; Kojima, M.; Matsuo, H.;
- Eto, T.: Cloning and characterization of cDNA encoding a precursor
- for human adrenomedullin. Biochem. Biophys. Res. Commun. 194: 720-725,
- 1993.
-
- 3. Richards, A. M.; Nicholls, M. G.; Lewis, L.; Lainchbury, J. G.
- : Adrenomedullin. Clin. Sci. 91: 3-16, 1996.
-
- *FIELD* CD
- Victor A. McKusick: 9/23/1993
-
- *FIELD* ED
- terry: 11/11/1996
- terry: 10/31/1994
- carol: 10/26/1993
- carol: 9/23/1993
-
- *RECORD*
- *FIELD* NO
- 103280
- *FIELD* TI
- *103280 ADULT SKELETAL MUSCLE GENE
- ASM;;
- ASM1;;
- H19 GENE;;
- D11S813E
- *FIELD* TX
- Leibovitch et al. (1991) used a rat skeletal muscle probe originating
- from a rhabdomyosarcoma to isolate a cDNA probe from a human placental
- cDNA library. In the rat, while the corresponding mRNA and protein were
- not expressed in fetal muscle, an increasing accumulation of the
- corresponding mRNA and protein were observed during postnatal
- development of skeletal muscle, and this accumulation was maximal in
- adulthood. As no expression was found in any other tissue, the gene was
- referred to as the adult skeletal muscle (ASM) gene. Leibovitch et al.
- (1991) mapped the human gene to 11p15 by a combination of somatic hybrid
- cell analysis and in situ hybridization. (D11S813E was the designation
- assigned by HGM11 (Nguyen et al., 1991).)
-
- A gene coding for an abundant fetal transcript in mice had been
- identified by Bartolomei et al. (1991), who designated it H19. The H19
- gene is expressed in a number of organs during a restricted period of
- fetal development, and in embryonal carcinoma cells after induction of
- differentiation. The gene shows a restricted pattern of expression in
- adult tissues; expression is confined to skeletal and cardiac muscle.
- Leibovitch et al. (1991) presented evidence that the human H19 gene has
- a transcript that gives rise to a 29-kD protein.
-
- The human H19 gene is 2.7 kb long and includes 4 small introns. Zhang
- and Tycko (1992) found restriction site polymorphisms in the human H19
- gene and, by examination of the representation of these polymorphisms in
- cDNAs from fetal organs, demonstrated that H19 expression was largely or
- exclusively from a single allele. Expression of the WT1 gene (194070),
- which, like H19, maps to 11p and shows fetal expression, was found to
- have biallelic expression. In the context of previous studies of allelic
- losses in 11p15 in human embryonal tumors, the findings of Zhang and
- Tycko (1992) supported the possibility of single-step inactivation of
- monoallelically expressed growth-regulating genes in human oncogenesis.
- It was not determined in this study whether the expression was
- uniparental to indicate parental imprinting. The H19 gene and 2 other
- genes, insulinlike growth factor II (147470) and insulinlike growth
- factor II receptor (147280), show monoallelic expression in mice. IGF2
- is, like H19, located in 11p15. Zhang and Tycko (1992) commented that,
- if IGF2 also shows monoallelic expression, it may indicate that that
- region is a 'hot spot' for this phenomenon.
-
- In the mouse, the H19 gene is located on chromosome 7 in a region of
- conservation of synteny with human 11p (Jones et al., 1992). Like the
- H19 gene, the Igf2 gene is imprinted in the mouse, although in the
- opposite parents, one paternally imprinted, the other maternally. Zemel
- et al. (1992) showed that the Igf2 gene lies about 90 kb 5-prime to H19,
- in the same transcriptional orientation. Based on similar pulsed field
- gel analysis, they showed that this physical proximity is conserved in
- humans. Both genes hybridized to a fragment of about 200 kb. Zemel et
- al. (1992) proposed a model to account for the imprinting of 2 linked
- genes in opposite directions, i.e., one (H19) being paternally imprinted
- and the other (IGF2) maternally imprinted. They pointed out that the
- IGF2/H19 domain is a candidate for the Beckwith-Wiedemann syndrome (BWS;
- 130650) since the genes show imprinting and chimeric mouse embryos that
- are paternally disomic for distal mouse chromosome 7 show an overgrowth
- phenotype similar to that of BWS (Ferguson-Smith et al., 1991).
-
- From the study of the androgenetic complete hydatidiform mole,
- Rachmilewitz et al. (1992) presented strong evidence of parental
- imprinting of the human H19 gene, with the maternally derived allele as
- the active one. Furthermore, they showed that the paternally derived
- allele of the IGF2 is expressed. Thus, the situation in the human is the
- same as that in the mouse. Rainier et al. (1993) found that both H19 and
- IGF2 show monoallelic expression in human tissues and that, as in mouse,
- H19 is expressed from the maternal allele and IGF2 from the paternal
- allele. In contrast, 69% of Wilms tumors not undergoing loss of
- heterozygosity at 11p showed biallelic expression of one or both genes,
- suggesting that relaxation or loss of imprinting may represent a new
- epigenetic mutational mechanism in carcinogenesis.
-
- Mutter et al. (1993) found that normal gestations express H19 only from
- the maternal allele and express IGF2 from the paternal allele, whereas
- neither is expressed from the maternal genome of gynogenetic gestations,
- and both are expressed from the paternal genome of androgenetic
- gestations. Coexpression of H19 and IGF2 in the androgenetic tissues was
- in a single population of cells, mononuclear trophoblast--the same cell
- type expressing these genes in biparental placentas. These results
- demonstrated that a biparental genome may be required for expression of
- the reciprocal IGF2/H19 imprint.
-
- In the mouse, the imprinted H19 gene, which encodes an untranslated RNA,
- lies at the end of a cluster of imprinted genes. Leighton et al. (1995)
- found that imprinting of the insulin-2 gene and the insulin-like growth
- factor 2 gene, which lie about 100 kb upstream of H19, can be disrupted
- by maternal inheritance of a targeted deletion of the H19 gene and its
- flanking sequence. Animals inheriting the H19 mutation from their
- mothers were 27% heavier than those inheriting from their fathers.
- Paternal inheritance of the disruption had no effect, which presumably
- reflects the normally silent state of the paternal gene. The somatic
- overgrowth of heterozygotes for the maternal deletion was attributed to
- a gain-of-function of the Igf2 gene rather than a loss of function of
- H19.
-
- H19 is abundantly expressed in both extraembryonic and fetal tissues.
- Jinno et al. (1995) found that H19 is monoallelically (maternally)
- expressed in the human placenta after 10 weeks of gestation, whereas it
- is biallelically expressed at earlier stages. Regardless of H19
- biallelic or monoallelic expression, IGF2 (147470) is monoallelically
- (paternally) expressed in the placenta. Furthermore, with in situ mRNA
- hybridization using placenta showing H19 biallelic and IGF2 monoallelic
- expression, they demonstrated that defined cell types simultaneously
- contained both H19 and IGF2 transcripts. Therefore, the reciprocal
- linkage of H19 and IGF2 expression demonstrated in Wilms tumors is not
- observed in placentas. Furthermore, Jinno et al. (1995) found that,
- unlike methylation analyses of the human H19 gene, the promoter region
- of the human H19 gene is hypomethylated at all stages of placental
- development. In contrast, allele-dependent methylation of the 3-prime
- portion of the gene increases with gestational age.
-
- H19 is a developmentally regulated gene with putative tumor suppressor
- activity; loss of H19 expression may be involved in Wilms tumorigenesis.
- Han et al. (1996) performed in situ hybridization analysis of H19
- expression during normal rabbit development and in human atherosclerotic
- plaques. They found that H19 expression in developing skeletal and
- smooth muscles correlated with specific differentiation events in these
- tissues. Expression of H19 in skeletal muscle correlated with
- nonproliferative, actin-positive muscle cells. In the prenatal blood
- vessel, H19 expression was both temporally and spatially regulated with
- initial loss of expression in the inner smooth muscle layers adjacent to
- the lumen. Han et al. (1996) also identified H19-positive cells in adult
- atherosclerotic lesions, suggesting that these cells may recapitulate
- early developmental events. These results, along with the identification
- of the insulin family of growth factors as potent regulatory molecules
- for H19 expression, provided additional clues toward understanding the
- physiologic regulation and function of H19.
-
- Pfeifer et al. (1996) stated that the product of the H19 gene is an
- untranslated RNA that is expressed exclusively from the maternal
- chromosome during mammalian development. The H19 gene and its
- 5-prime-flanking sequence are required for the genomic imprinting of 2
- paternally expressed genes in mice, Ins2 and Igf2, that lie 90 and 115
- kb 5-prime to the H19 gene, respectively. Pfeifer et al. (1996)
- investigated the role of the H19 gene in its own imprinting by
- introducing a Mus spretus H19 gene into heterologous locations in the
- mouse genome. They found that multiple copies of the transgene were
- sufficient for its paternal silencing and DNA methylation. Replacing the
- H19 structural gene with a luciferase reporter gene resulted in loss of
- imprinting of the transgene; that is, high expression and low levels of
- DNA methylation were observed with both paternal and maternal
- inheritance. Removal of 701 bp at the 5-prime end of the structural H19
- gene resulted in a similar loss of paternal-specific DNA methylation,
- arguing that those sequences are required for both the establishment and
- maintenance of the sperm-specific gametic mark. The M. spretus H19
- transgene could not rescue the loss of IGF-2 imprinting in trans in H19
- deletion mice, implying a cis requirement for the H19 gene. In contrast
- to a previous report (Brunkow and Tilghman, 1991) in which
- overexpression of a marked H19 gene was a prenatal lethal, Pfeifer et
- al. (1996) found that expression of the M. spretus transgene had no
- deleterious effect, leading them to conclude that the 20-bp insertion in
- the marked gene created a neomorphic mutation.
-
- *FIELD* RF
- 1. Bartolomei, M. S.; Zemel, S.; Tilghman, S. M.: Parental imprinting
- of the mouse H19 gene. Nature 351: 153-155, 1991.
-
- 2. Brunkow, M. E.; Tilghman, S. M.: Ectopic expression of the H19
- gene in mice causes prenatal lethality. Genes Dev. 5: 1092-1101,
- 1991.
-
- 3. Ferguson-Smith, A. C.; Cattanach, B. M.; Barton, S. C.; Beechey,
- C. V.; Surani, M. A.: Embryological and molecular investigations
- of parental imprinting on mouse chromosome 7. Nature 351: 667-670,
- 1991.
-
- 4. Han, D. K. M.; Khaing, Z. Z.; Pollock, R. A.; Haudenschild, C.
- C.; Liau, G.: H19, a marker of developmental transition, is reexpressed
- in human atherosclerotic plaques and is regulated by the insulin family
- of growth factors in cultured rabbit smooth muscle cells. J. Clin.
- Invest. 97: 1276-1285, 1996.
-
- 5. Jinno, Y.; Ikeda, Y.; Yun, K.; Maw, M.; Masuzaki, H.; Fukuda, H.;
- Inuzuka, K.; Fujishita, A.; Ohtani, Y.; Okimoto, T.; Ishimaru, T.;
- Niikawa, N.: Establishment of functional imprinting of the H19 gene
- in human developing placentae. Nature Genet. 10: 318-324, 1995.
-
- 6. Jones, J. M.; Meisler, M. H.; Seldin, M. F.; Lee, B. K.; Eicher,
- E. M.: Localization of insulin-2 (Ins-2) and the obesity mutant tubby
- (tub) to distinct regions of mouse chromosome 7. Genomics 14: 197-199,
- 1992.
-
- 7. Leibovitch, M. P.; Nguyen, V. C.; Gross, M. S.; Solhonne, B.; Leibovitch,
- S. A.; Bernheim, A.: The human ASM (adult skeletal muscle) gene:
- expression and chromosomal assignment to 11p15. Biochem. Biophys.
- Res. Commun. 180: 1241-1250, 1991.
-
- 8. Leighton, P. A.; Ingram, R. S.; Eggenschwiler, J.; Efstratiadis,
- A.; Tilghman, S. M.: Disruption of imprinting caused by deletion
- of the H19 gene region in mice. Nature 375: 34-39, 1995.
-
- 9. Mutter, G. L.; Stewart, C. L.; Chaponot, M. L.; Pomponio, R. J.
- : Oppositely imprinted genes H19 and insulin-like growth factor 2
- are coexpressed in human androgenetic trophoblast. Am. J. Hum. Genet. 53:
- 1096-1102, 1993.
-
- 10. Nguyen, V. C.; Leibovitch, M.; Gross, M.; Solhonne, B.; Leibovitch,
- S. A.; Bernheim, A.: Assignment of ASM (adult skeletal muscle) to
- chromosome 11 (somatic hybrid cell analysis), region 11p15 (in situ
- hybridization). (Abstract) Cytogenet. Cell Genet. 58: 1968, 1991.
-
- 11. Pfeifer, K.; Leighton, P. A.; Tilghman, S. M.: The structural
- H19 gene is required for transgene imprinting. Proc. Nat. Acad. Sci. 93:
- 13876-13883, 1996.
-
- 12. Rachmilewitz, J.; Goshen, R.; Ariel, I.; Schneider, T.; de Groot,
- N.; Hochberg, A.: Parental imprinting of the human H19 gene. FEBS
- Lett. 309: 25-28, 1992.
-
- 13. Rainier, S.; Johnson, L. A.; Dobry, C. J.; Ping, A. J.; Grundy,
- P. E.; Feinberg, A. P.: Relaxation of imprinted genes in human cancer. Nature 362:
- 747-749, 1993.
-
- 14. Zemel, S.; Bartolomei, M. S.; Tilghman, S. M.: Physical linkage
- of two mammalian imprinted genes, H19 and insulin-like growth factor
- 2. Nature Genet. 2: 61-65, 1992.
-
- 15. Zhang, Y.; Tycko, B.: Monoallelic expression of the human H19
- gene. Nature Genet. 1: 40-44, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 1/9/1992
-
- *FIELD* ED
- terry: 01/23/1997
- terry: 1/10/1997
- mark: 5/2/1996
- terry: 4/24/1996
- mark: 7/20/1995
- carol: 11/8/1993
- carol: 9/24/1993
- carol: 10/22/1992
- carol: 10/13/1992
- carol: 10/7/1992
-
- *RECORD*
- *FIELD* NO
- 103285
- *FIELD* TI
- 103285 ADULT SYNDROME
- ACRO-DERMATO-UNGUAL-LACRIMAL-TOOTH SYNDROME
- *FIELD* TX
- Propping and Zerres (1993) described a family with at least 7 living
- members who were affected by a hitherto undescribed syndrome with
- variable expression, which bore a close resemblance to the EEC syndrome
- (129900). The main manifestations were hypodontia and/or early onset of
- permanent teeth, ectrodactyly, obstruction of lacrimal ducts,
- onychodysplasia, and excessive freckling. Another finding was
- hypoplastic breasts.
-
- *FIELD* RF
- 1. Propping, P.; Zerres, K.: ADULT-syndrome: an autosomal-dominant
- disorder with pigment anomalies, ectrodactyly, nail dysplasia, and
- hypodontia. Am. J. Med. Genet. 45: 642-648, 1993.
-
- *FIELD* CS
-
- Teeth:
- Hypodontia;
- Early onset of permanent teeth
-
- Limbs:
- Ectrodactyly
-
- Eyes:
- Lacrimal duct obstruction
-
- Nails:
- Onychodysplasia
-
- Skin:
- Excessive freckling
-
- Thorax:
- Hypoplastic breasts
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 3/24/1993
-
- *FIELD* ED
- mimadm: 3/11/1994
- carol: 3/24/1993
-
- *RECORD*
- *FIELD* NO
- 103300
- *FIELD* TI
- 103300 AGLOSSIA-ADACTYLIA
- PEROMELIA WITH MICROGNATHISM;;
- OROMANDIBULAR LIMB HYPOPLASIA
- HANHART SYNDROME, INCLUDED
- *FIELD* TX
- The features are indicated by the name, although it is to be noted that
- both the aglossia and the adactylia may be only partial. In Turkey,
- Tuncbilek et al. (1977) observed 3 sporadic cases, each with
- consanguineous parents, and espoused autosomal recessive inheritance;
- the general consanguinity rate may be high in the population in
- question, however. Epicanthus was a feature of the case I saw with
- Shokeir (1978). Robinow et al. (1978) observed discordant monozygotic
- twins; it is noteworthy, although perhaps coincidental, that the parents
- were second cousins. They also described a case with associated 'apple
- peel' bowel (243600) which is thought to arise through obliteration of
- the superior mesenteric artery. This suggested to them that the
- aglossia-adactylia syndrome might likewise be the result of vascular
- occlusion, as in the embryopathy experimentally induced by Jost and
- Poswillo. Hanhart (1950) described 3 cases of the same disorder; 2 were
- related and, in the third, the parents were consanguineous. The disorder
- is a nonmendelian developmental disturbance (Opitz, 1982). Buttiens and
- Fryns (1986) described Hanhart syndrome in brother and sister. These
- persons had retrognathia, microstomia and symmetric severe limb
- reduction defects but normal tongue. Thus, it is arguable whether it
- should be called Hanhart syndrome. Chandra Sekhar et al. (1987) reported
- with photographs 2 remarkable cases in which micrognathia was extreme.
- One patient was a male who died in the neonatal period. Structural
- abnormalities of the middle ear were described. The second case was a
- 14-year-old boy with bilateral conductive hearing loss and bilateral
- absent thumbs.
-
- *FIELD* SA
- Bokesoy et al. (1983); Falk and Murphree (1978); Nevin et al. (1975);
- Nevin et al. (1970)
- *FIELD* RF
- 1. Bokesoy, I.; Aksuyek, C.; Deniz, E.: Oromandibular limb hypogenesis/Hanhart's
- syndrome: possible drug influence on the malformation. Clin. Genet. 24:
- 47-49, 1983.
-
- 2. Buttiens, M.; Fryns, J.-P.: Hanhart syndrome in siblings. (Abstract) 7th
- Int. Cong. Hum. Genet., Berlin 274 only, 1986.
-
- 3. Chandra Sekhar, H. K.; Sachs, M.; Siverls, V. C.: Hanhart's syndrome
- with special reference to temporal bone findings. Ann. Otol. Rhinol.
- Laryng. 96: 309-314, 1987.
-
- 4. Falk, R. E.; Murphree, L.: Colobomatous microphthalmia in the
- hypoglossia-hypodactylia syndrome. (Abstract) Am. J. Hum. Genet. 30:
- 101A only, 1978.
-
- 5. Hanhart, E.: Ueber die Kombination von Peromelie mit Mikrognathie,
- ein neues Syndrom beim Menschen, entsprechend der Akroteriasis congenita
- von Wriedt und Mohr beim Rind. Arch. Klaus Stift. Vererbungsforsch. 25:
- 531-543, 1950.
-
- 6. Nevin, N. C.; Burrows, D.; Allen, G.; Kernohan, D. C.: Aglossia-adactylia
- syndrome. J. Med. Genet. 12: 89-93, 1975.
-
- 7. Nevin, N. C.; Dodge, J. A.; Kernohan, D. C.: Aglossia-adactylia
- syndrome. Oral Surg. 29: 443-446, 1970.
-
- 8. Opitz, J. M.: Personal Communication. Helena, Mont. 1982.
-
- 9. Robinow, M.; Marsh, J. L.; Edgerton, M. T.; Sabio, H.; Johnson,
- G. F.: Discordance in monozygotic twins for aglossia-adactylia, and
- possible clues to the pathogenesis of the syndrome. Birth Defects
- Orig. Art. Ser. XIV(6A): 223-230, 1978.
-
- 10. Shokeir, M. H. K.: Personal Communication. Saskatoon, Saskatchewan,
- Canada 10/3/1978.
-
- 11. Tuncbilek, E.; Yalcin, C.; Atasu, M.: Aglossia-adactylia syndrome
- (special emphasis on the inheritance pattern). Clin. Genet. 11:
- 421-423, 1977.
-
- *FIELD* CS
-
- Mouth:
- Aglossia/hypoglossia;
- Abnormal ventral frenulum;
- Retrognathia;
- Microstomia
-
- Limbs:
- Adactylia;
- Hypodactyly;
- Ectrodactyly
-
- Eyes:
- Epicanthus
-
- inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- warfield: 4/7/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- supermim: 3/9/1990
- carol: 3/6/1990
-
- *RECORD*
- *FIELD* NO
- 103320
- *FIELD* TI
- *103320 AGRIN; AGRN
- *FIELD* TX
- One of the important events in synapse formation is the accumulation of
- neurotransmitter receptors beneath the presynaptic nerve terminal. Agrin
- is a component of the synaptic basal lamina that induces the clustering
- (aggregation) of acetylcholine receptors (e.g., 100690) on cultured
- muscle fibers. Campanelli et al. (1991) showed that when a cDNA encoding
- a putative agrin protein is transfected into cells, the molecule is
- secreted and concentrated on the extracellular surface. Coculture of
- transfected cells with muscle fibers induced formation of receptor
- patches at contact sites. These results demonstrated that expression of
- a single gene encoding agrin confers receptor clustering that is
- restricted to specific sites of contact between the synthesizing cell
- and muscle.
-
- Rupp et al. (1991) isolated cDNAs from a rat embryonic spinal cord
- library using an agrin cDNA clone isolated from electromotor neurons of
- a marine ray. Analysis of a set of clones predicted a protein with 1,940
- amino acids, including 141 cysteine residues. The predicted protein had
- 9 domains homologous to protease inhibitors, a region similar to domain
- III of laminin, and 4 epidermal growth factor repeats. The gene was
- expressed in rat embryonic nervous system and muscle. The protein was
- concentrated at synapses, where it may play a role in development and
- regeneration. Rupp et al. (1992) described alternative RNA splicing in
- mammalian agrin resulting in many extracellular matrix protein isoforms.
-
- Rupp et al. (1992) mapped the human AGRN gene to 1pter-p32 by analysis
- of Chinese hamster/human somatic cell hybrids, including one that
- carried chromosome 1 region p32-qter (which was negative for the human
- signal). The mouse gene was mapped to chromosome 4 by study of Chinese
- hamster/mouse somatic cell hybrids. Thus, this is another example of
- extensive homology of synteny between 1pter-p32 and the distal half of
- mouse chromosome 4. Three neurologic mutants in that region of mouse
- chromosome 4 were pointed to as possible candidate diseases for
- mutations in the Agrn gene.
-
- Data on the structure, expression, and bioactivity of agrin all support
- the notion that it plays a central role in regulating postsynaptic
- differentiation. However, agrin is only one of several agents that can
- cause clustering of acetylcholine receptors in vitro. To test critically
- the 'agrin hypothesis' (McMahan, 1990), Gautam et al. (1996) generated
- knockout mice deficient for agrin and showed that neuromuscular
- differentiation is grossly defective in these mice. Some postsynaptic
- differentiation occurred in the mutant, suggesting the existence of a
- second nerve-derived synaptic organizing signal.
-
- Formation of the neuromuscular junction depends upon reciprocal
- inductive interactions between the developing nerve and muscle,
- resulting in the precise juxtaposition of a differentiated nerve
- terminal with a highly specialized patch on the muscle membrane, termed
- the motor endplate. Agrin is a nerve-derived factor involved in
- induction of the molecular reorganization at the motor endplate. Glass
- et al. (1996) found that mice lacking either agrin or the receptor
- tyrosine kinase they called MuSK (601296) exhibit similar profound
- defects at the neuromuscular junction. DeChiara et al. (1996) showed in
- knockout mice that MuSK is required for formation of the neuromuscular
- junction in vivo.
-
- *FIELD* RF
- 1. Campanelli, J. T.; Hoch, W.; Rupp, F.; Kreiner, T.; Scheller, R.
- H.: Agrin mediates cell contact-induced acetylcholine receptor clustering.
- Cell 67: 909-916, 1991.
-
- 2. DeChiara, T. M.; Bowen, D. C.; Valenzuela, D. M.; Simmons, M. V.;
- Poueymirou, W. T.; Thomas, S.; Kinetz, E.; Compton, D. L.; Rojas,
- E.; Park, J. S.; Smith, C.; DiStefano, P. S.; Glass, D. J.; Burden,
- S. J.; Yancopoulos, G. D.: The receptor tyrosine kinase MuSK is required
- for neuromuscular junction formation in vivo. Cell 85: 501-512,
- 1996.
-
- 3. Gautam, M.; Noakes, P. G.; Moscoso, L.; Rupp, F.; Scheller, R.
- H.; Merlie, J. P.; Sanes, J. R.: Defective neuromuscular synaptogenesis
- in agrin-deficient mutant mice. Cell 85: 525-535, 1996.
-
- 4. Glass, D. J.; Bowen, D. C.; Stitt, T. N.; Radziejewski, C.; Bruno,
- J.; Ryan, T. E.; Gies, D. R.; Shah, S.; Mattsson, K.; Burden, S. J.;
- DiStefano, P. S.; Valenzuela, D. M.; DeChiara, T. M.; Yancopoulos,
- G. D.: Agrin acts via a MuSK receptor complex. Cell 85: 513-523,
- 1996.
-
- 5. McMahan, U. J.: The agrin hypothesis Cold Spring Harb. Symp.
- Quant. Biol. 50: 407-418, 1990.
-
- 6. Rupp, F.; Ozcelik, T.; Linial, M.; Peterson, K.; Francke, U.; Scheller,
- R.: Structure and chromosomal localization of the mammalian agrin
- gene. J. Neurosci. 12: 3535-3544, 1992.
-
- 7. Rupp, F.; Payan, D. G.; Magill-Solc, C.; Cowan, D. M.; Scheller,
- R. H.: Structure and expression of a rat agrin. Neuron 6: 811-823,
- 1991.
-
- *FIELD* CD
- Victor A. McKusick: 12/17/1991
-
- *FIELD* ED
- terry: 06/06/1996
- terry: 6/4/1996
- carol: 3/31/1994
- carol: 12/9/1993
- supermim: 3/16/1992
- carol: 2/17/1992
- carol: 12/17/1991
-
- *RECORD*
- *FIELD* NO
- ^103321
- *FIELD* TI
- ^103321 MOVED TO 128239
- *FIELD* TX
- This entry was incorporated into 128239 on 14 October 1996.
-
- *FIELD* CN
- Mark H. Paalman - edited: 10/14/1996
-
- *FIELD* CD
- Victor A. McKusick: 3/18/1994
- *FIELD* ED
- mark: 10/15/1996
- mark: 10/14/1996
- jason: 6/22/1994
- carol: 3/18/1994
- *RECORD*
- *FIELD* NO
- 103390
- *FIELD* TI
- *103390 AHNAK NUCLEOPROTEIN
- DESMOYOKIN
- *FIELD* TX
- Neuroblastoma represents the most primitive neoplasm originating from
- migratory neural crest cells and apparently arises as a result of
- arrested differentiation. To identify genes whose transcription might be
- repressed during the genesis of neuroblastomas, Shtivelman and Bishop
- (1991) used subtractive cDNA cloning to detect genes expressed in human
- melanomas and pheochromocytomas but not in neuroblastomas. The first of
- these genes identified encoded the cell surface protein CD44 (107269),
- an integral membrane glycoprotein that is the principal receptor for
- hyaluronate on the cell surface. A second gene, originally designated
- PM227, attracted their attention because its expression appeared to be
- coordinated with that of CD44. Shtivelman et al. (1992) reported that
- PM227 encodes a protein whose exceptionally large size of 700 kD caused
- them to rename the gene AHNAK (meaning 'giant' in Hebrew). The amino
- acid sequence of AHNAK suggested secondary structure with a periodicity
- of 2.33 residues per turn. Individual chains could associate to form a
- 7- or 8-stranded barrel. The resulting structure would be a polyionic
- rod with length as great as 1.2 microns. Preliminary evidence indicated
- that the protein resides predominantly within the nucleus, but no
- function had been discerned. The highly conserved repeated elements
- were, for the most part, 128 amino acids long.
-
- AHNAK is thought to be identical to desmoyokin (Hashimoto et al., 1993)
- which was first identified as a 680-kD desmosomal plaque protein in
- bovine muzzle epidermis. Using a panel of somatic cell hybrids and
- Southern blot analysis, Kudoh et al. (1995) mapped the human
- AHNAK/desmoyokin gene to chromosome 11. Fluorescence in situ
- hybridization experiments independently confirmed the chromosomal
- localization and refined it to band 11q12.
-
- *FIELD* RF
- 1. Hashimoto, T.; Amagai, M.; Parry, D. A. D.; Dixon, T. W.; Tsukita,
- S.; Tsukita, S.; Miki, K.; Sakai, K.; Inokuchi, Y.; Kudoh, J.; Shimizu,
- N.; Nishikawa, T.: Desmoyokin, a 680 kDa keratinocyte plasma membrane-associated
- protein, is homologous to the protein encoded by human gene AHNAK.
- J. Cell. Sci. 105: 275-286, 1993.
-
- 2. Kudoh, J.; Wang, Y.; Minoshima, S.; Hashimoto, T.; Amagai, M.;
- Nishikawa, T.; Shtivelman, E.; Bishop, J. M.; Shimizu, N.: Localization
- of the human AHNAK/desmoyokin gene (AHNAK) to chromosome band 11q12
- by somatic cell hybrid analysis and fluorescence in situ hybridization.
- Cytogenet. Cell Genet. 70: 218-220, 1995.
-
- 3. Shtivelman, E.; Bishop, J. M.: Expression of CD44 is repressed
- in neuroblastoma cells. Molec. Cell. Biol. 11: 5446-5453, 1991.
-
- 4. Shtivelman, E.; Cohen, F. E.; Bishop, J. M.: A human gene (AHNAK)
- encoding an unusually large protein with a 1.2-micron polyionic rod
- structure. Proc. Nat. Acad. Sci. 89: 5472-5476, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 7/7/1992
-
- *FIELD* ED
- mark: 10/17/1995
- carol: 7/7/1992
-
- *RECORD*
- *FIELD* NO
- 103400
- *FIELD* TI
- 103400 AINHUM
- *FIELD* TX
- A narrow strip of hardened skin, a constricting ring, forms on the
- little toe at the level of the digitoplantar fold and progresses to
- spontaneous amputation of the digit. Familial occurrence has been noted
- by Maass (1926) and by DaSilva Lima (1880). Simon (1921) reported ainhum
- in father and 2 sons. Ainhum-like constriction bands occur with
- neurogenic acroosteolysis (201300) and with mutilating keratoderma
- (124500, 244850).
-
- *FIELD* SA
- Horwitz and Tunick (1937)
- *FIELD* RF
- 1. DaSilva Lima, J. F.: On ainhum. Arch. Derm. Syph. 6: 367-376,
- 1880.
-
- 2. Horwitz, M. T.; Tunick, I.: Ainhum: report of six cases in New
- York. Arch. Derm. Syph. 36: 1058-1063, 1937.
-
- 3. Maass, E.: Beobachtungen ueber Ainhum. Arch. Schiffs-u. Tropenhygiene 30:
- 32-34, 1926.
-
- 4. Simon, K. M. B.: Ainhum, a family disease. J.A.M.A. 76: 560
- only, 1921.
-
- *FIELD* CS
-
- Limbs:
- Little toe spontaneous amputation
-
- inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 5/13/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 103420
- *FIELD* TI
- *103420 ALACRIMA, CONGENITAL
- ALACRIMIA CONGENITA
- *FIELD* TX
- Mondino and Brown (1976) described a family with 5 persons in 4
- generations showing markedly deficient lacrimation from infancy and
- punctate corneal epithelial erosions. Male-to-male transmission was
- observed. Hypoplasia of the lacrimal glands was suggested by
- pharmacologic tests and histopathology of the lacrimal gland. Alacrima
- occurs in anhidrotic ectodermal dysplasia (305100) and dysautonomia
- (223900) and in association with ocular and adnexal abnormalities.
- Krueger (1954) described brother and sister with ptosis, distichiasis,
- conjunctivitis, keratitis, and alacrimia congenita. The father and
- another brother were said to have defective lacrimation. A nuclear
- defect was postulated for this disorder, which may be distinct from that
- reported by Mondino and Brown (1976). Milunsky et al. (1990) described
- hypoplasia of both lacrimal glands and left nasolacrimal duct atresia in
- association with almost total absence of the parotid glands and marked
- hypofunction of both submandibular glands; see 180920.
-
- *FIELD* RF
- 1. Krueger, K. E.: Angeborenes Fehlen der Traenensekretion in einer
- Familie. Klin. Mbl. Augenheilk. 124: 711-713, 1954.
-
- 2. Milunsky, J. M.; Lee, V. W.; Siegel, B. S.; Milunsky, A.: Agenesis
- or hypoplasia of major salivary and lacrimal glands. Am. J. Med.
- Genet. 37: 371-374, 1990.
-
- 3. Mondino, B. J.; Brown, S. I.: Hereditary congenital alacrima.
- Arch. Ophthal. 94: 1478-1480, 1976.
-
- *FIELD* CS
-
- Eyes:
- Congenital alacrima;
- Punctate corneal erosions;
- Lacrimal gland hypoplasia
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 12/12/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 103470
- *FIELD* TI
- 103470 ALBINISM, OCULAR, WITH SENSORINEURAL DEAFNESS
- *FIELD* TX
- Lewis (1978) found 7 affected males and 5 affected females in 3
- consecutive generations of a Caucasian kindred. As in the X-linked
- Nettleship-Falls form of ocular albinism (300500) and in the autosomal
- recessive O'Donnell variety (203310), the patients showed reduced visual
- acuity, photophobia, nystagmus, translucent irides, strabismus,
- hypermetropic refractive errors, and albinotic fundus with foveal
- hypoplasia. The skin lesions showed macromelanosomes as in X-linked
- ocular albinism. Deafness, which was accompanied by vestibular
- hypofunction, lentigines even in unexposed areas, optic nerve dysplasia,
- and dominant inheritance distinguished this form of ocular albinism. (In
- the LEOPARD syndrome (151100) vestibular function is normal.) Lewis
- (1989) expressed the opinion that the family reported by Bard (1978) as
- an instance of Waardenburg syndrome in fact had this disorder. Lewis
- (1989) had also been told of 2 other small families with the syndrome.
-
- *FIELD* RF
- 1. Bard, L. A.: Heterogeneity in Waardenburg's syndrome: report of
- a family with ocular albinism. Arch. Ophthal. 96: 1193-1198, 1978.
-
- 2. Lewis, R. A.: Ocular albinism and deafness. (Abstract) Am. J.
- Hum. Genet. 30: 57A only, 1978.
-
- 3. Lewis, R. A.: Personal Communication. Houston, Texas 9/1989.
-
- *FIELD* CS
-
- Eyes:
- Reduced visual acuity;
- Photophobia;
- Nystagmus;
- Translucent irides;
- Strabismus;
- Hypermetropia;
- Albinotic fundus;
- Foveal hypoplasia;
- Optic nerve dysplasia
-
- Skin:
- Hypomelanosis;
- Lentigines
-
- Ears:
- Deafness;
- Vestibular hypofunction
-
- Lab:
- Macromelanosomes
-
- Inheritance:
- Autosomal dominant form;
- also X-linked
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- warfield: 4/14/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 2/29/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 103500
- *FIELD* TI
- 103500 ALBINISM-DEAFNESS OF TIETZ
- *FIELD* TX
- Tietz (1963) described 14 persons in 6 generations with albinism and
- complete nerve deafness. The albinism was generalized but did not affect
- the eyes. The irides were blue. Nystagmus and other ocular abnormalities
- were absent. The medial canthi and nasal bridge were normal. The
- eyebrows were almost totally lacking. The albinism in this trait is
- hypopigmentation and not true albinism; the affected individuals tan,
- for example. Reed et al. (1967) thought this might have been merely a
- dominant type of deafness in unusually blond persons. See 156845.0003
- for a description of a mutation in the MITF gene in mother and son with
- a syndrome resembling that reported by Tietz (1963).
-
- *FIELD* RF
- 1. Reed, W. B.; Stone, V. M.; Boder, E.; Ziprkowski, L.: Pigmentary
- disorders in association with congenital deafness. Arch. Derm. 95:
- 176-186, 1967.
-
- 2. Tietz, W.: A syndrome of deaf-mutism associated with albinism
- showing dominant autosomal inheritance. Am. J. Hum. Genet. 15:
- 259-264, 1963.
-
- *FIELD* CS
-
- Skin:
- Generalized hypopigmentaion
-
- Ears:
- Complete nerve deafness
-
- Eyes:
- Normal
-
- Hair:
- Absent eyebrows
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 03/12/1996
- terry: 3/5/1996
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 103580
- *FIELD* TI
- #103580 ALBRIGHT HEREDITARY OSTEODYSTROPHY; AHO
- PSEUDOHYPOPARATHYROIDISM, TYPE IA, INCLUDED; PHP;;
- PHP-IA, INCLUDED
- *FIELD* MN
- Albright hereditary osteodystrophy is characterized by ectopic
- calcification and ossification, rounded facies, 'absent 4th knuckles,'
- and short feet and hands with short metacarpals (particularly the 4th)
- and terminal phalanges (Weinberg and Stone, 1971).
- Pseudohypoparathyroidism type IA (PHP-IA) is caused by a defect in the
- alpha subunit of Gs (Levine et al., 1988). (Gs = stimulatory guanine
- nucleotide-binding protein of adenylate cyclase. PHP-IA = disorder in
- patients with decreased cell membrane Gs activity; PHP-IB = disorder in
- those with normal activity.) The stimulatory and inhibitory G molecules
- are composed of beta and gamma subunits common to the two, and alpha
- units genetically unique to each. The gene for the alpha subunit of Gs
- (GNAS1; 139320) has been mapped to the long arm of chromosome 20. PHP-IB
- is presumably a receptor defect. G unit activity is 50% in PHP-IA and
- 100% in PHP-IB. All cases of PHP-IA and only 15% of cases of PHP-IB have
- Albright hereditary osteodystrophy. All cases of both types have renal
- resistance to PTH, but thyroid resistance to TSH, hepatic resistance to
- glucagon, and gonadal dysfunction, which occur in most cases of PHP-IA,
- are rarely if ever seen in PHP-IB. The alpha subunit of Gs is probably
- encoded by the same gene in most, if not all, endocrine target cells,
- since patients with PHP-IA have reduced responsiveness to many hormones
- that act by stimulating adenylate cyclase (Stryer and Bourne, 1986).
-
- Levine et al. (1986) found reductions in red cell membrane Gs activity
- in cases of pseudopseudohypoparathyroidism comparable to those in
- pseudohypoparathyroidism type IA, but the patients with
- pseudopseudohypoparathyroidism did not have obvious endocrine
- dysfunction. Other factors must determine whether hormone resistance
- occurs with this genetic defect.
-
- In patients whose Gs-protein activity had been determined, 9 of 14
- patients with type IA and none of 11 patients with type IB
- pseudohypoparathyroidism had mental deficiency (Farfel and Friedman,
- 1986). Levine et al. (1988) determined that mRNA levels were
- approximately 50% reduced for the alpha subunit of the G protein in
- affected members of 6 pedigrees studied, whereas they were normal in
- affected members of 2 other pedigrees. Some cases of AHO are due to
- point mutations in the GNAS1 gene.
-
- Published reports of AHO involving 2 or more generations show a marked
- excess of maternal transmission. Furthermore, full expression of the
- gene (AHO plus hormone resistance in the form of
- pseudohypoparathyroidism) occurs in maternally transmitted cases, and
- only partial expression (AHO alone) occurs in paternally transmitted
- cases (Davies and Hughes, 1993). The suggestion that genomic imprinting
- is involved has not been substantiated (Wilson et al., 1994).
-
- *FIELD* ED
- carol: 07/26/1996 marlene: 7/25/1996 joanna: 7/11/1996
-
- *FIELD* CD
- F. Clarke Fraser: 5/9/1996
- *FIELD* TX
- A number sign (#) is used with this entry because the phenotype is known
- to be due to mutation in the GNAS1 gene (139320), located on chromosome
- 20.
-
- See 300800 and 139320. Weinberg and Stone (1971) described a family in
- which a brother and sister and a son and daughter of the brother had
- typical Albright osteodystrophy. The patients were of normal
- intelligence but showed ectopic calcification and ossification, rounded
- facies, 'absent 4th knuckles,' and short feet and hands with
- particularly short 4th metacarpals. (In subsequent studies of this
- family by Levine and Van Dop (1986), Ns (or Gs) was found to be normal.)
- Other families suggesting autosomal dominant inheritance were reviewed.
- In a large number of patients, Farfel et al. (1981) studied erythrocyte
- N-protein, the membrane-bound coupling protein required for stimulation
- of adenylate cyclase by hormones and by guanine nucleotides. (This
- protein was called 'N' by Bourne et al. (1981) and 'G' by Levine et al.
- (1981).) A group of 15 patients with N-protein activity of about 50% of
- normal included a mother and daughter and 2 sisters. The authors
- suggested that both dominant and recessive inheritance exist. They also
- observed families with normal erythrocyte N-protein in which
- pseudohypoparathyroidism and hypothyroidism were inherited as an
- autosomal dominant. Fitch (1982) favored autosomal dominant inheritance.
- She pointed out confusion with myositis ossificans. Short metacarpals
- and short terminal phalanges are typical. Izraeli et al. (1992)
- described a family in which 5 members of 3 successive generations had
- the clinical features of AHO associated with congenital osteoma cutis
- (166350). Zung et al. (1996) pictured subcutaneous nodules of the left
- heel in a 7-year-old girl with AHO. The mother, aged 38 years, had
- multiple subcutaneous masses of the limbs and bilateral shortening of
- the fourth metacarpals. A mammogram showed calcified breast nodules
- which were also palpable.
-
- The possibility of an anomalous parathormone in one form of PHP is
- suggested by observations of Loveridge et al. (1982) using a
- cytochemical bioassay in which plasma or a standard reference
- preparation of parathormone is added to segments of guinea pig kidney
- maintained in organ culture. When exogenous parathormone was added to
- plasma of normal subjects or those with hyperparathyroidism or
- hypoparathyroidism, response was commensurate with the amount added; 50
- to 90% of the exogenous hormone was 'recovered.' When this was done with
- the plasma of 10 PHP patients, recovery ranged from less than 1% up to
- 35%. This seemed to indicate an inhibitor in PHP plasma. Interestingly,
- it was not found in the plasma of a PHP patient who had previously
- undergone parathyroidectomy. Thus, the PHP patient appears to have an
- immunoreactive parathormone which lacks activity on the kidney, acting
- much as do certain synthetic parathyroid-hormone peptides, such as 3-34
- PTH; these bind to renal receptors without stimulating adenylate cyclase
- activity. Levine et al. (1986) found reductions in red cell membrane Gs
- activity in cases of pseudopseudohypoparathyroidism that were comparable
- to those in pseudohypoparathyroidism type IA. (Gs = stimulatory guanine
- nucleotide-binding protein of adenylate cyclase. Synonyms = G/F, G unit,
- and Ns. PHP IA = disorder in patients with decreased cell membrane Gs
- activity; PHP IB = disorder in those with normal activity.) Yet the
- patients with pseudopseudohypoparathyroidism did not have obvious
- endocrine dysfunction. Other factors, as yet undefined, must determine
- whether hormone resistance occurs with this genetic defect. Autosomal
- dominant inheritance is supported by the demonstration of father-to-son
- transmission of decreased Gs activity (Van Dop et al., 1984).
-
- Pseudohypoparathyroidism type IA (PHP-IA) is caused by a defect in the
- alpha subunit of Gs. (The stimulatory and inhibitory G molecules are
- composed of beta and gamma subunits common to the two, and alpha units
- genetically unique to each.) The gene for the alpha subunit of Gs has
- been mapped to chromosome 20 (see 139320). Thus, the quandary of
- autosomal vs X-linked inheritance (see discussion in 300800) was settled
- for this form of pseudohypoparathyroidism. PHP-IB is presumably a
- receptor defect. G unit activity is 50% in PHP-IA and 100% in PHP-IB.
- All cases of PHP-IA and only 15% of cases of PHP-IB have Albright
- hereditary osteodystrophy. All of both types have renal resistance to
- PTH, but thyroid resistance to TSH, hepatic resistance to glucagon, and
- gonadal dysfunction, which occur in most cases of PHP-IA, are rarely if
- ever seen in PHP-IB. Studies in frog neuroepithelium showed that the
- sense of smell is mediated by a G(s)-adenylate cyclase system. Weinstock
- et al. (1986) found that all G(s)-deficient patients (with type IA PHP)
- had impaired olfaction whereas all G(s)-normal PHP patients (type IB)
- had normal olfaction. This suggested that G(s)-deficient patients may be
- resistant or impaired in other cAMP-mediated actions in other
- nonendocrine systems. In type IA pseudohypoparathyroidism (PHP-IA), Gs
- activity, measured by in vitro complementation of the cyc(-) defect, is
- reduced by about 50% in red cells, skin fibroblasts, lymphoblasts, and
- renal cells. These findings are consistent with autosomal dominant
- inheritance (Spiegel et al., 1985). The cyc(-) complementation assay
- measures activity of the alpha subunit of Gs. This subunit is probably
- encoded by the same gene in most, if not all, endocrine target cells,
- since patients with PHP-IA have reduced responsiveness to many hormones
- that act by stimulating adenylate cyclase. Visual excitation is mediated
- by a related G protein, transducin (Stryer and Bourne, 1986); see
- 189970. Mental deficiency occurs in 47 to 75% of patients with
- pseudohypoparathyroidism type I.
-
- Because mutations in the adenylate cyclase-cAMP system may affect the
- learning ability of Drosophila, Farfel and Friedman (1986) assessed
- mental deficiency in 25 patients whose Ns-protein activity had been
- determined. Nine of 14 patients with type IA and none of 11 patients
- with type IB pseudohypoparathyroidism had mental deficiency. Farfel and
- Friedman (1986) concluded that Ns-protein deficiency, reduced cAMP
- levels, or both are involved in the mental deficiency of these patients.
- Levine et al. (1988) presented evidence that patients with type I
- pseudohypoparathyroidism associated with Albright hereditary
- osteodystrophy have deficiency of the alpha subunit of the G protein
- that stimulates adenylyl cyclase, and examined the nature of the
- molecular defect in 8 kindreds. Using a cDNA hybridization probe for
- GNAS (139320), they could show no abnormalities of restriction fragments
- or gene dosage on restriction analysis with several endonucleases. RNA
- blot and dot blot analysis of total RNA from cultured fibroblasts
- obtained from the patients revealed about 50% reduced mRNA levels for
- the alpha subunit of the G protein in affected members of 6 of the
- pedigrees but normal levels in affected members of the other 2
- pedigrees. By contrast, mRNA levels encoding the alpha subunit of the G
- protein that inhibits adenylyl cyclase (139310) were not altered in any.
- Allen et al. (1988) concluded that hypomagnesemia can prevent the
- elevation of parathyroid hormone concentrations in familial
- pseudohypoparathyroidism; the observation indicates that the parathyroid
- gland retains its physiologic response to hypomagnesemia in this
- disorder. Gejman et al. (1990) used a combination of PCR, denaturing
- gradient gel electrophoresis, and direct sequencing to detect a total of
- 5 allelic variants in the GNAS1 gene. Only 2 of these, both in exon 10,
- were present in AHO affected individuals exclusively. One neutral
- polymorphism in exon 5 creates a new FOK1 restriction site which was
- used for linkage mapping of the GNAS1 gene in the CEPH reference
- pedigrees. A maximal lod score of 9.31 was obtained at a theta of 0.042
- with the locus D20S15, previously reported to be on the long arm of
- chromosome 20 (Donis-Keller et al., 1987).
-
- Abnormalities of secretion of thyroid hormone (de Wijn and Steendijk,
- 1982) and gonadotropins (Shapiro et al., 1980) have been described in
- patients with pseudohypoparathyroidism. Stirling et al. (1991) described
- mother and son with deficiency in production of growth hormone-releasing
- factor (GHRH; 139190) in combination with other features characteristic
- of this syndrome.
-
- Davies and Hughes (1993) pointed out that published reports of AHO
- involving 2 or more generations show a marked excess of maternal
- transmission. Furthermore, full expression of the gene (AHO plus hormone
- resistance in the form of pseudohypoparathyroidism) occurs in maternally
- transmitted cases, and only partial expression (AHO alone) occurs in
- paternally transmitted cases. Davies and Hughes (1993) suggested that
- genomic imprinting is involved in the expression of this disorder. The
- region of chromosome 20 occupied by the Gs protein that is mutant in
- this disorder is homologous to an area of mouse chromosome 2 involved in
- both maternal and paternal imprinting. Hall (1990) had suggested that
- AHO may show imprinting by virtue of location in this area. Schuster et
- al. (1994), however, reported findings inconsistent with the imprinting
- hypothesis in a family with AHO and reduced GNAS1 activity reported by
- Schuster et al. (1993). PHP-Ia was inherited paternally as well as
- maternally, suggesting that mechanisms other than genomic imprinting are
- responsible for the full expression of hormone resistance, at least
- within this family. It may be that additional components of signal
- transduction (for example, calmodulin, cAMP phosphodiesterase, or
- protein kinase A) are responsible for the difference between PHP-Ia and
- PPHP. Along the same line, to establish if GNAS1 is indeed imprinted,
- Campbell et al. (1994) examined the parental origin of GNAS1
- transcription in human fetal tissues. Of 75 fetuses genotyped, at
- gestational ages ranging from 6 to 13 weeks, 13 heterozygous for an FokI
- polymorphism in exon 5 of GNAS1 were identified whose mothers were
- homozygous for one or another allele. RNA from up to 10 different
- tissues from each fetus was analyzed by reverse transcriptase-PCR. In
- all cases, expression from both parental alleles was shown by FokI
- digestion of RT-PCR products and quantification of the resulting
- fragments. No tissue-specific pattern of expression was discerned.
- Campbell et al. (1994) concluded that if genomic imprinting regulates
- the expression of the GNAS1 gene, the effect must either be subtle and
- quantitative or be confined to a small subset of specialized
- hormone-responsive cells within the target tissues. Wilson et al. (1994)
- likewise used an intragenic GNAS1 FokI polymorphism to determine the
- parental origin of the gene mutations in sporadic and familial AHO. A
- sporadic case of pseudo-pseudohypoparathyroidism was found to be
- associated with a de novo G-to-A substitution at the exon 5 donor splice
- junction; the mutation was paternally derived.
-
- *FIELD* SA
- Goeminne (1965); Patten et al. (1989); Winter and Hughes (1980)
- *FIELD* RF
- 1. Allen, D. B.; Friedman, A. L.; Greer, F. R.; Chesney, R. W.: Hypomagnesemia
- masking the appearance of elevated parathyroid hormone concentrations
- in familial pseudohypoparathyroidism. Am. J. Med. Genet. 31: 153-158,
- 1988.
-
- 2. Bourne, H. R.; Kaslow, H. R.; Brickman, A. S.; Farfel, Z.: Fibroblast
- defect in pseudohypoparathyroidism, type I: reduced activity of receptor-cyclase
- coupling protein. J. Clin. Endocr. Metab. 53: 636-640, 1981.
-
- 3. Campbell, R.; Gosden, C. M.; Bonthron, D. T.: Parental origin
- of transcription from the human GNAS1 gene. J. Med. Genet. 31: 607-614,
- 1994.
-
- 4. Davies, S. J.; Hughes, H. E.: Imprinting in Albright's hereditary
- osteodystrophy. J. Med. Genet. 30: 101-103, 1993.
-
- 5. de Wijn, E. M.; Steendijk, R.: Growth and development in a girl
- with pseudohypoparathyroidism and hypothyroidism. Acta Paediat. Scand. 71:
- 657-660, 1982.
-
- 6. Donis-Keller, H.; Green, P.; Helms, C.; Cartinhour, S.; Weiffenbach,
- B.; Stephens, K.; Keith, T. P.; Bowden, D. W.; Smith, D. R.; Lander,
- E. S.; et al.: A genetic linkage map of the human genome. Cell 51:
- 319-337, 1987.
-
- 7. Farfel, Z.; Brothers, V. M.; Brickman, A. S.; Conte, F.; Neer,
- R.; Bourne, H. R.: Pseudohypoparathyroidism: inheritance of deficient
- receptor-cyclase coupling activity. Proc. Nat. Acad. Sci. 78: 3098-3102,
- 1981.
-
- 8. Farfel, Z.; Friedman, E.: Mental deficiency in pseudohypoparathyroidism
- type I is associated with Ns-protein deficiency. Ann. Intern. Med. 105:
- 197-199, 1986.
-
- 9. Fitch, N.: Albright's hereditary osteodystrophy: a review. Am.
- J. Med. Genet. 11: 11-29, 1982.
-
- 10. Gejman, P. V.; Weinstein, L. S.; Martinez, M.; Spiegel, A. M.;
- Gershon, E. S.: Genetic mapping of the G(s)-alpha gene and detection
- of mutations in Albright hereditary osteodystrophy (AHO) by using
- polymerase chain reaction (PCR), denaturing gradient gel electrophoresis
- (DGGE) and direct sequencing. (Abstract) Am. J. Hum. Genet. 47 (suppl.):
- A217 only, 1990.
-
- 11. Goeminne, L.: Albright's hereditary poly-osteochondrodystrophy
- (pseudo-pseudo-hypoparathyroidism with diabetes, hypertension, arteritis
- and polyarthrosis). Acta Genet. Med. Gemellol. 14: 226-281, 1965.
-
- 12. Hall, J. G.: Genomic imprinting: review and relevance to human
- diseases. Am. J. Hum. Genet. 46: 857-873, 1990.
-
- 13. Izraeli, S.; Metzker, A.; Horev, G.; Karmi, D.; Merlob, P.; Farfel,
- Z.: Albright hereditary osteodystrophy with hypothyroidism, normocalcemia,
- and normal Gs protein activity: a family presenting with congenital
- osteoma cutis. Am. J. Med. Genet. 43: 764-767, 1992.
-
- 14. Levine, M. A.; Ahn, T. G.; Klupt, S. F.; Kaufman, K. D.; Smallwood,
- P. M.; Bourne, H. R.; Sullivan, K. A.; Van Dop, C.: Genetic deficiency
- of the alpha subunit of the guanine nucleotide-binding protein G(s)
- as the molecular basis for Albright hereditary osteodystrophy. Proc.
- Nat. Acad. Sci. 85: 617-621, 1988.
-
- 15. Levine, M. A.; Downs, R. W., Jr.; Lasker, R. D.; Marx, S. J.;
- Moses, A. M.; Aurbach, G. D.; Spiegel, A. M.: Resistance to multiple
- hormones in patients with pseudohyperparathyroidism and deficient
- guanine nucleotide regulatory protein. (Abstract) Clin. Res. 29:
- 412A only, 1981.
-
- 16. Levine, M. A.; Jap, T.-S.; Mauseth, R. S.; Downs, R. W.; Spiegel,
- A. M.: Activity of the stimulatory guanine nucleotide-binding protein
- is reduced in erythrocytes from patients with pseudohypoparathyroidism
- and pseudopseudohypoparathyroidism: biochemical, endocrine, and genetic
- analysis of Albright's hereditary osteodystrophy in six kindreds. J.
- Clin. Endocr. Metab. 62: 497-502, 1986.
-
- 17. Levine, M. A.; Van Dop, C.: Personal Communication. Baltimore,
- Md. 2/27/1986.
-
- 18. Loveridge, N.; Fischer, J. A.; Nagant de Deuxchaisnes, C.; Dambacher,
- M. A.; Tschopp, F.; Werder, E.; Devogelaer, J.-P.; De Meyer, R.; Bitensky,
- L.; Chayen, J.: Inhibition of cytochemical bioactivity of parathyroid
- hormone by plasma in pseudohypoparathyroidism type I. J. Clin. Endocr.
- Metab. 54: 1274-1275, 1982.
-
- 19. Patten, J. L.; Smallwood, P. M.; Eil, C.; Johns, D. R.; Valle,
- D.; Steel, G.; Levine, M. A.: An initiator codon mutation in the
- gene encoding the alpha subunit of Gs in pseudohypoparathyroidism
- type IA (PHP IA). (Abstract) Am. J. Hum. Genet. 45 (suppl.): A212
- only, 1989.
-
- 20. Schuster, V.; Eschenhagen, T.; Kruse, K.; Gierschik, P.; Kreth,
- H. W.: Endocrine and molecular biological studies in a German family
- with Albright hereditary osteodystrophy. Europ. J. Pediat. 152:
- 185-189, 1993.
-
- 21. Schuster, V.; Kress, W.; Kruse, K.: Paternal and maternal transmission
- of pseudohypoparathyroidism type Ia in a family with Albright hereditary
- osteodystrophy: no evidence of genomic imprinting. (Letter) J. Med.
- Genet. 31: 84-86, 1994.
-
- 22. Shapiro, M. S.; Bernheim, J.; Gutman, A.; Arber, I.; Spitz, I.
- M.: Multiple abnormalities of anterior pituitary hormone secretion
- in association with pseudohypoparathyroidism. J. Clin. Endocr. Metab. 51:
- 483-487, 1980.
-
- 23. Spiegel, A. M.; Gierschik, P.; Levine, M. A.; Downs, R. W., Jr.
- : Clinical implications of guanine nucleotide-binding proteins as
- receptor-effector couplers. New Eng. J. Med. 312: 26-33, 1985.
-
- 24. Stirling, H. F.; Barr, D. G. D.; Kelnar, C. J. H.: Familial growth
- hormone releasing factor deficiency in pseudopseudohypoparathyroidism. Arch.
- Dis. Child. 66: 533-535, 1991.
-
- 25. Stryer, L.; Bourne, H. R.: G proteins: a family of signal transducers. Annu.
- Rev. Cell Biol. 2: 391-419, 1986.
-
- 26. Van Dop, C.; Bourne, H. R.; Neer, R. M.: Father to son transmission
- of decreased N(s) activity in pseudohypoparathyroidism type Ia. J.
- Clin. Endocr. Metab. 59: 825-834, 1984.
-
- 27. Weinberg, A. G.; Stone, R. T.: Autosomal dominant inheritance
- in Albright's hereditary osteodystrophy. J. Pediat. 79: 996-999,
- 1971.
-
- 28. Weinstock, R. S.; Wright, H. N.; Spiegel, A. M.; Levine, M. A.;
- Moses, A. M.: Olfactory dysfunction in humans with deficient guanine
- nucleotide-binding protein. Nature 322: 635-636, 1986.
-
- 29. Wilson, L. C.; Oude Luttikhuis, M. E. M.; Clayton, P. T.; Fraser,
- W. D.; Trembath, R. C.: Parental origin of Gs-alpha gene mutations
- in Albright's hereditary osteodystrophy. J. Med. Genet. 31: 835-839,
- 1994.
-
- 30. Winter, J. S. D.; Hughes, I. A.: Familial pseudohypoparathyroidism
- without somatic anomalies. J. Canad. Med. Assoc. 123: 26-31, 1980.
-
- 31. Zung, A.; Herzenberg, J. E.; Chalew, S. A.: Radiological case
- of the month. Arch. Pediat. Adolesc. Med. 15: 643-644, 1996.
-
- *FIELD* CS
-
- Endocrine:
- Pseudohypoparathyroidism;
- Thyrotropin resistance;
- Gonadotropin resistance;
- Hypothyroidism;
- Deficient prolactin release;
- Partial resistance to antidiuretic hormone;
- Hypertension
-
- Growth:
- Short stature;
- Obesity
-
- Limbs:
- Brachydactyly;
- Short metacarpals
-
- GU:
- Oligomenorrhea
-
- Skin:
- Subcutaneous ossifications
-
- Neuro:
- Mental retardation;
- Hypocalcemic tetany;
- Seizures
-
- HEENT:
- Round face;
- Cataract;
- Calcified choroid plexus
-
- Teeth:
- Delayed tooth eruption;
- Enamel hypoplasia
-
- Lab:
- Hypocalcemia;
- Elevated serum parathyroid hormone (PTH) level;
- Parathyroid hyperplasia;
- Low urinary cyclic AMP response to PTH administration;
- Reduced Gs activity in PHP-IA;
- Normal Gs activity in PHP-IB;
- Decreased N protein activity in some patients with PHP-IA;
- Low serum estrogen;
- High LH and FSH;
- Abnormal parathormone-receptor-adenylate cyclase complex of the renal
- cortical cell plasma membrane;
- Abnormal nucleotide-binding regulatory protein activity
-
- Inheritance:
- Autosomal dominant type (20q);
- also X-linked and autosomal recessive varieties
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 11/27/1996
- terry: 11/12/1996
- terry: 11/4/1996
- carol: 7/26/1996
- carol: 3/2/1995
- davew: 8/18/1994
- terry: 7/18/1994
- mimadm: 4/12/1994
- warfield: 4/7/1994
- pfoster: 3/25/1994
-
- *RECORD*
- *FIELD* NO
- 103581
- *FIELD* TI
- 103581 ALBRIGHT HEREDITARY OSTEODYSTROPHY-2; AHO2
- *FIELD* TX
- Hedeland et al. (1992) described a mother and daughter with classic
- features of pseudohypoparathyroidism type I and Albright hereditary
- osteodystrophy in association with proximal deletion of 15q
- del(15)(q11q13) similar to that seen in Prader-Willi syndrome (176270)
- and Angelman syndrome (105830). Using a series of DNA probes that often
- show deletion or uniparental disomy in the latter 2 conditions, Hedeland
- et al. (1992) found no evidence for either in the mother or the
- daughter. One form of AHO has been demonstrated to be due to point
- mutations in the GNAS1 gene (139320) on chromosome 20. It is possible,
- of course, that there are other forms of AHO that map elsewhere.
-
- *FIELD* RF
- 1. Hedeland, H.; Berntorp, K.; Arheden, K.; Kristoffersson, U.: Pseudohypoparathyroidism
- type I and Albright's hereditary osteodystrophy with a proximal 15q
- chromosomal deletion in mother and daughter. Clin. Genet. 42: 129-134,
- 1992.
-
- *FIELD* CS
-
- Endocrine:
- Pseudohypoparathyroidism;
- Thyrotropin resistance;
- Gonadotropin resistance;
- Hypothyroidism;
- Deficient prolactin release;
- Partial resistance to antidiuretic hormone;
- Hypertension
-
- Growth:
- Short stature;
- Obesity
-
- Limbs:
- Brachydactyly;
- Short metacarpals
-
- GU:
- Oligomenorrhea
-
- Skin:
- Subcutaneous ossifications
-
- Neuro:
- Mental retardation;
- Hypocalcemic tetany;
- Seizures
-
- HEENT:
- Round face;
- Cataract;
- Calcified choroid plexus
-
- Teeth:
- Delayed tooth eruption;
- Enamel hypoplasia
-
- Lab:
- Hypocalcemia;
- Elevated serum parathyroid hormone (PTH) level;
- Parathyroid hyperplasia;
- Low urinary cyclic AMP response to PTH administration;
- Reduced Gs activity in PHP-IA;
- Normal Gs activity in PHP-IB;
- Decreased N protein activity in some patients with PHP-IA;
- Low serum estrogen;
- High LH and FSH;
- Abnormal parathormone-receptor-adenylate cyclase complex of the renal
- cortical cell plasma membrane;
- Abnormal nucleotide-binding regulatory protein activity
-
- Inheritance:
- ? Autosomal dominant type (?15q);
- also X-linked and autosomal recessive varieties
-
- *FIELD* CD
- Victor A. McKusick: 1/21/1993
-
- *FIELD* ED
- mimadm: 3/11/1994
- carol: 1/21/1993
-
- *RECORD*
- *FIELD* NO
- 103600
- *FIELD* TI
- *103600 ALBUMIN; ALB
- DYSALBUMINEMIC HYPERTHYROXINEMIA, INCLUDED;;
- HYPERTHYROXINEMIA, DYSALBUMINEMIC, INCLUDED;;
- ANALBUMINEMIA, INCLUDED;;
- BISALBUMINEMIA, INCLUDED
- *FIELD* MN
- Albumin is a soluble, globular, unglycosylated, monomeric protein of
- molecular weight 65,000, which comprises about one-half of the blood
- serum protein. Albumin functions primarily as a carrier protein for
- steroids, fatty acids, and thyroid hormones and plays a role in
- stabilizing extracellular fluid volume. The human albumin gene is 16,961
- nucleotides long. It is split into 15 exons which are symmetrically
- placed within 3 domains (Minghetti et al., 1986). Mutations in the ALB
- gene, located on chromosome 4q11-q13 (Harper and Dugaiczyk, 1983),
- result in various anomalous proteins (Madison et al., 1994).
- 'Alloalbuminemia' is the term suggested for the variant albumins
- (Blumberg et al., 1968).
-
- Analbuminemia is a rare autosomal recessive disorder in which serum
- albumin is absent (Ruffner and Dugaiczyk, 1988). The homozygotes have
- remarkably little inconvenience attributable to the lack of serum
- albumin. Some fetal hydrops may be caused by analbuminemia. The normal
- levels of albumin in heterozygotes may indicate that the mutation is at
- a regulatory locus independent of the albumin locus.
-
- The serum albumin locus on 4q is presumably the site of the mutation
- responsible for the condition called 'familial dysalbuminemic
- hyperthyroxinemia' (FDH) (Ruiz et al., 1982). Patients, who are
- euthyroid, show elevated serum thyroxine and free-thyroxine index caused
- by an abnormal serum albumin that preferentially binds thyroxine. FDH
- can be subdivided into 3 types, depending on the coexistence of T3 and
- rT3 excess with hyperthyroxinemia (Lalloz et al., 1985). Seemingly, the
- binding of drugs by albumin and the release of thyroid hormone to the
- tissues are not altered in ways that have clinical significance though
- some patients could mistakenly be treated for hyperthyroidism (Yeo et
- al., 1987). Point mutations have been found (Petersen et al., 1994).
-
- *FIELD* ED
- carol: 07/25/1996 marlene: 7/23/1996 joanna: 7/11/1996
-
- *FIELD* CD
- F. Clarke Fraser: 5/9/1996
- *FIELD* TX
-
- DESCRIPTION
-
- Albumin is a soluble, monomeric protein which comprises about one-half
- of the blood serum protein. Albumin functions primarily as a carrier
- protein for steroids, fatty acids, and thyroid hormones and plays a role
- in stabilizing extracellular fluid volume. Mutations in the ALB gene on
- chromosome 4 result in various anomalous proteins.
-
- MAPPING
-
- Weitkamp et al. (1966) concluded that the albumin locus is closely
- linked with the locus for GC type. Using the Naskapi variant, Kaarsalo
- et al. (1967) found close linkage of the albumin and GC loci. Work with
- somatic cell hybrids between human leukocytes and rat hepatoma cells
- suggested that nucleotide phosphorylase and a human serum albumin locus
- may be on the same chromosome (Darlington, 1974); however, these were
- subsequently assigned to chromosomes 14 and 4, respectively.
-
- Harper and Dugaiczyk (1983) mapped the albumin gene to chromosome 4 by
- in situ hybridization. Dextran sulfate was used to enhance labeling, and
- their technique permitted G-banding of the chromosomes with Wright's
- stain on the same preparations used for autoradiography without
- pretreatment. The regional localization (to 4q11-q13) agreed remarkably
- with that arrived at by indirect methods. Kao et al. (1982) assigned the
- albumin locus to chromosome 4 by using a human albumin cDNA probe in
- human/Chinese hamster somatic cell hybrids. The ALB and
- alpha-fetoprotein (AFP; 104150) genes are within 50 kb of each other
- (Urano et al., 1984) and show strong linkage disequilibrium (Murray et
- al., 1984). Magenis et al. (1989) used in situ hybridization to localize
- the ALB and AFP genes to orangutan chromosome 3q11-q15 and gorilla
- chromosome 3q11-q12 which are considered homologous to 4q11-q13.
-
- EVOLUTION
-
- The characteristic 3-domain structure of albumin and alpha-fetoprotein
- has been conserved throughout mammalian evolution. Thus, 35.2% amino
- acid homology is found between bovine serum albumin and murine AFP. Ohno
- (1981) addressed the vexing question of why this conservation occurs
- despite the nonessential nature of serum albumin as indicated by cases
- of analbuminemia. Minghetti et al. (1985) found a high rate of both
- silent substitutions and effective substitutions with amino acid changes
- in serum albumin. Although the rates of effective substitution in amino
- acid changes were not as high in albumin as in alpha-fetoprotein, they
- were still faster than those of either hemoglobin or cytochrome c. This
- high evolutionary change rate for albumin may be consistent with the
- fact that inherited analbuminemia produces surprisingly few symptoms
- despite the virtually complete absence of albumin.
-
- Vitamin D binding protein (GC; 139200) and serum protease inhibitor are
- linked not only in humans, but also in horse, cattle, and sheep in
- mammals, and chicken in avian species. Shibata and Abe (1996) added the
- Japanese quail to the group.
-
- GENE STRUCTURE
-
- Albumin is a globular unglycosylated serum protein of molecular weight
- 65,000. Minghetti et al. (1986) found that the human albumin gene is
- 16,961 nucleotides long from the putative 'cap' site to the first
- poly(A) addition site. It is split into 15 exons which are symmetrically
- placed within the 3 domains that are thought to have arisen by
- triplication of a single primordial domain.
-
- The albumin variant first described by Fraser et al. (1959) in a Welsh
- family was characterized as a dimer by Jamieson and Ganguly (1969). The
- amino acid sequence has been determined in fragments of serum albumin of
- man (Dayhoff, 1972). By 1980, at least 2 dozen electrophoretic variants
- of serum albumin had been reported but only 2 of them had been
- characterized with respect to their primary structure: albumin A (the
- common form) and albumin B (the variant found mainly in Europeans).
-
- GENE FUNCTION
-
- Albumin is synthesized in the liver as preproalbumin, which has an
- N-terminal peptide that is removed before the nascent protein is
- released from the rough endoplasmic reticulum. The product, proalbumin,
- is in turn cleaved in the Golgi vesicles to give the secreted albumin.
-
- Pinkert et al. (1987) used transgenic mice to locate a cis-acting DNA
- element, an enhancer, important for efficient, tissue-specific
- expression of the mouse albumin gene in the adult. Chimeric genes with
- up to 12 kb of mouse albumin 5-prime flanking region fused to a human
- growth hormone 'reporter' gene were tested. Whereas a region located 8.5
- to 10.4 kb upstream of the albumin promoter was essential for high-level
- expression in adult liver, the region between -8.5 and 0.3 kb was
- dispensable.
-
- GENETIC VARIABILITY
-
- - Protein Variations
-
- Fraser et al. (1959) found, on 2-dimensional electrophoresis (paper
- first, followed by starch), an anomalous plasma protein in 6 persons in
- 2 generations of a family. The electrophoretic properties on paper were
- the same in the anomalous albumin and in normal albumin. This
- distinguishes the protein from that in bisalbuminemia, as does the fact
- that the amount of the anomalous protein is much less than that of the
- normal albumin in presumably heterozygous persons. That the same locus
- as that which determines bisalbuminemia is involved here is suggested by
- the finding of Weitkamp et al. (1967) that the Fraser anomalous albumin
- is also linked to the GC locus.
-
- 'Alloalbuminemia' is the term suggested by Blumberg et al. (1968) for
- the variant albumins. Various alloalbuminemias occur relatively
- frequently in various American Indians (Arends et al., 1969). Melartin
- and Blumberg (1966) found an electrophoretic variant of albumin in high
- frequency in Naskapi Indians of Quebec and in lower frequency in other
- North American Indians. Homozygotes were found.
-
- Weitkamp et al. (1967), using 2 electrophoretic systems, compared the
- serum albumin variants of 19 unrelated families. Five distinct classes
- were found. One class of variants was found only in North American
- Indians. The others were found only in persons of European descent.
-
- In Punjab, North India, Kaur et al. (1982) found, by electrophoresis, 4
- cases of alloalbuminemia among 550 persons. Two appeared to be new
- variants. Another was albumin Naskapi. Since this variant has been found
- also in North American Indians and Eti Turks, the authors suggested that
- albumin Naskapi existed in a common ancestral population before the
- migrations eastward and westward.
-
- In describing a new human albumin variant, albumin Carlisle, Hutchinson
- et al. (1986) stated that more than 80 genetically inherited variants of
- human albumin were known. Fine et al. (1987) found a frequency of
- alloalbuminemia in the French population of 0.0004. There was a high
- occurrence of albumin B and of 2 proalbumin variants, Christchurch and
- Lille.
-
- - Bisalbuminemia
-
- Bisalbuminemia is an asymptomatic variation in serum albumin.
- Heterozygotes have 2 species of albumin, a normal type and one which
- migrates abnormally rapidly or slowly on electrophoresis. Acrocyanosis
- was present in 2 and probably 3 successive generations of the family
- reported by Williams and Martin (1960) but 4 other bisalbuminemic
- persons did not show acrocyanosis.
-
- Tarnoky and Lestas (1964) described a 'new' type of bisalbuminemia in 2
- sibs and the son of one of them. The usual type was demonstrable by
- filter paper electrophoresis. The new type was demonstrable by
- electrophoresis on cellulose acetate at pH 8.6, but not on filter paper
- or starch gel. The term 'paralbuminemia' was suggested by Earle et al.
- (1959) as preferable to 'bisalbuminemia' which is perhaps appropriate
- for the heterozygous state only.
-
- A phenocopy of hereditary bisalbuminemia, acquired bisalbuminemia,
- occurs with overdose of beta-lactam antibiotics (Arvan et al., 1968) and
- with pancreatic pseudocyst associated with pleural or ascitic effusion
- (Shashaty and Atamer, 1972). The anomalous albumin is anodal to the
- normal albumin in its electrophoretic mobility. Vaysse et al. (1981)
- described acquired trisalbuminemia in a patient with familial
- bisalbuminemia and pancreatic pseudocyst.
-
- - Proalbumin
-
- Rochu and Fine (1986) described a new method for identifying genetic
- variants of human proalbumin. Two genetic variants of proalbumin,
- proalbumin Christchurch (Brennan and Carrell, 1978) and proalbumin Lille
- (Abdo et al., 1981), have been shown to result from a substitution at 1
- of the 2 arginyl residues at the dibasic site at which the normal
- propeptide is cleaved. Both of these mutations prevent excision of this
- basic propeptide, and thus each of these proalbumin variants has a
- slower electrophoretic mobility than that of normal albumin. Two genetic
- variants, previously described as albumin Gainesville and albumin
- Pollibauer, were shown to be identical with proalbumin Christchurch
- (Fine et al., 1983) and proalbumin Lille (Galliano et al., 1984),
- respectively.
-
- Arai et al. (1989) found that the 2 types of proalbumins most common in
- Europe (Lille type, arginine-to-histidine at position -2; Christchurch
- type, arginine-to-glutamic acid at position -1) also occur in Japan. The
- clustering of these and of several other amino acid exchanges in certain
- regions of the albumin molecule, arising as independent mutations,
- suggests that certain sites are hypermutable and/or that mutants
- involving certain sites are more subject to selection than mutants
- involving others. In a study of 15,581 unrelated children in Hiroshima
- and Nagasaki, Arai et al. (1989) found 5 rare albumin variants and
- determined the single amino acid substitution in each. All of these were
- inherited and therefore unrelated to parental exposure at the time of
- the bombing. The 5 substitutions were: Nag-1, asp269-to-gly; Nag-2,
- asp375-to-asn; Nag-3, his3-to-gln; Hir-1, glu354-to-lys; and Hir-2,
- glu382-to-lys. Two of the substitutions (Nag-1 and Nag-2) had previously
- been reported (Takahashi et al., 1987). No instances of proalbumin
- variants or of albumin B (glu570-to-lys), which are the most common
- Caucasian alloalbumins, were found in the Hiroshima-Nagasaki study. Arai
- et al. (1989) found 2 instances of albumin B and 1 example of a variant
- proalbumin in Japanese from the vicinity of Tokyo. In a review of all
- reported mutations, Arai et al. (1989) noted that 7 independent
- substitution sites have been identified in the alloalbumins of diverse
- populations in a sequence of only 29 amino acids as compared to a total
- of 5 sites (excluding proalbumin variants) reported thus far for the
- first 353 amino acids. Such a cluster of substitutions may reflect
- vulnerability of the albumin gene to mutation in this region or the ease
- of accommodation to structural changes in the affected area of the
- protein. Arai et al. (1990) studied the albumin genetic variants that
- have been reported in Asian populations and listed a total of 26 point
- substitutions in diverse ethnic groups.
-
- In the family reported by Laurell and Nilehn (1966), a 'new' type of
- paralbuminemia was associated with connective tissue disorders,
- including systemic lupus erythematosus, ruptured knee meniscus,
- recurrent dislocation of shoulder, and back pain. The albumin variant
- was characterized by a broad band in agarose gel electrophoresis that
- indicated the presence of a slow component. A family study showed that
- the anomalous albumin was present in 9 of 23 members representing 3
- generations. Noticing a similarity of the electrophoretic pattern to
- that of an albumin with an arg(-2)-to-cys mutation which they described,
- Brennan et al. (1990) obtained plasma from 1 of the original subjects of
- Laurell and Nilehn (1966) and demonstrated that it indeed showed the
- same mutation that they had found in proalbumin Malmo (103600.0030).
- This anomalous albumin occurs in about 1 per 1,000 persons in Sweden.
-
- - Analbuminemia
-
- Analbuminemia, a rare autosomal recessive disorder in which serum
- albumin is absent, was first reported by Bennhold et al. (1954) of
- Tubingen. See review by Ott (1962). In some reported families
- analbuminemia is a completely recessive condition; serum albumin has a
- normal level in heterozygotes. The homozygotes have remarkably little
- inconvenience attributable to the lack of serum albumin. In the kindreds
- of Bennhold et al. (1954) and Boman et al. (1976), heterozygotes showed
- intermediate levels of serum albumin.
-
- Kallee (1996) reported 2 sibs with analbuminemia who were followed for
- 38 years. The female patient received replacement therapy with human
- serum albumin. Extreme lipodystrophy developed in this patient by the
- fourth decade of life. She had juvenile osteoporosis, which normalized
- under albumin replacement. She died from a granulosa cell cancer at age
- 69. Her brother never received albumin. He suffered from severe
- osteoporosis with gibbus formation, and died from a colon carcinoma at
- age 59. Both sibs had chronic insufficiency of the crural veins, with
- chronic ulcerations of both lower legs but no varicosities of the upper
- thighs. Despite high cholesterol values and high levels of several blood
- clotting factors, neither of the patients had severe atherosclerosis or
- thrombotic events. Kallee (1996) concluded that although patients often
- fail to exhibit serious clinical signs apart from pathologic laboratory
- findings, analbuminemia can no longer be regarded as a harmless anomaly.
-
- Boman et al. (1976) presented data consistent with linkage of the
- analbuminemia locus and the Gc locus. Cormode et al. (1975) found very
- low plasma tryptophan in a neonate with analbuminemia who was small for
- gestational age. Murray et al. (1983) restudied the family reported by
- Boman et al. (1976). The proposita showed trace amounts of
- immunologically normal serum albumin. With cDNA probes for the albumin
- gene, no deletion could be detected. They demonstrated DNA polymorphism
- of the albumin gene. In a review, Ruffner and Dugaiczyk (1988) stated
- that of 22 reported analbuminemic individuals, 8 were known to be from
- consanguineous matings. Dugaiczyk (1989) suggested that some fetal
- hydrops may be caused by analbuminemia. The main causes of hydrops
- fetalis are thalassemia and fetomaternal incompatibility; instances in
- which neither of these can be demonstrated should be investigated for an
- albumin defect.
-
- Analbuminemic rats, like analbuminemic humans, are healthy (Nagase et
- al., 1979). The use of cDNA probes failed to detect serum albumin gene
- transcripts in liver of these analbuminemic rats (Esumi et al., 1980).
- Thus, the disorder in the rat and perhaps the human may be the result of
- gene deletion. On the other hand, the normal levels of albumin in
- heterozygotes may indicate that the mutation is at a regulatory locus
- independent of the albumin locus. In the analbuminemic rat, Esumi et al.
- (1982) found albumin mRNA precursors in nuclei although such were
- missing from the cytoplasm. From this they concluded that analbuminemia
- in rats is caused by a unique type of mutation that affects albumin mRNA
- maturation. In analbuminemia of the rat, Esumi et al. (1983)
- demonstrated that a 7-bp deletion in an intron interferes with mRNA
- formation. Shalaby and Shafritz (1990) showed that exon H is skipped in
- the Nagase analbuminemic rat as a result of the 7-bp deletion at the
- splice donor site of intron H-I. Mendel et al. (1989) could find no
- abnormality of thyroxine transport and distribution in Nagase
- analbuminemic rats. Murray et al. (1983) found a frequency of DNA
- polymorphism in the ALB gene comparable to that in the globin system. No
- gross structural rearrangement was found in a case of human
- analbuminemia.
-
- - Familial Dysalbuminemic Hyperthyroxinemia
-
- The serum albumin locus on 4q is presumably the site of the mutation
- responsible for the condition called by Ruiz et al. (1982) 'familial
- dysalbuminemic hyperthyroxinemia.' Ruiz et al. (1982) studied 15
- euthyroid patients from 8 families who showed elevated serum thyroxine
- and free-thyroxine index, both due to an abnormal serum albumin that
- preferentially binds thyroxine. Since there are several different
- changes in the albumin molecule that can lead to increased binding of
- thyroxine, several types might be expected. Lalloz et al. (1985)
- subdivided FDH into 3 types, depending on the coexistence of T3 and rT3
- excess with hyperthyroxinemia. Seemingly, the binding of drugs by
- albumin and the release of thyroid hormone to the tissues are not
- altered in ways that have clinical significance. DeCosimo et al. (1987)
- presented evidence indicating that familial dysalbuminemic
- hyperthyroxinemia is unusually frequent in Hispanics of Puerto Rican
- origin. Yeo et al. (1987) reported the largest kindred with familial
- dysalbuminemic hyperthyroxinemia thus far reported. Two of the patients
- had mistakenly been treated for hyperthyroidism. Two women with the
- disorder were receiving oral contraceptives, which produced an increase
- in serum thyroxine-binding globulin (314200). Yeo et al. (1987) pointed
- out that the coexistence of acquired high TBG or significant thyroid
- malfunction may confound the diagnosis of dysalbuminemic
- hyperthyroxinemia. Yabu et al. (1987) described a form of variant
- albumin with a markedly enhanced binding activity for
- L-3,5,3-prime-triiodothyronine (T3), a somewhat increased activity for
- thyroxine (T4), and a normal activity for
- 3,3-prime,5-prime-triiodothyronine (rT3). The presence of the variant
- albumin was recognized in a patient with Graves disease after successful
- subtotal thyroidectomy. The findings could be misdiagnosed as T3
- toxicosis or peripheral resistance to thyroid hormones. Premachandra et
- al. (1988) commented that in patients with familial dysalbuminemic
- hyperthyroxinemia, treatment of hypothyroidism with thyroxine has
- special considerations because of binding of the drug to the atypical
- albumin, and raised the possibility that other forms of drug therapy may
- require custom tailoring. It appears that the molecular change in the
- ALD gene responsible for familial dysalbuminemic hyperthyroxinemia has
- not been determined in any instance (Putnam, 1993).
-
- The ALB gene shows much DNA polymorphism. Except for chain terminations
- in 2 Italian variants, all of the albumin mutations determined to that
- time had been single base changes, with a preponderance of transitions
- and purine mutations.
-
- - Mutation Information
-
- Takahashi et al. (1987) identified the amino acid substitutions in 3
- different types of proalbumins designated Gainesville, Taipei, and
- Takefu. The first 2 proalbumins were found to be identical to previously
- described proalbumins, Christchurch and Lille types, respectively. All
- of the variant proalbumins contain a basic propeptide that is not
- removed during posttranscriptional processing because of a mutation in
- the site of excision, an arg-arg sequence. Takefu resists tryptic
- cleavage because of the substitution of proline for arginine at the -1
- position. The substitution of glutamine for histidine at position 3 in
- the variant albumin Nagasaki-3 decreases metal-binding affinity;
- mutations farther down the polypeptide chain do not affect metal-binding
- affinity, nor is there any reduction of copper-binding affinity in
- albumin from patients with Wilson disease (277900). The variant
- proalbumins show a characteristically lowered metal-binding affinity.
-
- Takahashi et al. (1987) reported the amino acid substitution in 4
- albumin variants detected by 1-dimensional electrophoresis in population
- surveys involving tribal Amerindians and Japanese children. Albumin
- Maku, discovered in a Maku Indian woman living among the Yanomama,
- showed a substitution of glutamine for lysine at position 541. Albumin
- Yanomama-2 appears to represent a true private polymorphism, i.e., it is
- the product of an apparently unique allele within a single tribe that
- has a frequency well above the 1% allele minimum for a polymorphism. It
- has been found only in Yanomama Indians, was present in 491 of 3,504
- persons studied, and had the highest frequency of any polypeptide
- variant identified in 21 South American Indian tribes. It was found to
- have a substitution of glycine for arginine at position 114. This
- appears to represent a change from codon CGA to GGA. Albumin Nagasaki-2
- showed a substitution of asparagine for aspartic acid at position 375,
- corresponding to a single base change in codon GAT to AAT. Albumin
- Nagasaki-3 was found to have substitution of glutamine for histidine at
- position 3, corresponding with a 1-base change in the codon CAC to
- either CAA or CAG.
-
- Takahashi et al. (1987) pointed out that about one-half of the known
- mutations in the coding sections in the large albumin gene border an
- exonic junction, raising the possibility that hypermutable 'hot spots'
- may be clustered there. In Japan, surveys showed that hemoglobin and
- albumin variants were of roughly equal frequency and neither protein
- appeared exceptionally variable. Since albumin is a much larger protein,
- one might expect more genetic variability than in hemoglobin. This might
- suggest that selection is relatively active against variants of this
- molecule; yet total absence of this protein (analbuminemia) is
- consistent with apparently satisfactory health.
-
- Takahashi et al. (1987) tabulated the 13 amino acid substitutions
- identified at that time and pointed out that they are unequally
- distributed throughout the polypeptide chain. The slower delineation of
- the nature of point mutations in albumin variants as compared to
- hemoglobin variants can be attributed to 2 primary factors: first,
- alloalbumins are not associated with disease or a significant effect on
- physiologic function, and most are rare; second, the albumin molecule
- consists of a single polypeptide chain with 585 amino acids and 17
- disulfide bridges, a circumstance that magnifies the difficulty of
- determining the presence of a single substitution.
-
- Madison et al. (1994) provided a tabulation of the molecular changes in
- albumin variants.
-
- *FIELD* AV
- .0001
- ALBUMIN FUKUOKA-2
- ALBUMIN TAIPEI
- ALBUMIN LILLE
- ALBUMIN VARESE
- ALB, ARG-2HIS
- Substitution of histidine for arginine at position -2 was found in
- albumin Fukuoka-2 by Arai et al. (1989), in albumin Taipei by Takahashi
- et al. (1987), in albumin Lille by Abdo et al. (1981) and Galliano et
- al. (1988), and in albumin Varese by Galliano et al. (1990). A
- CGT-to-CAT change is responsible for the substitution.
-
- .0002
- ALBUMIN HONOLULU-2
- PROALBUMIN CHRISTCHURCH
- PROALBUMIN GAINESVILLE
- PROALBUMIN FUKUOKA-3
- ALB, ARG-1GLN
- This albumin has an arg(-1)-to-gln change in the preproprotein (Arai et
- al., 1990; Brennan and Carrell, 1978). Brennan and Carrell (1978) found
- a family with a circulating variant of proalbumin in members of 4
- generations. No clinical abnormality was discernible in any of them. The
- variant represents 50% of total albumin and shows an additional
- N-terminal sequence, arg-gly-val-phe-arg-gln. Called 'proalbumin
- Christchurch,' the variant appears to have a mutation of arginine to
- glutamine at the last amino acid of this sequence. Thus, 2 basic amino
- acids must be necessary for cleavage of proalbumin in the Golgi
- vesicles. Copper binding is expected to be absent in the variant albumin
- because of blocking of the high affinity binding site. This is a
- situation comparable to Ehlers-Danlos syndrome type VII-A (130060) in
- which an amino acid substitution at the site of cleavage of procollagen
- results in persistence of procollagen and, in that case, clinically
- important abnormalities in collagen fiber formation.
-
- .0003
- ALBUMIN HONOLULU-1
- PROALBUMIN TAKEFU
- ALB, ARG-1PRO
- Substitution of proline for arginine at position -1 (Takahashi et al.,
- 1987).
-
- .0004
- ALBUMIN BREMEN
- ALBUMIN BLENHEIM
- ALBUMIN IOWA CITY-2
- ALB, ASP1VAL
- See Arai et al. (1990) and Brennan et al. (1990). Brennan et al. (1990)
- suggested that hypermutability of 2 CpG dinucleotides in the codons for
- the diarginyl sequence may account for the frequency of mutations in the
- propeptide. Madison et al. (1991) showed that this mutation is caused by
- a GAT-to-GTT change.
-
- .0005
- ALBUMIN NAGASAKI-3
- ALB, HIS3GLN
- See Takahashi et al. (1987).
-
- .0006
- ALBUMIN YANOMAMA-2
- ALB, ARG114GLY
- See Takahashi et al. (1987).
-
- .0007
- ALBUMIN NAGOYA
- ALB, GLU119LYS
- See Arai et al. (1990).
-
- .0008
- ALBUMIN NAGASAKI-1
- ALBUMIN NIIGATA
- ALB, ASP269GLY
- See Arai et al. (1989).
-
- .0009
- ALBUMIN NEW GUINEA
- ALBUMIN TAGLIACOZZO
- ALBUMIN COOPERSTOWN
- ALB, LYS313ASN
- Huss et al. (1988) described an electrophoretically fast alloalbumin in
- a family in New York State and called it albumin Cooperstown. It was
- found to have a substitution of asparagine for lysine at residue 313 and
- was shown to be the same as albumins found in Italy and in New Zealand.
- A change from AAG to AAY is responsible for the substitution; Y = either
- T or C. Galliano et al. (1990) found this albumin variant in 49
- individuals in the Abruzzo region of Italy.
-
- .0010
- ALBUMIN REDHILL
- ALB, ALA320THR AND ARG-2CYS
- Brennan et al. (1990) characterized albumin Redhill, an albumin variant
- that does not bind nickel and has a molecular mass 2.5 kD higher than
- normal albumin. Its inability to bind nickel was explained by the
- finding of an additional residue of arginine at position -1 of the
- mature protein, but this did not explain the molecular basis of the
- increase in apparent molecular mass. Further studies showed an
- ala320-to-thr change, which introduced an asn-tyr-thr oligosaccharide
- attachment sequence centered at asn318 and explained the increase in
- molecular mass. DNA sequencing of PCR-amplified genomic DNA encoding the
- prepro sequence of albumin indicated an additional mutation at position
- -2 from arg to cys. Brennan et al. (1990) proposed that the new
- phe-cys-arg sequence (replacing -phe-arg-arg-) in the propeptide serves
- as an aberrant signal peptidase cleavage site and that the signal
- peptidase cleaves the propeptide of albumin Redhill in the lumen of the
- endoplasmic reticulum before it reaches the Golgi vesicles, which is the
- site of the diarginyl-specific proalbumin convertase. Thus, albumin
- Redhill is longer than normal by 1 amino acid at its NH2-terminus. The
- ARG-2CYS mutation is the basis of proalbumin Malmo (103600.0030), a
- relatively frequent variant.
-
- .0011
- ALBUMIN ROMA
- ALB, GLU321LYS
- Galliano et al. (1988) demonstrated that albumin Roma has a substitution
- of lysine for glutamic acid at position 321. A GAG-to-AAG change is
- responsible for the substitution. Galliano et al. (1990) found this
- albumin variant in 25 individuals from various parts of Italy.
-
- .0012
- ALBUMIN HIROSHIMA-1
- ALB, GLU354LYS
- See Arai et al. (1989).
-
- .0013
- ALBUMIN PORTO ALEGRE-1
- ALBUMIN COARI 1
- ALB, GLU358LYS
- Arai et al. (1989) reported on amino acid substitutions in albumin
- variants found in Brazil. A previously unreported amino acid
- substitution was found in albumins Coari I and Porto Alegre I
- (glu358-to-lys).
-
- .0014
- ALBUMIN PARKLANDS
- ALB, ASP365HIS
- See Brennan (1985).
-
- .0015
- ALBUMIN MERSIN
- ALBUMIN NASKAPI
- ALBUMIN MEXICO-1
- ALB, LYS372GLU
- Franklin et al. (1980) demonstrated apparent identity between the
- polymorphic albumin variants Naskapi, found chiefly in the Naskapi
- Indians of Quebec, and Mersin, found in the Eti Turks of southeastern
- Turkey. They suggested that these were derived from the same mutation
- occurring in Asia and spreading with the progenitors of the American
- Indians to the North American continent and with Asiatic invaders to
- Asia Minor. Takahashi et al. (1987) found that lysine-372 of normal
- (common) albumin A was changed to glutamic acid both in albumin Naskapi
- and in albumin Mersin. Identity of these albumins may have originated
- through descent from a common mid-Asiatic founder of the 2 migrating
- ethnic groups, or it may represent identical but independent mutations
- of the albumin gene.
-
- .0016
- ALBUMIN NAGASAKI-2
- ALB, ASP375ASN
- See Takahashi et al. (1987) and Arai et al. (1989).
-
- .0017
- ALBUMIN TOCHIGI
- ALB, GLU376LYS
- See Arai et al. (1989).
-
- .0018
- ALBUMIN HIROSHIMA-2
- ALB, GLU382LYS
- See Arai et al. (1989).
-
- .0019
- ALBUMIN LAMBADI
- ALBUMIN MANAUS-1
- ALBUMIN VANCOUVER
- ALBUMIN BIRMINGHAM
- ALBUMIN ADANA
- ALBUMIN PORTO ALEGRE-2
- ALB, GLU501LYS
- Franklin et al. (1980) found a new variant in Eti Turks, which they
- termed albumin Adana. By improved methods, Huss et al. (1988) identified
- a substitution of lysine for glutamic acid at position 501 in albumins
- Vancouver and Birmingham, both from families that migrated from northern
- India, and also in albumin Adana from Turkey. This is the first
- substitution reported in an alloalbumin originating from the Indian
- subcontinent. Albumin Porto Alegre II also contains a glutamic
- acid-to-lysine substitution at position 501.
-
- .0020
- ALBUMIN MAKU
- ALBUMIN ORIXIMINA-1
- ALB, LYS541GLU
- See Takahashi et al. (1987). The substitution in albumin Oriximina I is
- the same as that found in albumin Maku (lysine to glutamic acid at
- position 541) (Arai et al., 1989).
-
- .0021
- ALBUMIN MEXICO-2
- ALB, ASP550GLY
- Franklin et al. (1980) showed that albumin Mexico is in fact 2 separate,
- electrophoretically similar variants and that albumin Mexico-2 contains
- a substitution of glycine for aspartic acid at position 550.
- Substitution of aspartic acid-550 by glycine was found in albumin
- Mexico-2 from 4 persons of the Pima tribe (Takahashi et al., 1987).
-
- .0022
- ALBUMIN FUKUOKA-1
- ALB, ASP563ASN
- See Arai et al. (1990).
-
- .0023
- ALBUMIN OSAKA-1
- ALB, GLU565LYS
- See Arai et al. (1990).
-
- .0024
- ALBUMIN OSAKA-2
- ALBUMIN PHNOM PENH
- ALBUMIN B
- ALBUMIN OLIPHANT
- ALBUMIN NAGANO
- ALBUMIN VERONA B
- ALB, GLU570LYS
- Arai et al. (1989) identified the amino acid substitution characteristic
- of albumin B (glutamic acid-to-lysine at position 570) in alloalbumins
- from 6 unrelated persons of 5 different European descents and also in 2
- Japanese and 1 Cambodian. A GAG-to-AAG change is responsible for this
- substitution. Galliano et al. (1990) found this variant in 103
- individuals in the Veneto area of Italy.
-
- .0025
- ALBUMIN GHENT
- ALBUMIN MILANO FAST
- ALB, LYS573GLU
- An AAA-to-GAA change is responsible for this substitution. Galliano et
- al. (1990) found this variant in 80 individuals from the Lombardy area
- of Italy. Homozygotes have been identified.
-
- .0026
- ALBUMIN VANVES
- ALB, LYS574ASN
- See Galliano et al. (1988).
-
- .0027
- ANALBUMINEMIA, AMERICAN INDIAN TYPE
- ALB, IVS6, A-G, -2
- Ruffner and Dugaiczyk (1988) identified a structural defect in the serum
- albumin gene of an analbuminemic American Indian girl. Sequence
- determination of 1.1 kb of the 5-prime regulatory region and of 6 kb
- across exonic regions revealed a single AG-to-GG mutation within the
- 3-prime splice site of intron 6 in the defective gene of the
- analbuminemic person. In an in vitro assay on the RNA transcript, this
- mutation caused a defect in out-splicing of the intron 6 sequence and in
- the subsequent ligation of the exon 6/exon 7 sequences. Using
- polymerase-amplified genomic DNA and allele-specific
- oligodeoxynucleotide probes, Ruffner and Dugaiczyk (1988) also showed
- that the sequence of this intron 6/exon 7 splice junction was normal in
- a different, unrelated analbuminemic person.
-
- .0028
- ALBUMIN VENEZIA
- ALB, EX14DEL
- Minchiotti et al. (1989) described the molecular defect of an
- electrophoretically fast alloalbumin named Venezia, found in about 90
- seemingly unrelated families in Italy, mainly in the Veneto region. In
- heterozygous subjects the total albumin content was in the normal range,
- with the variant accounting for about 30% of the total protein. Reduced
- stability of the mutant was thought to account for the
- lower-than-expected percentage. Minchiotti et al. (1989) found that
- albumin Venezia possesses a shortened polypeptide chain, 578 residues
- instead of 585, completely variant from residue 572 to the
- COOH-terminus: 572 pro-thr-met-arg-ile-arg-578 glu. This extensive
- modification can be accounted for by deletion of exon 14 and translation
- to the first terminator codon of exon 15, which normally does not code
- for protein. The absence of a basic COOH-terminal dipeptide in the
- mature molecule can be explained by the probable action of serum
- carboxypeptidase N. The low serum level of the variant in heterozygous
- subjects suggests that the carboxy-terminus of the molecule is critical
- for albumin stability. Galliano et al. (1990) found this variant in 105
- individuals, particularly in the region of Veneto in Italy.
-
- .0029
- ALBUMIN CASTEL DI SANGRO
- ALB, LYS536GLU
- An AAG-to-GAG change is responsible for this substitution. Galliano et
- al. (1990) found this variant in 1 individual in Italy.
-
- .0030
- PROALBUMIN MALMO
- PROALBUMIN TRADATE
- ALB, ARG-2CYS
- In a collaborative effort involving laboratories at Malmo, Sweden;
- Bloomington, Indiana; Christchurch, New Zealand; Saitama, Japan; and
- Pavia, Italy, Brennan et al. (1990) studied the most common Swedish
- albumin variant, which is expressed in plasma as a broadened
- electrophoretic band indicative of a slow component at pH 8.6. Present
- in about 1 per 1,000 persons in Sweden, it was also found in a family of
- Scottish descent from Kaikoura, New Zealand, and in 5 families in
- Tradate, Italy. The major variant component was found to be
- arginyl-albumin, in which arginine at the -1 position of the propeptide
- is still attached to the processed albumin. A minor component with the
- amino-terminal sequence of proalbumin was also present as 3 to 6% of the
- total albumin. The mutation was found to involve a change of arginine to
- cysteine at the -2 position. (In albumin Redhill (103600.0010), the
- Malmo mutation is combined with another.) A CGT-to-TGT change is
- responsible for the substitution.
-
- .0031
- PROALBUMIN JAFFNA
- ALB, ARG-1LEU
- In 2 members of a Tamil family from Jaffna (northern Sri Lanka),
- Galliano et al. (1989) found an electrophoretically slow-moving variant
- of serum albumin. Sequence analysis demonstrated that the variant is an
- abnormal proalbumin arising from a substitution of leucine for arginine
- at position -1, which prevents the proteolytic removal of the N-terminal
- hexapeptide and allows the mutated proalbumin to enter the circulation.
-
- .0032
- ALBUMIN Ge/Ct
- ALBUMIN CATANIA
- ALB, GLN580LYS
- This was the fourth albumin variant to be characterized structurally.
- Galliano et al. (1986) found a shortened chain with deletion of a
- cytosine in codon 580, causing frameshift and termination after amino
- acid 582. The COOH-terminal sequence is leu-val-ala-ala-ser-lys-leu-pro.
- Galliano et al. (1990) found this mutation in 62 individuals in Sicily.
-
- .0033
- ALBUMIN TORINO
- ALB, GLU60LYS
- Galliano et al. (1990) found a substitution of lysine for glutamic acid
- at position 60 resulting from a GAA-to-AAA change in a single Italian
- patient.
-
- .0034
- ALBUMIN VIBO VALENTIA
- ALB, GLU82LYS
- In 2 Italian individuals Galliano et al. (1990) found a GAA-to-AAA
- change in codon 82 leading to substitution of lysine for glutamic acid.
-
- .0035
- ALBUMIN CASEBROOK
- ALB, ASP494ASN
- In albumin Casebrook, an electrophoretically slow albumin variant with a
- relative molecular mass of 2.5 kD higher than normal albumin, Peach and
- Brennan (1991) identified substitution of asparagine for aspartic
- acid-494. The mutation introduced an asn-glu-thr N-linked
- oligosaccharide attachment sequence centered on asn494, which explained
- the increase in molecular mass. The mutant albumin was associated with
- no apparent pathology and was detected in 2 unrelated individuals of
- Anglo-Saxon descent.
-
- .0036
- ALBUMIN IOWA CITY-1
- ALB, ASP365VAL
- In a survey of alloalbumins in patients at 2 major medical centers in
- the United States and nearly 20,000 blood donors in Japan, Madison et
- al. (1991) identified 2 previously unreported alloalbumin types. In one
- type, found in a Caucasian family and designated Iowa City-1, aspartic
- acid at position 365 was replaced by valine. This was the second
- reported mutation at position 365; see albumin Parklands (103600.0014).
- The codon change was GAT-to-GTT. In the second type, found in a Japanese
- blood donor, histidine-128 was replaced by arginine (103600.0037). The
- codon change was CAT-to-CGT.
-
- .0037
- ALBUMIN KOMAGOME-2
- ALB, HIS128ARG
- See 103600.0036.
-
- .0038
- ALBUMIN RUGBY PARK
- ALB, IVS13DS, G-C, +1
- Peach et al. (1992) found that 3 members of a family were heterozygous
- for an electrophoretically fast albumin variant, designated albumin
- Rugby Park, which constituted only 8% of total serum albumin.
- Isoelectric focusing indicated an increased negative charge on the
- C-terminal CNBr peptide. Sequencing of PCR-amplified DNA indicated a
- G-to-C transversion at position 1 of the intron 13. The replacement of
- the obligate GT sequence by CT at the exon/intron boundary prevented
- splicing of intron 13, and translation continued for 21 nucleotides
- until a stop codon was reached. The new protein lacked the 14 amino
- acids encoded in exon 14, but these were replaced by 7 new residues,
- giving a truncated albumin of 578 residues.
-
- .0039
- ALBUMIN HERBORN
- ALB, LYS240GLU
- Minchiotti et al. (1993) found that albumin Herborn, a variant
- discovered in Germany, had a point mutation in codon 240 changing AAA
- (lys) to GAA (glu). The mutation was in the region implicated in
- bilirubin binding, but Minchiotti et al. (1993) found that the binding
- of bilirubin and biliverdin to albumin Herborn was not significantly
- reduced.
-
- .0040
- ANALBUMINEMIA ROMA
- ALB, 1-BP INS, AAT267AAAT, FS274TER
- Watkins et al. (1994) investigated analbuminemia in an Italian family by
- analysis of DNA from a mother and her daughter. The mother, whose
- parents were first cousins, was homozygous for the trait and had a serum
- albumin value of less than 0.01 g/dl (about 1/500 normal); the daughter
- was heterozygous for the trait and had a nearly normal albumin value.
- Molecular cloning and sequence analysis showed that the mutation, called
- analbuminemia Roma, was a nucleotide insertion in exon 8, producing a
- frameshift that led to a premature stop 7 codons downstream. Watkins et
- al. (1994) used heteroduplex hybridization and single-strand
- conformation polymorphism to compare the DNA of these 2 individuals with
- the DNA of 2 unrelated analbuminemic persons, 1 Italian (called Codogno)
- and 1 American (patient G.M.) and showed that each patient had a
- different mutation. These mutations also differed from the mutation in
- the only human case (in an American Indian) previously studied at the
- DNA level (103600.0027). Whereas the normal serum albumin gene has 4 A
- residues as nucleotides 9156-9159, the Roma allele had 5 A residues
- encompassing 9156-9160. The predicted translation product from the Roma
- allele would consist of only 273 amino acids instead of the normal 585
- amino acid residues found in mature serum albumin. The insertion of the
- additional adenine changed codon 267 from AAT (asn) to AAA (lys) and
- changed the reading frame in such a way that codon 274 was changed from
- AAA (lys) to TAA (stop).
-
- .0041
- DYSALBUMINEMIC HYPERTHYROXINEMIA
- ALB, ARG218HIS
- In 2 unrelated patients with dysalbuminemic hyperthyroxinemia, Petersen
- et al. (1994) identified an arg218-to-his substitution which was caused
- by a G (CGC)-to-A (CAG) transition at nucleotide 653. Abnormal affinity
- of the albumin from these patients for a thyroxine analog was verified
- by an adaptation of the procedure used in routine free T4 measurement.
- Both subjects were heterozygous. During the preparation of the
- manuscript, a third patient with the same mutation was found, suggesting
- that R218H may be the most frequent cause of this disorder. The mutation
- created a new HphI restriction site in exon 7 which was used
- diagnostically.
-
- .0042
- ALBUMIN LARINO
- ALB, HIS3TYR
- Madison et al. (1994) stated that of the more than 50 different genetic
- variants of human serum albumin that had been characterized by amino
- acid or DNA sequence analysis, almost half had been identified in Italy
- through a longterm electrophoretic survey of serum. They reported 4
- other Italian alloalbumins not previously recorded: Lorino, his3-to-tyr;
- Tradate-2, lys225-to-gln (103600.0043); Caserta, lys276-to-asn
- (103600.0044); and Bazzano, a carboxyl-terminal variant (103600.0045).
- The first 3 had point mutations that produced a single amino acid
- substitution; a nucleotide deletion caused a frameshift and an altered
- and truncated carboxy-terminal sequence in albumin Bazzano. In these 4
- instances, the expression of the alloalbumin was variable, ranging from
- 10 to 70% of the total albumin, in contrast to the usual 50% each for
- the normal and mutant albumin. Madison et al. (1994) commented that the
- distribution of point mutations in the albumin gene is nonrandom; most
- of the 47 reported point substitutions involved charged amino acid
- residues on the surface of the molecule that are not concerned with
- ligand-binding sites.
-
- .0043
- ALBUMIN TRADATE-2
- ALB, LYS225GLN
- See 103600.0042. In a patient from Tradate (Lombardy region), Madison et
- al. (1994) demonstrated a substitution of glutamine for lysine-225. An
- AAA-to-CAA change is responsible for the substitution. Albumin Tradate-2
- was present in equimolar ratio with albumin A and had a fast mobility.
-
- .0044
- ALBUMIN CASERTA
- ALB, LYS276ASN
- See 103600.0042. In 3 members of a family from Caserta near Naples,
- Madison et al. (1994) demonstrated a substitution of asparagine for
- lysine-276. An AAG-to-AAC change is responsible for the substitution.
- The alloalbumin was identified by its fast mobility. The 3 subjects were
- heterozygous, but the variant/normal ratio was 1.5/1 in the serum of the
- mother, whereas it was about 2/1 in both sibs. In all 3 cases, an
- increased total albumin content was observed.
-
- .0045
- ALBUMIN BAZZANO
- ALB, TGC567GC, FS583TER
- See 103600.0042. Madison et al. (1994) found albumin Bazzano in several
- families from Bazzano, a small town close to Bologna. At pH 8.6 the
- variant was much slower than normal and comprised only about 18% of the
- total albumin. In SDS/PAGE, the molecular weight of the variant appeared
- slightly lower than normal. Sequence analysis revealed deletion of the
- thymine nucleotide at position 15332 in the genomic sequence. This led
- to a frameshift and a divergent amino acid sequence of 16 residues
- beginning at position 567, with early termination after 582. The
- extensive modification caused an increase in positive charge, which
- explained the unusually slow mobility of the alloalbumin. The normal
- termination codon in albumin is 586. Other carboxy-terminal variants are
- albumin Venezia (103600.0028), albumin Rugby Park (103600.0038), and
- albumin Catania (103600.0032).
-
- .0046
- ALBUMIN ASOLA
- ALB, TYR140CYS
- In 2 members of a family living in Asola in Lombardia, Italy, Minchiotti
- et al. (1995) detected a slow migrating variant of human serum albumin
- present in lower amounts than the normal protein by routine clinical
- electrophoresis at pH 8.6. Isoelectric focusing analysis of CNBr
- fragments localized the mutation to fragment CNBr3 (amino acid residues
- 124-298). Amino acid sequence analysis showed a tyr140-to-cys
- substitution, confirmed by DNA sequence analysis, which resulted from a
- single transition of TAT to TGT at nucleotide 5074. Despite the presence
- of an additional cysteine residue, several lines of evidence indicated
- that albumin Asola had no free sulfhydryl group; therefore, Minchiotti
- et al. (1995) proposed that the mutant amino acid, cysteine, was
- involved in the formation of a new disulfide bond with cys34, the only
- free sulfydryl group present in the normal protein.
-
- .0047
- ALBUMIN MALMO-95
- ALB, ASP63ASN
- Carlson et al. (1992) demonstrated that albumin Malmo-95 has a
- substitution of asparagine for aspartic acid-63. A GAC-to-AAC change is
- responsible for the substitution.
-
- .0048
- ALBUMIN HAWKES BAY
- ALB, CYS177PHE
- Brennan and Fellowes (1993) demonstrated that albumin Hawkes Bay has a
- substitution of phenylalanine for cysteine-177. A TGC-to-TTC change is
- responsible for the substitution.
-
- .0049
- ALBUMIN MALMO-10
- ALB, GLN268ARG
- Carlson et al. (1992) demonstrated that albumin Malmo-10 has a
- substitution of arginine for glutamine-268. A CAA-to-CGA change is
- responsible for the substitution.
-
- .0050
- ALBUMIN MALMO-47
- ALB, ASN318LYS
- Carlson et al. (1992) demonstrated that albumin Malmo-47 has a
- substitution of lysine for asparagine-318. A change from AAC to AAA or
- AAG is responsible for the substitution.
-
- .0051
- ALBUMIN SONDRIA
- ALB, GLU333LYS
- Minchiotti et al. (1992) demonstrated that albumin Sondria has a
- substitution of lysine for glutamic acid-333. A GAA-to-AAA change is
- responsible for the substitution.
-
- .0052
- ALBUMIN MALMO-5
- ALB, GLU376ASN
- Carlson et al. (1992) demonstrated that albumin Malmo-5 has a
- substitution of glutamine for glutamic acid-376. A GAA-to-CAA change is
- responsible for the substitution.
-
- .0053
- ALBUMIN DUBLIN
- ALB, GLU479LYS
- Sakamoto et al. (1991) demonstrated that albumin Dublin has a
- substitution of lysine for glutamic acid-479. A GAA-to-AAA change is
- responsible for the substitution.
-
- .0054
- ALBUMIN ORTONOVO
- ALB, GLU505LYS
- Galliano et al. (1993) demonstrated that albumin Ortonovo has a
- substitution of lysine for glutamic acid-505. A GAA-to-AAA change is
- responsible for the substitution.
-
- *FIELD* SA
- Adams (1966); Arai et al. (1989); Arai et al. (1989); Au et al. (1984);
- Barlow et al. (1986); Barlow et al. (1982); Bennhold and Kallee (1959);
- Brennan and Herbert (1987); Brennan et al. (1990); Dammacco et al.
- (1980); Darlington et al. (1974); Dugaiczyk et al. (1982); Efremov
- and Braend (1964); Franklin et al. (1980); Galliano et al. (1988);
- Hawkins and Dugaiczyk (1982); Huss et al. (1988); Jensen and Faber
- (1987); Kueppers et al. (1969); Kurnit et al. (1982); Lalloz et al.
- (1983); Lau et al. (1972); Lavareda de Souza et al. (1984); Melartin
- (1967); Melartin et al. (1967); Murray et al. (1983); Prager et al.
- (1980); Rajatanavin et al. (1982); Rajatanavin et al. (1984); Sanders
- and Tarnoky (1979); Sarcione and Aungst (1962); Sargent et al. (1979);
- Sarich (1972); Schell et al. (1978); Schell and Blumberg (1977);
- Silverberg and Premachandra (1982); Swain et al. (1980); Takahashi
- et al. (1987); Takahashi et al. (1987); Vanzetti et al. (1979); Weitkamp
- (1978); Weitkamp and Buck (1972); Weitkamp and Chagnon (1968); Weitkamp
- et al. (1969); Weitkamp et al. (1970); Weitkamp et al. (1968); Weitkamp
- et al. (1973); Wieme (1960); Yabu et al. (1985); Ying et al. (1981)
- *FIELD* RF
- 1. Abdo, Y.; Rousseaux, J.; Dautrevaux, M.: Proalbumin Lille, a new
- variant of human serum albumin. FEBS Lett. 131: 286-288, 1981.
-
- 2. Adams, M. S.: Genetic diversity in serum albumin. J. Med. Genet. 3:
- 198-202, 1966.
-
- 3. Arai, K.; Huss, K.; Madison, J.; Putnam, F. W.; Salzano, F. M.;
- Franco, M. H. L. P.; Santos, S. E. B.; Freitas, M. J. M.: Amino acid
- substitutions in albumin variants found in Brazil. Proc. Nat. Acad.
- Sci. 86: 1821-1825, 1989.
-
- 4. Arai, K.; Ishioka, N.; Huss, K.; Madison, J.; Putnam, F. W.: Identical
- structural changes in inherited albumin variants from different populations. Proc.
- Nat. Acad. Sci. 86: 434-438, 1989.
-
- 5. Arai, K.; Madison, J.; Huss, K.; Ishioka, N.; Satoh, C.; Fujita,
- M.; Neel, J. V.; Sakurabayashi, I.; Putnam, F. W.: Point substitutions
- in Japanese alloalbumins. Proc. Nat. Acad. Sci. 86: 6092-6096, 1989.
-
- 6. Arai, K.; Madison, J.; Shimizu, A.; Putnam, F. W.: Point substitutions
- in albumin genetic variants from Asia. Proc. Nat. Acad. Sci. 87:
- 497-501, 1990.
-
- 7. Arends, T.; Gallango, M. L.; Layrisse, M.; Wilbert, J.; Heinen,
- H. D.: Albumin Warao: new type of human alloalbuminemia. Blood 33:
- 414-420, 1969.
-
- 8. Arvan, D.; Blumberg, B.; Melartin, L.: Transient bisalbuminemia
- induced by drugs. Clin. Chim. Acta 22: 211-218, 1968.
-
- 9. Au, H. Y. N.; Brand, S.; Hutchinson, D. W.; Matejtschuk, P.: Albumins
- Warwick 1 and Warwick 2, two human albumin variants. IRCS Med. Sci. 12:
- 56-57, 1984.
-
- 10. Barlow, J. W.; Csicsmann, J. M.; Meinhold, H.; Lim, C.-F.; Stockigt,
- J. R.: Familial dysalbuminaemic hyperthyroxinaemia: studies of albumin
- binding and implications for hormone action. Clin. Endocr. 24: 39-47,
- 1986.
-
- 11. Barlow, J. W.; Csicsmann, J. M.; White, E. L.; Funder, J. W.;
- Stockigt, J. R.: Familial euthyroid thyroxine excess: characterization
- of abnormal intermediate affinity thyroxine binding to albumin. J.
- Clin. Endocr. Metab. 55: 244-250, 1982.
-
- 12. Bennhold, H.; Kallee, E.: Comparative studies on the half-life
- of I(131) labelled albumins and nonradioactive human serum albumin
- in a case of analbuminemia. J. Clin. Invest. 38: 863-872, 1959.
-
- 13. Bennhold, H.; Peters, H.; Roth, E.: Uber einen Fall von kompletter
- Analbuminaemie ohne wesentliche klinische Krankheitszeichen. Verh.
- Dtsch. Ges. Inn. Med. 60: 630-634, 1954.
-
- 14. Blumberg, B. S.; Martin, J. R.; Melartin, L.: Alloalbuminemia:
- albumin Naskapi in Indians of the Ungava. J.A.M.A. 203: 180-185,
- 1968.
-
- 15. Boman, H.; Hermodson, M.; Hammond, C. A.; Motulsky, A. G.: Analbuminemia
- in an American Indian girl. Clin. Genet. 9: 513-526, 1976.
-
- 16. Brennan, S. O.: The molecular abnormality of albumin Parklands:
- 365 asp-to-his. Biochim. Biophys. Acta 830: 320-324, 1985.
-
- 17. Brennan, S. O.; Arai, K.; Madison, J.; Laurell, C.-B.; Galliano,
- M.; Watkins, S.; Peach, R.; Myles, T.; George, P.; Putnam, F. W.:
- Hypermutability of CpG dinucleotides in the propeptide-encoding sequence
- of the human albumin gene. Proc. Nat. Acad. Sci. 87: 3909-3913,
- 1990.
-
- 18. Brennan, S. O.; Carrell, R. W.: A circulating variant of human
- proalbumin. Nature 274: 908-909, 1978.
-
- 19. Brennan, S. O.; Fellowes, A. P.: Albumin Hawkes Bay; a low level
- variant caused by loss of a sulphydryl group at position 177. Biochim.
- Biophys. Acta 1182: 46-50, 1993.
-
- 20. Brennan, S. O.; Herbert, P.: Albumin Canterbury (313 lys-to-asn):
- a point mutation in the second domain of serum albumin. Biochim.
- Biophys. Acta 912: 191-197, 1987.
-
- 21. Brennan, S. O.; Myles, T.; Peach, R. J.; Donaldson, D.; George,
- P. M.: Albumin Redhill (-1 arg, ala320-to-thr): a glycoprotein variant
- of human serum albumin whose precursor has an aberrant signal peptidase
- cleavage site. Proc. Nat. Acad. Sci. 87: 26-30, 1990.
-
- 22. Carlson, J.; Sakamoto, Y.; Laurell, C.-B.; Madison, J.; Watkins,
- S.; Putnam, F. W.: Alloalbuminemia in Sweden: structural study and
- phenotypic distribution of nine albumin variants. Proc. Nat. Acad.
- Sci. 89: 8225-8229, 1992.
-
- 23. Cormode, E. J.; Lyster, D. M.; Israels, S.: Analbuminemia in
- a neonate. J. Pediat. 86: 862-867, 1975.
-
- 24. Dammacco, F.; Miglietta, A.; D'Addabbo, A.; Fratello, A.; Moschetta,
- R.; Bonomo, L.: Analbuminemia: report of a case and review of the
- literature. Vox Sang. 39: 153-161, 1980.
-
- 25. Darlington, G.: Personal Communication. New Haven, Conn. and
- New York, N. Y. 9/17/1974.
-
- 26. Darlington, G. J.; Bernhard, H. P.; Ruddle, F. H.: Human serum
- albumin phenotype activation in mouse hepatoma-human leukocyte cell
- hybrids. Science 185: 859-862, 1974.
-
- 27. Dayhoff, M. O.: Serum albumin. Atlas of Protein Sequence and
- Structure. Washington, D. C.: National Biomedical Research Foundation
- (pub.) 5: 1972. Pp. D316.
-
- 28. DeCosimo, D. R.; Fang, S.-L.; Braverman, L. E.: Prevalence of
- familial dysalbuminemic hyperthyroxinemia in Hispanics. (Letter) Ann.
- Intern. Med. 107: 780-781, 1987.
-
- 29. Dugaiczyk, A.: Personal Communication. Riverside, Calif. 4/29/1989.
-
- 30. Dugaiczyk, A.; Law, S. W.; Dennison, O. E.: Nucleotide sequence
- and the encoded amino acids of human serum albumin mRNA. Proc. Nat.
- Acad. Sci. 79: 71-75, 1982.
-
- 31. Earle, D. P.; Hutt, M. P.; Schmid, K.; Gitlin, D.: Observations
- on double albumin: a genetically transmitted serum protein anomaly. J.
- Clin. Invest. 38: 1412-1420, 1959.
-
- 32. Efremov, G. D.; Braend, M.: Serum albumin: polymorphism in man. Science 146:
- 1679-1680, 1964.
-
- 33. Esumi, H.; Okui, M.; Sato, S.; Sugimura, T.; Nagase, S.: Absence
- of albumin mRNA in the liver of analbuminemic rats. Proc. Nat. Acad.
- Sci. 77: 3215-3219, 1980.
-
- 34. Esumi, H.; Takahashi, Y.; Sato, S.; Nagase, S.; Sugimura, T.:
- A seven-base-pair deletion in an intron of the albumin gene of analbuminemic
- rats. Proc. Nat. Acad. Sci. 80: 95-99, 1983.
-
- 35. Esumi, H.; Takahashi, Y.; Sekiya, T.; Sato, S.; Nagase, S.; Sugimura,
- T.: Presence of albumin mRNA precursors in nuclei of analbuminemic
- rat liver lacking cytoplasmic albumin mRNA. Proc. Nat. Acad. Sci. 79:
- 734-738, 1982.
-
- 36. Fine, J. M.; Abdo, Y.; Rochu, D.; Rousseaux, J.; Dautrevaux, M.
- : Identification of the human albumin variant 'Gainesville' with proalbumin
- 'Christchurch'. Blood Transf. Immunohemat. 26: 341-346, 1983.
-
- 37. Fine, J. M.; Marneux, M.; Rochu, D.: Human albumin genetic variants:
- an attempt at a classification of European allotypes. Am. J. Hum.
- Genet. 40: 278-286, 1987.
-
- 38. Franklin, S. G.; Wolf, S. I.; Ozdemir, Y.; Yuregir, G. T.; Isbir,
- T.; Blumberg, B. S.: Albumin Naskapi variant in North American Indians
- and Eti Turks. Proc. Nat. Acad. Sci. 77: 5480-5482, 1980.
-
- 39. Franklin, S. G.; Wolf, S. I.; Zweidler, A.; Blumberg, B. S.:
- Localization of the amino acid substitution site in a new variant
- of human serum albumin, albumin Mexico-2. Proc. Nat. Acad. Sci. 77:
- 2505-2509, 1980.
-
- 40. Fraser, G. R.; Harris, H.; Robson, E. B.: A new genetically determined
- plasma-protein in man. Lancet I: 1023-1024, 1959.
-
- 41. Galliano, M.; Minchiotti, L.; Ferri, G.; Iadarola, P.; Zapponi,
- M. C.; Fine, J. M.: Structural characterization of the human albumin
- variant 'Pollibauer'. Blood Transf. Immunohemat. 27: 597-602, 1984.
-
- 42. Galliano, M.; Minchiotti, L.; Iadarola, P.; Ferri, G.; Zapponi,
- M. C.; Castellani, A. A.: The amino acid substitution in albumin
- Roma: 321 glu-to-lys. FEBS Lett. 233: 100-104, 1988.
-
- 43. Galliano, M.; Minchiotti, L.; Iadarola, P.; Porta, F.; Stoppini,
- M.; Zapponi, M. C.; Ferri, G.; Castellani, A. A.: Genetic variants
- of human serum albumin. Prog. Med. Lab. 2: 475-477, 1988.
-
- 44. Galliano, M.; Minchiotti, L.; Iadarola, P.; Stoppini, M.; Giagnoni,
- P.; Watkins, S.; Madison, J.; Putnam, F. W.: Protein and DNA sequence
- analysis of a 'private' genetic variant: albumin Ortonovo (glu505-to-lys). Biochim.
- Biophys. Acta 1225: 27-32, 1993.
-
- 45. Galliano, M.; Minchiotti, L.; Iadarola, P.; Zapponi, M. C.; Ferri,
- G.; Castellani, A. A.: Structural characterization of a chain termination
- mutant of human serum albumin. J. Biol. Chem. 261: 4283-4287, 1986.
-
- 46. Galliano, M.; Minchiotti, L.; Porta, F.; Rossi, A.; Ferri, G.;
- Madison, J.; Watkins, S.; Putnam, F. W.: Mutations in genetic variants
- of human serum albumin found in Italy. Proc. Nat. Acad. Sci. 87:
- 8721-8725, 1990.
-
- 47. Galliano, M.; Minchiotti, L.; Stoppini, M.; Tarnoky, A. L.: A
- new proalbumin variant: albumin Jaffna (-1 arg-to-leu). FEBS Lett. 255:
- 295-299, 1989.
-
- 48. Harper, M. E.; Dugaiczyk, A.: Linkage of the evolutionarily-related
- serum albumin and alpha-fetoprotein genes within q11-22 of human chromosome
- 4. Am. J. Hum. Genet. 35: 565-572, 1983.
-
- 49. Hawkins, J. W.; Dugaiczyk, A.: The human serum albumin gene:
- structure of a unique locus. Gene 19: 55-58, 1982.
-
- 50. Huss, K.; Madison, J.; Ishioka, N.; Takahashi, N.; Arai, K.; Putnam,
- F. W.: The same substitution, glutamic acid-to-lysine at position
- 501, occurs in three alloalbumins of Asiatic origin: albumins Vancouver,
- Birmingham, and Adana. Proc. Nat. Acad. Sci. 85: 6692-6696, 1988.
-
- 51. Huss, K.; Putnam, F. W.; Takahashi, N.; Takahashi, Y.; Weaver,
- G. A.; Peters, T., Jr.: Albumin Cooperstown: a serum albumin variant
- with the same (313 lys-to-asn) mutation found in albumins in Italy
- and New Zealand. Clin. Chem. 34: 183-187, 1988.
-
- 52. Hutchinson, D. W.; Matejtschuk, P.; Lord, C.: Albumin Carlisle:
- occurrence and properties of a new human albumin variant. IRCS Med.
- Sci. 14: 1095-1096, 1986.
-
- 53. Jamieson, G. A.; Ganguly, P.: Studies on a genetically determined
- albumin dimer. Biochem. Genet. 3: 403-416, 1969.
-
- 54. Jensen, I. W.; Faber, J.: Familial dysalbuminemic hyperthyroxinemia. Acta
- Med. Scand. 221: 469-473, 1987.
-
- 55. Kaarsalo, E.; Melartin, L.; Blumberg, B. S.: Autosomal linkage
- between the albumin and GC loci in humans. Science 158: 123-125,
- 1967.
-
- 56. Kallee, E.: Bennhold's analbuminemia: a follow-up study of the
- first two cases (1953-1992). J. Lab. Clin. Med. 127: 470-480, 1996.
-
- 57. Kao, F.-T.; Hawkins, J. W.; Law, M. L.; Dugaiczyk, A.: Assignment
- of the structural gene coding for albumin to human chromosome 4. Hum.
- Genet. 62: 337-341, 1982.
-
- 58. Kaur, H.; Franklin, S. G.; Shrivastava, P. K.; Blumberg, B. S.
- : Alloalbuminemia in North India. Am. J. Hum. Genet. 34: 972-979,
- 1982.
-
- 59. Kueppers, F.; Holland, P. V.; Weitkamp, L. R.: Albumin Santa
- Ana: a new inherited variant. Hum. Hered. 19: 378-384, 1969.
-
- 60. Kurnit, D. M.; Philipp, B. W.; Bruns, G. A. P.: Confirmation
- of the mapping assignment of human serum albumin to chromosome 4 using
- a cloned human albumin gene. Cytogenet. Cell Genet. 34: 282-288,
- 1982.
-
- 61. Lalloz, M. R. A.; Byfield, P. G. H.; Himsworth, R. L.: Hyperthyroxinaemia:
- abnormal binding of T4 by an inherited albumin variant. Clin. Endocr. 18:
- 11-24, 1983.
-
- 62. Lalloz, M. R. A.; Byfield, P. G. H.; Himsworth, R. L.: A new
- and distinctive albumin variant with increased affinities for both
- triiodothyronines and causing hyperthyroxinaemia. Clin. Endocr. 22:
- 521-529, 1985.
-
- 63. Lau, T. J.; Sunderman, F. W., Jr.; Weitkamp, L. R.; Agarwal, S.
- S.; Sutnick, A. I.; Blumberg, B. S.; De Jimenez, R. B. C.: Albumin
- Cartago: a 'new' slow-moving alloalbumin. Am. J. Clin. Path. 57:
- 247-251, 1972.
-
- 64. Laurell, C. B.; Nilehn, J. E.: A new type of inherited serum
- albumin anomaly. J. Clin. Invest. 45: 1935-1945, 1966.
-
- 65. Lavareda de Souza, S.; Frain, M.; Mornet, E.; Sala-Trepat, J.
- M.; Lucotte, G.: Polymorphisms of human albumin gene after DNA restriction
- by HaeIII endonuclease. Hum. Genet. 67: 48-51, 1984.
-
- 66. Madison, J.; Arai, K.; Sakamoto, Y.; Feld, R. D.; Kyle, R. A.;
- Watkins, S.; Davis, E.; Matsuda, Y.; Amaki, I.; Putnam, F. W.: Genetic
- variants of serum albumin in Americans and Japanese. Proc. Nat. Acad.
- Sci. 88: 9853-9857, 1991.
-
- 67. Madison, J.; Galliano, M.; Watkins, S.; Minchiotti, L.; Porta,
- F.; Rossi, A.; Putnam, F. W.: Genetic variants of human serum albumin
- in Italy: point mutants and a carboxyl-terminal variant. Proc. Nat.
- Acad. Sci. 91: 6476-6480, 1994.
-
- 68. Magenis, R. E.; Luo, X. Y.; Dugaiczyk, A.; Ryan, S. C.; Oosterhuis,
- J. E.: Chromosomal localization of the albumin and alpha-fetoprotein
- genes in the orangutan (Pongo pygmaeus) and gorilla (Gorilla gorilla).
- (Abstract) Cytogenet. Cell Genet. 51: 1037 only, 1989.
-
- 69. Melartin, L.: Albumin polymorphism in man: studies on albumin
- variants in North American native populations. Acta Path. Microbiol.
- Scand. 191 (suppl.): 1-50, 1967.
-
- 70. Melartin, L.; Blumberg, B. S.: Albumin Naskapi: a new variant
- of serum albumin. Science 153: 1664-1666, 1966.
-
- 71. Melartin, L.; Blumberg, B. S.; Lisker, R.: Albumin Mexico, a
- new variant of serum albumin. Nature 215: 1288-1289, 1967.
-
- 72. Mendel, C. M.; Cavalieri, R. R.; Gavin, L. A.; Pettersson, T.;
- Inoue, M.: Thyroxine transport and distribution in Nagase analbuminemic
- rats. J. Clin. Invest. 83: 143-148, 1989.
-
- 73. Minchiotti, L.; Galliano, M.; Iadarola, P.; Meloni, M. L.; Ferri,
- G.; Porta, F.; Castellani, A. A.: The molecular defect in a COOH-terminal-modified
- and shortened mutant of human serum albumin. J. Biol. Chem. 264:
- 3385-3389, 1989.
-
- 74. Minchiotti, L.; Galliano, M.; Kragh-Hansen, U.; Watkins, S.; Madison,
- J.; Putnam, F. W.: A genetic variant of albumin (albumin Asola; tyr140-to-cys)
- with no free -SH group but with an additional disulfide bridge. Europ.
- J. Biochem. 228: 155-159, 1995.
-
- 75. Minchiotti, L.; Galliano, M.; Stoppini, M.; Ferri, G.; Crespeau,
- H.; Rochu, D.; Porta, F.: Two alloalbumins with identical electrophoretic
- mobility are produced by differently charged amino acid substitutions. Biochim.
- Biophys. Acta 1119: 232-238, 1992.
-
- 76. Minchiotti, L.; Galliano, M.; Zapponi, M. C.; Tenni, R.: The
- structural characterization and bilirubin-binding properties of albumin
- Herborn, a lys240-to-glu albumin mutant. Europ. J. Biochem. 214:
- 437-444, 1993.
-
- 77. Minghetti, P. P.; Law, S. W.; Dugaiczyk, A.: The rate of molecular
- evolution of alpha-fetoprotein approaches that of pseudogenes. Molec.
- Biol. Evol. 2: 347-358, 1985.
-
- 78. Minghetti, P. P.; Ruffner, D. E.; Kuang, W.-J.; Dennison, O. E.;
- Hawkins, J. W.; Beattie, W. G.; Dugaiczyk, A.: Molecular structure
- of the human albumin gene is revealed by nucleotide sequence within
- q11-22 of chromosome 4. J. Biol. Chem. 261: 6747-6757, 1986.
-
- 79. Murray, J. C.; Demopulos, C. M.; Lawn, R. M.; Motulsky, A. G.
- : Restriction endonuclease study of analbuminemia and polymorphisms
- at the albumin locus. (Abstract) Clin. Res. 31: 456A only, 1983.
-
- 80. Murray, J. C.; Demopulos, C. M.; Lawn, R. M.; Motulsky, A. G.
- : Molecular genetics of human serum albumin: restriction enzyme fragment
- length polymorphisms and analbuminemia. Proc. Nat. Acad. Sci. 80:
- 5951-5955, 1983.
-
- 81. Murray, J. C.; Mills, K. A.; Demopulos, C. M.; Hornung, S.; Motulsky,
- A. G.: Linkage disequilibrium and evolutionary relationships of DNA
- variants (restriction enzyme fragment length polymorphisms) at the
- serum albumin locus. Proc. Nat. Acad. Sci. 81: 3486-3490, 1984.
-
- 82. Nagase, S.; Shimamune, K.; Shumiya, S.: Albumin-deficient rat
- mutant. Science 205: 590-591, 1979.
-
- 83. Ohno, S.: Original domain for the serum albumin family arose
- from repeated sequences. Proc. Nat. Acad. Sci. 78: 7657-7661, 1981.
-
- 84. Ott, H.: Analbuminemia.In: Linneweh, F.: Erbliche Stoffwechselkrankheiten.
- Munich: Urban und Schwarzenberg (pub.) 1962. Pp. 44.
-
- 85. Peach, R. J.; Brennan, S. O.: Structural characterization of
- a glycoprotein variant of human serum albumin: albumin Casebrook (494
- asp-to-asn). Biochim. Biophys. Acta 1097: 49-54, 1991.
-
- 86. Peach, R. J.; Fellowes, A. P.; Brennan, S. O.; George, P. M.:
- Albumin Rugby Park: a truncated albumin variant caused by a G-to-C
- splice-site mutation in intron 13. Biochim. Biophys. Acta 1180:
- 107-110, 1992.
-
- 87. Petersen, C. E.; Scottolini, A. G.; Cody, L. R.; Mandel, M.; Reimer,
- N.; Bhagavan, N. V.: A point mutation in the human serum albumin
- gene results in familial dysalbuminaemic hyperthyroxinaemia. J. Med.
- Genet. 31: 355-359, 1994.
-
- 88. Pinkert, C. A.; Ornitz, D. M.; Brinster, R. L.; Palmiter, R. D.
- : An albumin enhancer located 10 kb upstream functions along with
- its promoter to direct efficient, liver-specific expression in transgenic
- mice. Genes Dev. 1: 268-276, 1987.
-
- 89. Prager, E. M.; Wilson, A. C.; Lowenstein, J. M.; Sarich, V. M.
- : Mammoth albumin. Science 209: 287-289, 1980.
-
- 90. Premachandra, B. N.; Wolfe, B.; Williams, I. K.: Coexistence
- of familial dysalbuminemic hyperthyroxinemia with familial hypercholesterolemia
- and multiple lipoprotein type hyperlipidemia. Am. J. Med. 84: 345-351,
- 1988.
-
- 91. Putnam, F. W.: Personal Communication. Bloomington, Ind. 8/4/1993.
-
- 92. Rajatanavin, R.; Fournier, L.; DeCosimo, D.; Abreau, C.; Braverman,
- L. E.: Elevated serum free thyroxine by thyroxine analog radioimmunoassays
- in euthyroid patients with familial dysalbuminemic hyperthyroxinemia. Ann.
- Intern. Med. 97: 865-866, 1982.
-
- 93. Rajatanavin, R.; Young, R. A.; Braverman, L. E.: Effect of chloride
- on serum thyroxine binding in familial dysalbuminemic hyperthyroxinemia. J.
- Clin. Endocr. Metab. 58: 388-391, 1984.
-
- 94. Rochu, D.; Fine, J. M.: New method for identifying genetic variants
- of human proalbumin. Clin. Chem. 32: 2063-2065, 1986.
-
- 95. Ruffner, D. E.; Dugaiczyk, A.: Splicing mutation in human hereditary
- analbuminemia. Proc. Nat. Acad. Sci. 85: 2125-2129, 1988.
-
- 96. Ruiz, M.; Rajatanavin, R.; Young, R. A.; Taylor, C.; Brown, R.;
- Braverman, L. E.; Ingbar, S. H.: Familial dysalbuminemic hyperthyroxinemia:
- a syndrome that can be confused with thyrotoxicosis. New Eng. J.
- Med. 306: 635-639, 1982.
-
- 97. Sakamoto, Y.; Davis, E.; Madison, J.; Watkins, S.; McLaughlin,
- H.; Leahy, D. T.; Putnam, F. W.: Purification and structural study
- of two albumin variants in an Irish population. Clin. Chim. Acta 204:
- 179-188, 1991.
-
- 98. Sanders, G. T. B.; Tarnoky, A. L.: Albumin Amsterdam: a new European
- albumin variant. IRCS Med. Sci. 7: 581 only, 1979.
-
- 99. Sarcione, E. J.; Aungst, C. W.: Studies in bisalbuminemia: binding
- properties of the two albumins. Blood 20: 156-164, 1962.
-
- 100. Sargent, T. D.; Wu, J.-R.; Sala-Trepat, J. M.; Wallace, R. B.;
- Reyes, A. A.; Bonner, J.: The rat serum albumin gene: analysis of
- cloned sequences. Proc. Nat. Acad. Sci. 76: 3256-3260, 1979.
-
- 101. Sarich, V. M.: Generation time and albumin evolution. Biochem.
- Genet. 7: 205-212, 1972.
-
- 102. Schell, L. M.; Agarwal, S. S.; Blumberg, B. S.; Levy, H.; Bennett,
- H.; Laughlin, W. S.; Martin, J. P.: Distribution of albumin variants
- Naskapi and Mexico among Aleuts, Frobisher Bay Eskimos, and Micmac,
- Naskapi, Mohawk, Omaha and Apache Indians. Am. J. Phys. Anthrop. 49:
- 111-118, 1978.
-
- 103. Schell, L. M.; Blumberg, B. S.: The genetics of human serum
- albumin.In: Rosenoer, V. M.; Oratz, M.; Rothschild, M. A.: Albumin
- Structure, Function and Uses. Oxford: Pergamon Press (pub.) 1977.
- Pp. 113-141.
-
- 104. Shalaby, F.; Shafritz, D. A.: Exon skipping during splicing
- of albumin mRNA precursors in Nagase analbuminemic rats. Proc. Nat.
- Acad. Sci. 87: 2652-2656, 1990.
-
- 105. Shashaty, G.; Atamer, M.: Acquired bisalbuminemia with hyperamylasemia. Digest.
- Dis. 17: 59-67, 1972.
-
- 106. Shibata, T.; Abe, T.: Linkage between the loci for serum albumin
- and vitamin D binding protein (GC) in the Japanese quail. Animal
- Genet. 27: 195-197, 1996.
-
- 107. Silverberg, J. D. H.; Premachandra, B. N.: Familial hyperthyroxinemia
- due to abnormal thyroid hormone binding. Ann. Intern. Med. 96: 183-186,
- 1982.
-
- 108. Swain, B. K.; Talukder, G.; Sharma, A.: Bisalbuminaemia: reports
- from Calcutta. Biomedicine 33: 172-173, 1980.
-
- 109. Takahashi, N.; Takahashi, Y.; Blumberg, B. S.; Putnam, F. W.
- : Amino acid substitutions in genetic variants of human serum albumin
- and in sequences inferred from molecular cloning. Proc. Nat. Acad.
- Sci. 84: 4413-4417, 1987.
-
- 110. Takahashi, N.; Takahashi, Y.; Isobe, T.; Putnam, F. W.; Fujita,
- M.; Satoh, C.; Neel, J. V.: Amino acid substitutions in inherited
- albumin variants from Amerindian and Japanese populations. Proc.
- Nat. Acad. Sci. 84: 8001-8005, 1987.
-
- 111. Takahashi, N.; Takahashi, Y.; Putnam, F. W.: Structural changes
- and metal binding by proalbumins and other amino-terminal genetic
- variants of human serum albumin. Proc. Nat. Acad. Sci. 84: 7403-7407,
- 1987.
-
- 112. Tarnoky, A. L.; Lestas, A. N.: A new type of bisalbuminaemia. Clin.
- Chim. Acta 9: 551-558, 1964.
-
- 113. Urano, Y.; Sakai, M.; Watanabe, K.; Tamaoki, T.: Tandem arrangement
- of the albumin and alpha-fetoprotein genes in the human genome. Gene 32:
- 255-261, 1984.
-
- 114. Vanzetti, G.; Porta, F.; Prencipe, L.; Scherini, A.; Fraccaro,
- M.: A homozygote for a serum albumin variant of the fast type. Hum.
- Genet. 46: 5-9, 1979.
-
- 115. Vaysse, J.; Pilardeau, P.; Garnier, M.: Trisalbuminemia. (Letter) New
- Eng. J. Med. 305: 833-834, 1981.
-
- 116. Watkins, S.; Madison, J.; Galliano, M.; Minchiotti, L.; Putnam,
- F. W.: A nucleotide insertion and frameshift cause analbuminemia
- in an Italian family. Proc. Nat. Acad. Sci. 91: 2275-2279, 1994.
-
- 117. Weitkamp, L. R.: Comparative gene mapping: linkage between the
- albumin and Gc loci in the horse. (Abstract) Am. J. Hum. Genet. 30:
- 128A only, 1978.
-
- 118. Weitkamp, L. R.; Buck, A. A.: Phenotype frequencies for four
- serum proteins in Afghanistan: two 'new' albumin variants. Humangenetik 15:
- 335-340, 1972.
-
- 119. Weitkamp, L. R.; Chagnon, N. A.: Albumin Maku: a new variant
- of human serum albumin. Nature 217: 759-760, 1968.
-
- 120. Weitkamp, L. R.; Franglen, G.; Rokala, D. A.; Polesky, H. F.;
- Simpson, N. E.; Sunderman, F. W., Jr.; Bell, H. E.; Saave, J.; Lisker,
- R.; Bohls, S. W.: An electrophoretic comparison of human serum albumin
- variants: eight distinguishable types. Hum. Hered. 19: 159-169,
- 1969.
-
- 121. Weitkamp, L. R.; Renwick, J. H.; Berger, J. P.; Shreffler, D.
- C.; Drachmann, O.; Wuhrmann, F.; Braend, M.; Franglen, G.: Additional
- data and summary for albumin-GC linkage in man. Hum. Hered. 20:
- 1-7, 1970.
-
- 122. Weitkamp, L. R.; Robson, E. B.; Shreffler, D. C.; Corney, G.
- : An unusual human serum albumin variant: further data on genetic
- linkage between loci for human serum albumin and group-specific component
- (GC). Am. J. Hum. Genet. 20: 392-397, 1968.
-
- 123. Weitkamp, L. R.; Rucknagel, D. L.; Gershowitz, H.: Genetic linkage
- between structural loci for albumin and group specific component (GC). Am.
- J. Hum. Genet. 18: 559-571, 1966.
-
- 124. Weitkamp, L. R.; Salzano, F. M.; Neel, J. V.; Porta, F.; Geerdink,
- R. A.; Tarnoky, A. L.: Human serum albumin: twenty-three genetic
- variants and their population distribution. Ann. Hum. Genet. 36:
- 381-392, 1973.
-
- 125. Weitkamp, L. R.; Shreffler, D. C.; Robbins, J. L.; Drachmann,
- O.; Adner, P. L.; Weime, R. J.; Simon, N. M.; Cooke, K. B.; Sandor,
- G.; Wuhrmann, F.; Braend, M.; Tarnoky, A. L.: An electrophoretic
- comparison of serum albumin variants from nineteen unrelated families. Acta
- Genet. Statist. Med. 17: 399-405, 1967.
-
- 126. Wieme, R. J.: On the presence of two albumins in certain normal
- human sera and its genetic determination. Clin. Chim. Acta 5: 443-445,
- 1960.
-
- 127. Williams, D. I.; Martin, N. H.: Bisalbuminemia with curious
- acrocyanotic skin changes (two cases). Proc. Roy. Soc. Med. 53:
- 566-568, 1960.
-
- 128. Yabu, Y.; Amir, S. M.; Ruiz, M.; Braverman, L. E.; Ingbar, S.
- H.: Heterogeneity of thyroxine binding by serum albumins in normal
- subjects and patients with familial dysalbuminemic hyperthyroxinemia. J.
- Clin. Endocr. Metab. 60: 451-459, 1985.
-
- 129. Yabu, Y.; Miyai, K.; Kobayashi, A.; Miki, K.; Doi, K.; Takamatsu,
- J.; Mozai, T.; Matsuzuka, F.; Kuma, K.: A new type of albumin with
- predominantly increased binding affinity for 3,3-prime,5-triiodothyronine
- in a patient with Graves' disease. J. Endocr. Invest. 10: 163-169,
- 1987.
-
- 130. Yeo, P. P. B.; Yabu, Y.; Etzkorn, J. R.; Rajatanavin, R.; Braverman,
- L. E.; Ingbar, S. H.: A four generation study of dysalbuminemic hyperthyroxinemia:
- diagnosis in the presence of an acquired excess of thyroxine-binding
- globulin. J. Endocr. Invest. 10: 33-38, 1987.
-
- 131. Ying, Q.; Liang, Z.; Wu, H.; Wang, L.: The gene frequency of
- serum albumin variants in Chinese and the electrophoretic characterization
- of several serum albumin variants. Scientia Sinica 24: 1597-1602,
- 1981.
-
- *FIELD* CN
- Jon B. Obray - updated: 8/27/1996
- Stylianos E. Antonarakis - updated: 7/25/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 10/28/1996
- terry: 10/22/1996
- carol: 8/27/1996
- joanna: 8/26/1996
- carol: 8/13/1996
- carol: 7/27/1996
- carol: 7/25/1996
- mark: 6/27/1995
- jason: 7/13/1994
- davew: 8/10/1994
- terry: 6/3/1995
- carol: 8/30/1994
- warfield: 4/7/1994
-
- *RECORD*
- *FIELD* NO
- 103700
- *FIELD* TI
- *103700 ALCOHOL DEHYDROGENASE-1; ADH1
- ADH, ALPHA SUBUNIT
- *FIELD* TX
- Polymorphism of alcohol dehydrogenase was investigated by Smith et al.
- (1971), who concluded that 3 ADH loci are responsible for 3 distinct
- polypeptide subunits--alpha, beta and gamma. No electrophoretic or other
- allelic variants of ADH1 are known. At each of the ADH2 (103720) and
- ADH3 (103730) loci, the evidence indicated that 2 different common
- alleles occur. The ADH isozymes are dimers. Any particular isozyme may
- be made up of 2 identical subunits coded by a specific allele at one of
- the loci, or of 2 nonidentical subunits coded by alleles at 2 separate
- loci, or of 2 nonidentical subunits coded by different alleles at the
- same locus. At least 3 autosomal gene loci may, they concluded, be
- concerned with determining the structure of alcohol dehydrogenase in
- man. ADH1, ADH2 and ADH3 show differential tissue and developmental
- expression. (Class I ADH isozymes are pyrazole-sensitive and basic.
- Class II isozymes are less pyrazole-sensitive and less basic. Class III
- isozymes show anodal electrophoretic mobility and low ethanol
- dehydrogenase activity.) ADH1 is primarily active in the liver in early
- fetal life, becoming less active later in gestation and only weakly
- active during adult life when beta subunits and, to a lesser extent,
- gamma subunits predominate in liver. With the coenzyme NAD, this enzyme
- catalyzes the reversible conversion of organic alcohols to ketones or
- aldehydes. The physiologic function for alcohol dehydrogenase in the
- liver is the removal of ethanol formed by microorganisms in the
- intestinal tract. The enzyme from horse liver is a dimer with 2 very
- similar chains, one called E for ethanol-active and the other S for
- steroid active. Sequence data are not available in man but the data on
- the horse liver enzyme are given in the atlas by Dayhoff (1972). An
- atypical liver ADH was described by Von Wartburg and Schuerch (1968) in
- 2 of 50 English livers and in 12 of 59 Swiss livers. The difference
- studied concerned the ratio of activity at pH 10.8 and pH 8.8. About 1%
- of protein in horse liver is alcohol dehydrogenase. The list of
- substrates on which ADH operates is large. Important drug-ethanol
- interactions, e.g., digitalis-ethanol, probably have their basis in this
- fact (Vallee, 1979).
-
- Using a cDNA clone from an adult cDNA library in somatic hybrid cell
- studies, Smith et al. (1984) concluded that the class I ADH genes are
- located distal to 4q21. DNA polymorphism was found in both the ADH2 and
- ADH3 genes and Oriental/Caucasian differences were found. By Southern
- blot analysis of somatic hybrid cell DNAs, Smith et al. (1985) assigned
- the genes for alpha, beta and gamma ADH gene products (ADH1, ADH2, and
- ADH3) to chromosome 4 (4q21-4q25). This represents an exception to the
- rule that the subunits of heteromeric proteins are coded by separate
- chromosomes. The progression from fetal alpha to adult beta (and gamma)
- subunits as the predominant ones in adult life may represent an example
- of switching between linked genes similar to the changes in the
- beta-like globin genes during development. Von Bahr-Lindstrom et al.
- (1986) provided information on the cDNA and protein sequence of the
- alpha subunit. Smith (1986) stated the location of the class I ADH genes
- as 4q21-q24. In situ hybridization permitted a narrowing of the
- localization of the cluster to 4q22 (Tsukahara and Yoshida, 1989).
- Yasunami et al. (1989) described the organization of the human class I
- alcohol dehydrogenase gene cluster on chromosome 4q22. The cluster
- includes ADH1, ADH2, and ADH3, which are arranged in the same
- head-to-tail transcriptional orientation at intervals of approximately
- 15 kb. By genomic cloning using a cosmid vector, Yasunami et al. (1990)
- showed that the genes for the 3 subunits of class I ADH lie in an 80-kb
- segment in the following order: 5-prime--ADH3--ADH2--ADH1--3-prime.
- Perhaps significantly, the order of transcriptional activation in
- hepatic development, alpha-to-beta-to-gamma, is opposite to the order of
- gene arrangement.
-
- *FIELD* SA
- Adinolfi and Hopkinson (1978); Adinolfi and Hopkinson (1979); Harada
- et al. (1980); Ikuta et al. (1985); Lange et al. (1976); Murray and
- Price (1972); Smith et al. (1972); Smith et al. (1973); Smith et al.
- (1974)
- *FIELD* RF
- 1. Adinolfi, A.; Hopkinson, D. A.: Blue sepharose chromatography
- of human alcohol dehydrogenase: evidence for interlocus and interallelic
- differences in affinity characteristics. Ann. Hum. Genet. 41: 399-407,
- 1978.
-
- 2. Adinolfi, A.; Hopkinson, D. A.: Affinity electrophoresis of human
- alcohol dehydrogenase (ADH) isozymes. Ann. Hum. Genet. 43: 109-119,
- 1979.
-
- 3. Dayhoff, M. O.: Atlas of Protein Sequence and Structure. Dehydrogenases.
- Washington: National Biomedical Research Foundation (pub.) 5:
- 1972. Pp. D141-D144.
-
- 4. Harada, S.; Misawa, S.; Agarwal, D. P.; Goedde, H. W.: Liver alcohol
- dehydrogenase and aldehyde dehydrogenase in the Japanese: isozyme
- variation and its possible role in alcohol intoxication. Am. J.
- Hum. Genet. 32: 8-15, 1980.
-
- 5. Ikuta, T.; Fujiyoshi, T.; Kurachi, K.; Yoshida, A.: Molecular
- cloning of a full-length cDNA for human alcohol dehydrogenase. Proc.
- Nat. Acad. Sci. 82: 2703-2707, 1985.
-
- 6. Lange, L. G.; Sytkowski, A. J.; Vallee, B. L.: Human liver alcohol
- dehydrogenase: purification, composition, and catalytic features.
- Biochemistry 15: 4687-4693, 1976.
-
- 7. Murray, R. F., Jr.; Price, P. H.: Ontogenetic, polymorphic, and
- interethnic variation in the isoenzymes of human alcohol dehydrogenase.
- Ann. N.Y. Acad. Sci. 197: 68-72, 1972.
-
- 8. Smith, M.: Genetics of human alcohol and aldehyde dehydrogenases.
- Adv. Hum. Genet. 15: 249-290, 1986.
-
- 9. Smith, M.; Duester, G.; Bilanchone, V.; Carlock, L.; Hatfield,
- W.: Derivation of probes for molecular genetic analysis of human
- class I alcohol dehydrogenase (ADH), a polymorphic gene family on
- chromosome 4. (Abstract) Am. J. Hum. Genet. 36: 153S only, 1984.
-
- 10. Smith, M.; Duester, G.; Carlock, L.; Wasmuth, J.: Assignment
- of ADH1, ADH2 and ADH3 genes (class I ADH) to human chromosome 4q21-4q25,
- through use of DNA probes. (Abstract) Cytogenet. Cell Genet. 40:
- 748 only, 1985.
-
- 11. Smith, M.; Hopkinson, D. A.; Harris, H.: Developmental changes
- and polymorphism in human alcohol dehydrogenase. Ann. Hum. Genet. 34:
- 251-272, 1971.
-
- 12. Smith, M.; Hopkinson, D. A.; Harris, H.: Alcohol dehydrogenase
- isozymes in adult human stomach and liver: evidence for activity of
- the ADH(3) locus. Ann. Hum. Genet. 35: 243-253, 1972.
-
- 13. Smith, M.; Hopkinson, D. A.; Harris, H.: Studies on the subunit
- structure and molecular size of the human dehydrogenase isozymes determined
- by the different loci, ADH(1), ADH(2), and ADH(3). Ann. Hum. Genet. 36:
- 401-414, 1973.
-
- 14. Smith, M.; Hopkinson, D. A.; Harris, H.: Studies on the properties
- of the human alcohol dehydrogenase isozymes determined by the different
- loci ADH(1), ADH(2) and ADH(3). Ann. Hum. Genet. 37: 49-67, 1974.
-
- 15. Tsukahara, M.; Yoshida, A.: Chromosomal assignment of the alcohol
- dehydrogenase cluster locus to human chromosome 4q21-23 by in situ
- hybridization. Genomics 4: 218-220, 1989.
-
- 16. Vallee, B.: Personal Communication. Boston, Mass. 1979.
-
- 17. von Bahr-Lindstrom, H.; Hoog, J.-O.; Heden, L.-O.; Kaiser, R.;
- Fleetwood, L.; Larsson, K.; Lake, M.; Holmquist, B.; Holmgren, A.;
- Hempel, J.; Vallee, B. L.; Jornvall, H.: cDNA and protein structure
- for the alpha subunit of human liver alcohol dehydrogenase. Biochemistry 25:
- 2465-2470, 1986.
-
- 18. Von Wartburg, J. P.; Schuerch, P. M.: Atypical human liver alcohol
- dehydrogenase. Ann. N.Y. Acad. Sci. 151: 936-947, 1968.
-
- 19. Yasunami, M.; Kikuchi, I.; Sarapata, D.; Yoshida, A.: The human
- class I alcohol dehydrogenase gene cluster: three genes are tandemly
- organized in an 80-kb-long segment of the genome. Genomics 7: 152-158,
- 1990.
-
- 20. Yasunami, M.; Kikuchi, I.; Sarapata, D. E.; Yoshida, A.: The
- organization of human class I alcohol dehydrogenase gene cluster.
- (Abstract) Cytogenet. Cell Genet. 51: 1113 only, 1989.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- davew: 6/8/1994
- warfield: 4/7/1994
- carol: 4/6/1994
- pfoster: 4/4/1994
- mimadm: 2/11/1994
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 103710
- *FIELD* TI
- *103710 ALCOHOL DEHYDROGENASE 5, CHI POLYPEPTIDE; ADH5
- ALCOHOL DEHYDROGENASE, CHI ISOZYME;;
- ADH, CLASS III; ADHX
- *FIELD* TX
- See 103720. Adinolfi et al. (1984) purified the chi isozyme of ADH (EC
- 1.1.1.1) from human liver and used it to raise immune sera. Its
- immunologic properties suggested that it has no structural similarity to
- either class I (ADH1, ADH2, ADH3) or class II (ADH4) isozymes. The chi
- isozyme was found in most human tissues including fetal specimens of 16
- weeks gestational age and showed a preference for long chain primary
- alcohols with a double bond in the beta position. Adinolfi et al. (1984)
- concluded that the locus, designated ADH5, has a separate evolutionary
- origin from other ADH genes. (The class I ADH isozymes are virtually
- indistinguishable immunologically; the genes that determine them
- presumably originated by gene duplication.) Class III or chi ADH has
- specificity for complex alcohols of high molecular weight such as
- cinnamyl alcohol. Beisswenger et al. (1985) showed that ADH-chi is the
- only ADH isozyme in brain. It oxidizes ethanol very poorly; its function
- in brain is unknown. Since its gene is expressed constitutively in
- somatic cell hybrids, Carlock et al. (1985) could assign the locus to
- chromosome 4, specifically 4q21-q25, by analysis of gene products in
- starch gel electrophoresis. Smith (1986) gave the regional assignment as
- 4q21-q24. Goldman et al. (1989) isolated and sequenced a full-length
- cDNA for the class III alcohol dehydrogenase ADH5. By analysis of
- human/hamster hybrid cell lines, ADH5 was mapped to chromosome 4 where
- other ADH genes have been located, including class I genes and a class
- II gene, all of which metabolize ethanol, and the unusual class III ADH,
- which does not. Analysis of mouse/hamster hybrid cell lines showed that
- the corresponding gene maps to mouse chromosome 3, which carries the
- other murine ADH genes. The sequence of ADH5 indicated that it is about
- equidistant between class I and class II ADHs. In contrast to other ADHs
- whose expression is more restricted, class III ADH was found to be
- expressed ubiquitously in human and rodent tissues. Giri et al. (1989)
- also mapped the gene to mouse chromosome 3. Matsuo and Yokoyama (1990)
- demonstrated a processed pseudogene derived from the ADH5 gene. Engeland
- et al. (1993) reported the kinetic characterization of human class III
- ADH altered at position 115 to asp and to ala by in vitro mutagenesis.
- The results indicated that the arg115 residue is a component of the
- binding site for activating fatty acids and is critical for the binding
- of S-hydroxymethylglutathione in glutathione-dependent formaldehyde
- dehydrogenase activity.
-
- *FIELD* RF
- 1. Adinolfi, A.; Adinolfi, M.; Hopkinson, D. A.: Immunological and
- biochemical characterization of the human alcohol dehydrogenase chi-ADH
- isozyme. Ann. Hum. Genet. 48: 1-10, 1984.
-
- 2. Beisswenger, T. B.; Holmquist, B.; Vallee, B. L.: Chi-ADH is the
- sole alcohol dehydrogenase isozyme of mammalian brains: implications
- and inferences. Proc. Nat. Acad. Sci. 82: 8369-8373, 1985.
-
- 3. Carlock, L.; Hiroshige, S.; Wasmuth, J.; Smith, M.: Assignment
- of the gene coding for class III ADH to human chromosome 4: 4q21-4q25.
- (Abstract) Cytogenet. Cell Genet. 40: 598 only, 1985.
-
- 4. Engeland, K.; Hoog, J.-O.; Holmquist, B.; Estonius, M.; Jornvall,
- H.; Vallee, B. L.: Mutation of arg-115 of human class III alcohol
- dehydrogenase: a binding site required for formaldehyde dehydrogenase
- activity and fatty acid activation. Proc. Nat. Acad. Sci. 90: 2491-2494,
- 1993.
-
- 5. Giri, P.; Krug, J. F.; Kozak, C.; Moretti, T.; O'Brien, S. J.;
- Seuanez, H. N.; Goldman, D.: Cloning and comparative mapping of a
- human class III (chi) alcohol dehydrogenase cDNA. Biochem. Biophys.
- Res. Commun. 164: 453-460, 1989.
-
- 6. Goldman, D.; RathnaGiri, P.; Moretti, T. R.; Krug, J. F.; Kozak,
- C.; Dean, M.; Seuanez, H. N.; O'Brien, S. J.: Class III alcohol dehydrogenase
- (ADH5): widespread expression and synteny with other ADHs in both
- mouse and man. (Abstract) Am. J. Hum. Genet. 45 (suppl.): A141
- only, 1989.
-
- 7. Matsuo, Y.; Yokoyama, S.: Cloning and sequencing of a processed
- pseudogene derived from a human class III alcohol dehydrogenase gene.
- Am. J. Hum. Genet. 46: 85-91, 1990.
-
- 8. Smith, M.: Genetics of human alcohol and aldehyde dehydrogenases.
- Adv. Hum. Genet. 15: 249-290, 1986.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 06/25/1996
- carol: 10/21/1993
- carol: 10/15/1993
- carol: 4/28/1993
- supermim: 3/16/1992
- supermim: 3/20/1990
- supermim: 2/2/1990
-
- *RECORD*
- *FIELD* NO
- 103720
- *FIELD* TI
- *103720 ALCOHOL DEHYDROGENASE-2; ADH2
- ADH, BETA SUBUNIT
- *FIELD* TX
- See 103700 for evidence on the mapping of the ADH2 gene in the cluster
- of related genes on 4q22. According to the conclusion of Smith et al.
- (1973), locus ADH2 is expressed in the lung in early fetal life and
- remains active in this tissue throughout life. It is active also in
- liver after about the first trimester and gradually increases in
- activity so that in adults this locus is responsible for most of the
- liver ADH activity. It is active in the adult kidney. The 'atypical pH
- ratio' phenotype is probably determined by a variant allele at the ADH2
- locus. Stamatoyannopoulos et al. (1975) found that 85% of Japanese carry
- an atypical liver ADH (ADH2 type). About the same proportion have
- alcohol sensitivity, which they suggest may be due to increased
- formation of acetaldehyde by persons with the atypical ADH. Bosron et
- al. (1980) found new molecular forms of human ADH, collectively
- designated ADH(Indianapolis), in 29% of liver specimens from black
- Americans. Three different Indianapolis ADH phenotypes were identified
- by starch gel electrophoresis and 4 isolated by affinity and
- ion-exchange chromatography. One is a homodimer of a newly discovered
- subunit. The other 3 are heterodimers of this new subunit and the known
- subunits, alpha, beta-1, and gamma-1. Agarwal et al. (1981) could find
- no instance of the Indianapolis variant in Germany or Japan; it may be
- confined to American blacks. Bosron et al. (1983) concluded that the
- Indianapolis phenotypes reflect polymorphism at the ADH2 locus with the
- variant ADH(Indianapolis) allele coding for the beta-Indianapolis
- subunit. The frequency of this allele was 0.16 in black Americans and
- was not found in any of 63 livers from white Americans. The frequency of
- alleles at the ADH3 locus also differs in these 2 populations.
-
- The ADH1, ADH2, and ADH3 loci code for 3 closely related polypeptides:
- alpha, beta, and gamma, respectively. Two additional ADH isozymes, pi
- and chi, encoded by the ADH4 and ADH5 loci, respectively, differ from
- the first three in a number of properties and are not related to them.
- The primary structure of the beta subunit (Hempel et al., 1985) and the
- nucleotide sequence of the cDNA corresponding to beta mRNA (Heden et
- al., 1986) have been determined. Yokoyama et al. (1987) cloned the gene
- coding for the beta-1 subunit of human ADH, the 'typical' subunit
- encoded by the ADH2*1 allele. A phylogenetic tree for the class I human
- ADHs, alpha, beta, and gamma, showed that the alpha and beta subunits
- diverged most recently and that their common ancestor diverged from the
- ancestor of the gamma subunit earlier. The evolutionary rates of
- nucleotide substitution for the 3 subunits showed that the gamma subunit
- is evolving at the slowest rate, followed by beta and alpha, in that
- order, implying that the gamma subunit may be providing the original
- function of ethanol metabolism. Trezise et al. (1989) cloned and
- sequenced cDNA encoding baboon liver alcohol dehydrogenase. From the
- sequence they concluded that baboon liver class I ADH is of the same
- ancestral lineage as human ADH-beta; 363 of 374 residues were identical
- in the 2 amino acid sequences. They estimated that the primate class I
- ADH gene duplication predated the primate radiation and that the
- alpha/beta-gamma separation of human ADH genes occurred about 60 million
- years ago. Goedde et al. (1992) presented extensive data on population
- frequencies of the ADH2 and ALDH2 (100650) genes.
-
- Muramatsu et al. (1995) used the PCR/RFLP method to determine the
- genotypes of the ADH2 and ALDH2 loci of alcoholic and nonalcoholic
- Chinese living in Shanghai. They found that the alcoholics had
- significantly lower frequencies of the ADH2*2 and ALDH2*2 alleles than
- did the nonalcoholics, suggesting the inhibitory effects of these
- alleles for the development of alcoholism. In the nonalcoholic subjects,
- ADH2*2 had little, if any, effect, despite the significant effect of the
- ALDH2*2 allele in decreasing the alcohol consumption of the individual.
- Taken together, these results were considered consistent with the
- proposed hypothesis for the development of alcoholism, i.e., drinking
- behavior is greatly influenced by the individual's genotype of
- alcohol-metabolizing enzymes and the risk of becoming alcoholic is
- proportionate with the ethanol consumption of the individual.
-
- Takeshita et al. (1996) evaluated the effects of the ADH2 polymorphism
- in 524 Japanese individuals who had previously been typed for the ALDH2
- polymorphism. In the ALDH2*1/ALDH2*2 heterozygotes, the frequency of
- facial flushing following consumption of one glass of beer was
- significantly higher in the presence of the ADH2*2 alleles in homozygous
- or heterozygous form. The proportion of individuals with ethanol-induced
- cutaneous erythema was also higher depending on the presence of the ADH2
- allele in ALDH2*1 homozygotes or ALDH2*1/ALDH2*2 heterozygotes.
- Takeshita et al. (1996) presented evidence that drinking habits were not
- significantly associated with the ADH2 genotype.
-
- Higuchi et al. (1996) reported that higher ADH2*1 and ADH3*2 allele
- frequencies were observed in alcoholics than in controls. Their results
- suggested that genetic variations in ethanol oxidizing activities are
- involved in the development of alcoholism but that these variations do
- not have a specific effect in alcoholics with inactive ALDH2, a group at
- low genetic risk for alcoholism.
-
- Data on gene frequencies of allelic variants were tabulated by
- Roychoudhury and Nei (1988).
-
- *FIELD* AV
- .0001
- ALCOHOL DEHYDROGENASE, BETA SUBUNIT, 'TYPICAL'/'ATYPICAL'
- ADH2*1/ADH2*2
- ADH2, ARG47HIS
- Matsuo et al. (1989) showed that the typical and atypical forms of ADH-2
- differ by only a single amino acid. In the ADH2*2 ('atypical') allele,
- CAC codes for histidine at residue 47; in the corresponding codon of the
- ADH2*1 (typical) allele, CGC codes for arginine. Surprisingly, no silent
- substitutions were found between the coding regions of the 2 alleles
- over the 1,122 nucleotide sites. The kinetic properties of human alcohol
- dehydrogenases with various substitutions at residue 47 in the coenzyme
- binding site differ considerably. The V(max) of ethanol oxidation
- differs by 100-fold between beta-1/beta-1 and beta-2/beta-2 (i.e., the
- homozygotes for the ADH2*1 and ADH2*2 alleles, respectively). Using
- site-directed mutagenesis, Hurley et al. (1990) studied the effects of
- substitution of lysine, histidine, glutamine, and glycine for
- arginine-47 in beta-1/beta-1. They expressed the enzymes in E. coli and
- compared their kinetics.
-
- .0002
- ALCOHOL DEHYDROGENASE, BETA SUBUNIT, INDIANAPOLIS
- ADH2*3
- ADH2, ARG369CYS
- Burnell et al. (1987) demonstrated that in the homozygote for the beta*3
- allele, formerly called beta(Indianapolis), the only difference from the
- homozygote for the beta*1 allele was a single nucleotide change that
- resulted in substitution of cysteine for arginine at position 369.
- Burnell et al. (1987) predicted that arg369 interacts with the
- nicotinamide phosphate moiety of NAD(H) and that this accounts for the
- effect of the arg369-to-cys substitution in decreasing the isoenzyme's
- affinity for coenzyme.
-
- *FIELD* SA
- Duester et al. (1984); Xu et al. (1988); Yin et al. (1984)
- *FIELD* RF
- 1. Agarwal, D. P.; Meier-Tackmann, D.; Harada, S.; Goedde, H. W.:
- A search for the Indianapolis-variant of human alcohol dehydrogenase
- in liver autopsy samples from North Germany and Japan. Hum. Genet. 59:
- 170-171, 1981.
-
- 2. Bosron, W. F.; Li, T.-K.; Vallee, B. L.: New molecular forms of
- human liver alcohol dehydrogenase: isolation and characterization
- of ADH (Indianapolis). Proc. Nat. Acad. Sci. 77: 5784-5788, 1980.
-
- 3. Bosron, W. F.; Magnes, L. J.; Li, T.-K.: Human liver alcohol dehydrogenase:
- ADH(Indianapolis) results from genetic polymorphism at the ADH-2 gene
- locus. Biochem. Genet. 21: 735-744, 1983.
-
- 4. Burnell, J. C.; Carr, L. G.; Dwulet, F. E.; Edenberg, H. J.; Li,
- T.-K.; Bosron, W. F.: The human beta(3) alcohol dehydrogenase subunit
- differs from beta-1 by a cys for arg-369 substitution which decreases
- NAD(H) binding. Biochem. Biophys. Res. Commun. 146: 1227-1233,
- 1987.
-
- 5. Duester, G.; Hatfield, G. W.; Buhler, R.; Hempel, J.; Jornvall,
- H.; Smith, M.: Molecular cloning and characterization of cDNA for
- the beta subunit of human alcohol dehydrogenase. Proc. Nat. Acad.
- Sci. 81: 4055-4059, 1984.
-
- 6. Goedde, H. W.; Agarwal, D. P.; Fritze, G.; Meier-Tackmann, D.;
- Singh, S.; Beckmann, G.; Bhatia, K.; Chen, L. Z.; Fang, B.; Lisker,
- R.; Paik, Y. K.; Rothhammer, F.; Saha, N.; Segal, B.; Srivastava,
- L. M.; Czeizel, A.: Distribution of ADH-2 and ALDH2 genotypes in
- different populations. Hum. Genet. 88: 344-346, 1992.
-
- 7. Heden, L.-O.; Hoog, J.-O.; Larsson, K.; Lake, M.; Lagerholm, E.;
- Holmgren, A.; Vallee, B. L.; Jornvall, H.; von Bahr-Lindstrom, H.
- : cDNA clones coding for the beta-subunit of human liver alcohol dehydrogenase
- have differently sized 3-prime-non-coding regions. FEBS Lett. 194:
- 327-332, 1986.
-
- 8. Hempel, J.; Holmquist, B.; Fleetwood, L.; Kaiser, R.; Barros-Soderling,
- J.; Buhler, R.; Vallee, B. L.; Jornvall, H.: Structural relationships
- among class I isozymes of human liver alcohol dehydrogenase. Biochemistry 24:
- 5303-5307, 1985.
-
- 9. Higuchi, S.; Muramatsu, T.; Matsushita, S.; Murayama, M.; Hayashida,
- M.: Polymorphisms of ethanol-oxidizing enzymes in alcoholics with
- inactive ALDH2. Hum. Genet. 97: 413-434, 1996.
-
- 10. Hurley, T. D.; Edenberg, H. J.; Bosron, W. F.: Expression and
- kinetic characterization of variants of human beta-1/beta-1 alcohol
- dehydrogenase containing substitutions at amino acid 47. J. Biol.
- Chem. 265: 16366-16372, 1990.
-
- 11. Matsuo, Y.; Yokoyama, R.; Yokoyama, S.: The genes for human alcohol
- dehydrogenases beta-1 and beta-2 differ by only one nucleotide. Europ.
- J. Biochem. 183: 317-320, 1989.
-
- 12. Muramatsu, T.; Zu-Cheng, W.; Yi-Ru, F.; Kou-Bao, H.; Heqin, Y.;
- Yamada, K.; Higuchi, S.; Harada, S.; Kono, H.: Alcohol and aldehyde
- dehydrogenase genotypes and drinking behavior of Chinese living in
- Shanghai. Hum. Genet. 96: 151-154, 1995.
-
- 13. Roychoudhury, A. K.; Nei, M.: Human Polymorphic Genes: World
- Distribution. New York: Oxford Univ. Press (pub.) 1988.
-
- 14. Smith, M.; Hopkinson, D. A.; Harris, H.: Studies on the subunit
- structure and molecular size of the human dehydrogenase isozymes determined
- by the different loci, ADH(1), ADH(2), and ADH(3). Ann. Hum. Genet. 36:
- 401-414, 1973.
-
- 15. Stamatoyannopoulos, G.; Chen, S.-H.; Fukui, M.: Liver alcohol
- dehydrogenase in Japanese: high population frequency of atypical form
- and its possible role in alcohol sensitivity. Am. J. Hum. Genet. 27:
- 789-796, 1975.
-
- 16. Takeshita, T.; Mao, X.-Q.; Morimoto, K.: The contribution of
- polymorphism in the alcohol dehydrogenase beta subunit to alcohol
- sensitivity in a Japanese population. Hum. Genet. 97: 409-413, 1996.
-
- 17. Trezise, A. E. O.; Godfrey, E. A.; Holmes, R. S.; Beacham, I.
- F.: Cloning and sequencing of cDNA encoding baboon liver alcohol
- dehydrogenase: evidence for a common ancestral lineage with the human
- alcohol dehydrogenase beta subunit and for class I ADH gene duplications
- predating primate radiation. Proc. Nat. Acad. Sci. 86: 5454-5458,
- 1989.
-
- 18. Xu, Y.; Carr, L. G.; Bosron, W. F.; Li, T.-K.; Edenberg, H. J.
- : Genotyping of human alcohol dehydrogenases at the ADH2 and ADH3
- loci following DNA sequence amplification. Genomics 2: 209-214,
- 1988.
-
- 19. Yin, S.-J.; Bosron, W. F.; Li, T.-K.; Ohnishi, K.; Okuda, K.;
- Ishii, H.; Tsuchiya, M.: Polymorphism of human liver alcohol dehydrogenase:
- identification of ADH(2)2-1 and ADH(2)2-2 phenotypes in the Japanese
- by isoelectric focusing. Biochem. Genet. 22: 169-180, 1984.
-
- 20. Yokoyama, S.; Yokoyama, R.; Rotwein, P.: Molecular characterization
- of cDNA clones encoding the human alcohol dehydrogenase beta-1 and
- the evolutionary relationship to the other class I subunits alpha
- and gamma. Jpn. J. Genet. 62: 241-256, 1987.
-
- *FIELD* CN
- Moyra Smith - updated: 03/13/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 03/13/1996
- terry: 3/13/1996
- mark: 3/13/1996
- mark: 8/22/1995
- pfoster: 4/5/1994
- warfield: 3/31/1994
- mimadm: 2/11/1994
- carol: 6/9/1992
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 103730
- *FIELD* TI
- *103730 ALCOHOL DEHYDROGENASE-3; ADH3
- ADH, GAMMA SUBUNIT
- *FIELD* TX
- See 103700 for evidence on the mapping of the ADH3 gene to the cluster
- of related genes on 4q22. According to the conclusion of Smith et al.
- (1973), the ADH3 locus is active in intestine and kidney in fetal and
- early postnatal life. Two alleles at the ADH3 locus, called 1 and 2,
- have a frequency of about 0.63 and 0.37, respectively. Hoog et al.
- (1986) determined the cDNA and amino acid structures of the gamma-1 and
- gamma-2 subunits of human liver alcohol dehydrogenase. These subunits
- are determined by allelic genes at the ADH3 locus, just as the beta-1
- and beta-2 and beta-Indianapolis subunits are determined by alleles at
- the ADH2 locus (103720). Morris et al. (1989) described a polymorphic
- anonymous DNA marker, D4S138, which is closely linked to the ADH3 locus.
-
- Data on gene frequencies of allelic variants were tabulated by
- Roychoudhury and Nei (1988).
-
- *FIELD* AV
- .0001
- ALCOHOL DEHYDROGENASE, GAMMA-1 TYPE
- ADH3*1
- ADH3, ARG271,ILE349
- Hoog et al. (1986) found 2 amino acid differences between gamma-1 and
- gamma-2: at position 349, isoleucine was found in gamma-1 and valine in
- gamma-2; at position 271, arginine was found in gamma-1 and glutamine in
- gamma-2. Xu et al. (1988) used the ile349-to-val substitution to
- distinguish ADH3*1 from ADH3*2 by means of allele-specific
- oligonucleotide probes.
-
- .0002
- ALCOHOL DEHYDROGENASE, GAMMA-2 TYPE
- ADH3*2
- ADH3, GLN271,VAL349
- See 103730.0001.
-
- *FIELD* SA
- Azevedo et al. (1976)
- *FIELD* RF
- 1. Azevedo, E. S.; Da Silva, M. C. B. O.; Tavares-Neto, J.: Human
- alcohol dehydrogenase ADH 1, ADH 2 and ADH 3 loci in a mixed population
- of Bahia, Brazil. Ann. Hum. Genet. 39: 321-327, 1976.
-
- 2. Hoog, J.-O.; Heden, L.-O.; Larsson, K.; Jornvall, H.; von Bahr-Lindstrom,
- H.: The gamma-1 and gamma-2 subunits of human liver alcohol dehydrogenase:
- cDNA structures, two amino acid replacements, and compatibility with
- changes in the enzymatic properties. Europ. J. Biochem. 159: 215-218,
- 1986.
-
- 3. Morris, D. J.; Willem, P.; dos Santos, M.; Povey, S.; Jenkins,
- T.: A new chromosome 4q marker, D4S138, closely linked to the ADH3
- locus. (Abstract) Cytogenet. Cell Genet. 51: 1047-1048, 1989.
-
- 4. Roychoudhury, A. K.; Nei, M.: Human Polymorphic Genes: World Distribution.
- New York: Oxford Univ. Press (pub.) 1988.
-
- 5. Smith, M.; Hopkinson, D. A.; Harris, H.: Studies on the subunit
- structure and molecular size of the human dehydrogenase isozymes determined
- by the different loci, ADH(1), ADH(2), and ADH(3). Ann. Hum. Genet. 36:
- 401-414, 1973.
-
- 6. Xu, Y.; Carr, L. G.; Bosron, W. F.; Li, T.-K.; Edenberg, H. J.
- : Genotyping of human alcohol dehydrogenases at the ADH2 and ADH3
- loci following DNA sequence amplification. Genomics 2: 209-214,
- 1988.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 2/11/1994
- carol: 3/20/1992
- supermim: 3/16/1992
- carol: 2/29/1992
- carol: 1/27/1992
- carol: 12/3/1990
-
- *RECORD*
- *FIELD* NO
- 103735
- *FIELD* TI
- *103735 ALCOHOL DEHYDROGENASE-6; ADH6
- *FIELD* TX
- Yasunami et al. (1991) used cross-hybridization with the ADH2 cDNA probe
- to isolate a 'new' ADH gene. cDNA clones corresponding to the gene were
- derived from PCR-amplified libraries as well. The coding sequence of a
- 368-amino acid long open reading frame was interrupted by introns into 8
- exons and spanned approximately 17 kb of genome. The gene contains a
- glucocorticoid response element at the 5-prime region. The transcript
- was detected in stomach and liver. The deduced amino acid sequence of
- the open reading frame showed about 60% positional identity with known
- human ADHs. This extent of homology is comparable to interclass
- similarity within the human ADH family. Thus, the newly identified gene,
- designated ADH6, governs synthesis of an enzyme that belongs to another
- class of ADHs, presumably with a distinct physiologic function.
-
- *FIELD* RF
- 1. Yasunami, M.; Chen, C.-S.; Yoshida, A.: A human alcohol dehydrogenase
- gene (ADH6) encoding an additional class of isozyme. Proc. Nat.
- Acad. Sci. 88: 7610-7614, 1991.
-
- *FIELD* CD
- Victor A. McKusick: 9/27/1991
-
- *FIELD* ED
- supermim: 3/16/1992
- carol: 9/27/1991
-
- *RECORD*
- *FIELD* NO
- 103740
- *FIELD* TI
- *103740 ALCOHOL DEHYDROGENASE, PI ISOZYME
- ALCOHOL DEHYDROGENASE-4; ADH4;;
- ADH, CLASS II
- *FIELD* TX
- Li et al. (1977) described a functionally distinct form of human liver
- alcohol dehydrogenase and termed it Pi-alcohol dehydrogenase.
- Variability from person to person was found, suggesting genetic
- variability. At intoxicating levels of alcohol, this enzyme may account
- for as much as 40% of the total ethanol oxidation rate. Unlike the other
- alcohol dehydrogenases, this type is not inhibited by pyrazole; hence,
- its name. It is called into operation at high levels of ethanol. It
- differs immunologically from other alcohol dehydrogenases and also has
- different substrate specificities; e.g., ethylene glycol is digested by
- other alcohol dehydrogenases but not by the Pi form. ADH4 (pi) isozyme,
- characteristic of adult liver, was termed class II by Vallee and Bazzone
- (1983), who referred to ADH5 (chi; 103710) as class III. In addition to
- the distinct loci determining alcohol dehydrogenase listed here, there
- are probably several others as yet not characterized. Mardh et al.
- (1986) presented evidence that Pi-ADH has a physiological role in the
- degradation of circulating epinephrine and norepinephrine. McPherson et
- al. (1989) used a combination of somatic cell hybrid DNA analysis and in
- situ hybridization to localize the ADH4 gene locus to human chromosome
- 4q22 in the cluster of alcohol dehydrogenase genes. Edman and Maret
- (1992) described RFLPs for the ADH4 and ADH5 genes. Linkage
- disequilibrium was detected between RFLPs in several of the 5 genes in
- the ADH cluster on chromosome 4. The disequilibrium between ADH4 and
- ADH5 indicated a hitherto unknown physical proximity of these 2 genes of
- different ADH classes, class II and class III, respectively.
-
- *FIELD* RF
- 1. Edman, K.; Maret, W.: Alcohol dehydrogenase genes: restriction
- fragment length polymorphisms for ADH4 (pi-ADH) and ADH5 (chi-ADH)
- for construction of haplotypes among different ADH classes. Hum.
- Genet. 90: 395-401, 1992.
-
- 2. Li, T.-K.; Bosron, W. F.; Dafeldecker, W. P.; Lange, L. G.; Vallee,
- B. L.: Isolation of PI-alcohol dehydrogenase of human liver: is it
- a determinant of alcoholism?. Proc. Nat. Acad. Sci. 74: 4378-4381,
- 1977.
-
- 3. Mardh, G.; Dingley, A. L.; Auld, D. S.; Vallee, B. L.: Human class
- II (pi) alcohol dehydrogenase has a redox-specific function in norepinephrine
- metabolism. Proc. Nat. Acad. Sci. 83: 8908-8912, 1986.
-
- 4. McPherson, J. D.; Smith, M.; Wagner, C.; Wasmuth, J. J.; Hoog,
- J.-O.: Mapping of the class II alcohol dehydrogenase gene locus to
- 4q22. (Abstract) Cytogenet. Cell Genet. 51: 1043 only, 1989.
-
- 5. Vallee, B. L.; Bazzone, T. J.: Isozymes of human liver alcohol
- dehydrogenase. In: Rattazzi, M. C.; Scandalios, J. G.; Whitt, G. S.
- : Isozymes. Current Topics in Biological and Medical Research.
- New York: Alan R. Liss (pub.) 8: 1983. Pp. 219-244.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- davew: 8/9/1994
- jason: 6/16/1994
- carol: 5/27/1994
- pfoster: 3/31/1994
- mimadm: 2/11/1994
- carol: 4/2/1993
-
- *RECORD*
- *FIELD* NO
- 103780
- *FIELD* TI
- 103780 ALCOHOLISM
- *FIELD* TX
- The tendency for drinking patterns of children to resemble those of
- their parents has been recognized since antiquity, e.g., in the
- observations of Plato and Aristotle (Warner and Rosett, 1975).
- Alcoholism is probably a multifactorial, genetically influenced disorder
- (Goodwin, 1976). The genetic influence is indicated by studies showing
- that (1) there is a 25 to 50% lifetime risk for alcoholism in sons and
- brothers of severely alcoholic men; (2) alcohol preference can be
- selectively bred for in experimental animals; (3) there is a 55% or
- higher concordance rate in monozygotic twins with only a 28% rate for
- like-sex dizygotic twins; and (4) half-brothers with different fathers
- and adopted sons of alcoholic men show a rate of alcoholism more like
- that of the biologic father than that of the foster father. A possible
- biochemical basis is a metabolic difference such that those prone to
- alcoholism have higher levels of a metabolite giving pleasurable effects
- or those not prone to alcoholism have higher levels of a metabolite
- giving unpleasant effects. Schuckit and Rayses (1979) found that, after
- a moderate dose of alcohol, blood acetaldehyde levels were elevated more
- in young men with alcoholic parents or sibs than in controls. A certain
- degree of organ specificity in the pathologic effects of alcohol is
- observed. For example, patients have cardiomyopathy, cirrhosis or
- pancreatitis but rarely more than one of these. A genetic basis of organ
- specificity is evident in Wernicke-Korsakoff syndrome (277730) and
- pancreatitis from type V hyperlipidemia (238400). Cloninger (1987)
- identified 2 separate heritable types of alcoholism. Type 1 alcohol
- abuse had its usual onset after the age of 25 years and was
- characterized by severe psychological dependence and guilt. It occurred
- in both men and women and required both genetic and environmental
- factors to become manifest. By contrast, type 2 alcohol abuse had its
- onset before the age of 25; persons with this type of alcoholism were
- characterized by their inability to abstain from alcohol and by frequent
- aggressive and antisocial behavior. Type 2 alcoholism was rarely found
- in women and was much more heritable. Abnormalities in platelet
- monoamine oxidase activity were found only in type 2 alcoholics (Von
- Knorring et al., 1985). See comments by Omenn (1988). Crabb (1990)
- reviewed biologic markers for increased risk of alcoholism. Aston and
- Hill (1990) performed complex segregation analysis of 35
- multigenerational families ascertained through a pair of male
- alcoholics. They concluded that liability to alcoholism is, in part,
- controlled by a major effect with or without additional multifactorial
- effects. However, mendelian transmission of this major effect was
- rejected, as was the hypothesis that the major effect is due to a single
- major locus. The candidate gene approach was used by Blum et al. (1990)
- and by Bolos et al. (1990) to investigate a possible relationship of the
- dopamine D2 receptor (DRD2; 126450) to alcoholism. Although Blum et al.
- (1990) suggested an association between a particular allele at the DRD2
- locus, Bolos et al. (1990) could not confirm this. In family studies,
- Bolos et al. (1990) excluded linkage between alcoholism and the DRD2
- locus.
-
- *FIELD* SA
- Propping et al. (1981)
- *FIELD* RF
- 1. Aston, C. E.; Hill, S. Y.: Segregation analysis of alcoholism
- in families ascertained through a pair of male alcoholics. Am. J.
- Hum. Genet. 46: 879-887, 1990.
-
- 2. Blum, K.; Noble, E. P.; Sheridan, P. J.; Montgomery, A.; Ritchie,
- T.; Jagadeeswaran, P.; Nogami, H.; Briggs, A. H.; Cohn, J. B.: Allelic
- association of human dopamine D(2) receptor gene in alcoholism. J.A.M.A. 263:
- 2055-2060, 1990.
-
- 3. Bolos, A. M.; Dean, M.; Lucas-Derse, S.; Ramsburg, M.; Brown, G.
- L.; Goldman, D.: Population and pedigree studies reveal a lack of
- association between the dopamine D(2) receptor gene and alcoholism.
- J.A.M.A. 264: 3156-3160, 1990.
-
- 4. Cloninger, C. R.: Neurogenetic adaptive mechanisms in alcoholism.
- Science 236: 410-416, 1987.
-
- 5. Crabb, D. W.: Biological markers for increased risk of alcoholism
- and for quantitation of alcohol consumption. J. Clin. Invest. 85:
- 311-315, 1990.
-
- 6. Goodwin, D.: Is Alcoholism Hereditary?. New York: Oxford Univ.
- Press (pub.) 1976.
-
- 7. Omenn, G. S.: Genetic investigations of alcohol metabolism and
- of alcoholism. Am. J. Hum. Genet. 43: 579-581, 1988.
-
- 8. Propping, P.; Kruger, J.; Mark, N.: Genetic disposition to alcoholism:
- an EEG study in alcoholics and their relatives. Hum. Genet. 59:
- 51-59, 1981.
-
- 9. Schuckit, M. A.; Rayses, V.: Ethanol ingestion: differences in
- blood acetaldehyde concentrations in relatives of alcoholics and controls.
- Science 203: 54-55, 1979.
-
- 10. Von Knorring, A.-L.; Bohman, M.; Von Knorring, L.; Oreland, L.
- : Platelet MAO activity as a biological marker in subgroups of alcoholism.
- Acta Psychiat. Scand. 72: 51-58, 1985.
-
- 11. Warner, R. H.; Rosett, H. L.: The effects of drinking on offspring:
- an historical survey of the American and British literature. J.
- Studies Alcohol 36: 1395-1420, 1975.
-
- *FIELD* CS
-
- Neuro:
- Alcoholism
-
- Misc:
- 25 to 50% lifetime risk for sons and brothers of severely alcoholic
- men
-
- Inheritance:
- Probably multifactorial, genetically influenced
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 4/14/1994
- carol: 4/6/1994
- supermim: 3/16/1992
- carol: 1/10/1991
- carol: 6/4/1990
- carol: 6/1/1990
-
- *RECORD*
- *FIELD* NO
- 103800
- *FIELD* TI
- *103800 ALDER ANOMALY
- *FIELD* TX
- Azurophilic cytoplasmic inclusions of the polymorphonuclear leukocytes
- were thought to be inherited as an autosomal dominant. Alder (1939)
- originally described the anomaly in a brother and sister who later at
- puberty developed changes in their hip joints. The brother was said to
- be in good health at age 28 (Davidson, 1961). This was, in fact, not
- true (Steinmann, 1994). Alder (1939) described the granules in a
- 9-year-old girl with scarlet fever. They persisted after recovery and
- were also detectable in the healthy 7-year-old brother, R.W., but not in
- 3 other sibs and not in the consanguineous parents. Gitzelmann et al.
- (1987) had the opportunity to examine R. W. and to study his fibroblasts
- which had only 2 to 3% residual arylsulfatase B activity but normal
- alpha-iduronidase activity. Thus, he clearly suffered from MPS VI
- (253200). Initially, Alder (1939) considered the granules as
- constitutional and harmless until the brother (R.W.) developed a
- waddling gait. Alder (1939) found in both sibs bony destruction in the
- shoulders, hips, and skull, and later in the knees and spine. R.W. had
- herniotomy at the age of 36 years, a decompressive laminectomy C1 to C7
- at age 50, hip replacement at age 51, and operation for aortic stenosis
- at age 60. He was very intelligent and a dedicated violin maker. His
- sister died early from an unknown cause. Thus, the granules that Alder
- (1939) first described are inherited as an autosomal recessive. They are
- part of MPS VI which is the mucopolysaccharidosis that shows the most
- striking leukocyte inclusions.
-
- Jordans (1947) reported a Dutch family showing a dominant inheritance
- pattern--9 affected persons in 3 generations with male-to-male
- transmission. The inclusions are probably morphologically
- indistinguishable from the Reilly granulations observed in
- mucopolysaccharidoses (Reilly, 1941).
-
- Francois et al. (1960) observed Alder anomaly and Fuchs atrophia gyrata
- chorioideae et retinae in the offspring of first-cousin parents, both of
- whom had the Alder anomaly. They suggested that the eye disorder is the
- homozygous expression of the Alder anomaly gene. It is possible, of
- course, that the eye disorder was merely an unrelated recessive disorder
- and indeed later observations (see Fuchs atrophia gyrata, 229900)
- supported this view.
-
- *FIELD* RF
- 1. Alder, A.: Ueber konstitutionell bedingte Granulationsveraenderungen
- der Leukocyten. Dtsch. Arch. Klin. Med. 183: 372-378, 1939.
-
- 2. Davidson, W. M.: Inherited variations in leucocytes. Brit. Med.
- Bull. 17: 190-195, 1961.
-
- 3. Francois, J.; Barbier, F.; De Rouck, A.: Les conducteurs du gene
- de l'atrophia gyrata chorioideae et retinae de Fuchs (anomalie d'Alder).
- Acta Genet. Med. Gemellol. 9: 74-91, 1960.
-
- 4. Gitzelmann, R.; Steinmann, B.; Wiesmann, U.; Spycher, M.; Herschkowitz,
- N.; Marti, H.-R.: Aldersche Granulationsanomalie: Albert Alders Patienten
- litten nicht an M. Pfaundler-Hurler. (Abstract) Helv. Paediat. Acta 42:
- 90 only, 1987.
-
- 5. Jordans, G. H. W.: Hereditary granulation anomaly of the leucocytes
- (Alder). Acta Med. Scand. 129: 348-351, 1947.
-
- 6. Reilly, W. A.: The granules in the leukocytes in gargoylism. Am.
- J. Dis. Child. 62: 489-491, 1941.
-
- 7. Steinmann, B.: Personal Communication. Zurich, Switzerland
- 12/9/1994.
-
- *FIELD* CS
-
- Heme:
- Azurophilic cytoplasmic neutrophil inclusions
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 1/19/1995
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- supermim: 2/17/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 103830
- *FIELD* TI
- *103830 ALDEHYDE REDUCTASE; ALR
- *FIELD* TX
- Petrash et al. (1981) studied aldose reductase (AR), aldose reductase M
- (ARM), and aldehyde reductase (ALR) in a variety of human tissues. Lens
- aldose reductase is composed of a single subunit with molecular weight
- 35K, and liver aldehyde reductase is composed of a single subunit of
- molecular weight 32K. Liver aldose reductase M is composed of 2
- nonidentical subunits of molecular weights 35K and 42K. Lens has only
- AR, liver has ARM and ALR, red cells have only ALR, while brain and
- placenta have all three enzymes. Petrash et al. (1981) suggested that
- three loci--alpha, beta, and delta--code for these enzymes, and that AR
- is a monomer of alpha polypeptide, ARM a dimer of alpha and beta
- subunits, and ALR a monomer of delta polypeptide.
-
- *FIELD* RF
- 1. Petrash, J. M.; Ansari, N. H.; Sadana, I.; Srivastava, S. K.:
- Biochemical and genetic interrelationship between aldose reductase,
- aldose reductase M and aldehyde reductase in human tissues. (Abstract) Am.
- J. Hum. Genet. 33: 52A only, 1981.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- carol: 10/24/1990
- supermim: 3/20/1990
- carol: 12/13/1989
- ddp: 10/27/1989
- root: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 103850
- *FIELD* TI
- *103850 ALDOLASE A, FRUCTOSE-BISPHOSPHATE; ALDOA
- FRUCTOSE-1,6-BISPHOSPHATE ALDOLASE A;;
- ALDOLASE A; ALDA;;
- ALDOLASE-1;;
- FRUCTOALDOLASE A
- ALDOLASE A DEFICIENCY, INCLUDED
- *FIELD* TX
- Fructose-1,6-bisphosphate aldolase (EC 4.1.2.13) is a glycolytic enzyme
- that catalyzes the reversible conversion of fructose-1,6-bisphosphate to
- glyceraldehyde 3-phosphate and dihydroxyacetone phosphate. The enzyme is
- a tetramer of identical 40,000-dalton subunits. Vertebrates have 3
- aldolase isozymes which are distinguished by their electrophoretic and
- catalytic properties. Electrophoretic variants were found by
- Charlesworth (1972). The amino acid sequence of the aldolases around the
- active site lysine is greatly conserved in evolution. Differences
- indicate that aldolases A, B (229600), and C (103870) are distinct
- proteins, the products of a family of related genes. Study of the genes
- is of interest because expression of the isozymes is regulated during
- development and because they represent the poorly characterized class of
- 'housekeeping genes' which are expressed in all cells. The developing
- embryo produces aldolase A, which is produced in even greater amounts in
- adult muscle where it can be as much as 5% of total cellular protein. In
- adult liver, kidney and intestine, aldolase A expression is repressed
- and aldolase B is produced. In brain and other nervous tissue, aldolase
- A and C are expressed about equally. In transformed liver cells,
- aldolase A replaces aldolase B.
-
- Freemont et al. (1988) presented the complete amino acid sequence of
- human skeletal muscle fructose-bisphosphate aldolase, comprising 363
- residues. Izzo et al. (1988) found that the cloned gene sequence of
- ALDOA spans 7,530 base pairs, includes 12 exons, and occurs as a single
- copy per haploid human genome. Eight exons containing the coding
- sequence were found to be common to all mRNAs extracted from several
- mammalian sources; 4 additional exons were found in the 5-prime
- untranslated region, 1 of which was contained in the ubiquitous type of
- mRNA, the second in the muscle-specific type of mRNA, and the third and
- fourth in a minor species of mRNA found in human liver tissue.
- S(1)-nuclease-protection analysis of the 5-prime end of mRNA from
- cultured fibroblasts, muscle, and hepatoma cell lines showed the
- existence of 4 different transcription-initiation sites. Also, the
- presence of conventional sequences for 4 eukaryotic promoters was
- demonstrated. The nucleotide similarities in the coding region and the
- intron-exon organization of aldolases A, B, and C confirm that they
- arose from a common ancestral gene and that aldolase B diverged first.
-
- Harris (1974) concluded that 3 loci determine aldolase. One group
- (Cohen-Haguenauer et al., 1985) assigned aldolase A to chromosome 16 and
- a second group (Kukita et al., 1985) assigned it to chromosome 22. The
- better evidence supported chromosome 16. Kukita et al. (1985) used
- Northern blot analysis of RNA isolated from human-mouse somatic cell
- hybrids and a cDNA clone for human aldolase mRNA. Later, however, Kukita
- et al. (1987) mapped ALDOA to chromosome 16 by 3 different methods:
- molecular hybridization to hybrid cell DNA, molecular hybridization to
- DNA of sorted metaphase chromosomes, and in situ hybridization. In situ
- hybridization indicated that the gene is located on the 16q22-q24 band.
- Serero et al. (1988) also assigned the aldolase A gene to chromosome 16
- by Southern blot analysis of human genomic DNA with a cDNA probe.
- Aldolase A pseudogenes were found on chromosomes 3 and 10. The map
- location of the 3 aldolase genes (ALDOA, ALDOB, ALDOC) and the aldolase
- pseudogene (see 229600) is of considerable interest from the point of
- view of chromosome evolution. The 4 genes are found on 2 pairs of
- morphologically similar chromosomes, 9 and 10, and 16 and 17. These
- homeologous (i.e., of similar origin) chromosome pairs may have arisen
- from 1 or 2 tetraploidization events (Comings, 1972; Ohno, 1973). As
- predicted by the chromosomal locations, the coding sequences of the
- expressed aldolase-A and -C genes (on chromosomes 16 and 17) are more
- homologous to each other than either of them is to the expressed
- aldolase-B gene (on chromosome 9).
-
- Beutler et al. (1973) described a son of first-cousin parents who had
- nonspherocytic hemolytic anemia, mental retardation and increased
- hepatic glycogen due, apparently, to deficiency of red cell aldolase.
- Puzzlingly, both parents had normal levels of red cell aldolase. The
- patient was presented again at the Birth Defects Conference in Vancouver
- in 1976 (Lowry and Hanson, 1977). He showed many dysmorphic features,
- some of which (ptosis, epicanthi, short neck, low posterior hairline)
- were reminiscent of the Noonan syndrome. The patient reported by Beutler
- et al. (1973) had an unstable enzyme which became depleted in enucleated
- erythrocytes. Consequently, energy production was impaired and membrane
- stability decreased with declining ion-transport activity. Hurst et al.
- (1987) described brother and sister with mental retardation, short
- stature, delayed puberty, hemolytic anemia, and an abnormal facial
- appearance. The similarities to the boy reported by Beutler et al.
- (1973) were striking.
-
- Kreuder et al. (1996) described a boy with aldolase deficiency who
- presented with predominantly myopathic symptoms, including muscle
- weakness and premature muscle fatigue. He had episodes of anemia and
- jaundice and was also prone to episodes of rhabdomyolysis during febrile
- illness. Biochemical assays revealed a profound reduction in muscle and
- red cell aldolase levels and a decrease in thermostability of residual
- enzyme. The aldolase A coding sequence was examined following RT-PCR of
- mRNA from peripheral blood mononuclear cells and muscle. The patient was
- found to be homozygous for a germline mutation in which a negatively
- charged glutamic acid is changed to a positively charged lysine at
- residue 206, a residue that is highly conserved within the subunit
- interface region.
-
- *FIELD* AV
- .0001
- ALDOLASE DEFICIENCY OF RED CELLS
- ALDOA, ASP128GLY
- Kishi et al. (1987) studied a case of red cell aldolase deficiency and
- found an A-G transversion at nucleotide 386 in the codon for the 128th
- amino acid, leading to a change from aspartic acid (GAU) to glycine
- (GGU) in the aldolase protein. The patient's enzyme from red cells and
- from cultured lymphoblastoid cells was found to be highly thermolabile,
- and the enzyme expressed in E. coli was likewise thermolabile. Since
- asp128 is conserved in aldolase A, -B, and -C of eukaryotes, including
- Drosophila, this residue is likely to have a crucial role in maintaining
- the correct spatial structure or in performing the catalytic function of
- the enzyme. The parents had intermediate levels of red cell aldolase A.
- The change in the second letter of the aspartic acid codon extinguished
- an Fok1 restriction site (GGATG to GGGTG). Southern blot analysis of the
- genomic DNA showed the patient to be homozygous for a mutation that was
- heterozygous in both parents.
-
- *FIELD* SA
- Miwa et al. (1981); Penhoet et al. (1966); Rottmann et al. (1984);
- Sakakibara et al. (1985); Tolan et al. (1987)
- *FIELD* RF
- 1. Beutler, E.; Scott, S.; Bishop, A.; Margolis, N.; Matsumoto, F.;
- Kuhl, W.: Red cell aldolase deficiency and hemolytic anemia: a new
- syndrome. Trans. Assoc. Am. Phys. 86: 154-166, 1973.
-
- 2. Charlesworth, D.: Starch-gel electrophoresis of four enzymes from
- human red blood cells: glyceraldehyde-3-phosphate dehydrogenase, fructoaldolase,
- glyoxalase II and sorbitol dehydrogenase. Ann. Hum. Genet. 35:
- 477-484, 1972.
-
- 3. Cohen-Haguenauer, O.; Van Cong, N.; Mennecier, F.; Kahn, A.; Frezal,
- J.: The human aldolase A gene is on chromosome 16.(Abstract) Cytogenet.
- Cell Genet. 40: 605, 1985.
-
- 4. Comings, D. E.: Evidence of ancient tetraploidy and conservation
- of linkage groups in mammalian chromosomes. Nature 238: 455-457,
- 1972.
-
- 5. Freemont, P. S.; Dunbar, B.; Fothergill-Gilmore, L. A.: The complete
- amino acid sequence of human skeletal-muscle fructose-bisphosphate
- aldolase. Biochem. J. 249: 779-788, 1988.
-
- 6. Harris, H.: Personal Communication. London, England 1974.
-
- 7. Hurst, J. A.; Baraitser, M.; Winter, R. M.: A syndrome of mental
- retardation, short stature, hemolytic anemia, delayed puberty, and
- abnormal facial appearance: similarities to a report of aldolase A
- deficiency. Am. J. Med. Genet. 28: 965-970, 1987.
-
- 8. Izzo, P.; Costanzo, P.; Lupo, A.; Rippa, E.; Paolella, G.; Salvatore,
- F.: Human aldolase A gene: structural organization and tissue-specific
- expression by multiple promoters and alternate mRNA processing. Europ.
- J. Biochem. 174: 569-578, 1988.
-
- 9. Kishi, H.; Mukai, T.; Hirono, A.; Fujii, H.; Miwa, S.; Hori, K.
- : Human aldolase A deficiency associated with a hemolytic anemia:
- thermolabile aldolase due to a single base mutation. Proc. Nat.
- Acad. Sci. 84: 8623-8627, 1987.
-
- 10. Kreuder, J.; Borkhardt, A.; Repp, R.; Pekrun, A.; Gottsche, B.;
- Gottschalk, U.; Reichmann, H.; Schachenmayr, W.; Schlegel, K.; Lampert,
- F.: Inherited metabolic myopathy and hemolysis due to a mutation
- in aldolase A. New Eng. J. Med. 334: 1100-1104, 1996.
-
- 11. Kukita, A.; Yoshida, M. C.; Fukushige, S.; Sakakibara, M.; Joh,
- K.; Mukai, T.; Hori, K.: Molecular gene mapping of human aldolase
- A (ALDOA) gene to chromosome 16. Hum. Genet. 76: 20-26, 1987.
-
- 12. Kukita, A.; Yoshida, M. C.; Sakakibara, M.; Mukai, T.; Hori, K.
- : Molecular gene mapping of the structural gene for human aldolase
- A (ALDOA) to chromosome 22.(Abstract) Cytogenet. Cell Genet. 40:
- 674, 1985.
-
- 13. Lowry, R. B.; Hanson, J. W.: Aldolase A deficiency with syndrome
- of growth and developmental retardation, midfacial hypoplasia, hepatomegaly,
- and consanguineous parents. Birth Defects Orig. Art. Ser. XIII(3B):
- 222-228, 1977.
-
- 14. Miwa, S.; Fujii, H.; Tani, K.; Takahashi, K.; Takegawa, S.; Fujinami,
- N.; Sakurai, M.; Kubo, M.; Tanimoto, Y.; Kato, T.; Matsumoto, N.:
- Two cases of red cell aldolase deficiency associated with hereditary
- hemolytic anemia in a Japanese family. Am. J. Hemat. 11: 425-437,
- 1981.
-
- 15. Ohno, S.: Ancient linkage groups and frozen accidents. Nature 244:
- 259-262, 1973.
-
- 16. Penhoet, E.; Rajkumar, T.; Rutter, W. I.: Multiple forms of fructose
- diphosphate aldolase in mammalian tissues. Proc. Nat. Acad. Sci. 56:
- 1275-1282, 1966.
-
- 17. Rottmann, W. H.; Tolan, D. R.; Penhoet, E. E.: Complete amino
- acid sequence for human aldolase B derived from cDNA and genomic clones.
- Proc. Nat. Acad. Sci. 81: 2738-2742, 1984.
-
- 18. Sakakibara, M.; Mukai, T.; Hori, K.: Nucleotide sequence of a
- cDNA clone for human aldolase: a messenger RNA in the liver. Biochem.
- Biophys. Res. Commun. 131: 413-420, 1985.
-
- 19. Serero, S.; Maire, P.; Van Cong, N.; Cohen-Haguenauer, O.; Gross,
- M. S.; Jegou-Foubert, C.; de Tand, M. F.; Kahn, A.; Frezal, J.: Localization
- of the active gene of aldolase on chromosome 16, and two aldolase
- A pseudogenes on chromosomes 3 and 10. Hum. Genet. 78: 167-174,
- 1988.
-
- 20. Tolan, D. R.; Niclas, J.; Bruce, B. D.; Lebo, R. V.: Evolutionary
- implications of the human aldolase-A, -B, -C, and -pseudogene chromosome
- locations. Am. J. Hum. Genet. 41: 907-924, 1987.
-
- *FIELD* CS
-
- Heme:
- Congenital nonspherocytic hemolytic anemia;
- Normocytic anemia;
- Normochromic anemia;
- Normal red cell osmotic fragility
-
- Skin:
- Jaundice
-
- GI:
- Splenomegaly;
- Cholelithiasis;
- Cholecystitis
-
- Neuro:
- Mental retardation reported
-
- Eyes:
- Ptosis;
- Epicanthus
-
- Neck:
- Short neck;
- Low posterior hairline
-
- Growth:
- Short stature
-
- Endocrine:
- Delayed puberty
-
- Lab:
- Aldolase A deficiency
-
- Inheritance:
- Autosomal recessive (16q22-q24)
-
- *FIELD* CN
- Moyra Smith - updated: 6/3/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 06/04/1996
- carol: 6/3/1996
- davew: 6/8/1994
- warfield: 4/7/1994
- carol: 4/6/1994
- mimadm: 3/11/1994
- supermim: 3/16/1992
- carol: 1/27/1992
-
- *RECORD*
- *FIELD* NO
- 103870
- *FIELD* TI
- *103870 ALDOLASE-3
- ALDOLASE C;;
- FRUCTOALDOLASE C; ALDC; ALDOC
- *FIELD* TX
- See aldolase-1 (103850). Rottmann et al. (1987) determined the complete
- amino acid sequence of aldolase C from recombinant genomic clones.
- Aldolase C was found to share 81% amino acid identity with aldolase A
- and 70% identity with aldolase B. The gene structure was found to be the
- same as that in other aldolase genes in birds and mammals, having 9
- exons separated by 8 introns, all in precisely the same positions, with
- only the intron sizes being different. Eight of the exons contained the
- protein coding region comprised of 363 amino acids. The entire gene is
- approximately 4 kb long. Tolan et al. (1987) reported the mapping of
- ALDOC to chromosome 17 by spot-blot analysis of sorted chromosomes.
- Rocchi et al. (1989) also mapped the gene and narrowed the assignment to
- 17cen-q21 by in situ hybridization. In addition, they corroborated the
- assignment of ALDOA (103850) to chromosome 16, and of ALDOB (229600) to
- chromosome 9. Buono et al. (1988) presented the complete nucleotide
- sequence of ALDOC.
-
- *FIELD* RF
- 1. Buono, P.; Paolella, G.; Mancini, F. P.; Izzo, P.; Salvatore, F.
- : The complete nucleotide sequence of the gene coding for the human
- aldolase C. Nucleic Acids Res. 16: 4733 only, 1988.
-
- 2. Rocchi, M.; Vitale, E.; Covone, A.; Romeo, G.; Santamaria, R.;
- Buono, P.; Paolella, G.; Salvatore, F.: Assignment of human aldolase
- C gene to chromosome 17, region cen--q21.1. Hum. Genet. 82: 279-282,
- 1989.
-
- 3. Rottmann, W. H.; Deselms, K. R.; Niclas, J.; Camerato, T.; Holman,
- P. S.; Green, C. J.; Tolan, D. R.: The complete amino acid sequence
- of the human aldolase C isozyme derived from genomic clones. Biochimie 69:
- 137-145, 1987.
-
- 4. Tolan, D. R.; Niclas, J.; Bruce, B. D.; Lebo, R. V.: Evolutionary
- implications of the human aldolase-A, -B, -C, and -pseudogene chromosome
- locations. Am. J. Hum. Genet. 41: 907-924, 1987.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 10/13/1993
- carol: 3/31/1992
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- root: 8/7/1989
-
- *RECORD*
- *FIELD* NO
- 103880
- *FIELD* TI
- *103880 ALDEHYDE REDUCTASE, ALDR1
- ALDOSE REDUCTASE, LOW Km
- *FIELD* TX
- See aldehyde reductase (103830). Aldose reductase (EC 1.1.1.21) is a
- member of the monomeric, NADPH-dependent aldoketoreductase family. It
- catalyzes the reduction of a number of aldehydes, including the aldehyde
- form of glucose, which is reduced to the corresponding sugar alcohol,
- sorbitol. Sorbitol is subsequently metabolized to fructose by sorbitol
- dehydrogenase. Under normal conditions, this pathway plays a minor role
- in glucose metabolism in most tissues. In diabetic hyperglycemia,
- however, cells undergoing insulin-independent uptake of glucose produce
- significant quantities of sorbitol. The sorbitol accumulates in cells
- because of its poor penetration across cellular membranes and its slow
- metabolism by sorbitol dehydrogenase. The resulting hyperosmotic stress
- to cells may be a cause of diabetic complications such as neuropathy,
- retinopathy, and cataracts. Chung and LaMendola (1989) cloned and
- sequenced the aldose reductase gene from a human placental cDNA library
- using antibodies against the bovine lens aldose reductase. The deduced
- amino acid sequence indicated that maturation of aldose reductase
- involves removal of the N-terminal methionine. Nishimura et al. (1990)
- also cloned the aldose reductase gene using synthetic oligonucleotide
- probes based on partial amino acid sequences of purified human psoas
- muscle aldose reductase.
-
- Graham et al. (1991) determined the structure and sequence of the ALDR1
- gene by analysis of cDNA and genomic clones. The gene extends over
- approximately 18 kb and consists of 10 exons, giving rise to a 1,384
- nucleotide mRNA, excluding the poly(A) tail. The gene codes for a
- 316-amino acid protein with a molecular mass of 35,858 Da. The exons
- range in size from 82 to 168 bp, whereas the introns range from 325 to
- about 7,160 bp. A major site of transcription initiation in liver was
- mapped to an adenine residue 31 nucleotides upstream from the the A of
- the ATG initiation codon. The promoter region of the gene contains a
- TATA (TATTTA) box and a CCAAT box, located 37 and 104 nucleotides
- upstream, respectively, from the transcription initiation site. Graham
- et al. (1991) found 4 Alu elements in the ALDR1 gene: two in intron 1
- and one each in introns 4 and 9. Using the PCR to amplify specifically
- the human AR sequence in hamster/human hybrid DNA and also in
- mouse/human monochromosome hybrids, Graham et al. (1991) assigned the
- gene to chromosome 7. The assignment was confirmed and regionalized to
- 7q35 by in situ hybridization to human metaphase chromosomes using a
- novel, rapid method.
-
- Brown et al. (1992) identified a putative pseudogene (ALDRP1) that
- contained no intronic sequences; the functional aldose reductase has 9
- introns. In addition, the homology was absent in the region 5-prime to
- the transcription start site for the cDNA, implying that regulatory
- elements such as the promoter were missing from the pseudogene. They
- mapped the pseudogene to chromosome 3 by PCR, using amplimers specific
- for it to amplify DNA from somatic cell hybrids.
-
- Using a cDNA clone encoding human aldose reductase, Bateman et al.
- (1993) mapped gene sequences to human chromosomes 1, 3, 7, 9, 11, 13,
- 14, and 18 by analysis of somatic cell hybrids. By in situ
- hybridization, sequences were localized to 1q32-q42, 3p12, 7q31-q35,
- 9q22, 11p14-p15, and 13q14-q21. As a putative functional ALDR1 gene had
- been mapped to chromosome 7 and a putative pseudogene (ALDRP1) to
- chromosome 3, the sequences on the other 7 chromosomes were thought to
- represent other active genes, non-aldose reductase homologous sequences,
- or pseudogenes.
-
- *FIELD* SA
- Graham et al. (1991)
- *FIELD* RF
- 1. Bateman, J. B.; Kojis, T.; Heinzmann, C.; Klisak, I.; Diep, A.;
- Carper, D.; Nishimura, C.; Mohandas, T.; Sparkes, R. S.: Mapping
- of aldose reductase gene sequences to human chromosomes 1, 3, 7, 9,
- 11, and 13. Genomics 17: 560-565, 1993.
-
- 2. Brown, L.; Hedge, P. J.; Markham, A. F.; Graham, A.: A human aldehyde
- dehydrogenase (aldose reductase) pseudogene: nucleotide sequence analysis
- and assignment to chromosome 3. Genomics 13: 465-468, 1992.
-
- 3. Chung, S.; LaMendola, J.: Cloning and sequence determination of
- human placental aldose reductase gene. J. Biol. Chem. 264: 14775-14777,
- 1989.
-
- 4. Graham, A.; Brown, L.; Hedge, P. J.; Gammack, A. J.; Markham, A.
- F.: Structure of the human aldose reductase gene. J. Biol. Chem. 266:
- 6872-6877, 1991.
-
- 5. Graham, A.; Heath, P.; Morten, J. E. N.; Markham, A. F.: The human
- aldose reductase gene maps to chromosome region 7q35. Hum. Genet. 86:
- 509-514, 1991.
-
- 6. Nishimura, C.; Matsuura, Y.; Kokai, Y.; Akera, T.; Carper, D.;
- Morjana, N.; Lyons, C.; Flynn, T. G.: Cloning and expression of human
- aldose reductase. J. Biol. Chem. 265: 9788-9792, 1990.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 11/27/1996
- mark: 2/26/1996
- carol: 4/6/1994
- carol: 9/21/1993
- carol: 12/21/1992
- carol: 6/3/1992
- supermim: 3/16/1992
- carol: 8/19/1991
-
- *RECORD*
- *FIELD* NO
- 103890
- *FIELD* TI
- *103890 ALDOSE REDUCTASE M; ARM
- *FIELD* TX
- See aldehyde reductase (103830).
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
- *FIELD* ED
- supermim: 3/16/1992
- carol: 10/24/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- root: 1/11/1988
- *RECORD*
- *FIELD* NO
- 103900
- *FIELD* TI
- #103900 ALDOSTERONISM, SENSITIVE TO DEXAMETHASONE
- GLUCOCORTICOID-SUPPRESSIBLE HYPERALDOSTERONISM; GSH;;
- GLUCOCORTICOID-REMEDIABLE ALDOSTERONISM; GRA;;
- ACTH-DEPENDENT HYPERALDOSTERONISM, SYNDROME OF;;
- HYPERALDOSTERONISM, FAMILIAL, TYPE 1
- *FIELD* TX
- A number sign (#) is used with this entry because of evidence that
- glucocorticoid-remediable aldosteronism is the result of an
- anti-Lepore-type fusion of the CYP11B2 (124080) and CYP11B1 (202010)
- genes. (The various hemoglobins Lepore (e.g., 142000.0019) have a fusion
- beta-type subunit that is delta globin at the NH2 end and beta globin at
- the COOH end. This chimeric structure results from nonhomologous pairing
- and unequal crossing-over between the contiguous delta and beta globin
- genes. The hemoglobins Lepore result from delta-beta fusion because the
- delta globin gene (142000) is located upstream from the beta globin gene
- (141900). The hemoglobins anti-Lepore, e.g., Hb Miyada (141900.0179) and
- Hb P(Nilotic) (141900.0215), are the reciprocal product of nonhomologous
- pairing and unequal crossing-over between the HBD and HBB genes; they
- are beta-delta fusion globins. In GRA, the 5-prime portion of the
- downstream gene is the 5-prime portion of the fusion gene; hence, it is
- an anti-Lepore fusion.)
-
- Sutherland et al. (1966) and Salti et al. (1969) described a father and
- son with hypertension, low plasma renin activity, and increased
- aldosterone secretion responsive to dexamethasone. Growth and sexual
- development were normal. At laparotomy the father was found to have
- multiple adrenocortical adenomas. This appears to be distinct from Conn
- syndrome (primary aldosteronism) which is not sensitive to
- dexamethasone. New and Peterson (1967) described 2 cases in a family.
- Giebink et al. (1973) studied 2 brothers and their mother who had
- glucocorticoid-remediable aldosteronism. Ganguly et al. (1981) showed
- that the paradoxic decline in plasma aldosterone when the patient is in
- the upright posture, usually observed in aldosterone-producing adenoma,
- is also seen in GSH. Thus, in patients with primary aldosteronism in
- whom GSH is suspected on the basis of young age and family history and a
- postural decline in plasma aldosterone is demonstrated, treatment with
- glucocorticoid should be given for 4 to 6 weeks before localization
- procedures are begun. Ganguly et al. (1981) studied 2 families, each
- with 3 affected persons. The diagnosis of hyperaldosteronism was
- established by failure of saline infusion to suppress plasma aldosterone
- normally and by the failure of furosemide or a low sodium diet to
- stimulate plasma renin activity. One family had basal serum potassium
- levels below 3.5 mmol per liter, whereas values were normal in the
- second family. Although primary aldosteronism is rare (about 2% of
- hypertensives have it), it has been subdivided into 3 types:
- aldosterone-producing adenoma (50-90% of cases), idiopathic form thought
- to be due to bilateral adrenal hyperplasia, and GSH (the rarest form).
- Mulrow (1981) speculated that the primary defect in GSH resides in the
- anterior pituitary gland. Experiments in animals have hinted at the
- existence of another aldosterone-regulating hormone, possibly
- originating in the pituitary. Mulrow (1981) asked: 'Is it possible that
- in the familial disorder of glucocorticoid-suppressible
- hyperaldosteronism, the pituitary gland is synthesizing or processing a
- more potent form of (a fragment of proopiomelanocortin) that enhances
- the response of the adrenal glomerulosa cell to normal concentrations of
- ACTH?' If the answer is 'yes,' GSH might appropriately be discussed in
- entry 176830. This hypothesis proved untrue, however.
-
- Aldosterone synthase (124080), like steroid 11-beta-hydroxylase
- (202010), is expressed in both adrenal fasciculata and glomerulosa; they
- are 95% identical (Mornet et al., 1989) and lie on chromosome 8q (Mornet
- et al., 1989; Chua et al., 1987). That they are immediately adjacent is
- indicated by the fact that a chimeric, anti-Lepore-like gene has been
- identified as the cause of glucocorticoid-remediable aldosteronism. In
- glucocorticoid-remediable aldosteronism (GRA, an alternative acronym for
- GSH) there are high levels of the abnormal adrenal steroids
- 18-oxocortisol and 18-hydroxycortisol. The hypertension, variable
- hyperaldosteronism, and abnormal steroid production are all under the
- control of ACTH and suppressible by glucocorticoids. The fusion gene has
- the promoter and some other 5-prime parts of the CYP11B2 gene. As is the
- practice with other hybrid genes, the details are given as an allelic
- variant of the gene that contributes the 5-prime portion; therefore, see
- 202010.0002.
-
- Glucocorticoid suppressible hyperaldosteronism is the result of CYP11B2
- activity under the control of ACTH (which normally regulates CYP11B1)
- and results from a unequal crossing-over involving the CYP11B1 and
- CYP11B2 genes. Normally, these genes are in the following orientation:
- 5-prime--CYP11B2--CYP11B1--3-prime; the hybrid anti-Lepore gene lies
- between CYP11B2 and CYP11B1 and has B1 sequence at its 5-prime end and
- B2 sequence at its 3-prime end. The breakpoints of the various hybrid
- genes that have been studied have been found to be 5-prime of intron 4.
- Pascoe et al. (1992) demonstrated that hybrid cDNAs containing 5-prime
- sequences from CYP11B1 and 3-prime sequences from CYP11B2, when
- transfected into COS-1 cells, resulted in aldosterone synthesis at near
- normal levels when the constructs contained up to the first 3 exons of
- CYP11B1, while those with 5 or more exons from CYP11B1 produced no
- detectable aldosterone.
-
- Gordon (1995) stated that 'in a study on approximately 1,000 descendants
- of an English convict transported to Australia in 1837 for highway
- robbery in Northamptonshire,' his colleagues and he had confirmed, in 21
- affected members thus far identified, the extraordinary phenotypic
- heterogeneity in glucocorticoid-remediable aldosteronism. The affected
- members were often normokalemic, and some remained normotensive until
- late in life. This disorder, which he referred to as familial
- hyperaldosteronism type 1, is associated with hybrid genes showing
- somewhat different crossover points linking the CYP11B1 and CYP11B2
- portions. To that extent, the disorder shows genetic heterogeneity;
- however, no other gene has been implicated in the syndrome of
- ACTH-dependent hyperaldosteronism.
-
- Pascoe et al. (1995) studied a French kindred in which 7 members had
- GSH; of the 7, 2 also had adrenal tumors and 2 other members of the
- family had micronodular adrenal hyperplasia. One of the adrenal tumors
- and the surrounding adrenal tissue had been removed, giving a rare
- opportunity to study the regulation and action of the hybrid
- CYP11B1/CYP11B2 gene causing the disease. The hybrid gene was
- demonstrated to be expressed at higher levels than either CYP11B1 or
- CYP11B2 in the cortex of the adrenal by RT-PCR and Northern blot
- analysis. In situ hybridization showed that both CYP11B1 and the hybrid
- chain were expressed in all 3 zones of the cortex. In cell culture
- experiments, hybrid gene expression was stimulated by ACTH, leading to
- increased production of aldosterone and the hybrid steroids
- characteristic of GSH. The genetic basis of the tumors and hyperplasia
- in this family was not known but may have been related to the
- duplication causing the hyperaldosteronism.
-
- Gates et al. (1996) described 2 large pedigrees with many subjects who
- had the abnormal chimeric gene associated with glucocorticoid remediable
- aldosteronism. Most of the affected members, who had only mild
- hypertension and normal biochemistry, were clinically indistinguishable
- from patients with essential hypertension. This suggested to the authors
- that GRA is an underdiagnosed condition.
-
- *FIELD* SA
- Ganguly et al. (1981); Grim and Weinberger (1980)
- *FIELD* RF
- 1. Chua, S. C.; Szabo, P.; Vitek, A.; Grzeschik, K.-H.; John, M.;
- White, P. C.: Cloning of cDNA encoding steroid 11-beta-hydroxylase
- (P450C11). Proc. Nat. Acad. Sci. 84: 7193-7197, 1987.
-
- 2. Ganguly, A.; Grim, C. E.; Bergstein, J.; Brown, R. D.; Weinberger,
- M. H.: Genetic and pathophysiologic studies of a new kindred with
- glucocorticoid-suppressible hyperaldosteronism manifest in three generations.
- J. Clin. Endocr. Metab. 53: 1040-1046, 1981.
-
- 3. Ganguly, A.; Grim, C. E.; Weinberger, M. H.: Anomalous postural
- aldosterone response in glucocorticoid-suppressible hyperaldosteronism.
- New Eng. J. Med. 305: 991-993, 1981.
-
- 4. Gates, L. J.; MacConnachie, A. A.; Lifton, R. P.; Haites, N. E.;
- Benjamin, N.: Variation of phenotype in patients with glucocorticoid
- remediable aldosteronism. J. Med. Genet. 33: 25-28, 1996.
-
- 5. Giebink, G. S.; Gotlin, R. W.; Biglieri, E. G.; Katz, F. H.: A
- kindred with familial glucocorticoid-suppressible aldosteronism. J.
- Clin. Endocr. 36: 715-723, 1973.
-
- 6. Gordon, R. D.: Heterogeneous hypertension. Nature Genet. 11:
- 6-9, 1995.
-
- 7. Grim, C. E.; Weinberger, M. H.: Familial, dexamethasone-suppressible,
- normokalemic hyperaldosteronism. Pediatrics 65: 597-604, 1980.
-
- 8. Mornet, E.; Dupont, J.; Vitek, A.; White, P. C.: Characterization
- of two genes encoding human steroid 11-beta-hydroxylase (P-45011-beta).
- J. Biol. Chem. 264: 20961-20967, 1989.
-
- 9. Mulrow, P. J.: Glucocorticoid-suppressible hyperaldosteronism:
- a clue to the missing hormone?. (Editorial) New Eng. J. Med. 305:
- 1013-1014, 1981.
-
- 10. New, M. I.; Peterson, R. E.: A new form of congenital adrenal
- hyperplasia. J. Clin. Endocr. 27: 300-305, 1967.
-
- 11. Pascoe, L.; Curnow, K. M.; White, P. C.: Mutations in the CYP11B1
- (11-beta-hydroxylase) and CYP11B2 (aldosterone synthase) genes causing
- CMOII deficiency, 11-hydroxylase deficiency and glucocorticoid suppressible
- hyperaldosteronism. (Abstract) Am. J. Hum. Genet. 51 (suppl.):
- A28, 1992.
-
- 12. Pascoe, L.; Jeunemaitre, X.; Lebrethon, M.-C.; Curnow, K. M.;
- Gomez-Sanchez, C. E.; Gasc, J.-M.; Saez, J. M.; Corvol, P.: Glucocorticoid-suppressible
- hyperaldosteronism and adrenal tumors occurring in a single French
- pedigree. J. Clin. Invest. 96: 2236-2246, 1995.
-
- 13. Salti, I. S.; Stiefel, M.; Ruse, J. L.; Laidlaw, J. C.: Non-tumorous
- 'primary' aldosteronism. I. Type relieved by glucocorticoid (glucocorticoid-remediable
- aldosteronism). Canad. Med. Assoc. J. 101: 1-10, 1969.
-
- 14. Sutherland, D. J.; Ruse, J. L.; Laidlaw, J. C.: Hypertension,
- increased aldosterone secretion and low plasma renin activity relieved
- by dexamethasone. Canad. Med. Assoc. J. 95: 1109-1119, 1966.
-
- *FIELD* CS
-
- Endocrine:
- Hypertension;
- Low plasma renin activity;
- Increased aldosterone secretion responsive to dexamethasone
-
- Growth:
- Normal growth
-
- GU:
- Normal sexual development
-
- Oncology:
- Multiple adrenocortical adenomas;
- Hyperaldosteronism;
- Failure of saline infusion to suppress plasma aldosterone;
- Failure of furosemide or low sodium diet to stimulate plasma renin
- activity;
- Low/normal basal serum potassium;
- High levels of 18-oxocortisol and 18-hydroxycortisol
-
- Inheritance:
- Autosomal dominant resulting from unequal crossing-over between CYP11B1
- and CYP11B2 genes
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 02/17/1996
- terry: 2/12/1996
- mark: 2/2/1996
- terry: 1/26/1996
- mark: 8/31/1995
- carol: 4/8/1994
- mimadm: 3/11/1994
- carol: 11/19/1992
- carol: 11/18/1992
-
- *RECORD*
- *FIELD* NO
- 103920
- *FIELD* TI
- 103920 ALLERGIC BRONCHOPULMONARY ASPERGILLOSIS
- *FIELD* TX
- Graves et al. (1979) described 2 brothers with identical HLA haplotypes
- and allergic bronchopulmonary aspergillosis. A barn near the residence
- of the brothers was identified as the probable source. Vithayasai et al.
- (1973) also reported familial allergic aspergillosis. However, in 35
- unrelated cases no HLA association was found (Flaherty et al., 1978).
-
- *FIELD* RF
- 1. Flaherty, D. K.; Surfus, J. E.; Geller, M.; Rosenberg, M.; Patterson,
- R.; Reed, C. E.: HLA frequencies in allergic bronchopulmonary aspergillosis.
- Clin. Allergy 8: 73-76, 1978.
-
- 2. Graves, T. S.; Fink, J. N.; Patterson, R.; Kurup, V. P.; Scanlon,
- G. T.: A familial occurrence of allergic bronchopulmonary aspergillosis.
- Ann. Intern. Med. 91: 378-382, 1979.
-
- 3. Vithayasai, V.; Hydes, J. S.; Florio, L.: Allergic aspergillosis
- in a family. Indian Med. J. 144: 564-566 and 600 only, 1973.
-
- *FIELD* CS
-
- Immunology:
- Allergic bronchopulmonary aspergillosis
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- pfoster: 3/30/1994
- mimadm: 3/11/1994
- carol: 3/31/1992
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 103950
- *FIELD* TI
- *103950 AL-M
- ALPHA-2-MACROGLOBULIN; A2M;;
- MACROGLOBULIN, ALPHA-2
- ALPHA-2-MACROGLOBULIN DEFICIENCY, INCLUDED
- *FIELD* TX
- This polymorphism, which has been demonstrated in Japanese persons, is
- distinct from Gm, Am, and haptoglobins. It is likewise distinct from Xm
- (314900), also a macroglobulin, as indicated by the autosomal
- inheritance and specific tests. Gene frequency of the allele whose
- product is demonstrated by the antiserum is about 0.16 in Japanese.
- Using a rabbit antihuman serum, Gallango and Castillo (1974) also
- described a polymorphism of alpha-2-macroglobulin. This may be separate
- from that described by Leikola et al. (1972). From comparison of the
- sequence of the subunit of human alpha-2-macroglobulin with those of C3
- (120700) and C4 (120810, 120820), Sottrup-Jensen et al. (1985) concluded
- that these 3 proteins, which all contain a unique activatable
- beta-cysteinyl-gamma-glutamyl thiol ester, have a common evolutionary
- origin. C5 (120900) also shows sequence homology to A2M. A2M maps,
- however, to chromosome 12 (Kan et al., 1985). Kan et al. (1985) isolated
- A2M cDNA clones from a human liver cDNA library by using synthetic
- oligonucleotides as hybridization probes. They then assigned the A2M
- locus to chromosome 12 by Southern blot analysis of DNA from a panel of
- mouse/human somatic cell hybrids, using A2M cDNA as a hybridization
- probe. Fukushima et al. (1988) assigned the A2M locus to 12p13.3-p12.3
- by in situ hybridization. Assignment of the A2M gene to human chromosome
- 12p13-p12.2 was confirmed by Marynen et al. (1989) by use of in situ
- hybridization and somatic cell hybrid DNA analysis. Devriendt et al.
- (1989) also assigned A2M to 12p13-p12 by analysis of somatic cell
- hybrids and in situ hybridization. They showed, furthermore, that a
- closely related gene for pregnancy-zone protein (PZP; 176420) and an A2M
- pseudogene map to the same region.
-
- Umans et al. (1994) found that the homologous gene in the mouse contains
- 36 exons, coding for a 4.8-kb cDNA. Including putative control elements
- in the 5-prime flanking region, the gene covers about 45 kb. The
- promoter region of the mouse A2m gene differed considerably from the
- known promoter sequences of the human and rat genes. Hilliker et al.
- (1992) showed that the gene is located on mouse chromosome 6 band F1-G3
- in a syntenic group that has its human counterpart on 12p13-p12.
-
- Matthijs et al. (1992) demonstrated that the A2M gene spans
- approximately 48 kb and consists of 36 exons, from 21 to 229 bp in size
- and with consensus splice sites. Intron sizes range from 125 bp to 7.5
- kb. The A2M gene is present in single copy in the haploid genome.
-
- By the electroimmunoassay of Laurell, Bergqvist and Nilsson (1979) found
- deficient alpha-2-macroglobulin in a 37-year-old man, his mother, and
- one daughter. Alpha-2-macroglobulin is, like alpha-1-antitrypsin,
- alpha-2-antiplasmin, and antithrombin III, a protease inhibitor. It
- inhibits many proteases, including trypsin, thrombin and collagenase.
- The deficient persons were apparently heterozygotes. No clinical
- disadvantage resulted from the deficiency. Poller et al. (1989) detected
- an alteration in the A2M gene in a patient with serum A2M deficiency and
- chronic lung disease since childhood. The alteration involved
- restriction sites detected with 10 different enzymes and was thought to
- have been caused by major deletion or rearrangement in the gene. Nine of
- the restriction enzymes used detected no polymorphism in 40 healthy
- control subjects and 39 patients with chronic obstructive pulmonary
- disease. The patient was heterozygous for the A2M alteration; Poller et
- al. (1989) suggested that this was responsible for the pulmonary
- disease.
-
- *FIELD* AV
- .0001
- ALPHA-2-MACROGLOBULIN POLYMORPHISM
- A2M, VAL1000ILE
- By direct genomic sequencing of the 2 exons encoding the bait region and
- the exon encoding the thiolester site in 30 healthy individuals and in
- 30 patients with chronic lung disease, Poller et al. (1992) found a
- sequence polymorphism near the thiolester site of the gene, changing
- val1000 (GTC) to ile (ATC); the 2 alleles had frequencies of 0.30 and
- 0.70, respectively. No difference of A2M serum levels was observed for
- these 2 alleles.
-
- .0002
- ALPHA-2-MACROGLOBULIN POLYMORPHISM
- A2M, CYS972TYR
- In 1 of the 30 patients and in none of the 30 healthy persons studied by
- Poller et al. (1992), a mutation within the thiolester site, changing
- cys972 (TGT) to tyr (TAT), was found. Since activation of the internal
- thiolester formed between cys972 and gln975 in each of the subunits of
- the tetrameric A2M molecule is involved in the covalent crosslinking of
- the activating proteinase, this mutation was predicted to interfere with
- A2M function. The A2M serum level was within the normal range in this
- patient.
-
- .0003
- ALPHA-2-MACROGLOBULIN POLYMORPHISM
- A2M, IVS1 DEL
- In 1 healthy individual, Poller et al. (1992) found a deletion of the
- intron that ordinarily separates exons 1 and 2. As a result, the 2 exons
- that code the bait domain of the alpha-2-macroglobulin gene were fused.
-
- .0004
- ALPHA-2-MACROGLOBULIN POLYMORPHISM
- A2M, ARG681HIS
- Matthijs et al. (1992) demonstrated an amino acid polymorphism in the
- bait domain of the alpha-2-macroglobulin molecule which defines the
- specific interaction of the molecule with proteinases. A G-to-A
- transition in exon 17 was detected in 1 person out of a group of 132
- tested. The change predicted an arginine-to-his substitution at position
- 681. In the mutant allele an MaeII restriction site was lost and a new
- NspHI site was created.
-
- *FIELD* SA
- Bell et al. (1985); David et al. (1987); Marynen et al. (1985)
- *FIELD* RF
- 1. Bell, G. I.; Rall, L. B.; Sanchez-Pescador, R.; Merryweather, J.
- P.; Scott, J.; Eddy, R. L.; Shows, T. B.: Human alpha-2-macroglobulin
- gene is located on chromosome 12. Somat. Cell Molec. Genet. 11:
- 285-289, 1985.
-
- 2. Bergqvist, D.; Nilsson, I. M.: Hereditary alpha-2-macroglobulin
- deficiency. Scand. J. Haemat. 23: 433-436, 1979.
-
- 3. David, F.; Kan, C. C.; Lucotte, G.: Two Taq I RFLPs for human
- alpha-2 macroglobulin (alpha-2M) using a full length cDNA probe. Nucleic
- Acids Res. 15: 374 only, 1987.
-
- 4. Devriendt, K.; Zhang, J.; van Leuven, F.; van den Berghe, H.; Cassiman,
- J. J.; Marynen, P.: A cluster of alpha 2-macroglobulin-related genes
- (alpha 2 M) on human chromosome 12p: cloning of the pregnancy-zone
- protein gene and an alpha 2M pseudogene. Gene 81: 325-334, 1989.
-
- 5. Fukushima, Y.; Bell, G. I.; Shows, T. B.: The polymorphic human
- alpha-2-macroglobulin gene (A2M) is located in chromosome region 12p12.3-p13.3.
- Cytogenet. Cell Genet. 48: 58-59, 1988.
-
- 6. Gallango, M. L.; Castillo, O.: Alpha-2-macroglobulin polymorphism:
- a new genetic system detected by immuno-electrophoresis. J. Immunogenet. 1:
- 147-151, 1974.
-
- 7. Hilliker, C.; Overbergh, L.; Petit, P.; Van Leuven, F.; Van den
- Berghe, H.: Assignment of mouse alpha-2-macroglobulin gene to chromosome
- 6 band F1-G3. Mammalian Genome 3: 469-471, 1992.
-
- 8. Kan, C.-C.; Solomon, E.; Belt, K. T.; Chain, A. C.; Hiorns, L.
- R.; Fey, G.: Nucleotide sequence of cDNA encoding human alpha-2-macroglobulin
- and assignment of the chromosomal locus. Proc. Nat. Acad. Sci. 82:
- 2282-2286, 1985.
-
- 9. Leikola, J.; Fudenberg, H. H.; Kasukawa, R.; Milgrom, F.: A new
- genetic polymorphism of human serum: alpha(2) macroglobulin (AL-M).
- Am. J. Hum. Genet. 24: 134-144, 1972.
-
- 10. Marynen, P.; Bell, G. I.; Cavalli-Sforza, L. L.: Three RFLPs
- associated with the human alpha-2-macroglobulin gene (A2M). Nucleic
- Acids Res. 13: 8287 only, 1985.
-
- 11. Marynen, P.; Zhang, J.; Devriendt, K.; Cassiman, J.-J.: Alpha-2-macroglobulin,
- pregnancy zone protein and an alpha-2-macroglobulin pseudogene map
- to chromosome 12p12.2-p13. (Abstract) Cytogenet. Cell Genet. 51:
- 1040 only, 1989.
-
- 12. Matthijs, G.; Devriendt, K.; Cassiman, J.-J.; van den Berghe,
- H.; Marynen, P.: Structure of the human alpha-2 macroglobulin gene
- and its promotor (sic). Biochem. Biophys. Res. Commun. 184: 596-603,
- 1992.
-
- 13. Poller, W.; Barth, J.; Voss, B.: Detection of an alteration of
- the alpha-2-macroglobulin gene in a patient with chronic lung disease
- and serum alpha-2-macroglobulin deficiency. Hum. Genet. 83: 93-96,
- 1989.
-
- 14. Poller, W.; Faber, J.-P.; Klobeck, G.; Olek, K.: Cloning of the
- human alpha-2-macroglobulin gene and detection of mutations in two
- functional domains: the bait region and the thiolester site. Hum.
- Genet. 88: 313-319, 1992.
-
- 15. Sottrup-Jensen, L.; Stepanik, T. M.; Kristensen, T.; Lonblad,
- P. B.; Jones, C. M.; Wierzbicki, D. M.; Magnusson, S.; Domdey, H.;
- Wetsel, R. A.; Lundwall, A.; Tack, B. F.; Fey, G. H.: Common evolutionary
- origin of alpha-2-macroglobulin and complement components C3 and C4.
- Proc. Nat. Acad. Sci. 82: 9-13, 1985.
-
- 16. Umans, L.; Serneels, L.; Hilliker, C.; Stas, L.; Overbergh, L.;
- De Strooper, B.; Van Leuven, F.; Van den Berghe, H.: Molecular cloning
- of the mouse gene coding for alpha-2-macroglobulin and targeting of
- the gene in embryonic stem cells. Genomics 22: 519-529, 1994.
-
- *FIELD* CS
-
- Pulmonary:
- Chronic lung disease
-
- Lab:
- Serum A2M deficiency
-
- Inheritance:
- Autosomal dominant (12p13.3-p12.3)
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 9/12/1994
- mimadm: 3/11/1994
- carol: 1/15/1993
- carol: 6/19/1992
- supermim: 3/16/1992
- carol: 2/29/1992
-
- *RECORD*
- *FIELD* NO
- 104000
- *FIELD* TI
- 104000 ALOPECIA AREATA
- *FIELD* TX
- Lubowe (1959) described a family with affected mother and affected
- daughter and son. Evidence suggests an autoimmune mechanism in this
- disorder. See autoimmune diseases (109100). Stankler (1979) observed
- onset in brother and sister at age 2, with regular and periodic
- synchronous exacerbation thereafter. One exacerbation was after mumps.
- In a white American family, Hordinsky et al. (1984) found alopecia
- universalis in 2 brothers and alopecia areata in the son of one of them.
- Valsecchi et al. (1985) found 6 cases in 3 generations and showed that
- all affected persons had the same haplotype, HLA-Aw32,B18. In 2 Israeli
- families, Zlotogorski et al. (1990) could find no linkage to HLA.
- Galbraith and Pandey (1989) suggested an association between the gene
- encoding the Km1 allotype of the immunoglobulin kappa light chain
- determinant and a chromosome 2 gene encoding susceptibility to alopecia
- areata, based on a significantly higher frequency of this allotype in
- patients with the disorder than in a reference population of 105 healthy
- subjects. Within the patient population, an association between the
- absence of detectable serum antibody and the Km1 allotype was observed.
-
- Among first-degree relatives of 348 severely affected patients, van der
- Steen et al. (1992) found that one of the parents was affected in 7%.
- Among the sibs, 3% had developed alopecia areata (AA), while AA was
- present in 2% of the children. Taking into account the age of the
- children, they estimated that the lifetime risk approached 6%. They
- concluded that the degree of involvement observed in the probands did
- not influence the frequency and type of AA present in their first-degree
- relatives.
-
- Galbraith and Pandey (1995) studied 2 polymorphic systems of tumor
- necrosis factor alpha (TNFA; 191160) in 50 patients with alopecia
- areata. The first bi-allelic TNFA polymorphism was detected in humans by
- Wilson et al. (1992); this involved a single base change from G to A at
- position -308 in the promoter region of the gene. The less common
- allele, A at -308 (called T2), shows an increased frequency in patients
- with IDDM, but this depends on the concurrent increase in HLADR3 with
- which T2 is associated. A second TNFA polymorphism, described by
- D'Alfonso and Richiardi (1994), also involves a G-to-A transition at
- position -238 of the gene. In alopecia areata, Galbraith and Pandey
- (1995) found that the distribution of T1/T2 phenotypes differed between
- patients with the patchy form of the disease and patients with
- totalis/universalis disease. There was no significant difference in the
- distribution of the phenotypes for the second system. The results
- suggested genetic heterogeneity between the 2 forms of alopecia areata
- and suggested that the TNFA gene is a closely linked locus within the
- major histocompatibility complex on chromosome 6 where this gene maps
- and may play a role in the pathogenesis of the patchy form of the
- disease.
-
- *FIELD* RF
- 1. D'Alfonso, S.; Richiardi, P. M.: A polymorphic variation in a
- putative regulation box of the TNFA promoter region. Immunogenetics 39:
- 150-154, 1994.
-
- 2. Galbraith, G. M. P.; Pandey, J. P.: Km1 allotype association with
- one subgroup of alopecia areata. Am. J. Hum. Genet. 44: 426-428,
- 1989.
-
- 3. Galbraith, G. M. P.; Pandey, J. P.: Tumor necrosis factor alpha
- (TNF-alpha) gene polymorphism in alopecia areata. Hum. Genet. 96:
- 433-436, 1995.
-
- 4. Hordinsky, M. K.; Hallgren, H.; Nelson, D.; Filipovich, A. H.:
- Familial alopecia areata: HLA antigens and autoantibody formation
- in an American family. Arch. Derm. 120: 464-468, 1984.
-
- 5. Lubowe, I. I.: The clinical aspects of alopecia areata, totalis,
- and universalis. Ann. N.Y. Acad. Sci. 83: 458-462, 1959.
-
- 6. Stankler, L.: Synchronous alopecia areata in two siblings: a possible
- viral aetiology. (Letter) Lancet I: 1303-1304, 1979.
-
- 7. Valsecchi, R.; Vicari, O.; Frigeni, A.; Foiadelli, L.; Naldi, L.;
- Cainelli, T.: Familial alopecia areata--genetic susceptibility or
- coincidence?. Acta Derm. Venerol. 65: 175-177, 1985.
-
- 8. van der Steen, P.; Traupe, H.; Happle, R.; Boezeman, J.; Strater,
- R.; Hamm, H.: The genetic risk for alopecia areata in first degree
- relatives of severely affected patients: an estimate. Acta Derm.
- Venerol. 72: 373-375, 1992.
-
- 9. Wilson, A. G.; di Giovine, F. S.; Blakemore, A. I. F.; Duff, G.
- W.: Single base polymorphism in the human tumour necrosis factor
- alpha (TNF-alpha) gene detectable by NcoI restriction of PCR product.
- Hum. Molec. Genet. 1: 353 only, 1992.
-
- 10. Zlotogorski, A.; Weinrauch, L.; Brautbar, C.: Familial alopecia
- areata: no linkage with HLA. Tissue Antigens 35: 40-41, 1990.
-
- *FIELD* CS
-
- Hair:
- Alopecia areata
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- terry: 11/17/1995
- mark: 10/6/1995
- mimadm: 3/11/1994
- carol: 1/19/1993
- carol: 3/25/1992
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 104100
- *FIELD* TI
- 104100 ALOPECIA CONGENITA WITH KERATOSIS PALMOPLANTARIS
- *FIELD* TX
- Stevanovic (1959) described a family with a dominant pattern of
- inheritance who had hyperkeratosis of the palms and soles and mild
- dystrophic changes of the fingernails.
-
- *FIELD* RF
- 1. Stevanovic, D. V.: Alopecia congenita. The incomplete dominant
- form of inheritance with varying expressivity. Acta Genet. Statist.
- Med. 9: 127-132, 1959.
-
- *FIELD* CS
-
- Hair:
- Alopecia congenita
-
- Skin:
- Hyperkeratosis of palms and soles
-
- Nails:
- Mildly dystrophic fingernails
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 6/4/1986
-
- *RECORD*
- *FIELD* NO
- 104110
- *FIELD* TI
- 104110 ALOPECIA, FAMILIAL FOCAL
- *FIELD* TX
- Headington and Astle (1987) described a 14-year-old girl and her mother
- who had patchy hair loss present from early childhood. When studied in
- transverse section, biopsy specimens from both women showed marked
- anagen-telogen transformation that appeared to be irreversible.
- Preservation of telogen epithelium with absence of inflammation and
- scarring distinguished familial focal alopecia from pseudopelade
- (alopecia cicatrisata) and from localized alopecia areata. They could
- find no description of similar cases.
-
- ('Anagen' refers to the growth phase of the cycle of activity of the
- hair follicle. 'Telogen' refers to the resting phase of the cycle of
- activity of the hair follicle. 'Catagen' refers to the involutional
- phase of the cycle of activity of the hair follicle.)
-
- *FIELD* RF
- 1. Headington, J. T.; Astle, N.: Familial focal alopecia: a new disorder
- of hair growth clinically resembling pseudopelade. Arch. Derm. 123:
- 234-237, 1987.
-
- *FIELD* CS
-
- Hair:
- Patchy hair loss
-
- Lab:
- Marked irreversible anagen-telogen transformation
-
- Inheritance:
- Autosomal dominant
-
- *FIELD* CN
- Victor A. McKusick - updated: 02/20/1997
-
- *FIELD* CD
- Victor A. McKusick: 3/31/1987
-
- *FIELD* ED
- mark: 02/20/1997
- terry: 2/12/1997
- mimadm: 3/11/1994
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- carol: 3/31/1987
-
- *RECORD*
- *FIELD* NO
- 104130
- *FIELD* TI
- *104130 ALOPECIA, PSYCHOMOTOR EPILEPSY, PYORRHEA, AND MENTAL SUBNORMALITY
- *FIELD* TX
- Shokeir (1977) observed this combination of abnormalities in 12 persons
- in 4 generations with male-to-male transmission. The alopecia was
- congenital, permanent, and universal. In those with alopecia, mental
- subnormality was noted in 8 and psychomotor epilepsy in 7. Periodontal
- disease was present in all. Timar et al. (1993) described a case that
- presumably represented a new mutation. In addition to congenital total
- permanent alopecia, psychomotor epilepsy, pyorrhea, and mental
- retardation, the child had a giant pigmented nevus over the lower back
- area on the left. Timar et al. (1993) suggested the designation Shokeir
- syndrome, but this runs the risk of confusion with the 2 Pena-Shokeir
- syndromes (208150, 214150) that already exist.
-
- *FIELD* RF
- 1. Shokeir, M. H. K.: Universal permanent alopecia, psychomotor epilepsy,
- pyorrhea and mental subnormality. Clin. Genet. 11: 13-17, 1977.
-
- 2. Timar, L.; Czeizel, A. E.; Koszo, P.: Association of Shokeir syndrome
- (congenital universal alopecia, epilepsy, mental subnormality and
- pyorrhea) and giant pigmented nevus. Clin. Genet. 44: 76-78, 1993.
-
- *FIELD* CS
-
- Hair:
- Congenital alopecia totalis
-
- Skin:
- Giant pigmented nevus
-
- Mouth:
- Periodontitis
-
- Neuro:
- Psychomotor seizures;
- Mental retardation
-
- Inheriance:
- Autosomal dominant
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mimadm: 3/11/1994
- carol: 10/19/1993
- supermim: 3/16/1992
- carol: 2/27/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
-
- *RECORD*
- *FIELD* NO
- 104145
- *FIELD* TI
- *104145 AFAMIN; AFM
- ALPHA-ALBUMIN; ALBA; ALB2
- *FIELD* TX
- Belanger et al. (1994) identified a fourth member of the albumin gene
- family, the others being albumin (ALB; 103600), alpha-fetoprotein (AFP;
- 104150), and vitamin D-binding protein (DBP; 139200). The 'new' gene,
- called alpha-albumin, was located 10 kb downstream from the AFP locus.
- The gene is selectively expressed in the liver at late stages of
- development. The mRNA sequence encodes a predicted secreted protein with
- the typical triple domain disulfide cross-linked structure. Comparisons
- of coding and promoter sequences suggested that ALBA could be a
- phylogenetic intermediate between the ALB and AFP genes. The
- developmental switch between ALBA gene activation and AFP gene
- repression suggested new regulatory interplays at the albumin locus and
- adult stage-specific ligand binding functions carried out by the ALBA
- gene product.
-
- The ALB and DBP genes diverged before the emergence of amphibians 500
- Myr ago, while the AFP gene evolved slowly after the amphibian/reptile
- separation 350 Myr ago. The fact that the 3 genes have remained closely
- linked in single copy per haploid genome suggests a selective advantage
- to their proximity, plausibly provided by shared cis-regulatory
- elements. The sequence of the 3 most closely related genes is
- 5-prime--ALB--AFP--ALBA--3-prime.
-
- Lichenstein et al. (1994) described the initial characterization of
- afamin and its cDNA and provided evidence that AFM is a novel member of
- the albumin family. This serum protein, with a molecular mass of 87,000
- Da, was purified to homogeneity and subjected to amino acid sequence
- analyses. These sequences were used to design oligonucleotide primers
- and to isolate a full-length cDNA. The amino acid sequence encoded by
- the cDNA was found to share strong similarity to albumin family members,
- including the characteristic pattern of cys residues observed in that
- family. The gene maps to chromosome 4 as do other members of the albumin
- gene family. The mapping was performed by PCR applied to a panel of
- somatic cell hybrids.
-
- Noteworthy distinctions among ALB family members include the following:
- concentrations in adult serum are 50 ng/ml for AFP, 350 microg/ml for
- DBP, 40 mg/ml for ALB, and 30 microg/ml for AFM. ALB is not
- N-glycosylated, AFP and DBP each have 1 potential N-glycosylation site,
- and AFM has 4 potential sites. ALB expresses 1 free thiol group that has
- been implicated in complex formation with cysteine, glutathione, and
- mercurial and gold compounds. In contrast, the other 3 have an even
- number of cys residues, are thought not to have a free thiol, and may
- not bind glutathione and mercurials as does ALB.
-
- Nishio and Dugaiczyk (1996) showed that the approximately 23-kb
- alpha-albumin gene contains 15 exons, the last of which is untranslated.
- The predicted protein has a 21-amino acid leader sequence followed by a
- 578-residue mature polypeptide. The exon structure is similar to that of
- the related genes for albumin, alpha-fetoprotein, and vitamin D-binding
- protein.
-
- *FIELD* RF
- 1. Belanger, L.; Roy, S.; Allard, D.: New albumin gene 3-prime adjacent
- to the alpha-1-fetoprotein locus. J. Biol. Chem. 269: 5481-5484,
- 1994.
-
- 2. Lichenstein, H. S.; Lyons, D. E.; Wurfel, M. M.; Johnson, D. A.;
- McGinley, M. D.; Leidli, J. C.; Trollinger, D. B.; Mayer, J. P.; Wright,
- S. D.; Zukowski, M. M.: Afamin is a new member of the albumin, alpha-fetoprotein,
- and vitamin D-binding protein gene family. J. Biol. Chem. 269: 18149-18154,
- 1994.
-
- 3. Nishio, H.; Dugaiczyk, A.: Complete structure of the human alpha-albumin
- gene, a new member of the serum albumin multigene family. Proc. Nat.
- Acad. Sci. 93: 7557-7561, 1996.
-
- *FIELD* CN
- Alan F. Scott - updated: 08/21/1996
-
- *FIELD* CD
- Victor A. McKusick: 9/22/1994
-
- *FIELD* ED
- mark: 08/21/1996
- marlene: 8/19/1996
- carol: 9/22/1994
-
- *RECORD*
- *FIELD* NO
- 104150
- *FIELD* TI
- *104150 ALPHA-FETOPROTEIN; AFP
- ALPHA-FETOPROTEIN, HEREDITARY PERSISTENCE OF, INCLUDED;;
- HPAFP, INCLUDED;;
- AFP DEFICIENCY, INCLUDED
- *FIELD* MN
- Alpha-fetoprotein (AFP) is a major plasma protein in the fetus, where it
- is produced by the yolk sac and liver. It is the fetal counterpart of
- serum albumin. The AFP gene maps to 4q11-q22, the same region as the
- albumin gene (Harper and Dugaiczyk, 1983). In the adult the plasma
- concentration of AFP is very low except when a tumor such as hepatoma or
- teratoma is present.
-
- In congenital nephrosis (256300), an autosomal recessive disorder
- frequent in Finland, alpha-fetoprotein is increased in the maternal
- blood and amniotic fluid--an expression of renal loss of protein. Loss
- into the amniotic fluid in cases of spina bifida and anencephaly is the
- basis of a screening test. AFP deficiency, which appears to be a benign
- genetic trait like analbuminemia, has been recorded in infants
- (Greenberg et al., 1992).
-
- Autosomal dominant hereditary persistence of alpha-fetoprotein is a
- clinically benign autosomal dominant condition characterized by
- continued expression of the AFP gene in adult life. Such elevated
- alpha-fetoprotein levels complicate the interpretation of findings in
- patients being screened for malignancy (e.g., hepatocellular carcinoma
- or teratoma) or in pregnant women being screened for neural tube defects
- or Down syndrome in the fetus. In 1 family there was a G-to-A transition
- at position -119 in a potential HNF1 (hepatocyte nuclear factor) binding
- site, highlighting the importance of this HNF1 binding site in the
- developmental regulation of the AFP gene (McVey et al., 1993).
-
- *FIELD* ED
- carol: 07/23/1996 marlene: 7/23/1996 joanna: 7/11/1996
-
- *FIELD* CD
- F. Clarke Fraser: 5/9/1996
- *FIELD* TX
- Alpha-fetoprotein is a major plasma protein in the fetus, where it is
- produced by the yolk sac and liver. In the adult its concentration is
- very low except when a tumor such as hepatoma or teratoma is present.
- The similarity in physical properties of AFP and albumin (103600) and
- the fact that their presence is inversely related suggested that AFP is
- the fetal counterpart of serum albumin. In the mouse, the
- alpha-fetoprotein and albumin genes are syntenic; presumably the same is
- true in man and this may represent an ontogenically significant
- arrangement with switch from AFP to albumin, comparable to the
- hemoglobin F to hemoglobin A switch. Mammalian AFP and serum albumin
- genes arose through duplication of an ancestral gene 300-500 Myr ago. By
- means of restriction endonuclease mapping, Ingram et al. (1981) showed
- that the AFP and albumin genes in the mouse are in tandem, 13.5 kb pairs
- apart, with the albumin gene on the 5-prime side of the AFP gene. Thus,
- they are transcribed from the same strand of DNA. The order is, however,
- different from that expected by analogy with the gamma and beta globin
- genes; with the presumed switch from AFP to albumin, one might expect
- their position to be reversed from that observed. An overall
- conservatism of 32% exists for DNA sequence of the 2 genes in the mouse
- and probably about the same in man (Ruoslahti and Terry, 1976). In mice,
- Tilghman and Belayew (1982) found a parallel accumulation of AFP and
- albumin mRNAs before birth, followed by a selective decrease in AFP mRNA
- after birth. The decrease in AFP mRNA was the result of decrease in
- transcription of the AFP gene, as measured by an in vitro nuclear
- transcription assay. They suggested a model for hepatic expression of
- the AFP and albumin gene cluster in which transcription of the 2 genes
- is activated simultaneously during differentiation and each gene is
- thereafter modulated independently in committed cells. Minghetti et al.
- (1985) found a high rate of silent substitutions for both
- alpha-fetoprotein and albumin genes, perhaps the highest so far reported
- for an expressed nuclear gene. The rates of effective substitution and
- amino acid changes were also very high but, in contrast to silent
- substitutions, they were found to be higher for alpha-fetoprotein than
- for albumin by about 70%. For alpha-fetoprotein, the rate of effective
- substitution may approach that for nonfunctional pseudogenes. This high
- rate suggests that alpha-fetoprotein can tolerate a great deal of
- molecular variation without its function being impaired. Hammer et al.
- (1986) described enhancer elements in the 5-prime flanking region of the
- mouse AFP gene. Gibbs et al. (1987) identified 4 types of repetitive
- sequence elements in the introns and flanking regions of the human AFP
- gene. One of these was apparently a novel structure designated Xba. The
- others were Alu, X, and Kpn elements. X, Xba, and Kbn elements are not
- present in the human albumin gene and Alu sequences are present in
- different positions. From phylogenetic evidence, it appears that Alu
- elements were inserted into the AFP gene at some time postdating the
- mammalian radiation, 85 million years ago.
-
- Direct confirmation of the assignment of the AFP gene to chromosome 4 by
- in situ hybridization was provided by Harper and Dugaiczyk (1983), who
- placed the gene in the q11-q22 region, the same region as the albumin
- gene. Magenis et al. (1989) used in situ hybridization to localize the
- ALB and the AFP genes to orangutan chromosome 3q11-q15 and gorilla
- chromosome 3q11-q22. Beattie and Dugaiczyk (1982) found extensive DNA
- sequence homology between human AFP and the third domain of serum
- albumin. AFP appears to have evolved more rapidly than albumin.
-
- In congenital nephrosis (256300), an autosomal recessive disorder
- frequent in Finland, alpha-fetoprotein is increased in the maternal
- blood and amniotic fluid--an expression of renal loss of protein. Loss
- into the amniotic fluid in cases of spina bifida and anencephaly is the
- basis of a screening test. Whether AFP increases in patients with
- analbuminemia is apparently not known. (AFP was not increased (Motulsky,
- 1983) in the instance of analbuminemia reported by Boman et al. (1976).)
- See 208900 for a discussion of the use of AFP in the diagnosis of
- ataxia-telangiectasia. Voigtlander and Vogel (1985) commented on the
- fact that not only is AFP low in maternal serum and amniotic fluid in
- pregnancies with a Down syndrome fetus but also serum albumin is low
- (according to most reports) in Down syndrome patients of all ages.
- (Total serum protein may be normal because of an increase in gamma
- globulins.) A defect in a regulatory mechanism common to the 2 proteins
- was suggested. Faucett et al. (1989) and Greenberg et al. (1992)
- documented AFP deficiency in 2 infants. One was found in the case of a
- 36-year-old woman who had amniocentesis for genetic indications;
- amniotic fluid AFP levels were undetectable and chromosome analysis
- showed a 46,XX pattern. The maternal serum AFP level was likewise
- undetectable. A healthy, term female infant was delivered. In the cord
- blood, AFP level was undetectable. The second mother had an
- amniocentesis because of low maternal serum AFP levels. Amniotic fluid
- AFP level was undetectable. Chromosome analysis showed a 46,XY pattern;
- a normal, term male infant was delivered. This appears to be a situation
- analogous to analbuminemia, and it is presumably a benign genetic trait
- like analbuminemia.
-
- Ferguson-Smith et al. (1984) reported an autosomal dominant hereditary
- persistence of alpha-fetoprotein. The proband was a 38-year-old woman
- found to have elevated AFP during pregnancy, as part of screening for
- neural tube defects. The level of AFP in the amniotic fluid was normal;
- the mother's elevation persisted after delivery. The infant and 2 of 3
- other children also had elevated serum AFP. Subsequently, 21 members of
- her family, including 9 males, were found to have elevated values.
- Although close linkage of HPAFP with GC (139200) was originally excluded
- (Ferguson-Smith et al., 1984), repeat GC typing with an improved
- technique of isoelectric focusing showed several misclassifications in
- the earlier study and the new calculations were consistent with linkage
- (lod, 1.7; theta, 0.0) (Ferguson-Smith et al., 1985). Ferguson-Smith et
- al. (1985) used a cDNA albumin probe which recognizes RFLPs at the ALB
- locus. No recombination was found between an ALB polymorphism and HPAFP
- (lod = 6.02; theta = 0). With the same ALB probe, in situ hybridization
- confirmed assignment to 4q11-q21.
-
- In the mouse liver, the adult basal levels of AFP mRNA is determined by
- a gene called raf (regulator of alpha-fetoprotein) (Olsson et al.,
- 1977), and the inducibility of AFP mRNA during regeneration is regulated
- by a gene termed rif (regulator of induction of alpha-fetoprotein)
- (Belayew and Tilghman, 1982). (The raf regulatory locus must not be
- confused with the RAF oncogene; see 164760.) The raf and rif genes are
- not linked to the AFP gene or to each other (Vogt et al., 1987); it is
- possible that these regulatory genes function through trans-acting
- regulatory factors that interact with cis-acting elements of the AFP
- gene. Watanabe et al. (1987) described experiments showing that the
- 5-prime flanking region of the human AFP gene contains transcription
- control elements with characteristics of enhancers. Vogt et al. (1987)
- identified in the mouse the transacting locus termed raf. The authors
- suggested that the mutation in the Scottish kindred reported by
- Ferguson-Smith et al. (1985) involves a DNA-binding sequence for the raf
- product. This sequence must be contained within the proximal 7.6 kb of
- DNA 5-prime to the AFP gene, as demonstrated in transgenic mouse strains
- in which integrated AFP gene constructs exhibited raf regulation. The
- evolutionarily related and closely linked albumin gene is not affected
- by raf, nor is another oncofetal protein, gamma-glutamyl transpeptidase
- (231950). However, raf does regulate the level of at least one other
- structural gene termed H19 (103280). Tilghman (1992) indicated that
- homologs of raf and rif had not been identified in humans. The only
- regulatory mutation in AFP of which she was aware mapped to the
- structural gene and resulted in persistence of AFP expression in adults.
-
- Staples (1986) demonstrated high serum AFP in 6 members of 2 generations
- of the family of a man with testicular carcinoma. Hereditary
- spherocytosis (182900) was segregating independently in this family.
- Staples (1986) also indicated that alcoholic steatosis of the liver can
- cause reversible elevation of AFP. Rose et al. (1989) reported a third
- family ascertained through a 42-year-old male who had 2 sibs and a
- daughter with elevated serum alpha-fetoprotein levels. Such elevated
- alpha-fetoprotein levels complicate the interpretation of findings in
- patients being screened for malignancy (e.g., hepatocellular carcinoma
- or teratoma) or in pregnant women being screened for neural tube defects
- or Down syndrome in the fetus. Greenberg et al. (1990) reported another
- family. A 33-year-old man, 2 of his sibs, and a daughter showed elevated
- serum AFP levels.
-
- *FIELD* AV
- .0001
- HEREDITARY PERSISTENCE OF ALPHA-FETOPROTEIN
- HPAFP
- AFP, G-A, -119
- As part of an extensive screening program for spina bifida, a large
- Scottish kindred spanning 5 generations was identified as having
- hereditary persistence of alpha-fetoprotein, a clinically benign
- autosomal dominant condition characterized by continued expression of
- the AFP gene in adult life. Affected persons had mean serum AFP levels
- 23-fold higher than normal controls. These raised levels were, however,
- far below the levels seen in the fetus. McVey et al. (1993) showed by
- sequence analysis of the 5-prime flanking sequences of the AFP gene that
- in this family a G-to-A transition at position -119 was associated with
- the trait. This substitution occurs in a potential HNF I binding site
- and increases the similarity of the sequence to a consensus HNF I
- recognition site. In a competitive gel retardation assay, the mutant
- sequence bound HNF-1-alpha (142410) more tightly than the wildtype
- sequence. Furthermore, 5-prime-flanking sequences of the human AFP gene
- containing the G-to-A substitution directed a higher level of
- chloramphenicol acetyltransferase (CAT) expression in transfected human
- hepatoma cells than the wildtype sequences. The findings not only
- provide an explanation for the findings in this family, but also
- highlight the importance of this HNF I binding site in the developmental
- regulation of the AFP gene. The substitution is similar to those that
- cause hereditary persistence of fetal hemoglobin (e.g., 142200.0026; a
- G-A substitution at -117 of the HBG1 gene).
-
- *FIELD* SA
- D'Eustachio et al. (1981); Eiferman et al. (1981); Gorin and Tilghman
- (1980); Jagodzinski et al. (1981); Morinaga et al. (1983); Sakai et
- al. (1985); Szpirer et al. (1984); Urano et al. (1984)
- *FIELD* RF
- 1. Beattie, W. G.; Dugaiczyk, A.: Structure and evolution of human
- alpha-fetoprotein deduced from partial sequence of cloned cDNA. Gene 20:
- 415-422, 1982.
-
- 2. Belayew, A.; Tilghman, S. M.: Genetic analysis of alpha-fetoprotein
- synthesis in mice. Molec. Cell. Biol. 2: 1427-1435, 1982.
-
- 3. Boman, H.; Hermodson, M.; Hammond, C. A.; Motulsky, A. G.: Analbuminemia
- in an American Indian girl. Clin. Genet. 9: 513-526, 1976.
-
- 4. D'Eustachio, P.; Ingram, R. S.; Tilghman, S. M.; Ruddle, F. H.
- : Murine alpha-fetoprotein and albumin: two evolutionarily linked
- proteins encoded on the same mouse chromosome. Somat. Cell Genet. 7:
- 289-294, 1981.
-
- 5. Eiferman, F. A.; Young, P. R.; Scott, R. W.; Tilghman, S. M.:
- Intragenic amplification and divergence in the mouse alpha-fetoprotein
- gene. Nature 294: 713-718, 1981.
-
- 6. Faucett, W. A.; Greenberg, F.; Rose, E.; Alpert, E.; Bancalari,
- L.; Kardon, N. B.; Mizjewski, G.; Knight, G.; Haddow, J. E.: Congenital
- deficiency of alpha-fetoprotein. (Abstract) Am. J. Hum. Genet. 45
- (suppl.): A259, 1989.
-
- 7. Ferguson-Smith, M. A.; May, H. M.; Aitken, D. A.; O'Hare, E.; Yates,
- J. R. W.; Gallagher, J.; Krumlauf, R.; Tilghman, S. M.: Hereditary
- persistence of alphafetoprotein (HPAFP); linkage studies with chromosome
- 4 markers. (Abstract) Cytogenet. Cell Genet. 37: 469, 1984.
-
- 8. Ferguson-Smith, M. A.; Yates, J. R. W.; Kelly, D.; Aitken, D. A.;
- May, H. M.; Krumlauf, R.; Tilghman, S. M.: Hereditary persistence
- of alphafetoprotein maps to the long arm of chromosome 4. (Abstract) Cytogenet.
- Cell Genet. 40: 628, 1985.
-
- 9. Gibbs, P. E. M.; Zielinski, R.; Boyd, C.; Dugaiczyk, A.: Structure,
- polymorphism, and novel repeated DNA elements revealed by a complete
- sequence of the human alpha-fetoprotein gene. Biochemistry 26:
- 1332-1343, 1987.
-
- 10. Gorin, M. B.; Tilghman, S. M.: Structure of the alpha-fetoprotein
- gene in the mouse. Proc. Nat. Acad. Sci. 77: 1351-1355, 1980.
-
- 11. Greenberg, F.; Faucett, A.; Rose, E.; Bancalari, L.; Kardon, N.
- B.; Mizejewski, G.; Haddow, J. E.; Alpert, E.: Congenital deficiency
- of alpha-fetoprotein. Am. J. Obstet. Gynec. 167: 509-511, 1992.
-
- 12. Greenberg, F.; Rose, E.; Alpert, E.: Hereditary persistence of
- alpha-fetoprotein. Gastroenterology 98: 1083-1085, 1990.
-
- 13. Hammer, R. E.; Krumlauf, R.; Camper, S. A.; Brinster, R. L.; Tilghman,
- S. M.: Diversity of alpha-fetoprotein gene expression in mice is
- generated by a combination of separate enhancer elements. Science 235:
- 53-58, 1986.
-
- 14. Harper, M. E.; Dugaiczyk, A.: Linkage of the evolutionarily-related
- serum albumin and alpha-fetoprotein genes within q11-22 of human chromosome
- 4. Am. J. Hum. Genet. 35: 565-572, 1983.
-
- 15. Ingram, R. S.; Scott, R. W.; Tilghman, S. M.: Alpha-fetoprotein
- and albumin genes are in tandem in the mouse genome. Proc. Nat.
- Acad. Sci. 78: 4694-4698, 1981.
-
- 16. Jagodzinski, L. L.; Sargent, T. D.; Yang, M.; Glackin, C.; Bonner,
- J.: Sequence homology between RNAs encoding rat alpha-fetoprotein
- and rat serum albumin. Proc. Nat. Acad. Sci. 78: 3521-3525, 1981.
-
- 17. Magenis, R. E.; Luo, X. Y.; Dugaiczyk, A.; Ryan, S. C.; Oosterhuis,
- J. E.: Chromosomal localization of the albumin and alpha-fetoprotein
- genes in the orangutan (Pongo pygmaeus) and gorilla (Gorilla gorilla).
- (Abstract) Cytogenet. Cell Genet. 51: 1037, 1989.
-
- 18. McVey, J. H.; Michaelides, K.; Hansen, L. P.; Ferguson-Smith,
- M.; Tilghman, S.; Krumlauf, R.; Tuddenham, E. G. D.: A G-to-A substitution
- in an HNF I binding site in the human alpha-fetoprotein gene is associated
- with hereditary persistence of alpha-fetoprotein (HPAFP). Hum. Molec.
- Genet. 2: 379-384, 1993.
-
- 19. Minghetti, P. P.; Law, S. W.; Dugaiczyk, A.: The rate of molecular
- evolution of alpha-fetoprotein approaches that of pseudogenes. Molec.
- Biol. Evol. 2: 347-358, 1985.
-
- 20. Morinaga, T.; Sakai, M.; Wegmann, T. G.; Tamaoki, T.: Primary
- structures of human alpha-fetoprotein and its mRNA. Proc. Nat. Acad.
- Sci. 80: 4604-4608, 1983.
-
- 21. Motulsky, A. G.: Personal Communication. Seattle, Wash. 1983.
-
- 22. Olsson, M.; Lindahl, G.; Ruoslahti, E.: Genetic control of alpha-fetoprotein
- synthesis in the mouse. J. Exp. Med. 145: 819-827, 1977.
-
- 23. Rose, E.; Greenberg, F.; Alpert, E.: Hereditary persistence of
- alpha fetoprotein. (Abstract) Am. J. Hum. Genet. 45 (suppl.): A61,
- 1989.
-
- 24. Ruoslahti, E.; Terry, W. D.: Alpha fetoprotein and serum albumin
- show sequence homology. Nature 260: 804-805, 1976.
-
- 25. Sakai, M.; Morinaga, T.; Urano, Y.; Watanabe, K.; Wegmann, T.
- G.; Tamaoki, T.: The human alpha-fetoprotein gene: sequence organization
- and the 5-prime flanking region. J. Biol. Chem. 260: 5055-5060,
- 1985.
-
- 26. Staples, J.: Alphafetoprotein, cancer, and benign conditions.
- (Letter) Lancet II: 1277, 1986.
-
- 27. Szpirer, J.; Levan, G.; Thorn, M.; Szpirer, C.: Gene mapping
- in the rat by mouse-rat somatic cell hybridization: synteny of the
- albumin and alpha-fetoprotein genes and assignment to chromosome 14.
- Cytogenet. Cell Genet. 38: 142-149, 1984.
-
- 28. Tilghman, S. M.: Personal Communication. Princeton, N. J.
- 8/12/1992.
-
- 29. Tilghman, S. M.; Belayew, A.: Transcriptional control of the
- murine albumin/alpha-fetoprotein locus during development. Proc.
- Nat. Acad. Sci. 79: 5254-5257, 1982.
-
- 30. Urano, Y.; Sakai, M.; Watanabe, K.; Tamaoki, T.: Tandem arrangement
- of the albumin and alpha-fetoprotein genes in the human genome. Gene 32:
- 255-261, 1984.
-
- 31. Vogt, T. F.; Solter, D.; Tilghman, S. M.: Raf, a trans-acting
- locus, regulates the alpha-fetoprotein gene in a cell-autonomous manner.
- Science 236: 301-303, 1987.
-
- 32. Voigtlander, T.; Vogel, F.: Low alpha-fetoprotein and serum albumin
- levels in Morbus Down may point to a common regulatory mechanism.
- Hum. Genet. 71: 276-277, 1985.
-
- 33. Watanabe, K.; Saito, A.; Tamaoki, T.: Cell-specific enhancer
- activity in a far upstream region of the human alpha-fetoprotein gene.
- J. Biol. Chem. 262: 4812-4818, 1987.
-
- *FIELD* CS
-
- Misc:
- Major fetal plasma protein produced by yolk sac and liver
-
- Lab:
- Elevated serum AFP with: Hepatoma;
- Teratoma;
- Alcoholic steatosis of the liver;
- Hereditary persistence of alpha-fetoprotein;
- Ataxia telangiectasia (208900);
- Elevated maternal serum and amniotic fluid AFP in: Congenital nephrosis
- (256300) pregnancy;
- Spina bifida or anencephalic pregnancy;
- Elevated maternal serum but normal amniotic fluid AFP in: Maternal
- hereditary persistence of AFP and normal fetus;
- Low maternal serum and amniotic fluid AFP in: Down syndrome pregnancy
-
- Inheritance:
- Autosomal dominant (4q11-q21)
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- carol: 07/23/1996
- carol: 7/18/1996
- marlene: 7/18/1996
- carol: 5/18/1996
- davew: 7/19/1994
- jason: 7/5/1994
- mimadm: 4/14/1994
- carol: 4/11/1994
- warfield: 4/7/1994
- carol: 10/14/1993
-
- *RECORD*
- *FIELD* NO
- 104155
- *FIELD* TI
- *104155 ALPHA-FETOPROTEIN ENHANCER-BINDING PROTEIN
- AT MOTIF-BINDING FACTOR; ATBF1
- *FIELD* TX
- Tissue-specific expression of the human alpha-fetoprotein (AFP) gene
- (104150) is strongly stimulated by an enhancer present 3.3 to 4.9 kb
- upstream of the transcription initiation site. One of the enhancer
- elements contains an AT-rich core sequence (AT motif). To determine the
- nuclear factor in hepatoma cell lines that interacts with the human AFP
- enhancer AT motif, Morinaga et al. (1991) screened a hepatoma cDNA
- expression library with an AFP enhancer fragment that bore the AT motif.
- They succeeded in isolating a cDNA that can code for an AT motif-binding
- factor, termed ATBF1. This was the largest DNA-binding protein
- identified to that time and the first protein shown to contain multiple
- homeodomains and multiple zinc finger motifs. The protein had a
- predicted mass of 306 kD and contained 4 homeodomains and 17 zinc finger
- motifs.
-
- By fluorescence in situ hybridization, Yamada et al. (1995) mapped the
- ATBF1 gene to 16q22.3-q23.1. Yamada et al. (1996) used fluorescence in
- situ hybridization to assign the Atbf1 gene to mouse chromosome 8E1.
-
- *FIELD* RF
- 1. Morinaga, T.; Yasuda, H.; Hashimoto, T.; Higashio, K.; Tamaoki,
- T.: A human alpha-fetoprotein enhancer-binding protein, ATBF1, contains
- four homeodomains and seventeen zinc fingers. Molec. Cell. Biol. 11:
- 6041-6049, 1991.
-
- 2. Yamada, K.; Ma, D.; Miura, Y.; Ido, A.; Tamaoki, T.; Yoshida, M.
- C.: Assignment of the ATBF1 transcription factor gene (Atbf1) to
- mouse chromosome band 8E1 by in situ hybridization. Cytogenet. Cell
- Genet. 75: 30-31, 1996.
-
- 3. Yamada, K.; Mirua, Y.; Scheidl, T.; Yoshida, M. C.; Tamaoki, T.
- : Assignment of the human ATBF1 transcription factor gene to chromosome
- 16q22.3-q23.1. Genomics 29: 552-553, 1995.
-
- *FIELD* CD
- Victor A. McKusick: 1/22/1992
-
- *FIELD* ED
- terry: 01/15/1997
- mark: 10/25/1995
- supermim: 3/16/1992
- carol: 1/22/1992
-
- *RECORD*
- *FIELD* NO
- 104160
- *FIELD* TI
- *104160 ALPHA-GLUCOSIDASE, NEUTRAL, AB FORM; GANAB
- *FIELD* TX
- Human tissues contain 2 isozymes of neutral alpha-glucosidase designated
- AB (GANAB) and C (GANC). Initially distinguished on the basis of
- differences in electrophoretic mobility in starch gel, the two have been
- shown to have other differences including those of substrate
- specificity. Martiniuk et al. (1982, 1983) assigned the GANAB locus to
- 11q13-qter by study of mouse-man hybrid cells. Since the AB form of
- mouse is not different electrophoretically from that in man, these
- workers used rocket immunoelectrophoresis to distinguish the enzymes
- from the 2 species.
-
- *FIELD* RF
- 1. Martiniuk, F.; Smith, M.; Desnick, R.; Astrin, K.; Mitra, J.; Hirschhorn,
- R.: Assignment of the gene for neutral alpha-glucosidase AB to chromosome
- 11. (Abstract) Am. J. Hum. Genet. 34: 173A only, 1982.
-
- 2. Martiniuk, F.; Smith, M.; Ellenbogen, A.; Desnick, R. J.; Astrin,
- K.; Mitra, J.; Hirschhorn, R.: Assignment of the gene for neutral
- alpha-glucosidase AB to chromosome 11. Cytogenet. Cell Genet. 35:
- 110-116, 1983.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- jason: 6/16/1994
- supermim: 3/16/1992
- carol: 8/23/1990
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
-
- *RECORD*
- *FIELD* NO
- 104170
- *FIELD* TI
- *104170 ALPHA-GALACTOSIDASE B; GALB
- N-ACETYL-ALPHA-D-GALACTOSAMINIDASE; NAGA
- LYSOSOMAL ALPHA-N-ACETYLGALACTOSAMINIDASE DEFICIENCY, INCLUDED;;
- SCHINDLER DISEASE, INCLUDED;;
- NEUROAXONAL DYSTROPHY, SCHINDLER TYPE, INCLUDED;;
- KANZAKI DISEASE, INCLUDED
- *FIELD* TX
- In a study of man-rodent somatic cell hybrids, de Groot et al. (1978)
- assayed human N-acetyl-alpha-D-galactosaminidase activity and concluded
- that alpha-galactosidase B and mitochondrial aconitase (ACO2; 100850),
- known to be on chromosome 22, are syntenic. They also obtained evidence
- for direct assignment of alpha-galactosidase B to chromosome 22.
- Alpha-NAGA was thought to be a more appropriate designation for this
- enzyme than alpha-galactosidase B by de Groot et al. (1978), who claimed
- that there was no structural relationship between alpha-gal A (on the X
- chromosome; GLA; 301500) and so-called alpha-gal B. However, DNA studies
- (Wang et al., 1990; Wang and Desnick, 1991), described later, led to a
- different conclusion. In man-rodent cell hybrids, Geurts van Kessel et
- al. (1979, 1980) studied chronic myeloid leukemia cells to determine the
- site of the break on 22q relative to markers assigned to chromosomes 22
- and 9. Alpha-NAGA remained with the Ph-1 chromosome, whereas ACO2 went
- with chromosome 9. Thus, the former is probably in band 22q11, whereas
- the latter is between it and 22qter.
-
- Wang et al. (1990) isolated a full-length 2.2-kb cDNA and a genomic
- cosmid clone containing the entire NAGA gene. Sequence analysis revealed
- striking similarities between the NAGA locus and exons 1-6 of
- alpha-galactosidase A, suggesting that the 2 genes evolved by
- duplication and divergence from a common ancestral locus. Wang and
- Desnick (1991) also pointed to remarkable amino acid identity between
- the NAGA and GLA genes.
-
- In 2 sons of a German couple with remote consanguinity, van Diggelen et
- al. (1987, 1988) described the clinical and biochemical features of
- lysosomal alpha-N-acetylgalactosaminidase deficiency. The boys showed
- neurologic abnormalities starting at age 9 months, followed by
- progressive psychomotor deterioration. By the age of 2.5 and 4 years,
- they had 'largely lost their previously acquired motor and language
- skills.' Growth had been normal. Computerized tomographic scans were
- normal, and there was no organomegaly, obvious coarsening of the facies,
- or skeletal dysplasia. A uniquely abnormal pattern of urinary
- oligosaccharides was demonstrated by thin-layer chromatography. Among
- the carbohydrate-hydrolyzing lysosomal enzymes, only alpha-NAGA had not
- previously been associated with a disorder. The levels of this enzyme
- were very low in cultured fibroblasts, leukocytes and plasma, whereas
- these levels were normal in a healthy brother. Both parents had low
- normal or reduced activity. A major neutral oligosaccharide from the
- urine of 1 patient was identified as the blood group A determinant, a
- trisaccharide with terminal alpha-N-acetylgalactosamine. The
- concentration of this product in the urine of the older boy, who was a
- secretor and had blood group A, was 5 times normal. The younger boy, who
- had blood group O, did not excrete this trisaccharide. Schindler et al.
- (1988) described the clinical findings as consisting of severe
- psychomotor retardation with myoclonic seizures, decorticate posture,
- optic atrophy, blindness, marked long tract signs, and total loss of
- contact with the environment. No features of other lysosomal storage
- diseases were present. Ultrastructural examination of peripheral nerves
- was unremarkable, whereas the rectal mucosa contained dystrophic
- autonomic axons with 'tubulovesicular' material. A unique pattern of
- abnormal urinary oligosaccharides/glycopeptides was found by thin layer
- chromatography. Wang et al. (1988) pointed out that the brothers
- reported by van Diggelen et al. (1987) had a clinical course and
- neuropathologic findings similar to those in Seitelberger disease, the
- infantile form of neuroaxonal dystrophy (256600). The characteristic
- 'spheroids' were observed histologically and ultrastructurally in
- terminal exons in gray matter. This disorder, which they referred to as
- Schindler disease, must represent, therefore, a form of infantile axonal
- dystrophy, the first in which a specific enzymatic defect has been
- identified. The disorder is autosomal recessive. Schindler et al. (1989)
- also characterized the disorder as a neuroaxonal dystrophy. They pointed
- out that although the disorder is caused by deficiency of a lysosomal
- enzyme, no lysosomal storage could be identified. It has been proposed
- that the dystrophic axons in infantile neuroaxonal dystrophy result from
- defective retrograde axonal transport. How deficiency of
- alpha-N-acetylgalactosaminidase might lead to a similar problem is not
- clear. Using PCR amplification and sequence analysis of PCR product from
- type I and type II offspring of consanguineous matings, Wang et al.
- (1990) demonstrated single basepair mutations in the homozygous state in
- both type I and type II. (Type I is classic Schindler disease; type II
- is an adult disorder with angiokeratoma as a prominent feature
- (104170.0002). Type II might appropriately be called Kanzaki disease
- (Kanzaki et al., 1989).)
-
- Keulemans et al. (1996) reported the genotypes of 5 more patients with
- NAGA deficiency. One of them, related to the first reported German
- family (van Diggelen et al., 1987), had classic Schindler disease and
- the same homozygous mutation, i.e., glu325to-lys (104170.0001). The only
- manifestations in another patient, a 5-year-old Dutch girl whose family
- was clinically described by de Jong et al. (1994), were convulsions
- during fever and psychomotor retardation starting after the age of 1
- year. She had 2 different mutations: glu325-to-lys inherited from her
- father and ser160-to-cys (104170.0004) inherited from her mother. The
- same genotype was found in a clinically unaffected 3.5-year-old brother
- of the proband. Keulemans et al. (1996) suggested that the brother might
- be a preclinical case of NAGA deficiency detected through screening. A
- homozygous nonsense mutation, glu193-to-ter, was found in 2 adult
- Spanish sibs who had angiokeratoma, lymphedema, and vacuolization in
- dermal cells, but no neurologic signs. These sibs, previously reported
- by Chabas et al. (1994), were clinically similar to the original patient
- described by Kanzaki et al. (1989). Although at the metabolic level the
- patients with NAGA deficiency are similar, extreme differences between
- the infantile form(s) and the adult form (Kanzaki disease) suggested to
- Keulemans et al. (1996) that other factors or genes contribute to the
- clinical heterogeneity.
-
- *FIELD* AV
- .0001
- SCHINDLER DISEASE
- NAGA, GLU325LYS
- In the first cases described with Schindler disease (van Diggelen et
- al., 1987, 1988), Wang et al. (1990) found a G-to-A transition at
- nucleotide 973 of the NAGA gene, resulting in substitution of lysine for
- glutamic acid as residue 325.
-
- .0002
- KANZAKI DISEASE
- SCHINDLER DISEASE, TYPE II
- LYSOSOMAL GLYCOAMINOACID STORAGE DISEASE WITH ANGIOKERATOMA CORPORIS
- DIFFUSUM
- NAGA, ARG329TRP
- In a 46-year-old Japanese woman with disseminated angiokeratoma, Kanzaki
- et al. (1989) demonstrated numerous cytoplasmic vacuoles in cells of the
- kidney and skin. Enzyme activities against synthetic and natural
- substrates were normal in leukocytes and fibroblasts. Her urine
- contained a large amount of sialylglycoaminoacids, with predominant
- excretion of an O-glycoside-linked glycoaminoacid. No information was
- provided on the patient's family. The enzyme studies excluded Fabry
- disease (301500), fucosidosis (230000), galactosialidosis (256540), and
- the various mucolipidoses and mucopolysaccharidoses. Desnick (1991)
- recounted reading an abstract by Kanzaki et al. (1988) in which the
- presence of angiokeratoma attracted his attention because of his
- longtime work with Fabry disease; the possibility that this disorder was
- related to Schindler disease was suggested by the excretion of large
- amounts of glycopeptides in the urine. A collaboration thereafter led to
- the demonstration that indeed there is deficiency of alpha-galactosidase
- B in Kanzaki disease also (Wang et al., 1990). Even though the disorder
- was much milder, with no neurodegeneration and no neuroaxonal dystrophy,
- the deficiency of enzymes seemed to be of the same order as in type I
- Schindler disease. In the laboratory of Desnick (1991), a substitution
- of tryptophan for arginine-329 was demonstrated as the basic defect
- (Wang et al., 1994). Again, it is remarkable that a change so close to
- that in Schindler disease could cause such a different phenotype. This
- situation is comparable to that of the Hurler and Scheie forms of
- mucopolysaccharidosis I and to the allelic mild and severe forms of many
- lysosomal storage diseases. Kanzaki et al. (1991) provided further
- evidence that there are 2 forms of alpha-N-acetylgalactosaminidase
- deficiency with sialopeptiduria: a severe infantile-onset form of
- neuroaxonal dystrophy without angiokeratoma or visceral lysosomal
- inclusions, and an adult-onset form with angiokeratoma, extensive
- lysosomal accumulation of sialoglycopeptides, and the absence of
- detectable neurologic involvement. Kanzaki et al. (1993) gave an
- extensive description of the 46-year-old Japanese woman with the adult
- form of lysosomal alpha-N-acetylgalactosaminidase deficiency. The
- angiokeratomas first appeared on her lower torso when she was 28 years
- old and later became diffusely distributed. Her 2 unaffected children
- had half-normal enzyme levels, consistent with autosomal recessive
- inheritance. The woman had mild intellectual impairment and peripheral
- neuroaxonal degeneration. She was the product of a first-cousin marriage
- and worked in a hospital as a nurse's aide. Endoscopic examination
- demonstrated telangiectasia on the gastric mucosa. Dilated blood vessels
- were present on the ocular conjunctiva and dilated vessels with
- corkscrewlike tortuosity were observed in the fundi.
-
- To identify the mutation causing this phenotypically distinct
- adult-onset form of NAGA deficiency, Wang et al. (1994) used reverse
- transcription, amplification, and sequencing of the NAGA transcript. The
- change was a C-to-T transition at nucleotide 985, resulting in an R329W
- amino acid substitution. The base substitution was confirmed by
- hybridization of PCR-amplified genomic DNA from family members with
- allele-specific oligonucleotides. Wang et al. (1994) showed that in
- transiently expressed COS-1 cells, both the E325K (infantile-onset) and
- R329W (adult-onset) precursors were processed to the mature form;
- however, the E325K mutant polypeptide was more rapidly degraded than the
- R329W subunit, thereby providing a basis for the distinctly different
- infantile- and adult-onset phenotypes.
-
- .0003
- KANZAKI DISEASE
- SCHINDLER DISEASE, TYPE II
- NAGA, GLU193TER
- Keulemans et al. (1996) showed by PCR and sequence analysis that the
- Spanish brother and sister with manifestations of Kanzaki disease
- described by Chabas et al. (1994) were homozygous for an E193X mutation
- in exon 5 leading to complete loss of NAGA protein.
-
- .0004
- NAGA DEFICIENCY, MILD FORM
- NAGA, SER160CYS
- Keulemans et al. (1996) reported that a Dutch girl with NAGA deficiency
- and mild neurologic manifestations was heterozygous for the E325K
- (104170.0001) mutation and a C-to-G change at nucleotide 11017
- (numbering according to Yamauchi et al., 1990) in exon 4, leading to a
- substitution of serine for cysteine at residue 160. The same genotype
- was found in the 3-year-old asymptomatic brother of the proband, who was
- presumed by the authors to be presymptomatic.
-
- *FIELD* SA
- Wang et al. (1990)
- *FIELD* RF
- 1. Chabas, A.; Coll, M. J.; Aparicio, M.; Rodriguez Diaz, E.: Mild
- phenotypic expression of alpha-N-acetylgalactosaminidase deficiency
- in two adult siblings. J. Inherit. Metab. Dis. 17: 724-731, 1994.
-
- 2. de Groot, P. G.; Westerveld, A.; Meera Khan, P.; Tager, J. M.:
- Localization of a gene for human alpha-galactosidase B (=N-acetyl-alpha-D-galactosaminidase)
- on chromosome 22. Hum. Genet. 44: 305-312, 1978.
-
- 3. de Jong, J; van den Berg, C; Wijburg, H.; Willemsen, R.; van Diggelen,
- O.; Schindler, D.; Hoevenaars, F.; Wevers, R.: Alpha-N-acetylgalactosaminidase
- deficiency with mild clinical manifestations and difficult biochemical
- diagnosis. J. Pediat. 125: 385-391, 1994.
-
- 4. Desnick, R. J.: Personal Communication. New York, N. Y. 1/15/1991.
-
- 5. Geurts van Kessel, A. H. M.; ten Brinke, H.; de Groot, P. G.; Hagemeijer,
- A.; Westerveld, A.; Meera Khan, P.; Pearson, P. L.: Regional localization
- of NAGA and ACO2 on human chromosome 22. (Abstract) Cytogenet. Cell
- Genet. 25: 161 only, 1979.
-
- 6. Geurts van Kessel, A. H. M.; Westerveld, A.; de Groot, P. G.; Meera
- Khan, P.; Hagemeijer, A.: Regional localization of the genes coding
- for human ACO2, ARSA, and NAGA on chromosome 22. Cytogenet. Cell
- Genet. 28: 169-172, 1980.
-
- 7. Kanzaki, T.; Wang, A. M.; Desnick, R. J.: Lysosomal alpha-N-acetylgalactosaminidase
- deficiency, the enzymatic defect in angiokeratoma corporis diffusum
- with glycopeptiduria. J. Clin. Invest. 88: 707-711, 1991.
-
- 8. Kanzaki, T.; Yokota, M.; Irie, F.; Hirabayashi, Y.; Wang, A. M.;
- Desnick, R. J.: Angiokeratoma corporis diffusum with glycopeptiduria
- due to deficient lysosomal alpha-N-acetylgalactosaminidase activity:
- clinical, morphologic, and biochemical studies. Arch. Derm. 129:
- 460-465, 1993.
-
- 9. Kanzaki, T.; Yokota, M.; Mizuno, N.: Clinical and ultrastructural
- studies of novel angiokeratoma corporis diffusum. (Abstract) Clin.
- Res. 36: 377A only, 1988.
-
- 10. Kanzaki, T.; Yokota, M.; Mizuno, N.; Matsumoto, Y.; Hirabayashi,
- Y.: Novel lysosomal glycoaminoacid storage disease with angiokeratoma
- corporis diffusum. Lancet I: 875-876, 1989.
-
- 11. Keulemans, J. L. M.; Reuser, A. J. J.; Kroos, M. A.; Willemsen,
- R.; Hermans, M. M. P.; van den Ouweland, A. M. W.; de Jong, J. G.
- N.; Wevers, R. A.; Renier, W. O.; Schindler, D.; Coll, M. J.; Chabas,
- A.; Sakuraba, H.; Suzuki, Y.; van Diggelen, O. P.: Human alpha-N-acetylgalactosaminidase
- (alpha-NAGA) deficiency: new mutations and the paradox between genotype
- and phenotype. J. Med. Genet. 33: 458-464, 1996.
-
- 12. Schindler, D.; Bishop, D. F.; Wallace, S.; Wolfe, D. E.; Desnick,
- R. J.: Characterization of alpha-N-acetylgalactosaminidase deficiency:
- a new neurodegenerative lysosomal disease. (Abstract) Pediat. Res. 23:
- 333A only, 1988.
-
- 13. Schindler, D.; Bishop, D. F.; Wolfe, D. E.; Wang, A. M.; Egge,
- H.; Lemieux, R. U.; Desnick, R. J.: Neuroaxonal dystrophy due to
- lysosomal alpha-N-acetylgalactosaminidase deficiency. New Eng. J.
- Med. 320: 1735-1740, 1989.
-
- 14. van Diggelen, O. P.; Schindler, D.; Kleijer, W. J.; Huijmans,
- J. G. M.; Galjaard, H.; Linden, H. U.; Peter-Katalinic, J.; Egge,
- H.; Dabrowski, U.; Cantz, M.: Lysosomal alpha-N-acetylgalactosaminidase
- deficiency: a new inherited metabolic disease. (Letter) Lancet II:
- 804 only, 1987.
-
- 15. van Diggelen, O. P.; Schindler, D.; Willemsen, R.; Boer, M.; Kleijer,
- W. J.; Huijmans, J. G. M.; Blom, W.; Galjaard, H.: Alpha-N-acetylgalactosaminidase
- deficiency, a new lysosomal storage disorder. J. Inherit. Metab.
- Dis. 11: 349-357, 1988.
-
- 16. Wang, A. M.; Bishop, D. F.; Desnick, R. J.: Human alpha-N-acetylgalactosaminidase-molecular
- cloning, nucleotide sequence, and expression of a full-length cDNA:
- homology with human alpha-galactosidase A suggests evolution from
- a common ancestral gene. J. Biol. Chem. 265: 21859-21866, 1990.
-
- 17. Wang, A. M.; Desnick, R. J.: Structural organization and complete
- sequence of the human alpha-N-acetylgalactosaminidase gene: homology
- with the alpha-galactosidase A gene provides evidence for evolution
- from a common ancestral gene. Genomics 10: 133-142, 1991.
-
- 18. Wang, A. M.; Kanzaki, T.; Desnick, R. J.: The molecular lesion
- in the alpha-N-acetylgalactosaminidase gene that causes angiokeratoma
- corporis diffusum with glycopeptiduria. J. Clin. Invest. 94: 839-845,
- 1994.
-
- 19. Wang, A. M.; Schindler, D.; Bishop, D. F.; Lemieux, R. U.; Desnick,
- R. J.: Schindler disease: biochemical and molecular characterization
- of a new neuroaxonal dystrophy due to alpha-N-acetylgalactosaminidase
- deficiency. (Abstract) Am. J. Hum. Genet. 43: A99 only, 1988.
-
- 20. Wang, A. M.; Schindler, D.; Desnick, R. J.: Schindler disease:
- the molecular lesion in the alpha-N-acetylgalactosaminidase gene that
- causes an infantile neuroaxonal dystrophy. J. Clin. Invest. 86:
- 1752-1756, 1990.
-
- 21. Wang, A. M.; Schindler, D.; Kanzaki, T.; Desnick, R. J.: Alpha-N-acetylgalactosaminidase
- gene: homology with human alpha-galactosidase A, and identification
- and confirmation of the mutations causing type I and II Schindler
- disease. (Abstract) Am. J. Hum. Genet. 47 (suppl.): A169 only, 1990.
-
- 22. Yamauchi, T.; Hiraiwa, M.; Kobayashi, H.; Uda, Y.; Miyatake, T.;
- Tsuji, S.: Molecular cloning of two species of cDNAs for human alpha-N-acetylgalactosaminidase
- and expression in mammalian cells. Biochem. Biophys. Res. Commun. 170:
- 231-237, 1990.
-
- *FIELD* CS
-
- Neuro:
- Progressive psychomotor deterioration;
- Loss of previously acquired motor and language skills;
- Abnormal pattern of urinary oligosaccharides;
- Myoclonic seizures;
- Decorticate posture;
- Marked long tract signs
-
- Skin:
- Angiokeratoma corporis diffusum (.0002 KANZAKI DISEASE)
-
- GI:
- Gastric mucosal telangiectasia
-
- Eyes:
- Optic atrophy;
- Blindness;
- Dilated conjunctival blood vessels;
- Dilated corkscrewlike tortuous fundal vessels
-
- Growth:
- Growth normal
-
- Misc:
- Onset about age 9 months
-
- Lab:
- Lysosomal alpha-N-acetylgalactosaminidase deficiency;
- Peripheral neuroaxonal degeneration;
- Rectal mucosal biopsy shows dystrophic autonomic axons with tubulovesicular
- material
-
- Inheritance:
- Autosomal recessive (22q11)
-
- *FIELD* CN
- Iosif W. Lurie - updated: 7/10/1996
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 11/27/1996
- carol: 7/22/1996
- carol: 7/10/1996
- carol: 10/10/1994
- jason: 6/9/1994
- warfield: 4/7/1994
- mimadm: 3/11/1994
- carol: 6/3/1993
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 104175
- *FIELD* TI
- *104175 ALPHA-1,3-GALACTOSYLTRANSFERASE
- GLYCOPROTEIN, ALPHA-GALACTOSYLTRANSFERASE 1; GGTA1
- *FIELD* TX
- Alpha-1,3-galactosyltransferase is a Golgi membrane-bound enzyme
- involved in the biosynthesis of the carbohydrate genes of glycoproteins
- and glycolipids. Enzyme levels are developmentally regulated and
- differentiation dependent. The enzyme is present in most mammals but
- cannot be detected in man, apes, or Old World monkeys. The carbohydrate
- structure produced by the enzyme is immunogenic in man, and most normal,
- healthy individuals have a significant titre of a natural antibody
- against the enzyme. Aberrant expression of the enzyme in man has been
- implicated in autoimmune disorders and in the occurrence of certain germ
- cell tumors. Joziasse et al. (1989) isolated 2 human homologs of the
- gene encoding the bovine enzyme. They concluded that these most likely
- represent a processed pseudogene and the inactivated remnant of the once
- functional source gene. The latter, referred to as HG-10 and symbolized
- GGTA1, was mapped to human chromosome 9 (Joziasse et al., 1991) by study
- of human-rodent somatic cell hybrids. The processed pseudogene,
- initially referred to as HGT-2 and later as GGTA1P, was mapped to human
- chromosome 12 by the same method. By in situ hybridization, Shaper et
- al. (1992) localized GGTA1 to 9q33-q34 and GGTA1P to 12q14-q15. It had
- previously been suggested (Joziasse et al., 1991) that this enzyme is
- evolutionarily related to the A and B blood group transferases; the
- location of the gene in distal 9q in the proximity of the ABO locus
- lends support to this hypothesis. The ABO and GGTA1 loci evolved from an
- ancestral locus through duplication. Subsequently, GGTA1 gave rise to an
- mRNA that, after reverse transcription, was incorporated into chromosome
- 12 as GGTA1P. In an even later event, the ancestral human alpha-1,3-GT
- became inactivated, possibly through a mutation in an upstream
- regulatory sequence, because its transcripts are no longer detected.
- This situation is comparable to the loss of vitamin C synthesizing
- capacity (240400) or uricase enzymatic activity (191540) in the human
- even though sequences for the relevant genes can be identified in the
- human genome.
-
- *FIELD* SA
- Joziasse et al. (1991)
- *FIELD* RF
- 1. Joziasse, D. H.; Shaper, J. H.; Jabs, E. W.; Shaper, N. L.: Characterization
- of an alpha-1,3-galactosyltransferase homologue on human chromosome
- 12 that is organized as a processed pseudogene. J. Biol. Chem. 266:
- 6991-6998, 1991.
-
- 2. Joziasse, D. H.; Shaper, J. H.; Van den Eijnden, D. H.; Van Tunen,
- A. J.; Shaper, N. L.: Bovine alpha-1,3-galactosyltransferase: isolation
- and characterization of a cDNA clone: identification of homologous
- sequences in human genomic DNA. J. Biol. Chem. 264: 14290-14297,
- 1989.
-
- 3. Joziasse, D. H.; Shaper, N. L.; Shaper, J. H.; Kozak, C. A.: The
- gene for murine alpha-1,3-galactosyltransferase is located in the
- centromeric region of chromosome 2. Somat. Cell Molec. Genet. 17:
- 201-205, 1991.
-
- 4. Shaper, N. L.; Lin, S.; Joziasse, D. H.; Kim, D.; Yang-Feng, T.
- L.: Assignment of two human alpha-1,3-galactosyltransferase gene
- sequences (GGTA1 and GGTA1P) to chromosomes 9q33-q34 and 12q14-q15.
- Genomics 12: 613-615, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 2/24/1992
-
- *FIELD* ED
- jason: 6/9/1994
- carol: 9/24/1993
- carol: 3/31/1992
- supermim: 3/16/1992
- carol: 3/6/1992
- carol: 2/26/1992
-
- *RECORD*
- *FIELD* NO
- 104180
- *FIELD* TI
- *104180 ALPHA-GLUCOSIDASE C, NEUTRAL; GANC
- *FIELD* TX
- Martiniuk et al. (1979, 1980) assigned a locus for this enzyme to
- chromosome 15. They also found a genetic polymorphism by starch gel
- electrophoresis, including a null allele. Martiniuk and Hirschhorn
- (1980) concluded that a combination of starch gel electrophoresis and
- isoelectric focusing permits recognition of 7 phenotypes resulting from
- 4 different alleles. The product of one of the alleles is 'silent,' with
- an unusually high gene frequency--0.174 in whites. About one-third of
- the population is heterozygous 'null.' It appears that the homozygous
- state does not result in disease.
-
- *FIELD* RF
- 1. Martiniuk, F.; Hirschhorn, R.: Human neutral alpha-glucosidase
- C: genetic polymorphism including a 'null' allele. Am. J. Hum. Genet. 32:
- 497-507, 1980.
-
- 2. Martiniuk, F.; Hirschhorn, R.; Smith, M.: Assignment of human
- neutral alpha-glucosidase C to chromosome 15. (Abstract) Cytogenet.
- Cell Genet. 25: 182 only, 1979.
-
- 3. Martiniuk, F.; Hirschhorn, R.; Smith, M.: Assignment of the gene
- for human neutral alpha-glucosidase C to chromosome 15. Cytogenet.
- Cell Genet. 27: 168-175, 1980.
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- supermim: 3/16/1992
- supermim: 3/20/1990
- ddp: 10/26/1989
- marie: 3/25/1988
- reenie: 2/9/1987
- marie: 1/7/1987
-
- *RECORD*
- *FIELD* NO
- 104200
- *FIELD* TI
- 104200 ALPORT SYNDROME
- HEREDITARY NEPHROPATHY AND DEAFNESS
- *FIELD* TX
- The classic phenotype as described by Alport (1927) is nephritis, often
- progressing to renal failure, and sensorineural hearing loss affecting
- both sexes in successive generations. The renal disease becomes evident
- as recurrent microscopic or gross hematuria as early as childhood,
- earlier in males than in females. Progression to renal failure is
- gradual and usually occurs in males by the fifth decade. The nephrotic
- syndrome is unusual but has been reported. The renal histology is
- nonspecific; both glomerular and interstitial abnormalities, including
- foam cells, occur. Some investigators (e.g., Churg and Sherman, 1973)
- believe the ultrastructural changes of the glomerular basement membrane,
- which is irregularly thickened and attenuated, are specific for this
- condition, but controversy exists on this point. Immunofluorescence
- studies have provided little evidence for an immunologic basis for renal
- damage. Hearing loss, which is sensorineural and primarily affects high
- tones, occurs in 30 to 50% of relatives with renal disease. The severity
- of auditory and renal features do not correlate in a given individual.
-
- Alport syndrome shows considerable heterogeneity. In addition to the
- existence of X-linked and autosomal forms (which often cannot be
- distinguished in individual kindreds), families differ in the age of
- end-stage renal disease (ESRD) and the occurrence of deafness. Hasstedt
- et al. (1986) tested for heterogeneity among 23 Utah kindreds using 2
- methods developed for assay of linkage heterogeneity: C.A.B. Smith's
- admixture test and Morton's predivided-sample test. The 3 phenotypes
- were juvenile Alport syndrome with deafness, adult Alport syndrome with
- deafness, and adult Alport syndrome without deafness or other defects.
- The age of 31 years for ESRD was taken as the divide between the
- juvenile and adult forms. Atkin et al. (1986) proposed the existence of
- 6 types of dominant Alport syndrome among kindreds reported: I, classic
- juvenile Alport syndrome with deafness; II, X-linked juvenile Alport
- syndrome with deafness (301050); III, X-linked adult Alport syndrome
- with deafness; IV, X-linked adult Alport syndrome without deafness or
- other defect, that is, purely renal disease; V, autosomal Alport
- syndrome with deafness and thrombocytopathia (153650); and VI, autosomal
- recessive juvenile Alport syndrome with deafness (203780). A possibly
- distinct entity is hereditary nephritis without deafness (161900)
- reported by Reyersbach and Butler (1954) and Dockhorn (1967). M'Rad et
- al. (1992) reviewed 31 families with Alport syndrome. Though there was
- clinical heterogeneity for ophthalmic signs and the age of development
- of end-stage renal disease, homogeneity tests failed to show evidence of
- genetic heterogeneity.
-
- Partial sex linkage (location of the gene on the part of the X and Y
- chromosomes that is homologous) was suggested on the basis of the large
- Mormon kindred reported by Perkoff et al. (1958). O'Neill et al. (1978)
- reexamined and extended this pedigree and provided convincing evidence
- for X-linked inheritance (see 301050), and Barker et al. (1990)
- demonstrated substitution of serine for cysteine in the alpha-5 chain of
- type IV collagen (303630.0002). Autosomal dominant inheritance with
- anomalous segregation was proposed by Shaw and Glover (1961).
- Heterozygous mothers transmit the gene to more than 50% of daughters and
- probably more than 50% of their sons. Evans et al. (1980) reported a
- family with male-to-male transmission. The kindred reported by Ohlsson
- (1963) differed from others reported in that myopia was a conspicuous
- feature and the impairment of renal function in the affected males was
- relatively mild even in 2 over age 30 years. Ocular abnormalities have
- been observed in some patients (Arnott et al., 1966). Stanbury and
- Castleman (1968) reported 7 persons in 3 generations; the proband had
- hypophosphatemia, nephrocalcinosis, and unilateral deafness; foam cells
- were demonstrated in the kidney. Miller et al. (1970) showed that the
- vestibular neuroepithelium is involved as well as that of the cochlea.
- Variability in histologic findings in the ear in Alport syndrome led
- Myers and Tyler (1972) to conclude that it is a heterogeneous category.
- They reported the temporal bone histology in 2 cases: both had severe
- deafness but one had a histologically normal inner ear whereas the other
- had a marked reduction in spinal ganglion cochlear neurons.
-
- Miyoshi et al. (1975) found antithyroid antibodies in the serum of
- multiple persons with Alport syndrome in 2 Japanese kindreds.
- Hyperthyroidism was present in one and histologic changes of thyroiditis
- in a second. They proposed that Alport syndrome may be an immunologic
- disorder. Spear (1973) suggested that a primary structural abnormality
- of basement membranes underlies the phenotype. Evidence from many
- sources suggests that the glomerular basement membrane of patients with
- Alport syndrome is different antigenically and therefore biochemically,
- as well as morphologically, from that of normal persons (review by
- Milliner et al., 1982); these authors reported successful results of
- kidney transplantation in most cases. Yoshikawa et al. (1982) emphasized
- 'basket weave' alteration in the lamina densa of the capillary basement
- membrane, demonstrated by electron microscopy, as the pathognomonic
- histologic feature of Alport syndrome. The change was, furthermore,
- found in all 3 children biopsied under 5 years of age. The finding
- served to differentiate benign familial hematuria (141200). Yoshikawa et
- al. (1982) found families with heavy proteinuria, segmented sclerosis,
- foam cells, and the 'basket weave' alteration, but no deafness (see
- 161900). They concluded that families with and without deafness 'fall
- within the spectrum of Alport syndrome, although the presence of
- deafness adversely affected the prognosis.' The report of Alport (1927),
- in which he first described deafness as a component of the syndrome, was
- the fourth concerning this signal pedigree. The first report (Dickinson,
- 1875) noted hematuria in 3 generations while 2 later studies (Guthrie,
- 1902; Kendall and Hertz, 1912) added albuminuria and azotemia to the
- spectrum of renal involvement. Patients with Alport syndrome constituted
- 2.3% of the transplant population at the Mayo Clinic (Milliner et al.,
- 1982). In the study Waldherr (1982), Alport syndrome comprised at least
- a sixth of familial glomerular disease, which itself was responsible for
- 6.3% of his biopsy material.
-
- In a retrospective, double-blind study, Savage et al. (1986) examined
- paraffin-embedded renal biopsy sections from 44 children with hematuria
- to see whether a mouse monoclonal antibody (MCA-P1) against glomerular
- basement membrane (GBM) could identify a subgroup of patients with
- Alport syndrome in which the Goodpasture antigen is abnormal. Strong
- linear binding of MCA-P1 to GBM was found in all 29 patients without
- evidence of hereditary nephritis and in 2 with possible but not definite
- hereditary nephritis. In contrast, 12 of 13 patients with strong
- evidence of hereditary nephritis showed no binding (9) or greatly
- reduced binding (3). Thus, abnormal antigenicity of the basement
- membrane in hereditary nephritis, as reported by McCoy et al. (1982), is
- confirmed. Savage et al. (1987) concluded that the inherited defect in
- hereditary nephritis affects Goodpasture antigen secondarily. Serum
- amyloid P component (SAP; 104770) has been found to be a constituent of
- normal glomerular basement membrane. Melvin et al. (1986) showed that
- SAP and Goodpasture antigen are closely associated in the glomerular
- basement membrane and that in patients with Alport-type hereditary
- nephritis who lack Goodpasture antigen, SAP is also uniformly absent.
-
- Yoshikawa et al. (1987) reviewed 48 children with ultrastructural
- changes of the glomerular basement membrane, a characteristic of
- hereditary nephritis. All had hematuria. In 30 cases, there was
- hematuria in at least 1 other member of the family; in the other 18
- cases, there was no familial incidence. There were no differences
- between the 2 groups with regard to clinical and pathologic findings. At
- the latest follow-up, 6 boys with familial hematuria and 3 boys with
- nonfamilial hematuria had reduced renal function, and 9 boys with
- familial hematuria and 4 boys and 1 girl with nonfamilial hematuria had
- sensorineural deafness. Yoshikawa et al. (1987) suggested that the
- disorder in patients with nonfamilial hematuria may represent new
- mutations for hereditary nephritis. Nielsen (1978) suggested that
- anterior lenticonus may be a specific sign of Alport syndrome, since all
- recently reported cases (e.g., Arnott et al., 1966) had been associated
- with hereditary nephropathy. Streeten et al. (1987) concluded that the
- anterior capsule of the lens 'is clearly fragile in this disease,
- forming the basis for the progressive lenticonus and anterior polar
- cataract. These abnormalities correlate well with a defect in the type
- IV collagen molecule.'
-
- The disorder that has come to be known as Alport syndrome is
- characterized by hematuria, progressive renal failure, and sensorineural
- hearing loss and is frequently associated with both ocular abnormalities
- (such as lenticonus and retinal anomalies) as well as the identification
- of mutations in the gene encoding the basement membrane specific type IV
- collagen alpha-5 chain (COL4A5; 303630), an X-linked gene. This syndrome
- was definitely proven to be an X-linked dominant disorder. The
- possibility of an autosomal dominant form became less likely, as most of
- the cases were shown to be X-linked. There was a possibility, however,
- that possible autosomal dominant Alport syndrome was, in fact,
- hereditary nephropathy and deafness in association with hematologic
- abnormalities, Epstein syndrome (153650), or Fechtner syndrome (153640).
- Although autosomal recessive transmission had been previously considered
- unlikely, this mode of transmission seemed likely in a remaining small
- percentage of kindreds in which there was parental consanguinity,
- absence of severe symptoms in parents, and equal severity of the disease
- in males and females; see 203780. The plausibility of an autosomal form
- of Alport syndrome was supported by the isolation of 2 autosomal type IV
- collagen genes, COL4A3 (120070) and COL4A4 (120131), which are located
- head-to-head on 2q35-q37 and are specifically expressed in the
- glomerular basement membrane and the specialized ocular and inner ear
- basement membranes. Demonstration of linkage analysis to chromosome 2q
- in consanguineous families and of mutations in one or the other of these
- 2 autosomal genes provided clear evidence of the existence of the
- autosomal recessive form of Alport syndrome. It remains to be determined
- whether mutations in either of these genes in heterozygous state cause
- abnormality.
-
- *FIELD* SA
- Beathard and Granholm (1977); Chazan et al. (1971); Chuang and Reuter
- (1974); Cohen et al. (1961); Crawfurd and Toghill (1968); DiBona
- (1983); Goyer et al. (1968); Kenya et al. (1977); Marin and Tyler
- (1961); Mulrow et al. (1963); Perrin et al. (1980); Preus and Fraser
- (1971); Purriel et al. (1970); Schneider (1963); Sherman et al. (1974);
- Spear (1984); Spear and Slusser (1972); Spear et al. (1970); Turner
- (1970); Westley (1970); Whalen et al. (1961); Williamson (1961)
- *FIELD* RF
- 1. Alport, A. C.: Hereditary familial congenital hemorrhagic nephritis.
- Brit. Med. J. 1: 504-506, 1927.
-
- 2. Arnott, E. J.; Crawfurd, M. D. A.; Toghill, P. J.: Anterior lenticonus
- and Alport's syndrome. Brit. J. Ophthal. 50: 390-403, 1966.
-
- 3. Atkin, C. L.; Gregory, M. C.; Border, W. A.: Alport syndrome.
- In: Schrier, R. W.; Gottschalk, C. W.: Strauss and Welt's Diseases
- of the Kidney. Boston: Little, Brown (pub.) (4th ed.): 1986.
-
- 4. Barker, D. F.; Hostikka, S. L.; Zhou, J.; Chow, L. T.; Oliphant,
- A. R.; Gerken, S. C.; Gregory, M. C.; Skolnick, M. H.; Atkin, C. L.;
- Tryggvason, K.: Identification of mutations in the COL4A5 collagen
- gene in Alport syndrome. Science 248: 1224-1227, 1990.
-
- 5. Beathard, G. A.; Granholm, N. A.: Development of the characteristic
- ultrastructural lesion of hereditary nephritis during the course of
- the disease. Am. J. Med. 62: 751-756, 1977.
-
- 6. Chazan, J. A.; Zacks, J.; Cohen, J. J.; Garella, S.: Hereditary
- nephritis: clinical spectrum and mode of inheritance in five new kindreds.
- Am. J. Med. 50: 764-771, 1971.
-
- 7. Chuang, V. P.; Reuter, S. R.: Angiographic features of Alport's
- syndrome: hereditary nephritis. Am. J. Roentgen. 121: 539-543,
- 1974.
-
- 8. Churg, J.; Sherman, R. L.: Pathology of hereditary nephritis.
- Arch. Path. 95: 374-379, 1973.
-
- 9. Cohen, M. M.; Cassady, G.; Hanna, B. L.: A genetic study of hereditary
- renal dysfunction with associated nerve deafness. Am. J. Hum. Genet. 13:
- 379-389, 1961.
-
- 10. Crawfurd, M. D. A.; Toghill, P. J.: Alport's syndrome of hereditary
- nephritis and deafness. Quart. J. Med. 37: 563-576, 1968.
-
- 11. DiBona, G. F.: Alport's syndrome: a genetic defect in biochemical
- composition of basement membrane of glomerulus, lens, and inner ear?.
- (Editorial) J. Lab. Clin. Med. 101: 817-820, 1983.
-
- 12. Dickinson, W. H.: Disease of the Kidney and Urinary Derangements.
- Part 2.. London: Longmans, Green (pub.) 1875. Pp. 379 only.
-
- 13. Dockhorn, R. J.: Hereditary nephropathy without deafness. Am.
- J. Dis. Child. 114: 135-138, 1967.
-
- 14. Evans, S. H.; Erickson, R. P.; Kelsch, R.; Pierce, J. C.: Apparently
- changing patterns of inheritance in Alport's hereditary nephritis:
- genetic heterogeneity versus altered diagnostic criteria. Clin.
- Genet. 17: 285-292, 1980.
-
- 15. Goyer, R. A.; Reynolds, J., Jr.; Burke, J.; Burkholder, P.: Hereditary
- renal disease with neurosensory hearing loss, prolinuria and ichthyosis.
- Am. J. Med. Sci. 256: 166-179, 1968.
-
- 16. Guthrie, L. B.: 'Idiopathic' or congenital, hereditary and family
- haematuria. Lancet I: 1243-1246, 1902.
-
- 17. Hasstedt, S. J.; Atkin, C. L.; San Juan, A. C., Jr.: Genetic
- heterogeneity among kindreds with Alport syndrome. Am. J. Hum. Genet. 38:
- 940-953, 1986.
-
- 18. Kendall, G.; Hertz, A. F.: Hereditary familial congenital hemorrhagic
- nephritis. Guy's Hosp. Rep. 66: 137-141, 1912.
-
- 19. Kenya, P. R.; Asal, N. R.; Pederson, J. A.; Lindeman, R. D.:
- Hereditary (familial) renal disease: clinical and genetic studies.
- Sth. Med. J. 70: 1049-1051, 1977.
-
- 20. M'Rad, R.; Sanak, M.; Deschenes, G.; Zhou, J.; Bonaiti-Pellie,
- C.; Holvoet-Vermaut, L.; Heuertz, S.; Gubler, M.-C.; Broyer, M.; Grunfeld,
- J.-P.; Tryggvason, K.; Hors-Cayla, M.-C.: Alport syndrome: a genetic
- study of 31 families. Hum. Genet. 90: 420-426, 1992.
-
- 21. Marin, O. S. M.; Tyler, H. R.: Hereditary interstitial nephritis
- associated with polyneuropathy. Neurology 11: 999-1005, 1961.
-
- 22. McCoy, R. C.; Johnson, K. H.; Stone, W. J.; Wilson, C. B.: Absence
- of nephritogenic GBM antigen(s) in some patients with hereditary nephritis.
- Kidney Int. 21: 642-652, 1982.
-
- 23. Melvin, T.; Kim, Y.; Michael, A. F.: Amyloid P component is not
- present in the glomerular basement membrane in Alport-type hereditary
- nephritis. Am. J. Path. 125: 460-464, 1986.
-
- 24. Miller, G. W.; Joseph, D. J.; Cozad, R. L.; McCabe, B. F.: Alport's
- syndrome. Arch. Otolaryng. 92: 419-432, 1970.
-
- 25. Milliner, D. S.; Pierides, A. M.; Holley, K. E.: Renal transplantation
- in Alport's syndrome: anti-glomerular basement membrane glomerulonephritis
- in the allograft. Mayo Clin. Proc. 57: 35-43, 1982.
-
- 26. Miyoshi, K.; Suzuki, M.; Ohno, F.; Yamano, T.; Yagi, F.; Khono,
- H.: Antithyroid antibodies in Alport's syndrome. Lancet II: 480-482,
- 1975.
-
- 27. Mulrow, P. J.; Aron, A. M.; Gathman, G. E.; Yesner, R.; Lubs,
- H. A.: Hereditary nephritis: report of a kindred. Am. J. Med. 35:
- 737-748, 1963.
-
- 28. Myers, G. J.; Tyler, H. R.: The etiology of deafness in Alport's
- syndrome. Arch. Otolaryng. 96: 333-340, 1972.
-
- 29. Nielsen, C. E.: Lenticonus anterior and Alport's syndrome. Acta
- Ophthal. 56: 518-530, 1978.
-
- 30. O'Neill, W. M., Jr.; Atkin, C. L.; Bloomer, H. A.: Hereditary
- nephritis: a re-examination of its clinical and genetic features.
- Ann. Intern. Med. 88: 176-182, 1978.
-
- 31. Ohlsson, L.: Congenital renal disease, deafness and myopia in
- one family. Acta Med. Scand. 174: 77-84, 1963.
-
- 32. Perkoff, G. T.; Nugent, C. A., Jr.; Dolowitz, D. A.; Stephens,
- F. E.; Carnes, W. H.; Tyler, F. H.: A follow-up study of hereditary
- chronic nephritis. Arch. Intern. Med. 102: 733-746, 1958.
-
- 33. Perrin, D.; Jungers, P.; Grunfeld, J. P.; Delons, S.; Noel, L.-H.;
- Zenatti, C.: Perimacular changes in Alport's syndrome. Clin. Nephrol. 13:
- 163-167, 1980.
-
- 34. Preus, M.; Fraser, F. C.: Genetics of hereditary nephropathy
- with deafness (Alport's disease). Clin. Genet. 2: 331-337, 1971.
-
- 35. Purriel, P.; Drets, M.; Pascale, E.; Cestau, R. S.; Borras, A.;
- Ferreira, W. A.; Delucca, A.; Fernandez, L.: Familial hereditary
- nephropathy (Alport's syndrome). Am. J. Med. 49: 753-773, 1970.
-
- 36. Reyersbach, G. C.; Butler, A. M.: Congenital hereditary hematuria.
- New Eng. J. Med. 251: 377-380, 1954.
-
- 37. Savage, C. O. S.; Noel, L. H.; Cashman, S.; Grunfeld, J. P.; Lockwood,
- C. M.: Characterisation by immunoblotting of the glomerular basement
- membrane defect in hereditary nephritis. (Abstract) Clin. Res. 35:
- 663A only, 1987.
-
- 38. Savage, C. O. S.; Reed, A.; Kershaw, M.; Pincott, J.; Pusey, C.
- D.; Dillon, M. J.; Barratt, T. M.; Lockwood, C. M.: Use of a monoclonal
- antibody in differential diagnosis of children with haematuria and
- hereditary nephritis. Lancet I: 1459-1461, 1986.
-
- 39. Schneider, R. G.: Congenital hereditary nephritis with nerve
- deafness. New York J. Med. 63: 2644-2648, 1963.
-
- 40. Shaw, R. F.; Glover, R. A.: Abnormal segregation in hereditary
- renal disease with deafness. Am. J. Hum. Genet. 13: 89-97, 1961.
-
- 41. Sherman, R. L.; Churg, J.; Yudis, M.: Hereditary nephritis with
- a characteristic renal lesion. Am. J. Med. 56: 44-51, 1974.
-
- 42. Spear, G. S.: Alport's syndrome: a consideration of pathogenesis.
- Clin. Nephrol. 1: 336-337, 1973.
-
- 43. Spear, G. S.: Hereditary nephritis (Alport's syndrome)--1983.
- Clin. Nephrol. 21: 3-6, 1984.
-
- 44. Spear, G. S.; Slusser, R. J.: Alport's syndrome: emphasizing
- electron microscopic studies of the glomerulus. Am. J. Path. 69:
- 213-224, 1972.
-
- 45. Spear, G. S.; Whitworth, J. M.; Konigsmark, B. W.: Hereditary
- nephritis with nerve deafness: immunofluorescent studies on the kidney,
- with a consideration of discordant immunoglobulin-complement immunofluorescent
- reactions. Am. J. Med. 49: 52-63, 1970.
-
- 46. Stanbury, S. W.; Castleman, B.: Nephrocalcinosis and azotemia
- in a young man. New Eng. J. Med. 278: 839-846, 1968.
-
- 47. Streeten, B. W.; Robinson, M. R.; Wallace, R.; Jones, D. B.:
- Lens capsule abnormalities in Alport's syndrome. Arch. Ophthal. 105:
- 1693-1697, 1987.
-
- 48. Turner, J. S., Jr.: Hereditary hearing loss with nephropathy
- (Alport's syndrome). Acta Otolaryng. 271 (suppl.): 7-26, 1970.
-
- 49. Waldherr, R.: Familial glomerular disease. Contrib. Nephrol. 33:
- 104-121, 1982.
-
- 50. Westley, C. R.: Familial nephritis and associated deafness in
- a southwestern Apache Indian family. Sth. Med. J. 63: 1415-1419,
- 1970.
-
- 51. Whalen, R. E.; Huang, S.-S.; Peschel, E.; McIntosh, H. D.: Hereditary
- nephropathy, deafness and renal foam cells. Am. J. Med. 31: 171-186,
- 1961.
-
- 52. Williamson, D. A. J.: Alport's syndrome of hereditary nephritis
- with deafness. Lancet II: 1321-1323, 1961.
-
- 53. Yoshikawa, N.; Matsuyama, S.; Ito, H.; Hajikano, H.; Matsuo, T.
- : Nonfamilial hematuria associated with glomerular basement membrane
- alterations characteristic of hereditary nephritis: comparison with
- hereditary nephritis. J. Pediat. 111: 519-524, 1987.
-
- 54. Yoshikawa, N.; White, R. H. R.; Cameron, A. H.: Familial hematuria:
- clinico-pathological correlations. Clin. Nephrol. 17: 172-182,
- 1982.
-
- *FIELD* CS
-
- GU:
- Nephritis;
- Renal failure;
- Nephrotic syndrome
-
- Ears:
- Sensorineural hearing loss
-
- Eyes:
- Fragile anterior lens capsule;
- Lenticonus;
- Anterior polar cataract;
- Myopia
-
- Lab:
- Hematuria;
- Renal foam cells;
- Hypophosphatemia;
- Nephrocalcinosis;
- Proteinuria;
- Azotemia;
- Ultrastructural glomerular basement membrane changes;
- Antithyroid antibodies
-
- Inheritance:
- Autosomal dominant form;
- 6 types including X-linked form
-
- *FIELD* CD
- Victor A. McKusick: 6/4/1986
-
- *FIELD* ED
- mark: 6/9/1995
- davew: 8/18/1994
- jason: 6/16/1994
- carol: 6/9/1994
- mimadm: 4/17/1994
- carol: 10/14/1993
-
- *RECORD*
- *FIELD* NO
- 104210
- *FIELD* TI
- *104210 ALPHA-2A-ADRENERGIC RECEPTOR; ADRA2A; ADRAR; ADRA2
- ALPHA-2-ADRENERGIC RECEPTOR, PLATELET TYPE;;
- ADRENOCEPTOR, ALPHA-2A
- *FIELD* TX
- Hormones and drugs exert their physiologic and pharmacologic effects by
- interacting with specific plasma membrane receptors of responsive cells.
- Adrenergic receptors fall into two major classes, alpha and beta, each
- of which is subdivided into 2 subclasses, termed alpha-1 and alpha-2 and
- beta-1 and beta-2. The beta-adrenergic receptors, which stimulate, and
- the alpha-2 adrenergic receptors, which often inhibit adenylate cyclase,
- are coupled to guanine nucleotide regulatory proteins. Using an
- alpha-2-adrenergic receptor clone, Yang-Feng et al. (1987) mapped the
- ADRAR locus to 10q23-q25 by somatic cell hybridization and in situ
- hybridization. Kobilka et al. (1987) cloned the gene for the human
- platelet alpha-2-adrenergic receptor using oligonucleotides
- corresponding to the partial amino acid sequence of the purified
- receptor. The deduced amino acid sequence was most similar to those of
- human beta-2 and beta-1 adrenergic receptors. Similarities to the
- muscarinic cholinergic receptors were also evident. Two related genes
- were identified by low stringency Southern blot analysis. Hoehe et al.
- (1988) identified a DraI RFLP of the ADRAR gene. By study of
- interspecific backcrosses, Oakey et al. (1991) assigned the Adra2r gene
- to the distal region of mouse chromosome 19.
-
- An aspartic acid residue at position 79 is highly conserved among G
- protein-coupled receptors. Surprenant et al. (1992) found that when
- asp-79 was mutated to asparagine, cells transfected with the mutant
- adrenoceptor showed inhibition of adenylyl cyclase and calcium currents
- by agonists but did not increase potassium currents. Because distinct G
- proteins appear to couple adrenoceptors to potassium and calcium
- currents, the findings suggested that the mutant adrenoceptor could not
- achieve the conformation necessary to activate G proteins that mediate
- potassium channel activation.
-
- *FIELD* SA
- Hoehe et al. (1989)
- *FIELD* RF
- 1. Hoehe, M.; Berrettini, W.; Leppert, M.; Lalouel, J.-M.; Byerley,
- W.; Gershon, E.; White, R.: Genetic mapping of adrenergic receptor
- genes. (Abstract) Am. J. Hum. Genet. 45 (suppl.): A143 only, 1989.
-
- 2. Hoehe, M. R.; Berrettini, W. H.; Lentes, K.-U.: Dra I identifies
- a two allele DNA polymorphism in the human alpha-2-adrenergic receptor
- gene (ADRAR), using a 5.5 kb probe (p ADRAR). Nucleic Acids Res. 16:
- 9070 only, 1988.
-
- 3. Kobilka, B. K.; Matsui, H.; Kobilka, T. S.; Yang-Feng, T. L.; Francke,
- U.; Caron, M. G.; Lefkowitz, R. J.; Regan, J. W.: Cloning, sequencing,
- and expression of the gene coding for the human platelet alpha-2-adrenergic
- receptor. Science 238: 650-656, 1987.
-
- 4. Oakey, R. J.; Caron, M. G.; Lefkowitz, R. J.; Seldin, M. F.: Genomic
- organization of adrenergic and serotonin receptors in the mouse: linkage
- mapping of sequence-related genes provides a method for examining
- mammalian chromosome evolution. Genomics 10: 338-344, 1991.
-
- 5. Surprenant, A.; Horstman, D. A.; Akbarali, H.; Limbird, L. E.:
- A point mutation of the alpha-2-adrenoceptor that blocks coupling
- to potassium but not calcium currents. Science 257: 977-980, 1992.
-
- 6. Yang-Feng, T. L.; Kobilka, B. K.; Caron, M. G.; Lefkowitz, R. J.;
- Francke, U.: Chromosomal assignment of genes for an alpha-adrenergic
- receptor (ADRAR) and for another member of this receptor family coupled
- to guanine nucleotide regulatory proteins (RG21). (Abstract) Cytogenet.
- Cell Genet. 46: 722-723, 1987.
-
- *FIELD* CD
- Victor A. McKusick: 8/31/1987
-
- *FIELD* ED
- carol: 9/9/1992
- carol: 9/8/1992
- carol: 4/1/1992
- supermim: 3/19/1992
- supermim: 3/16/1992
- carol: 3/5/1992
-
- *RECORD*
- *FIELD* NO
- 104219
- *FIELD* TI
- *104219 ALPHA-1A-ADRENERGIC RECEPTOR; ADRA1A
- *FIELD* TX
- Lomasney et al. (1991) demonstrated that there are at least 3
- alpha-1-adrenergic receptors. From in situ hybridization studies, they
- concluded that the gene for the alpha-1A receptor is located on
- chromosome 5 in the region q23-q32, the same region that contains the
- ADRA1B gene (104220). The ADRB2 gene (109690) is also in the same area.
- The close proximity of 3 adrenergic receptors on the same chromosome
- suggested that this family of proteins arose by gene duplication.
- However, Schwinn and Lomasney (1992) concluded from its pharmacologic
- characteristics that the clone represents a further subtype designated
- alpha-1D (see ADRA1D; 104222). Loftus et al. (1994) found by PCR
- analysis of somatic cell hybrids that ADRA1A is in fact located on
- chromosome 20. They cited work of others confirming the assignment of
- ADRA1A to chromosome 20 by FISH.
-
- Bruno et al. (1991) also cloned a human alpha-1A-adrenergic receptor.
- The homologous gene in the mouse is located on chromosome 11 (Wilkie et
- al., 1993), which shows homology of synteny with 5q, not chromosome 20.
-
- *FIELD* RF
- 1. Bruno, J. F.; Whittaker, J.; Song, J.; Berelowitz, M.: Molecular
- cloning and sequencing of a cDNA encoding a human alpha-1A adrenergic
- receptor. Biochem. Biophys. Res. Commun. 179: 1485-1490, 1991.
-
- 2. Loftus, S. K.; Shiang, R.; Warrington, J. A.; Bengtsson, U.; McPherson,
- J. D.; Wasmuth, J. J.: Genes encoding adrenergic receptors are not
- clustered on the long arm of human chromosome 5. Cytogenet. Cell
- Genet. 67: 69-74, 1994.
-
- 3. Lomasney, J. W.; Cotecchia, S.; Lorenz, W.; Leung, W.-Y.; Schwinn,
- D. A.; Yang-Feng, T. L.; Brownstein, M.; Lefkowitz, R. J.; Caron,
- M. G.: Molecular cloning and expression of the cDNA for the alpha-1A-adrenergic
- receptor: the gene for which is located on human chromosome 5. J.
- Biol. Chem. 266: 6365-6369, 1991.
-
- 4. Schwinn, D. A.; Lomasney, J. W.: Pharmacologic characterization
- of cloned alpha-1-adrenoceptor subtypes: selective antagonists suggest
- the existence of a fourth subtype. Europ. J. Pharm. 227: 433-436,
- 1992.
-
- 5. Wilkie, T. M.; Chen, Y.; Gilbert, D. J.; Moore, K. J.; Yu, L.;
- Simon, M. I.; Copeland, N. G.; Jenkins, N. A.: Identification, chromosomal
- location, and genome organization of mammalian G-protein-coupled receptors.
- Genomics 18: 175-184, 1993.
-
- *FIELD* CD
- Victor A. McKusick: 5/13/1991
-
- *FIELD* ED
- carol: 11/10/1994
- pfoster: 8/16/1994
- jason: 6/9/1994
- carol: 12/1/1993
- carol: 11/29/1993
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 104220
- *FIELD* TI
- *104220 ALPHA-1B-ADRENERGIC RECEPTOR; ADRA1B
- ALPHA-1-ADRENERGIC RECEPTOR; ADRA1
- *FIELD* TX
- The alpha-1B-adrenergic receptor is a member of the G-protein-coupled
- family of transmembrane receptors. See 104210. Yang-Feng et al. (1990)
- mapped the ADRA1 gene to chromosome 5 by Southern analysis of somatic
- cell hybrids and regionalized it to 5q32-q34 by in situ hybridization.
- From pulsed field gel electrophoresis, they concluded that the ADRA1R
- and ADRB2R (109690) loci are within 300 kb of each other. Lomasney et
- al. (1991) indicated that this alpha-1 receptor is alpha-1B and that the
- regional assignment is 5q23-q32. The corresponding gene in the mouse,
- symbolized Adra1r, is located on proximal chromosome 11 (Oakey et al.,
- 1991).
-
- From cloning and sequencing the ADRA1B gene, Ramarao et al. (1992) found
- that it comprises 2 exons and a single large intron of at least 20 kb
- that interrupts the coding region at the end of the putative sixth
- transmembrane domain. The genomic organization of this adrenergic
- receptor with a single large intron interrupting its coding region
- differs from those of other adrenergic receptors as well as muscarinic
- and 5-hydroxytryptamine receptors, which are intronless. The location of
- the intron is also unique among those members of the G-protein-coupled
- receptor family that do possess introns.
-
- When transfected into NIH 3T3 fibroblasts and other cell lines, the
- alpha-1B-adrenergic receptor induces neoplastic transformation which
- identifies this normal cellular gene as a protooncogene. Allen et al.
- (1991) demonstrated that mutational alteration of the receptor can lead
- to activation of this protooncogene in such a way that cell lines are
- constitutively activated, even though not stimulated by agonist. These
- cells demonstrate an enhanced ability for tumor generation in nude mice,
- with a decreased period of latency compared with cells expressing the
- wildtype receptor. From these observations, Allen et al. (1991)
- suggested that analogous spontaneously occurring mutations in this class
- of receptor proteins could play a key role in the induction or
- progression of neoplastic transformation and atherosclerosis. Indeed, a
- comparable situation was demonstrated in the case of the thyrotropin
- receptor, causing hyperfunctioning thyroid adenoma (275200.0002).
- Furthermore, a mutation in the luteinizing hormone receptor can result
- in its constitutive activation, resulting in familial male precocious
- puberty (152790.0001).
-
- Loftus et al. (1994) concluded that ADRA1B and ADRB2 are several Mb
- apart rather than a few hundred kb as reported by Yang-Feng et al.
- (1990).
-
- *FIELD* RF
- 1. Allen, L. F.; Lefkowitz, R. J.; Caron, M. G.; Cotecchia, S.: G-protein-coupled
- receptor genes as protooncogenes: constitutively activating mutation
- of the alpha-1B-adrenergic receptor enhances mitogenesis and tumorigenicity.
- Proc. Nat. Acad. Sci. 88: 11354-11358, 1991.
-
- 2. Loftus, S. K.; Shiang, R.; Warrington, J. A.; Bengtsson, U.; McPherson,
- J. D.; Wasmuth, J. J.: Genes encoding adrenergic receptors are not
- clustered on the long arm of human chromosome 5. Cytogenet. Cell
- Genet. 67: 69-74, 1994.
-
- 3. Lomasney, J. W.; Cotecchia, S.; Lorenz, W.; Leung, W.-Y.; Schwinn,
- D. A.; Yang-Feng, T. L.; Brownstein, M.; Lefkowitz, R. J.; Caron,
- M. G.: Molecular cloning and expression of the cDNA for the alpha-1A-adrenergic
- receptor: the gene for which is located on human chromosome 5. J.
- Biol. Chem. 266: 6365-6369, 1991.
-
- 4. Oakey, R. J.; Caron, M. G.; Lefkowitz, R. J.; Seldin, M. F.: Genomic
- organization of adrenergic and serotonin receptors in the mouse: linkage
- mapping of sequence-related genes provides a method for examining
- mammalian chromosome evolution. Genomics 10: 338-344, 1991.
-
- 5. Ramarao, C. S.; Kincade Denker, J. M.; Perez, D. M.; Gaivin, R.
- J.; Riek, R. P.; Graham, R. M.: Genomic organization and expression
- of the human alpha-1B-adrenergic receptor. J. Biol. Chem. 267:
- 21936-21945, 1992.
-
- 6. Yang-Feng, T. L.; Xue, F.; Zhong, W.; Cotecchia, S.; Frielle, T.;
- Caron, M. G.; Lefkowitz, R. J.; Francke, U.: Chromosomal organization
- of adrenergic receptor genes. Proc. Nat. Acad. Sci. 87: 1516-1520,
- 1990.
-
- *FIELD* CD
- Victor A. McKusick: 12/2/1987
-
- *FIELD* ED
- carol: 11/10/1994
- pfoster: 8/16/1994
- jason: 6/16/1994
- carol: 11/16/1993
- carol: 11/5/1993
- carol: 1/15/1993
-
- *RECORD*
- *FIELD* NO
- 104221
- *FIELD* TI
- *104221 ALPHA-1C-ADRENERGIC RECEPTOR; ADRA1C
- *FIELD* TX
- Schwinn et al. (1990) cloned the gene encoding the bovine
- alpha-1C-adrenergic receptor and localized its human counterpart to
- human chromosome 8 by somatic cell hybridization analysis. They used an
- interesting approach to demonstrate that the bovine gene is distinct
- from the hamster alpha-1B-adrenergic receptor; a human homolog of the
- latter gene is located on human chromosome 5 (104220). Despite the
- similarities in pharmacologic profile, the bovine alpha-1-adrenergic
- receptor showed differences in sensitivity to inhibition and lack of
- expression in some tissues in which the alpha-1A subtype (104219)
- existed. Hoehe et al. (1992) demonstrated a 2-allele PstI RFLP in the
- ADRA1C gene. Using this probe for the study of DNAs from the CEPH
- pedigrees, they concluded that the gene is closely linked (theta = 0.03)
- to NEFL (162280) on 8p21 (maximum lod = 12).
-
- *FIELD* RF
- 1. Hoehe, M. R.; Berrettini, W. H.; Schwinn, D. A.; Hsieh, W.-T.:
- A two-allele PstI RFLP for the alpha-1C adrenergic receptor gene (ADRA1C).
- Hum. Molec. Genet. 1: 349 only, 1992.
-
- 2. Schwinn, D. A.; Lomasney, J. W.; Lorenz, W.; Szklut, P. J.; Fremeau,
- R. T., Jr.; Yang-Feng, T. L.; Caron, M. G.; Lefkowitz, R. J.; Cotecchia,
- S.: Molecular cloning and expression of the cDNA for a novel alpha-1-adrenergic
- receptor subtype. J. Biol. Chem. 265: 8183-8189, 1990.
-
- *FIELD* CD
- Victor A. McKusick: 5/13/1991
-
- *FIELD* ED
- carol: 9/28/1992
- carol: 3/20/1992
- supermim: 3/16/1992
- carol: 10/1/1991
- carol: 5/13/1991
-
- *RECORD*
- *FIELD* NO
- 104222
- *FIELD* TI
- *104222 ALPHA-1D-ADRENERGIC RECEPTOR; ADRA1D
- *FIELD* TX
- As indicated in 104219, a receptor which was previously thought to
- represent the alpha-1A subtype of adrenergic receptor and to map to
- chromosome 5 was characterized pharmacologically as a distinct subtype,
- designated alpha-1D (Schwinn and Lomasney, 1992). Yang-Feng et al.
- (1994) mapped the ADRA1D gene to chromosome 20 by analysis of a
- mouse/human hybrid cell mapping panel and to 20p13 by isotopic in situ
- hybridization. Is it possible that this is, in fact, the same as ADRA1A
- (104219), which is located on chromosome 20?
-
- *FIELD* RF
- 1. Schwinn, D. A.; Lomasney, J. W.: Pharmacologic characterization
- of cloned alpha-1-adrenoceptor subtypes: selective antagonists suggest
- the existence of a fourth subtype. Europ. J. Pharm. 227: 433-436,
- 1992.
-
- 2. Yang-Feng, T. L.; Han, H.; Lomasney, J. W.; Caron, M. G.: Localization
- of the cDNA for an alpha-1-adrenergic receptor subtype (ADRA1D) to
- chromosome band 20p13. Cytogenet. Cell Genet. 66: 170-171, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 6/13/1994
-
- *FIELD* ED
- jason: 6/22/1994
- carol: 6/13/1994
-
- *RECORD*
- *FIELD* NO
- 104225
- *FIELD* TI
- *104225 LOW DENSITY LIPOPROTEIN-RELATED PROTEIN-ASSOCIATED PROTEIN 1; LRPAP1
- ALPHA-2-MACROGLOBULIN RECEPTOR-ASSOCIATED PROTEIN; A2; RAP; MRAP
- *FIELD* TX
- The alpha-2-macroglobulin receptor complex (107770), as purified by
- affinity chromatography, contains 3 polypeptides: a 515-kD heavy chain,
- an 85-kD light chain, and a 39-kD associated protein. The 515/85-kD
- components are derived from a 600-kD precursor whose complete sequence
- was determined by cDNA cloning (Herz et al., 1988). Strickland et al.
- (1991) determined the primary structure of the 39-kD polypeptide, termed
- alpha-2-macroglobulin receptor-associated protein (MRAP) by them, by
- cDNA cloning. The deduced amino acid sequence contains a putative signal
- sequence that precedes the 323-residue mature protein. The sequence
- showed 73% identity with a rat protein reported to be a pathogenic
- domain of the Heymann nephritis antigen gp 330 and 77% identity to a
- mouse heparin-binding protein termed HBP-44. There are also similarities
- between MRAP and apolipoprotein E (107741). Studies indicated that the
- molecule is present on the cell surface, forming a complex with the
- heavy and light chains of the alpha-2-macroglobulin receptor (103950).
-
- Using a human 1.5-kb cDNA clone encoding MRAP, Korenberg et al. (1994)
- performed fluorescence in situ hybridization to map the gene to human
- chromosome 4p16.3. This location is in the vicinity of the 2.5-Mb
- deletion associated with the Wolf-Hirschhorn syndrome (194190). The
- kidney hypoplasia associated with Wolf-Hirschhorn syndrome may be
- relevant in view of the high MRAP expression that is observed in this
- organ. The 39-kD MRAP has been shown to copurify and bind in vitro with
- high affinity to both LRP1 (107770) and LRP2 (600073). Although the
- function of MRAP remains to be established, MRAP can specifically
- inhibit ligand binding to both receptors. Although previous studies
- localized MRAP to the cell surface, Korenberg et al. (1994) stated: 'Its
- intracellular localization has led to suggestions that it might function
- in the biosynthesis of gp 330 and LRP, perhaps acting as a chaperone,
- preventing ligand binding during receptor trafficking.' The gene was
- symbolized also as LRPAP1 (low-density lipoprotein-associated
- protein-1).
-
- Jou et al. (1994) used the direct cDNA selection approach to isolate the
- LRPAP1 gene from cloned genomic DNA from the region of the Huntington
- disease gene (143100) located at 4p16.3. Van Leuven et al. (1995)
- assigned the LRPAP1 gene to chromosome 4 by PCR of human-hamster hybrid
- cell lines and to 4p16.3 by fluorescence in situ hybridization. Using an
- LRPAP1 genomic probe for fluorescence in situ hybridization, they
- studied 2 patients with deletions of 4p, resulting in the
- Wolf-Hirschhorn syndrome. One patient retained both copies of the gene,
- whereas the other patient displayed no signal for LRPAP1 on the deleted
- chromosome.
-
- Van Leuven et al. (1995) cloned the mouse gene coding for HBP-44 from a
- cosmid library and determined that its structure is very similar to that
- of the LRPAP1 gene: in both species, the known coding part of the cDNA
- is encoded by 8 exons and the position of the boundaries of the exons is
- conserved. (HBP-44 stands for 44-kD heparin-binding protein.)
-
- *FIELD* RF
- 1. Herz, J.; Hamann, U.; Rogne, S.; Myklebost, O.; Gausepohl, H.;
- Stanley, K. K.: Surface location and high affinity for calcium of
- a 500 kd liver membrane protein closely related to the LDL-receptor
- suggest a physiological role as lipoprotein receptor. EMBO J. 7:
- 4119-4127, 1988.
-
- 2. Jou, Y.-S.; Goold, R. D.; Myers, R. M.: Localization of the alpha-2-macroglobulin
- receptor-associated protein 1 gene (LRPAP1) and other gene fragments
- to human chromosome 4p16.3 by direct cDNA selection. Genomics 24:
- 410-413, 1994.
-
- 3. Korenberg, J. R.; Argraves, K. M.; Chen, X.-N.; Tran, H.; Strickland,
- D. K.; Argraves, W. S.: Chromosomal localization of human genes for
- the LDL receptor family member glycoprotein 330 (LRP2) and its associated
- protein RAP (LRPAP1). Genomics 22: 88-93, 1994.
-
- 4. Strickland, D. K.; Ashcom, J. D.; Williams, S.; Battey, F.; Behre,
- E.; McTigue, K.; Battey, J. F.; Argraves, W. S.: Primary structure
- of alpha-2-macroglobulin receptor-associated protein: human homologue
- of a Heymann nephritis antigen. J. Biol. Chem. 266: 13364-13369,
- 1991.
-
- 5. Van Leuven, F.; Hilliker, C.; Serneels, L.; Umans, L.; Overbergh,
- L.; De Strooper, B.; Fryns, J. P.; Van den Berghe, H.: Cloning, characterization,
- and chromosomal localization to 4p16 of the human gene (LRPAP1) coding
- for the alpha-2-macroglobulin receptor-associated protein and structural
- comparison with the murine gene coding for the 44-kDa heparin-binding
- protein. Genomics 25: 492-500, 1995.
-
- *FIELD* CD
- Victor A. McKusick: 4/12/1994
-
- *FIELD* ED
- mark: 12/31/1996
- mark: 12/6/1995
- carol: 3/6/1995
- terry: 1/9/1995
- jason: 6/16/1994
- mimadm: 4/12/1994
-
- *RECORD*
- *FIELD* NO
- 104230
- *FIELD* TI
- *104230 FUCOSYLTRANSFERASE-4; FUT4
- ALPHA-3-FUCOSYLTRANSFERASE; FCT3A;;
- CD15;;
- MYELOID-ASSOCIATED SURFACE ANTIGEN
- *FIELD* TX
- In human/mouse myeloid cell hybrids, Geurts van Kessel et al. (1984)
- tested for reactivity with monoclonal antibodies with known myelocytic,
- monocytic, or myelomonocytic specificity. Twenty antibodies, all of
- which bind specifically to the surface of human myeloid cells, exhibited
- similar reactivity patterns with the hybrid clones. Chromosomal analysis
- showed that the gene or genes involved in the expression of the one or
- more antigens recognized by these antibodies must be located on human
- 11q12-qter. This myeloid-associated surface antigen is designated CD15
- in the 'CD system.' Using panels of somatic cell and radiation hybrids
- which retained different rearrangements of chromosome 11, Reguigne et
- al. (1994) assigned this gene, which they symbolized FUT4, to 11q21
- between D11S388 and D11S919. Using fluorescence in situ hybridization
- and a cosmid containing FUT4 sequence, McCurley et al. (1995) confirmed
- the assignment of the FUT4 gene to 11q21.
-
- Tetteroo et al. (1987) found that alpha-3-fucosyltransferase activity is
- correlated with the presence of human chromosome 11 in human-mouse
- myeloid cell hybrids. Also, several other myeloid-associated
- carbohydrate antigens, e.g., Le(x), are associated with chromosome 11.
- Tetteroo et al. (1987) concluded, therefore, that the enzyme
- alpha-3-fucosyltransferase is responsible for the synthesis of these
- antigens. Using human/mouse hybrid cell lines, Couillin et al. (1991)
- mapped a human alpha-3-fucosyltransferase to 11q. Because the enzyme
- transfers fucose onto H type 2 more efficiently than onto
- sialyl-N-acetyllactosamine, Couillin et al. (1991) suggested that it is
- the myeloid type of alpha-3-fucosyltransferase which creates the
- 3-fucosyllactosamine epitope on polymorphonuclear cells and monocytes.
- (The Lewis enzyme (EC 2.4.1.65), alpha-3/4-fucosyltransferase, is coded
- by a gene on chromosome 19 (111100). It is never found in plasma but is
- found in human milk, digestive mucosa, and kidney. The plasma type of
- alpha-3-fucosyltransferase (EC 2.4.1.152) is found in hepatocytes and
- plasma; see 136835.)
-
- Gersten et al. (1995) demonstrated that the homolog of FUT4 maps to
- mouse chromosome 9 in a region of homology of synteny to 11q.
-
- *FIELD* RF
- 1. Couillin, P.; Mollicone, R.; Grisard, M. C.; Gibaud, A.; Ravise,
- N.; Feingold, J.; Oriol, R.: Chromosome 11q localization of one of
- the three expected genes for the human alpha-3-fucosyltransferases,
- by somatic hybridization. Cytogenet. Cell Genet. 56: 108-111, 1991.
-
- 2. Gersten, K. M.; Natsuka, S.; Trinchera, M.; Petryniak, B.; Kelly,
- R. J.; Hiraiwa, N.; Jenkins, N. A.; Gilbert, D. J.; Copeland, N. G.;
- Lowe, J. B.: Molecular cloning, expression, chromosomal assignment,
- and tissue-specific expression of a murine alpha-(1,3)-fucosyltransferase
- locus corresponding to the human ELAM-1 ligand fucosyl transferase. J.
- Biol. Chem. 270: 25047-25056, 1995.
-
- 3. Geurts van Kessel, A.; Tetteroo, P.; Van Agthoven, T.; Paulussen,
- R.; Van Dongen, J.; Hagemeijer, A.; Von dem Borne, A.: Localization
- of human myeloid-associated surface antigen detected by a panel of
- 20 monoclonal antibodies to the q12-qter region of chromosome 11.
- J. Immun. 133: 1265-1269, 1984.
-
- 4. McCurley, R. S.; Recinos, A., III; Olsen, A. S.; Gingrich, J. C.;
- Szczepaniak, D.; Cameron, H. S.; Krauss, R.; Weston, B. W.: Physical
- maps of human alpha(1,3)fucosyltransferase genes FUT3-FUT6 on chromosomes
- 19p13.3 and 11q21. Genomics 26: 142-146, 1995.
-
- 5. Reguigne, I.; James, M. R.; Richard, C. W., III; Mollicone, R.;
- Seawright, A.; Lowe, J. B.; Oriol, R.; Couillin, P.: The gene encoding
- myeloid alpha-3-fucosyltransferase (FUT4) is located between D11S388
- and D11S919 on 11q21. Cytogenet. Cell Genet. 66: 104-106, 1994.
-
- 6. Tetteroo, P. A. T.; de Heij, H. T.; Van den Eijnden, D. H.; Visser,
- F. J.; Schoenmaker, E.; Geurts van Kessel, A. H. M.: A GDP-fucose:(Gal-beta-1-to-4)GlcNAc
- alpha-1-to-3-fucosyltransferase activity is correlated with the presence
- of human chromosome 11 and the expression of the Le(x), Le(y), and
- sialyl-Le(x) antigens in human-mouse cell hybrids. J. Biol. Chem. 262:
- 15984-15989, 1987.
-
- *FIELD* CD
- Victor A. McKusick: 6/29/1988
-
- *FIELD* ED
- terry: 06/18/1996
- mark: 3/11/1996
- terry: 3/6/1996
- mark: 4/21/1995
- jason: 6/9/1994
- terry: 5/13/1994
- carol: 4/20/1994
- carol: 11/4/1992
- supermim: 3/16/1992
-
- *RECORD*
- *FIELD* NO
- 104240
- *FIELD* TI
- *104240 ALPHA-3-N-ACETYLNEURAMINYLTRANSFERASE
- CMP-N-ACETYLNEURAMINATE:BETA-GALACTOSIDASE ALPHA-2,3-SIALYLTRANSFERASE;;
- ; CGS23; NANTA3;;
- SIALYLTRANSFERASE 4; SIAT4
- *FIELD* TX
- Tetteroo et al. (1987) stated in an addendum that chromosome 11 codes
- for an alpha-3-N-acetylneuraminyltransferase involved in the sialylation
- of O-linked Gal-beta-1-to-3Gal-3GalNAc-alpha-to-R chains. The assignment
- to chromosome 11 was achieved by study of somatic cell hybrids (de Heij
- et al., 1988).
-
- *FIELD* RF
- 1. de Heij, H. T.; Tetteroo, P. A. T.; Geurts van Kessel, A. H. M.;
- Schoenmaker, E.; Visser, F. J.; van den Eijnden, D. H.: Specific
- expression of a myeloid-associated CMP-NeuAc:Gal-beta-1-3GalNAc-alpha-R-alpha-2-3-sialyltransferase
- and the sialyl-X determinant in myeloid human-mouse cell hybrids containing
- human chromosome 11. Cancer Res. 48: 1489-1493, 1988.
-
- 2. Tetteroo, P. A. T.; de Heij, H. T.; Van den Eijnden, D. H.; Visser,
- F. J.; Schoenmaker, E.; Geurts van Kessel, A. H. M.: A GDP-fucose:(Gal-beta-1-to-4)GlcNAc
- alpha-1-to-3-fucosyltransferase activity is correlated with the presence
- of human chromosome 11 and the expression of the Le(x), Le(y), and
- sialyl-Le(x) antigens in human-mouse cell hybrids. J. Biol. Chem. 262:
- 15984-15989, 1987.
-
- *FIELD* CD
- Victor A. McKusick: 6/29/1988
-
- *FIELD* ED
- jason: 6/13/1994
- carol: 1/11/1993
- supermim: 3/16/1992
- carol: 2/27/1992
- carol: 6/13/1990
- supermim: 3/20/1990
-
- *RECORD*
- *FIELD* NO
- 104250
- *FIELD* TI
- *104250 ALPHA-2C-ADRENERGIC RECEPTOR; ADRA2C
- ALPHA-2-ADRENERGIC RECEPTOR, RENAL TYPE
- *FIELD* TX
- Regan et al. (1988) cloned an alpha-2-adrenergic receptor subtype from a
- human kidney cDNA library using the gene for the human platelet
- alpha-2-adrenergic receptor as a probe. The deduced amino acid sequence
- resembled the human platelet alpha-2-adrenergic receptor. The gene for
- this receptor was found to be on human chromosome 4, whereas the gene
- for platelet receptor (104210) is on chromosome 10. (Curiously, the
- location of the gene on chromosome 4 was stated in the abstract but not
- documented by results reported in the paper.) In this work, Regan et al.
- (1988) achieved expression of the receptor in cultured cells, free of
- other adrenergic receptor subtypes; this approach should help in
- developing more selective alpha-adrenergic ligands for pharmaceutical
- purposes. Hoehe et al. (1989) found close linkage between the G8 (D4S10)
- marker of Huntington disease (HD; 143100) and a RFLP of the ADRA2C gene;
- thus, the ADRA2C gene is presumably in band 4p16.1.
-
- By studying cosmid clones covering the entire gene, Riess et al. (1994)
- found that the ADRA2C gene is intronless. Using 2 (GT)n repeats in close
- proximity to the ADRA2C gene, they analyzed its precise location.
- Linkage disequilibrium studies of one microsatellite in Huntington
- disease families showed strong nonrandom association to the HD mutation,
- indicating tight linkage to the HD gene. The investigation of families
- carrying recombinant chromosomes, pulsed-field analysis, and genomic
- walking mapped the ADRA2C gene adjacent to D4S81, 500 kb proximal to the
- HD gene.
-
- *FIELD* RF
- 1. Hoehe, M.; Berrettini, W.; Leppert, M.; Lalouel, J.-M.; Byerley,
- W.; Gershon, E.; White, R.: Genetic mapping of adrenergic receptor
- genes. (Abstract) Am. J. Hum. Genet. 45 (suppl.): A143 only, 1989.
-
- 2. Regan, J. W.; Kobilka, T. S.; Yang-Feng, T. L.; Caron, M. G.; Lefkowitz,
- R. J.; Kobilka, B. K.: Cloning and expression of a human kidney cDNA
- for an alpha-2-adrenergic receptor subtype. Proc. Nat. Acad. Sci. 85:
- 6301-6305, 1988.
-
- 3. Riess, O.; Thies, U.; Siedlaczck, I.; Potisek, S.; Graham, R.;
- Theilmann, J.; Grimm, T.; Epplen, J. T.; Hayden, M. R.: Precise mapping
- of the brain alpha-2-adrenergic receptor gene within chromosome 4p16.
- Genomics 19: 298-302, 1994.
-
- *FIELD* CD
- Victor A. McKusick: 9/15/1988
-
- *FIELD* ED
- carol: 2/10/1994
- supermim: 3/16/1992
- carol: 3/5/1992
- carol: 9/9/1990
- supermim: 3/20/1990
- carol: 12/14/1989
-
- *RECORD*
- *FIELD* NO
- 104260
- *FIELD* TI
- *104260 ALPHA-2B-ADRENERGIC RECEPTOR; ADRA2B
- ALPHA-2-ADRENERGIC RECEPTOR-LIKE 1;;
- ADRA2L1
- *FIELD* TX
- Regan et al. (1988) indicated that in addition to the platelet
- alpha-2-adrenergic receptor (encoded by chromosome 10; 104210) and the
- renal form of receptor (encoded by chromosome 4; 104250), a related
- protein is coded by chromosome 2. Lomasney et al. (1990) also cloned the
- ADRA2B gene. By hybridization with somatic cell hybrids, they showed
- that the gene for this receptor is located on chromosome 2. Northern
- blot analysis of various rat tissues showed expression in liver and
- kidney. Unique pharmacology and tissue localization suggested that this
- was a previously unidentified subtype.
-
- *FIELD* RF
- 1. Lomasney, J. W.; Lorenz, W.; Allen, L. F.; King, K.; Regan, J.
- W.; Yang-Feng, T. L.; Caron, M. G.; Lefkowitz, R. J.: Expansion of
- the alpha-2-adrenergic receptor family: cloning and characterization
- of a human alpha-2-adrenergic receptor subtype, the gene for which
- is located on chromosome 2. Proc. Nat. Acad. Sci. 87: 5094-5098,
- 1990.
-
- 2. Regan, J. W.; Kobilka, T. S.; Yang-Feng, T. L.; Caron, M. G.; Lefkowitz,
- R. J.; Kobilka, B. K.: Cloning and expression of a human kidney cDNA
- for an alpha-2-adrenergic receptor subtype. Proc. Nat. Acad. Sci. 85:
- 6301-6305, 1988.
-
- *FIELD* CD
- Victor A. McKusick: 9/20/1988
-
- *FIELD* ED
- jason: 6/16/1994
- supermim: 3/16/1992
- carol: 3/5/1992
- carol: 6/24/1991
- carol: 9/9/1990
- carol: 8/13/1990
-
- *RECORD*
- *FIELD* NO
- 104290
- *FIELD* TI
- 104290 ALTERNATING HEMIPLEGIA OF CHILDHOOD
- *FIELD* TX
- Alternating hemiplegia of childhood is a rare syndrome of episodic hemi-
- or quadriplegia lasting minutes to days. Most cases are accompanied by
- dystonic posturing, choreoathetoid movements, nystagmus, other ocular
- motor abnormalities, autonomic disturbances, and progressive cognitive
- impairment. Mikati et al. (1992) reported what appeared to be the first
- instance of familial occurrence. Inheritance appeared to be autosomal
- dominant. The proband, a 9-year-old boy, presented with developmental
- retardation, rare tonic-clonic seizures and frequent episodes of flaccid
- alternating hemiplegia that had been presumed to represent postictal
- paralysis. The hemiplegia spells, which started in his first year, did
- not respond to multiple antiepileptics. Between attacks, there was
- choreoathetosis and dystonic posturing. A brother, the father, a
- paternal uncle, and the maternal grandmother had similar histories of
- alternating hemiplegia. Investigations included negative CT and
- metabolic studies. EEG and SPECT scanning failed to reveal any
- significant slowing or major changes in cortical perfusion during
- hemiplegia as compared with nonhemiplegic periods. The karyotype
- demonstrated a balanced reciprocal translocation, 46,XY,t(3;9)(p26;q34)
- in the patient, in all the affected living relatives, and in 1
- apparently unaffected sib. The asymptomatic mother had a normal
- karyotype. Both affected sibs were treated with and responded to
- flunarizine, with a greater than 70% decrease in attack frequency.
-
- *FIELD* RF
- 1. Mikati, M. A.; Maguire, H.; Barlow, C. F.; Ozelius, L.; Breakefield,
- X. O.; Klauck, S. M.; Korf, B.; O'Tuama, S. L. A.; Dangond, F.: A
- syndrome of autosomal dominant alternating hemiplegia: clinical presentation
- mimicking intractable epilepsy; chromosomal studies; and physiologic
- investigations. Neurology 42: 2251-2257, 1992.
-
- *FIELD* CD
- Victor A. McKusick: 3/16/1994
-
- *FIELD* ED
- carol: 3/16/1994
-
- *RECORD*
- *FIELD* NO
- 104300
- *FIELD* TI
- #104300 ALZHEIMER DISEASE; AD
- PRESENILE AND SENILE DEMENTIA;;
- ALZHEIMER DISEASE, FAMILIAL; FAD
- *FIELD* MN
-
- Alzheimer disease is by far the most common cause of dementia.
- Clinically, it cannot be distinguished from Pick disease (172700).
-
- The histopathological picture is characterized by neurofibrillary
- tangles and amyloid plaques, which contain a novel amyloid protein, beta
- protein. It is suggested that the amyloid in Alzheimer disease (and Down
- syndrome) is formed from a precursor synthesized in neurons, where it
- produces neurofibrillary tangles, and in microglial cells and brain
- macrophages from which it is exuded and forms the extracellular amyloid
- plaques and vascular amyloid deposits (Gajdusek, 1986).
-
- In a study of 70 kindreds, Farrer et al. (1990) found evidence of 2
- categories of families: those with mean age of onset less than 58 years
- (early onset form) and those with mean age of onset greater than 58
- years (late-onset form).
-
- Early onset FAD is, in some families, due to a mutation of a gene, AD1,
- near the centromere on chromosome 21q, that codes for amyloid precursor
- protein (104760.0002) (Lawrence et al., 1992). Other early onset
- families show linkage to markers on 14q (Van Broeckhoven et al., 1992),
- and there may be a second locus on 21 (St. George-Hyslop et al., 1990).
-
- In one representative study (van Dujin et al., 1993) the lifetime risk
- (to age 90) of first degree relatives of early onset cases (less than 65
- years) was about 40%; higher in females than males (56 vs. 22%) and in
- parents than sibs (42 vs. 18%) compared to 14% for controls. The risk at
- age 70 was about 13% for first-degree relatives versus about 7% for
- controls.
-
- The situation for late-onset AD is even more complex, involving several
- loci, and perhaps polygenic and environmental contributions (Haines,
- 1991). Most, if not all, families with late-onset FAD have mutations on
- chromosomes other than 21, particularly AD2 (104310) on chromosome 19
- (Pericak-Vance et al., 1991). The recently discovered relationship of
- late-onset AD to the apolipoprotein E type 4 allele on chromosome 19 may
- clarify the picture (Corder et al., 1993). See 104310. In a series of 42
- late-onset families, 20% of affected members had no copies of E4, 47%
- had one, and 91% had two copies. Mean ages of onset were 84, 76, and 68
- years, respectively.
-
- *FIELD* TX
-
- DESCRIPTION
-
- A number sign (#) is used with this entry because of evidence that
- mutations in at least 4 genes can cause Alzheimer disease: AD1 is caused
- by mutations in the amyloid precursor gene (104760); AD2 is associated
- with the APOE*4 allele on chromosome 19 (107741); AD3 is caused by
- mutation in a chromosome 14 gene encoding a 7-transmembrane domain
- protein (104311); and AD4 is caused by mutation in a gene on chromosome
- 1 that encodes a similar 7-transmembrane domain protein (600759). In
- addition, evidence has been presented suggesting that mitochondrial DNA
- polymorphisms may be risk factors in Alzheimer disease (502500).
-
- Alzheimer disease, the most common cause of dementia, is inherited as an
- autosomal dominant trait in some families.
-
- Selkoe (1996) reviewed the pathophysiology, chromosomal loci, and
- pathogenetic mechanisms of Alzheimer disease as well as future research
- themes in the field.
-
- CLINICAL FEATURES
-
- Alzheimer disease is by far the most common cause of dementia. Terry and
- Davies (1980) pointed out that the presenile form (with onset before age
- 65) is identical to the most common form of senile dementia. Thus, they
- recommended the designation senile dementia of the Alzheimer type
- (SDAT). Clinically, Alzheimer disease cannot be distinguished from Pick
- disease (172700).
-
- Schottky (1932) described presenile dementia in 4 generations. The
- diagnosis was confirmed at autopsy in a patient in the fourth
- generation. Lowenberg and Waggoner (1934) reported a family with
- unusually early onset in the father and 4 of 5 children. Postmortem
- findings in 1 case were described. McMenemey et al. (1939) described 4
- affected males in 2 generations with pathologic confirmation in one.
-
- Heston et al. (1966) described a family with 19 affected in 4
- generations. Dementia was coupled with conspicuous parkinsonism and long
- tract signs. In a study of the families of Alzheimer disease patients,
- Heston (1977) found an excess of Down syndrome and of myeloproliferative
- disorders, e.g., lymphoma and leukemia. Although the mechanism is not
- clear, Heston (1977) speculated that a disorder of microtubules
- underlies the association. Microtubules are involved in the spatial
- orientation of chromosomes and their separation in meiosis and mitosis.
- Neurons of Alzheimer patients show a neurofibrillary tangle that is made
- up of disordered microtubules. An identical lesion occurs in the neurons
- of Down syndrome, at an earlier age than in Alzheimer disease. Leukemia
- and accelerated aging are also features of Down syndrome. In a large
- multicenter study of first-degree relatives of Alzheimer disease
- probands and nondemented spouse controls, Silverman et al. (1994) found
- only one case of Down syndrome, a relative of a spouse control. On the
- basis of a study of the families of 188 Down syndrome children and 185
- controls, Berr et al. (1989) found no evidence of an excess of dementia
- cases with insidious onset suggestive of dementia of Alzheimer type in
- the families of children with classic trisomy 21. One mechanism whereby
- Alzheimer disease might occur in a parent of a Down syndrome patient is
- somatic mosaicism in that parent.
-
- Harper et al. (1979) could not confirm that a systemic microtubular
- defect exists in Alzheimer disease. Cultured skin fibroblasts showed
- normal tubulin networks. Nordenson et al. (1980) found an increased
- frequency of acentric fragments in karyotypes from patients with
- Alzheimer disease. They viewed this as consistent with defective tubulin
- protein leading to erratic function of the spindle mechanism.
-
- Rice et al. (1980) and Ball (1980) reported a kindred in which members
- had the clinical features of familial Alzheimer disease but histologic
- changes of spongiform encephalopathy of the Creutzfeldt-Jakob type
- (123400) at autopsy. The clinical course, with dementia for as long as
- 10 years, was unusual for CJD. Masters et al. (1981) studied 52 families
- and compared them with familial Creutzfeldt-Jakob disease. The age at
- death and duration of illness was greater in AD. No maternal effect was
- evident in the pattern of autosomal dominant inheritance. In 4 families
- with AD, 1 or more members had died from CJD. In 17 other families with
- AD, 1 or more members presented with clinical features suggesting CJD.
- Although a virus causing an experimental spongiform encephalopathy was
- isolated from the brain of 2 cases of familial AD, brain tissue from
- most sporadic and familial cases of AD failed to cause disease when
- inoculated into nonhuman primates.
-
- In the families of 17 of 68 cases, Heyman et al. (1983) found secondary
- cases in parents and sibs. The cumulative incidence in these relatives
- was about 14% at age 75. A probable increase in the frequency of Down
- syndrome was noted: 3.6 per 1,000 as compared with an expected rate of
- 1.3 per 1,000. A history of thyroid disease was unusually frequent (9 of
- 46; 19.6%) in the female probands. No excess of hematologic malignancy
- was found in relatives. Parental age at time of birth of the probands
- did not differ from the normal. Corkin et al. (1983) also could find no
- difference in parental age from that in controls.
-
- Joachim et al. (1989) presented evidence suggesting that Alzheimer
- disease is not restricted to the brain but is a widespread systemic
- disorder with accumulation of amyloid beta protein in nonneuronal
- tissues.
-
- In a study of 70 kindreds containing 541 affected and 1,066 unaffected
- offspring of demented parents, Farrer et al. (1990) found evidence of 2
- categories of families: those with mean age of onset less than 58 years
- (early-onset form) and those with mean age of onset greater than 58
- years (late-onset form). At-risk offspring in early-onset families had
- an estimated lifetime risk for dementia of 53%, leading Farrer et al.
- (1990) to suggest autosomal dominant inheritance. The lifetime risk in
- late-onset families was 86%. Farrer et al. (1990) concluded that this
- form may have at least 2 causes: autosomal dominant inheritance in some
- families and other genetic or shared environmental factors in other
- families. Farrer et al. (1990) pointed out that some early-onset
- families show linkage to markers on chromosome 21, whereas there is
- evidence against linkage to the same group of markers in late-onset
- families. By the criteria of the analysis, the Volga Germans (Bird et
- al., 1988), who are among the unlinked families, were classified as the
- upper boundary of the early-onset group.
-
- In a complex segregation analysis on 232 nuclear families ascertained
- through a single proband who was referred for diagnostic evaluation of
- memory disorder, Farrer et al. (1991) concluded that susceptibility to
- AD is determined, in part, by a major autosomal dominant allele with an
- additional multifactorial component. The frequency of the AD
- susceptibility allele is estimated to be 0.038, but the major locus was
- thought to account for only 24% of the 'transmission variance,'
- indicating a substantial role for other genetic and nongenetic
- mechanisms.
-
- Silverman et al. (1994) used a standardized family history assessment to
- study first-degree relatives of Alzheimer disease probands and
- nondemented spouse controls. First-degree relatives of the probands with
- Alzheimer disease had a significantly greater cumulative risk of
- Alzheimer disease (24.8%) than did the relatives of spouse controls
- (15.2%). The cumulative risk for the disorder among female relatives of
- probands was significantly greater than that among male relatives.
-
- BIOCHEMICAL FEATURES
-
- Glenner and Wong (1984) identified a novel amyloid protein, called beta
- protein (APP; 104760), in Alzheimer disease. The 4.2-kD polypeptide was
- called beta protein because of its partial beta-pleated sheet structure.
- It was identified in both amyloid plaque core and in cerebral vascular
- amyloid; both have an identical 28-amino acid sequence. A cDNA for the
- beta protein suggested that it is derived from a larger protein
- expressed in a variety of tissues (Tanzi et al., 1987).
-
- Kang et al. (1987) isolated and sequenced an apparently full-length cDNA
- clone coding for the A4 polypeptide (the designation they used for the
- major protein subunit of the amyloid fibril of tangles, plaques, and
- blood vessel deposits in AD and Down syndrome). The predicted precursor
- consisted of 695 residues and contained features characteristic of
- glycosylated cell-surface receptors.
-
- Abraham et al. (1988) identified one of the components of the amyloid
- deposits seen in Alzheimer disease as the serine protease inhibitor
- alpha-1-antichymotrypsin. Carrell (1988) speculated that plaque
- formation in Alzheimer disease is a consequence of proteolysis of the
- precursor protein; self-aggregation of the cleaved A4 peptides explains
- the precipitated amyloid, while release of a trophic inhibitory domain
- explains the interwoven neuritic development. Zubenko et al. (1987)
- described a biophysical alteration of platelet membranes in Alzheimer
- disease. They concluded that increased platelet membrane fluidity
- identifies a subgroup of patients with early age of symptomatic onset
- and rapidly progressive course.
-
- Zubenko and Ferrell (1988) described monozygotic twins concordant for
- probable Alzheimer disease and for increased platelet membrane fluid.
- See 173560. Birchall and Chappell (1988) suggested that individual
- vulnerability to aluminum might depend on genetic factors influencing
- intake, transport or excretion, and might be a mechanism for familial
- Alzheimer disease. The inositol phosphate system may be particularly
- vulnerable.
-
- Ponte et al. (1988), Tanzi et al. (1988), and Kitaguchi et al. (1988)
- showed that the amyloid protein precursor contains a domain very similar
- to the Kunitz family of serine protease inhibitors. All 3 groups found
- the variable presence of a domain of 56 residues interpolated at residue
- 289, that is, in the proposed extracellular portion of the amyloid
- precursor protein. The best-studied member of the protease inhibitor
- family is bovine pancreatic trypsin inhibitor, also called aprotinin.
- The newly found amyloid protein sequence was 50% identical to aprotinin
- and also to the second inhibitory domain of the human plasma protein,
- inter-alpha-trypsin inhibitor.
-
- Yan et al. (1996) reported that the AGER protein (600214), called RAGE
- (receptor for advanced glycation end products) by them, is an important
- receptor for the amyloid beta peptide and that expression of this
- receptor increases in Alzheimer disease. They noted that expression of
- RAGE is particularly increased in neurons close to deposits of amyloid
- beta peptide and to neurofibrillary tangles.
-
- OTHER FEATURES
-
- Gajdusek (1986) suggested that the amyloid in Alzheimer disease and Down
- syndrome is formed from a precursor synthesized in neurons as well as in
- microglial cells and brain macrophages: that synthesized in neurons
- produces neurofibrillary tangles, and that synthesized in microglial
- cells and brain macrophages is exuded from the cell and forms the
- extracellular amyloid plaques and vascular amyloid deposits. Dying
- neurons may also contribute to extracellular deposits.
-
- Wolozin et al. (1988) performed immunocytochemical studies of cerebral
- cortex tissue sections from normal human fetal and neonatal brain, and
- of brain tissue from individuals with Down syndrome and patients with
- Alzheimer disease. They used the monoclonal antibody ALZ-50, which
- recognizes a 68-kD protein. The authors reported that ALZ-50-reactive
- neurons are found in normal fetal and neonatal human brain as well as in
- brain tissue from neonates with Down syndrome. The number of reactive
- neurons decreased sharply after age 2 years, but reappeared in older
- individuals with Down syndrome and in patients with Alzheimer disease.
-
- INHERITANCE
-
- From an extensive study in Sweden, Sjogren et al. (1952) suggested that
- whereas Pick disease may be dominant with important modifier genes,
- Alzheimer disease is multifactorial. However, a dominant pattern of
- inheritance, more common in presenile cases than in older patients, is
- well documented and accounts for about one-third of all cases of
- Alzheimer disease.
-
- Masters et al. (1981) found no maternal effect in the autosomal dominant
- inheritance pattern of 52 families.
-
- In 7 of 21 families, Powell and Folstein (1984) found evidence of
- 3-generation transmission. Paternal age was raised, they concluded, in
- the case of new mutation cases. Age of onset varied from 25 to 85 years.
- Breitner and Folstein (1984) suggested that most cases of Alzheimer
- disease are familial. Fitch et al. (1988) found a familial incidence of
- 43%. They could detect no clinical differences between the familial and
- sporadic cases. In one-third of the familial cases, the gene was not
- expressed until after age 70. In a continuing longitudinal study of
- family members of probands with Alzheimer disease, Breitner et al.
- (1988) found that the cumulative incidence of Alzheimer disease among
- relatives was 49% by age 87. The risk was similar among parents and
- siblings and did not differ significantly between relatives of
- presenile-onset versus senile-onset probands.
-
- Rao et al. (1996) carried out a complex segregation analysis in 636
- nuclear families of consecutively ascertained and rigorously diagnosed
- probands in the Multi-Institutional Research in Alzheimer Genetic
- Epidemiology study in order to derive models of disease transmission
- that account for the influences of the APOE genotype of the proband and
- gender. In the total group of families, models postulating sporadic
- occurrence, no major gene effect, random environmental transmission, and
- mendelian inheritance were rejected. Transmission of AD in families of
- probands with at least 1 APOE4 allele best fitted a dominant model.
- Moreover, single gene inheritance best explained clustering of the
- disorder in families of probands lacking APOE4, but a more complex
- genetic model or multiple genetic models may ultimately account for risk
- in this group of families. The results suggested to Rao et al. (1996)
- that susceptibility to AD differs between men and women regardless of
- the proband's APOE status. Assuming a dominant model, AD appeared to be
- completely penetrant in women, whereas only 62% to 65% of men with
- predisposing genotypes developed AD. However, parameter estimates from
- the arbitrary major gene model suggested that AD is expressed dominantly
- in women and additively in men. These observations, taken together with
- epidemiologic data, were considered consistent with the hypothesis of an
- interaction between genes and other biologic factors affecting disease
- susceptibility.
-
- CYTOGENETICS
-
- Percy et al. (1991) described 2 sisters thought to have Alzheimer
- disease of late onset who also had an unusual chromosome 22-derived
- marker with a greatly elongated short arm containing 2 well-separated
- nucleolus organizer regions. Eleven of 24 of their biological relatives
- were also found to have the marker. In the sisters' generation and in
- the previous generation, 7 persons with Alzheimer disease had died. The
- average age at onset of dementia was 65.8 years and the average age at
- death, 74.9 years.
-
- MAPPING
-
- Wheelan and Race (1959) studied a family in which the mother and 5 of 10
- children were affected. Possible linkage with the MNS locus was found.
-
- In the large kindred reported by Nee et al. (1983), Weitkamp et al.
- (1983) studied the transmission of HLA and Gm types and concluded that
- 'genes in the HLA region of chromosome 6 and perhaps also in the Gm
- region of chromosome 14 are determinants of susceptibility.' The
- association between immunoglobulins and the amyloid in the senile plaque
- of AD was thought to be significant in this connection. The peak lod
- score with Gm was 1.37 (at theta = 0.05).
-
- Nee et al. (1983) reported the most extensively affected kindred, with
- 51 affected persons in 8 generations. No preponderance of affected
- females and no increased incidence of Down syndrome or hematologic
- malignancy were found.
-
- Nerl et al. (1984) reported an increase in the frequency of the C4B
- (120820) allele C4B2 in patients with Alzheimer disease, but Eikelenboom
- et al. (1988) failed to find a significant association between C4B2
- allelic frequency and AD.
-
- Kang et al. (1987) showed by somatic cell hybrids that the gene for A4
- peptide is localized to chromosome 21. They commented on the fact that
- this protein shows similarities to the prion protein (PRNP; 176640)
- found in the amyloid of transmissible spongiform encephalopathies (Oesch
- et al., 1985). Membrane-spanning domains of both proteins may share an
- amyloid-forming or amyloid-inducing potential.
-
- St. George-Hyslop et al. (1987) studied 4 extensive kindreds with many
- members affected with familial Alzheimer disease (FAD). They found
- linkage to DNA markers on chromosome 21. The markers in band 21q22,
- critical to the development of Down syndrome, showed negative lod
- scores. Notably, the marker B21S58, which is tightly linked to SOD1
- (147450), was not tightly linked. The linked markers were found to lie
- on the centromere side of q22 in the region 21q11.2-21q21. Using a RFLP
- of SOD1 in the study of a large family with Alzheimer disease, David et
- al. (1988) concluded that SOD1 and AD are not closely linked. Goldgaber
- et al. (1987) used the first 20 of the 28 amino acids in the sequence to
- prepare an oligonucleotide probe for isolation of cDNA. They found that
- a 3.5-kb mRNA was detectable in mammalian brains and human thymus. The
- gene was found to be highly conserved in evolution and was mapped to
- chromosome 21 by somatic cell hybridization.
-
- The type of Alzheimer disease coded by chromosome 21 may be an
- early-onset type; families with late onset are said not to show linkage
- to chromosome 21 markers (HGM9) (Cheng et al., 1988).
-
- Using a RFLP of the A4-amyloid gene, Van Broeckhoven et al. (1987) found
- recombinants in 2 Alzheimer disease families. Two of their families were
- of early onset: one with 36 cases in 6 generations of which 10 had been
- histopathologically confirmed (mean age of onset, 33 years), and the
- second with 22 cases in 5 generations of which 4 had been
- histopathologically confirmed (mean age of onset, 34 years). All lod
- scores were negative in these 2 families. In 1 of 5 families of late
- onset, positive lod scores were observed. These data demonstrated that
- the gene for plaque core A4-amyloid cannot be the locus of the defect
- causing Alzheimer disease in these families. Tanzi et al. (1987) also
- found recombination between Alzheimer disease and the amyloid protein
- and came to the same conclusion.
-
- Haines et al. (1987), who studied 4 large families with FAD, found
- linkage with 2 DNA markers on chromosome 21 that had previously been
- shown to be linked to each other at a distance of 8 cM. However, the
- pair-wise linkage analysis showed a lod score of 2.37 at theta = 0.08
- for one and 2.32 at theta = 0.00 for the other. The use of multipoint
- analysis provided stronger evidence for linkage with a peak score of
- 4.25.
-
- Bird et al. (1988) described 7 families with autopsy-confirmed AD, all
- being descendants of a group of immigrants known as the Volga Germans,
- who came to the United States between 1870 and 1920. Their ancestors had
- moved from Germany to the southern Volga region of Russia in the 1760s.
- All 5 were descendants of persons who originally lived in 2 small
- adjacent Volga German villages and shared several surnames known to have
- been present in the census records of those villages. There are more
- than 300,000 American descendants of the Volga Germans. In a further
- study of the 7 Volga German kindreds and in 8 other kindreds, all with
- autopsy-proven AD (except for 1 of the German Volga families),
- Schellenberg et al. (1988) could demonstrate no linkage to chromosome 21
- markers. Other researchers have been unable to demonstrate linkage
- between late-onset Alzheimer disease and chromosome 21 markers, but the
- disorder in the families studied by Schellenberg et al. (1988) was of
- the early-onset type. The families studied by St. George-Hyslop et al.
- (1987) in which linkage with chromosome 21 markers was found had the
- early-onset type. The data strongly suggest that there is at least 1
- other genetically distinct form of Alzheimer disease. (Rogaev et al.
- (1995) demonstrated that the mutation in the Volga Germans is located in
- the presenilin-2 gene encoded by chromosome 1 (600759.0001).)
-
- By the study of linkage to DNA markers, Van Broeckhoven et al. (1988)
- concluded that the gene for early-onset familial Alzheimer disease is
- located close to the centromere of chromosome 21. Pulst et al. (1989)
- used a panel of aneuploid cell lines containing various regions of human
- chromosome 21 to map the physical order of DNA probes linked to the FAD
- locus. Van Camp et al. (1989) described the isolation of 35 chromosome
- 21 specific DNA probes for analysis in Alzheimer disease and Down
- syndrome. Ross et al. (1989) described the isolation of cDNAs from brain
- and spinal cord, mapping to chromosome 21, for investigation in
- Alzheimer disease. Pericak-Vance et al. (1988) found no linkage to
- chromosome 21 specific probes in studies of 13 families with FAD. The
- same group (Pericak-Vance et al., 1989, 1990) presented evidence for
- linkage to 2 markers on chromosome 19. When analysis was limited to the
- affecteds only, a lod score of 2.5 at theta = 0 was obtained for linkage
- with BCL3 (109560). Pericak-Vance et al. (1991) found evidence of both
- chromosome 19 linkage in their late-onset FAD families and chromosome 21
- linkage in their early-onset FAD families. When only affected persons
- were used in the analysis, a high lod score was obtained also with
- ATP1A3 (182350), which maps to 19q12-q13.2. Haines (1991) gave a review.
-
- Using the exclusion mapping method of Edwards (1987) and the
- affected-pedigree-member method (APM) of Weeks and Lange (1988), Roses
- et al. (1989) found some suggestion of implication of chromosome 19;
- predominantly late-onset families were studied.
-
- Van Broeckhoven et al. (1989) described linkage analysis of 2 families
- with Alzheimer disease by use of chromosome 21 DNA markers. With probe
- D21S13, they found a lod score of 1.52 at theta = 0.09 in 1 family.
- Further studies analyzing D21S13 with D21S16 and D21S1/S11, 2 markers
- that had previously been linked to Alzheimer disease, found D21S13 to be
- tightly linked to D21S16 with a peak lod score of 6.24 at theta = 0.
- Pulsed field gel electrophoresis confirmed that the loci are separated
- by a distance of approximately 400 kb.
-
- Using pulsed field gel electrophoresis to construct a physical map of
- the region of chromosome 21 around the FAD locus, Owen et al. (1989)
- suggested the following order:
- cen--D21S16--D21S48--D21S13--D21S46--(D21S52, D21S4)--(D21S1, D21S11).
- Using genetic linkage analysis, Goate et al. (1989) found a peak lod
- score of 3.3 between the FAD locus and locus D21S16.
-
- Pulst et al. (1991) excluded the proximal portion of the long arm of
- chromosome 21 as the site of the AD gene in 1 large kindred.
-
- Because of the conflicting findings concerning linkage to chromosome 21,
- St. George-Hyslop and many other members of the FAD collaborative study
- group undertook a study of 5 polymorphic chromosome 21 markers in a
- large unselected series of pedigrees with FAD. The results seemed to
- indicate that, in many families at least, early-onset Alzheimer disease
- is indeed due to a mutation on chromosome 21, whereas the late-onset
- form has other causes. From the work of Goate et al. (1991), it seems
- clear that 1 form of early-onset AD is caused by mutation in the gene
- for amyloid precursor protein (104760.0002). The families with Alzheimer
- disease mapping to chromosome 21 represent this form. Other families
- with early-onset AD and probably all families with late-onset AD have
- mutations on chromosomes other than chromosome 21.
-
- Lawrence et al. (1992) reviewed the reported data on multiplex Alzheimer
- pedigrees for which lod scores had been reported; the AD1 locus which
- mapped to the site of the APP locus (104760) on 21q accounted for 63 +/-
- 11% of these pedigrees. The AD1/APP locus was placed at approximately
- 27.7 Mb from pter, corresponding to genetic intervals of 10.9 cM in
- males and 33.9 cM in females, flanked proximally by D21S8 and distally
- by D21S111. Since a much smaller proportion of pedigrees than 63% have
- mutations in the cDNA for beta-amyloid, which corresponds to exons 16
- and 17 of APP, it is likely that the AD1 locus spans controlling
- elements near those exons. There was no evidence in this analysis for a
- second locus on chromosome 21.
-
- MOLECULAR GENETICS
-
- Delabar et al. (1986) analyzed DNA from 4 patients with Alzheimer
- disease and estimated the state of markers on chromosome 21. In all 4
- cases, duplication of the ETS2 locus (164740) was found, whereas SOD1
- (147450) was normal. These studies were undertaken because the patients
- had a phenotype of trisomy 21 but were found to have a normal karyotype;
- by chemical investigations and DNA analyses, they showed partial trisomy
- due to duplication of a short segment of chromosome 21, located at the
- interface between 21q21 and 21q22.1 and carrying the SOD1 and ETS2
- genes.
-
- Blanquet et al. (1987) found by molecular genetic methods that the
- Alzheimer amyloid protein gene and the ETS2 oncogene are distally
- located in the normal individual; surprisingly, 2 hybridization peaks
- were observed for ETS2 in the Alzheimer patient, 1 at the normal site of
- the oncogene and 1 at the site of the amyloid protein. Blanquet et al.
- (1987) interpreted these results as indicating that Alzheimer disease is
- associated with a complex rearrangement within chromosome 21, by which 2
- distantly related genes come to lie in the vicinity of each other.
-
- Overexpression of the gene in brain tissue from fetuses with Down
- syndrome is explained by dosage effect since the locus encoding the beta
- protein maps to chromosome 21. Regional localization of the gene by
- somatic cell hybridization and with linkage to DNA markers placed it in
- the vicinity of the genetic defect causing the inherited form of
- Alzheimer disease. This was done with somatic cell hybridization and
- with linkage to DNA markers (Tanzi et al., 1987). The 28-amino acid
- sequence has a variation at position 11: glutamine in the case of the
- cerebral vascular amyloid of Alzheimer disease, but glutamic acid in the
- case of cerebral vascular amyloid of Down syndrome and the amyloid
- plaque core of both disorders (Tanzi et al., 1987).
-
- St. George-Hyslop et al. (1987), Tanzi et al. (1987), and Podlisny et
- al. (1987) could demonstrate no evidence of duplication of chromosome 21
- genes, and the amyloid beta protein gene specifically, in patients with
- either familial or sporadic Alzheimer disease; thus, some other
- mechanism for the brain-specific deposition of the amyloid beta protein
- must be sought. Warren et al. (1987) and Murdoch et al. (1988) likewise
- found no duplication of the gene in autopsy-proved cases of Alzheimer
- disease.
-
- ANIMAL MODEL
-
- Selkoe et al. (1987) used a panel of antibodies against amyloid fibrils
- and their constituent vascular amyloid in 5 other species of aged
- mammals, including monkey, orangutan, polar bear, and dog. Antibodies to
- the 28-amino acid peptide recognized the cortical and microvascular
- amyloid of all the aged mammals examined (Selkoe et al., 1987).
-
- Cheng et al. (1988) described the comparative mapping of DNA markers in
- the region of familial Alzheimer disease on human chromosome 21 and
- mouse chromosome 16. The linkage group shared by mouse chromosome 16 and
- human chromosome 21 includes both the Alzheimer amyloid beta precursor
- protein and markers linked to familial Alzheimer disease. The linkage
- group of 6 loci extends from anonymous DNA marker D21S52 to ETS2, and
- spans 39% recombination in man but only 6.4% recombination in the mouse.
- A break in synteny occurs distal to ETS2, and the homolog of human
- marker D21S56 maps to mouse chromosome 17.
-
- To test whether the amyloid beta peptide in Alzheimer disease is
- neurotoxic, LaFerla et al. (1995) introduced a transgene, which included
- 1.8 kb of 5-prime flanking DNA from the mouse neurofilament-light (NF-L)
- gene, into mice to restrict expression of the peptide coding region of
- the APP gene to neuronal cells. In situ hybridization and immunostaining
- with amyloid beta antibodies detected extensive transgene expression and
- peptide in cerebral cortex and hippocampus, and limited expression in
- other areas of the brains of the transgenic mice. (Both the cerebral
- cortex and hippocampus are severely affected in Alzheimer disease.) The
- study showed that expression of amyloid beta is sufficient to induce a
- progressive series of changes within the brains of transgenic mice,
- initiating with neurodegeneration and apoptosis, followed by the
- activation of secondary events such as astrogliosis, and ultimately
- ending with spongiosis. Accompanying the cell death was the appearance
- of clinical features including seizures and premature death, both of
- which have been described in Alzheimer disease.
-
- HISTORY
-
- Bogerts (1993) provided a biographic sketch and photograph of Alois
- Alzheimer (1864-1915). Alzheimer was a neuropathologist, clinical
- psychiatrist, and chairman of psychiatry. He always considered himself a
- psychiatrist. He worked with Nissl in the application of the Nissl
- staining techniques for the study of the cerebral cortex in psychosis.
- Alzheimer discovered the disorder that bears his name in 1906 when he
- reported on 'a strange disease of the cerebral cortex' in a 56-year-old
- with presenile dementia who displayed diffuse cortical atrophy, nerve
- cell loss, plaques, and tangles. He was then working in Munich in the
- department of Kraepelin, who coined the term 'Alzheimer's disease.'
-
- In light of the findings of Tomita et al. (1997) concerning PS2 mutation
- and altered metabolism of APP (summarized in 600759.0001), Hardy (1997)
- reviewed the evidence that AD, or as he put it, the Alzheimer family of
- diseases, has many etiologies but one pathogenesis. Mutations in all
- known pathogenic genes have in common the fact that they alter
- processing of APP, thus lending strong support to the 'amyloid cascade
- hypothesis.' Hardy (1997) commented that 'genetics and molecular biology
- now are revealing credible drug targets' for effective therapy.
-
- O'Brien (1996) reported that the file on the case of Auguste D., who at
- the age of 51 came under the care of Alois Alzheimer, had come to light;
- it had been missing since 1910. Auguste D. came under the care of
- Alzheimer at a Frankfurt hospital in 1901. The eponym 'Alzheimer
- disease' was popularized by Emil Kraepelin, director of the Munich
- psychiatric clinic where Alzheimer moved in 1903. On the basis of the
- record some questions of whether Auguste D. had the disorder now called
- Alzheimer disease were raised; namely, that autopsy findings included
- arteriosclerosis noted in the smaller cerebral blood vessels. O'Brien
- (1996) noted that today this is a criterion for exclusion from a
- diagnosis of AD.
-
- *FIELD* SA
- Ball et al. (1985); Cohen et al. (1988); Cook and Austin (1978); Cook
- et al. (1979); Corder et al. (1993); Goate et al. (1989); Goudsmit
- et al. (1981); Grundke-Iqbal et al. (1979); Heston and Mastri (1977);
- Heston and White (1978); McKhann et al. (1984); Prusiner (1984);
- St. George-Hyslop et al. (1990); St. George-Hyslop et al. (1987);
- Tanzi et al. (1987); Tanzi et al. (1987); Van Broeckhoven et al. (1992);
- van Dujin et al. (1993); Ward et al. (1979); White et al. (1981);
- Wolstenholme and O'Connor (1970)
- *FIELD* RF
- 1. Abraham, C. R.; Selkoe, D. J.; Potter, H.: Immunochemical identification
- of the serine protease inhibitor alpha-1-antichymotrypsin in the brain
- amyloid deposits of Alzheimer's disease. Cell 52: 487-501, 1988.
-
- 2. Ball, M. J.: Features of Creutzfeldt-Jakob disease in brains of
- patients with familial dementia of Alzheimer type. Canad. J. Neurol.
- Sci. 7: 51-57, 1980.
-
- 3. Ball, M. J.; Fisman, M.; Hachinski, V.; Blume, W.; Fox, A.; Kral,
- V. A.; Kirshen, A. J.; Fox, H.; Merskey, H.: A new definition of
- Alzheimer's disease: a hippocampal dementia. Lancet I: 14-16, 1985.
-
- 4. Berr, C.; Borghi, E.; Rethore, M. O.; Lejeune, J.; Alperovitch,
- A.: Absence of familial association between dementia of Alzheimer
- type and Down syndrome. Am. J. Med. Genet. 33: 545-550, 1989.
-
- 5. Birchall, J. D.; Chappell, J. S.: Aluminum, chemical physiology,
- and Alzheimer's disease. Lancet II: 1008-1010, 1988.
-
- 6. Bird, T. D.; Lampe, T. H.; Nemens, E. J.; Miner, G. W.; Sumi, S.
- M.; Schellenberg, G. D.: Familial Alzheimer's disease in American
- descendants of the Volga Germans: probable genetic founder effect. Ann.
- Neurol. 23: 25-31, 1988.
-
- 7. Blanquet, V.; Turleau, C.; Stehelin, D.; Creau-Goldberg, N.; Delabar,
- J. M.; Sinet, P. M.; Davous, P.; de Grouchy, J.: Regional mapping
- of ETS 2 on chromosome 21 in normal Alzheimer disease individuals.
- (Abstract) Cytogenet. Cell Genet. 46: 583, 1987.
-
- 8. Bogerts, B.: Alois Alzheimer. Am. J. Psychiat. 150: 12, 1993.
-
- 9. Breitner, J. C. S.; Folstein, M. F.: Familial nature of Alzheimer's
- disease. (Letter) New Eng. J. Med. 311: 192, 1984.
-
- 10. Breitner, J. C. S.; Silverman, J. M.; Mohs, R. C.; Davis, K. L.
- : Familial aggregation in Alzheimer's disease: comparison of risk
- among relatives of early- and late-onset cases, and among male and
- female relatives in successive generations. Neurology 38: 207-212,
- 1988.
-
- 11. Carrell, R. W.: Alzheimer's disease: enter a protease inhibitor. Nature 331:
- 478-479, 1988.
-
- 12. Cheng, S. V.; Nadeau, J. H.; Tanzi, R. E.; Watkins, P. C.; Jagadesh,
- J.; Taylor, B. A.; Haines, J. L.; Sacchi, N.; Gusella, J. F.: Comparative
- mapping of DNA markers from the familial Alzheimer disease and Down
- syndrome regions of human chromosome 21 to mouse chromosomes 16 and
- 17. Proc. Nat. Acad. Sci. 85: 6032-6036, 1988.
-
- 13. Cohen, M. L.; Golde, T. E.; Usiak, M. F.; Younkin, L. H.; Younkin,
- S. G.: In situ hybridization of nucleus basalis neurons shows increased
- beta-amyloid mRNA in Alzheimer disease. Proc. Nat. Acad. Sci. 85:
- 1227-1231, 1988.
-
- 14. Cook, R. H.; Austin, J. H.: Precautions in familial transmissible
- dementia. Arch. Neurol. 35: 697-698, 1978.
-
- 15. Cook, R. H.; Ward, B. E.; Austin, J. H.: Studies in aging of
- the brain: IV. Familial Alzheimer disease: relation to transmissible
- dementia, aneuploidy, and microtubular defects. Neurology 29: 1402-1412,
- 1979.
-
- 16. Corder, E. H.; Sounder, W. J.; Strittmatter, W. J.; Scheckel,
- D. E.; Gaskell, P. C.; Small, G. W.; Roses, A. D.; Haines, J. L.;
- Periak-Vance, M. A.: Gene dose of apolipoprotein E type 4 allele
- and the risk of Alzheimer's disease in late onset families. Science 261:
- 921-923, 1993.
-
- 17. Corkin, S.; Growdon, J. H.; Rasmussen, S. L.: Parental age as
- a risk factor in Alzheimer's disease. Ann. Neurol. 13: 674-676,
- 1983.
-
- 18. David, F.; Intrator, S.; Foncin, J.-F.; Salmon, D.; Lucotte, G.
- : Absence de liaison etroite entre la maladie d'Alzheimer et la sonde
- polymorphe codant pour la superoxyde dismutase 1. C. R. Acad. Sci. 306
- (ser. III): 1-4, 1988.
-
- 19. Delabar, J. M.; Lamour, Y.; Gegonne, A.; Davous, P.; Roudier,
- M.; Nicole, A.; Ceballos, I.; Amouyel, P.; Stehelin, D.; Sinet, P.
- M.: Rearrangement of chromosome 21 in Alzheimer's disease. Ann.
- Genet. 29: 226-228, 1986.
-
- 20. Edwards, J. H.: Exclusion mapping. J. Med. Genet. 24: 539-543,
- 1987.
-
- 21. Eikelenboom, P.; Goetz, J.; Pronk, J. C.; Hauptmann, G.: Complement
- C4 phenotypes in dementia of the Alzheimer type. Hum. Hered. 38:
- 48-51, 1988.
-
- 22. Farrer, L. A.; Myers, R. H.; Connor, L.; Cupples, L. A.; Growdon,
- J. H.: Segregation analysis reveals evidence of a major gene for
- Alzheimer disease. Am. J. Hum. Genet. 48: 1026-1033, 1991.
-
- 23. Farrer, L. A.; Myers, R. H.; Cupples, L. A.; St. George-Hyslop,
- P. H.; Bird, T. D.; Rossor, M. N.; Mullan, M. J.; Polinsky, R.; Nee,
- L.; Heston, L.; Van Broeckhoven, C.; Martin, J.-J.; Crapper-McLachlan,
- D.; Growdon, J. H.: Transmission and age-at-onset patterns in familial
- Alzheimer's disease: evidence for heterogeneity. Neurology 40: 395-403,
- 1990.
-
- 24. Fitch, N.; Becker, R.; Heller, A.: The inheritance of Alzheimer's
- disease: a new interpretation. Ann. Neurol. 23: 14-19, 1988.
-
- 25. Gajdusek, D. C.: On the uniform source of amyloid in plaques,
- tangles and vascular deposits. Neurobiol. Aging 7: 453-454, 1986.
-
- 26. Glenner, G. G.; Wong, C. W.: Alzheimer's disease: initial report
- of the purification and characterization of a novel cerebrovascular
- amyloid protein. Biochem. Biophys. Res. Commun. 120: 885-890, 1984.
-
- 27. Goate, A.; Chartier-Harlin, M.-C.; Mullan, M.; Brown, J.; Crawford,
- F.; Fidani, L.; Giuffra, L.; Haynes, A.; Irving, N.; James, L.; Mant,
- R.; Newton, P.; Rooke, K.; Roques, P.; Talbot, C.; Pericak-Vance,
- M.; Roses, A.; Williamson, R.; Rossor, M.; Owen, M.; Hardy, J.: Segregation
- of a missense mutation in the amyloid precursor protein gene with
- familial Alzheimer's disease. Nature 349: 704-706, 1991.
-
- 28. Goate, A.; Haynes, A.; Mullan, M.; Owen, M.; James, L.; Farrall,
- M.; Rossor, M.; Williamson, R.; Hardy, J.: Alzheimer's disease locus
- maps close to D21S16 on the long arm of chromosome 21. (Abstract) Cytogenet.
- Cell Genet. 51: 1006, 1989.
-
- 29. Goate, A. M.; Haynes, A. R.; Owen, M. J.; Farrall, M.; James,
- L. A.; Lai, L. Y. C.; Mullan, M. J.; Roques, P.; Rossor, M. N.; Williamson,
- R.; Hardy, J. A.: Predisposing locus for Alzheimer's disease on chromosome
- 21. Lancet I: 352-355, 1989.
-
- 30. Goldgaber, D.; Lerman, M. I.; McBride, O. W.; Saffiotti, U.; Gajdusek,
- D. C.: Characterization and chromosomal localization of a cDNA encoding
- brain amyloid of Alzheimer's disease. Science 235: 877-880, 1987.
-
- 31. Goudsmit, J.; White, B. J.; Weitkamp, L. R.; Keats, B. J. B.;
- Morrow, C. H.; Gajdusek, D. C.: Familial Alzheimer's disease in two
- kindreds of the same geographic and ethnic origin: a clinical and
- genetic study. J. Neurol. Sci. 49: 79-89, 1981.
-
- 32. Grundke-Iqbal, I.; Johnson, A. B.; Wisniewski, H. M.; Terry, R.
- D.; Iqbal, K.: Evidence that Alzheimer neurofibrillary tangles originate
- from neurotubules. Lancet I: 578-581, 1979.
-
- 33. Haines, J.; St. George-Hyslop, P.; Tanzi, R.; Polinsky, R.; Nee,
- L.; Watkins, P.; Myers, R.; Feldman, R.; Pollen, D.; Drachman, D.;
- Growdon, J.; Bruni, A.; Foncin, J.-F.; Salmon, D.; Frommelt, P.; Amaducci,
- L.; Sorbi, S.; Piacentini, S.; Stewart, G.; Hobbs, W.; Gusella, J.;
- Conneally, M.: Linkage of familial Alzheimer disease to markers on
- chromosome 21. (Abstract) Cytogenet. Cell Genet. 46: 625, 1987.
-
- 34. Haines, J. L.: The genetics of Alzheimer disease--a teasing problem. Am.
- J. Hum. Genet. 48: 1021-1025, 1991.
-
- 35. Hardy, J.: The Alzheimer family of diseases: many etiologies,
- one pathogenesis? Proc. Nat. Acad. Sci. 94: 2095-2097, 1997.
-
- 36. Harper, C. G.; Buck, D.; Gonatas, N. K.; Guilbert, B.; Avrameas,
- S.: Skin fibroblast microtubular network in Alzheimer disease. Ann.
- Neurol. 6: 548-552, 1979.
-
- 37. Heston, L. L.: Alzheimer's disease, trisomy 21, and myeloproliferative
- disorders: associations suggesting a genetic diathesis. Science 196:
- 322-323, 1977.
-
- 38. Heston, L. L.; Lowther, D. L. W.; Leventhal, C. M.: Alzheimer's
- disease: a family study. Arch. Neurol. 15: 225-233, 1966.
-
- 39. Heston, L. L.; Mastri, A. R.: The genetics of Alzheimer's disease:
- associations with hematologic malignancy and Down's syndrome. Arch.
- Gen. Psychiat. 34: 976-981, 1977.
-
- 40. Heston, L. L.; White, J.: Pedigrees of 30 families with Alzheimer
- disease: associations with defective organization of microfilaments
- and microtubules. Behav. Genet. 8: 315-331, 1978.
-
- 41. Heyman, A.; Wilkinson, W. E.; Hurwitz, B. J.; Schmechel, D.; Sigmon,
- A. H.; Weinberg, T.; Helms, M. J.; Swift, M.: Alzheimer's disease:
- genetic aspects and associated clinical disorders. Ann. Neurol. 14:
- 507-515, 1983.
-
- 42. Joachim, C. L.; Mori, H.; Selkoe, D. J.: Amyloid beta-protein
- deposition in tissues other than brain in Alzheimer's disease. Nature 341:
- 226-230, 1989.
-
- 43. Kang, J.; Lemaire, H.-G.; Unterbeck, A.; Salbaum, J. M.; Masters,
- C. L.; Grzeschik, K.-H.; Multhaup, G.; Beyreuther, K.; Muller-Hill,
- B.: The precursor of Alzheimer's disease amyloid A4 protein resembles
- a cell-surface receptor. Nature 325: 733-736, 1987.
-
- 44. Kitaguchi, N.; Takahashi, Y.; Tokushima, Y.; Shiojiri, S.; Ito,
- H.: Novel precursor of Alzheimer's disease amyloid protein shows
- protease inhibitory activity. Nature 331: 530-532, 1988.
-
- 45. LaFerla, F. M.; Tinkle, B. T.; Bieberich, C. J.; Haudenschild,
- C. C.; Jay, G.: The Alzheimer's A-beta peptide induces neurodegeneration
- and apoptotic cell death in transgenic mice. Nature Genet. 9: 21-30,
- 1995.
-
- 46. Lawrence, S.; Keats, B. J.; Morton, N. E.: The AD1 locus in familial
- Alzheimer disease. Ann. Hum. Genet. 56: 295-301, 1992.
-
- 47. Lowenberg, K.; Waggoner, R. W.: Familial organic psychosis (Alzheimer's
- type). Arch. Neurol. Psychiat. 31: 737-754, 1934.
-
- 48. Masters, C. L.; Gajdusek, D. C.; Gibbs, C. J., Jr.: The familial
- occurrence of Creutzfeldt-Jakob disease and Alzheimer's disease. Brain 104:
- 535-558, 1981.
-
- 49. McKhann, G.; Drachman, D.; Folstein, M.; Katzman, R.; Price, D.;
- Stadlan, E. M.: Clinical diagnosis of Alzheimer's disease. Neurology 34:
- 939-944, 1984.
-
- 50. McMenemey, W. H.; Worster-Drought, C.; Flind, J.; Williams, H.
- G.: Familial presenile dementia: report of a case with clinical and
- pathological features of Alzheimer's disease. J. Neurol. Psychopath. 2:
- 293-302, 1939.
-
- 51. Murdoch, G. H.; Manuelidis, L.; Kim, J. H.; Manuelidis, E. E.
- : Beta-amyloid gene dosage in Alzheimer's disease. Nucleic Acids
- Res. 16: 357, 1988.
-
- 52. Nee, L. E.; Polinsky, R. J.; Eldridge, R.; Weingartner, H.; Smallberg,
- S.; Ebert, M.: A family with histologically confirmed Alzheimer's
- disease. Arch. Neurol. 40: 203-208, 1983.
-
- 53. Nerl, C.; Mayeux, R.; O'Neill, G. J.: HLA-linked complement markers
- in Alzheimer's and Parkinson's disease C4 variant (C4B2)--a possible
- marker for senile dementia of the Alzheimer type. Neurology 34:
- 310-314, 1984.
-
- 54. Nordenson, I.; Adolfsson, R.; Beckman, G.; Bucht, G.; Winblad,
- B.: Chromosomal abnormality in dementia of Alzheimer type. Lancet I:
- 481-482, 1980.
-
- 55. O'Brien, C.: Auguste D. and Alzheimer's disease. Science 273:
- 28 only, 1996.
-
- 56. Oesch, B.; Westaway, D.; Walchli, M.; McKinley, M. P.; Kent, S.
- B. H.; Aebersold, R.; Barry, R. A.; Tempst, P.; Teplow, D. B.; Hood,
- L. E.; Prusiner, S. B.; Weissmann, C.: A cellular gene encodes scrapie
- PrP 27-30 protein. Cell 40: 735-746, 1985.
-
- 57. Owen, M.; Hardy, J.; Williamson, R.; Goate, A.: Physical mapping
- of the long arm of chromosome 21. (Abstract) Cytogenet. Cell Genet. 51:
- 1056, 1989.
-
- 58. Percy, M. E.; Markovic, V. D.; Crapper McLachlan, D. R.; Berg,
- J. M.; Hummel, J. T.; Laing, M. E.; Dearie, T. G.; Andrews, D. F.
- : Family with 22-derived marker chromosome and late-onset dementia
- of the Alzheimer type. I. Application of a new model for estimation
- of the risk of disease associated with the marker. Am. J. Med. Genet. 39:
- 307-313, 1991.
-
- 59. Pericak-Vance, M. A.; Bebout, J. L.; Gaskell, P. C., Jr.; Yamaoka,
- L. H.; Hung, W.-Y.; Alberts, M. J.; Walker, A. P.; Bartlett, R. J.;
- Haynes, C. A.; Welsh, K. A.; Earl, N. L.; Heyman, A.; Clark, C. M.;
- Roses, A. D.: Linkage studies in familial Alzheimer disease: evidence
- for chromosome 19 linkage. Am. J. Hum. Genet. 48: 1034-1050, 1991.
-
- 60. Pericak-Vance, M. A.; Bebout, J. L.; Haynes, C. A.; Gaskell, P.
- C., Jr.; Yamaoka, L. A.; Hung, W.-Y.; Alberts, M. J.; Walker, A. P.;
- Bartlett, R. J.; Welsh, K. A.; Earl, N. L.; Heyman, A.; Clark, C.
- M.; Roses, A. D.: Linkage studies in familial Alzheimer's disease:
- evidence for chromosome 19 linkage. (Abstract) Am. J. Hum. Genet. 47
- (suppl.): A194, 1990.
-
- 61. Pericak-Vance, M. A.; Yamaoka, L. H.; Bebout, J.; Gaskell, P.
- C.; Clark, C.; Haynes, C. S.; Earl, N.; Welch, K.; Hung, W.-Y.; Alberts,
- M. J.; Heyman, A.; Roses, A. D.: Linkage studies in familial Alzheimer's
- disease. (Abstract) Cytogenet. Cell Genet. 51: 1058-1059, 1989.
-
- 62. Pericak-Vance, M. A.; Yamaoka, L. H.; Haynes, C. S.; Speer, M.
- C.; Haines, J. L.; Gaskell, P. C.; Hung, W.-Y.; Clark, C. M.; Heyman,
- A. L.; Trofatter, J. A.; Eisenmenger, J. P.; Gilbert, J. R.; Lee,
- J. E.; Alberts, M. J.; Dawson, D. V.; Bartlett, R. J.; Earl, N. L.;
- Siddique, T.; Vance, J. M.; Conneally, P. M.; Roses, A. D.: Genetic
- linkage studies in Alzheimer's disease families. Exp. Neurol. 102:
- 271-279, 1988.
-
- 63. Podlisny, M. B.; Lee, G.; Selkoe, D. J.: Gene dosage of the amyloid
- beta precursor protein in Alzheimer's disease. Science 238: 669-671,
- 1987.
-
- 64. Ponte, P.; Gonzalez-DeWhitt, P.; Schilling, J.; Miller, J.; Hsu,
- D.; Greenburg, B.; Davis, K.; Wallace, W.; Lieberburg, I.; Fuller,
- F.; Cordell, B.: A new A4 amyloid mRNA contains a domain homologous
- to serine proteinase inhibitors. Nature 331: 525-527, 1988.
-
- 65. Powell, D.; Folstein, M. F.: Pedigree study of familial Alzheimer
- disease. J. Neurogenet. 1: 189-197, 1984.
-
- 66. Prusiner, S. B.: Some speculatio