Entry - #301040 - ALPHA-THALASSEMIA/IMPAIRED INTELLECTUAL DEVELOPMENT SYNDROME, X-LINKED; ATRX - OMIM
# 301040

ALPHA-THALASSEMIA/IMPAIRED INTELLECTUAL DEVELOPMENT SYNDROME, X-LINKED; ATRX


Alternative titles; symbols

ALPHA-THALASSEMIA/MENTAL RETARDATION SYNDROME, X-LINKED
ALPHA-THALASSEMIA/MENTAL RETARDATION SYNDROME, NONDELETION TYPE
ATR-X SYNDROME
ATR, NONDELETION TYPE


Phenotype-Gene Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Gene/Locus Gene/Locus
MIM number
Xq21.1 Alpha-thalassemia/impaired intellectual development syndrome 301040 XLD 3 ATRX 300032
Clinical Synopsis
 

INHERITANCE
- X-linked dominant
GROWTH
Other
- Postnatal growth deficiency
HEAD & NECK
Head
- Microcephaly
- Absent frontal sinuses
Face
- Mid-face hypoplasia
Ears
- Small ears
- Low-set ears
- Posteriorly rotated ears
- Sensorineural hearing loss (less common)
Eyes
- Hypertelorism
- Epicanthal folds
Nose
- Low nasal bridge
- Small triangular nose
- Anteverted nares
Mouth
- 'Carp-like' mouth
- Full lips
- Protruding tongue
Teeth
- Widely-spaced upper incisors
CARDIOVASCULAR
Heart
- Perimembranous ventricular septal defect
ABDOMEN
External Features
- Umbilical hernia
Gastrointestinal
- Constipation
- Gastroesophageal reflux
GENITOURINARY
External Genitalia (Male)
- Shawl scrotum
- Small penis
- Hypospadias
Internal Genitalia (Male)
- Cryptorchidism
Kidneys
- Renal agenesis
- Hydronephrosis
SKELETAL
Spine
- Kyphoscoliosis
- Hemivertebra
Pelvis
- Coxa valga
Hands
- Tapering fingers
- Clinodactyly
Feet
- Talipes equinovarus
NEUROLOGIC
Central Nervous System
- Mental retardation
- Delayed developmental milestones
- Expressive speech absent
- Hypotonia early
- Spasticity later
- Seizures (in 35%)
- Cerebral atrophy
HEMATOLOGY
- Mild hypochromic microcytic anemia
- Mild form of hemoglobin H (Hb H) disease
LABORATORY ABNORMALITIES
- Hb H erythrocyte inclusions
MISCELLANEOUS
- Variable phenotype
- X-linked mental retardation-hypotonic facies syndrome (309580) is an allelic disorder without alpha-thalassemia
MOLECULAR BASIS
- Caused by mutation in the ATRX chromatin remodeler gene (ATRX, 300032.0001)

TEXT

A number sign (#) is used with this entry because X-linked alpha-thalassemia/impaired intellectual development syndrome is caused by mutation in the ATRX gene (300032) on Xq21.

The 'deletion' type of alpha-thalassemia/impaired intellectual development syndrome (141750) is a contiguous gene syndrome due to a deletion in chromosome 16p that involves the hemoglobin alpha-1 (HBA1; 141800) and alpha-2 (HBA2; 141850) genes.

The X-linked mental retardation-hypotonic facies syndrome (309580) is also caused by mutation in the ATRX gene.


Clinical Features

Weatherall et al. (1981) reported the association of hemoglobin H disease (Hb H; see alpha-thalassemias, 141800) and mental retardation in 3 unrelated patients of northern European descent.

Wilkie et al. (1990) reported 5 unrelated patients, 2 of whom were reported by Weatherall et al. (1981), with mental retardation and alpha-thalassemia without molecular abnormalities of the alpha-globin gene complex on chromosome 16p. The patients showed a strikingly uniform phenotype comprising severe mental handicap, characteristic dysmorphic facies, genital abnormalities, and an unusual, mild form of hemoglobin H disease. Facial features included microcephaly, hypertelorism, epicanthus, a small triangular upturned nose, and flat face. The degree of red blood cell hypochromia and Hb H levels, which varied from 0.7 to 6.7%, were milder than usually found in alpha-thalassemia. Although several approaches failed to find a defect in the alpha-globin genes, 3 patients tested had markedly reduced total mRNA levels of both HBA1 and HBA2. The authors suggested that the responsible locus encoded a trans-acting factor involved in the normal regulation of alpha-globin expression.

Harvey et al. (1990) described the syndrome in a 21-year-old male and his brother who had died earlier, suggesting X-linked inheritance. DNA analysis showed no deletions within the alpha-globin gene cluster. Hb H bodies were present at a low level (1.6%).

Porteous and Burn (1990) described a 6-year-old boy who had a maternal uncle with an X-linked mental retardation syndrome, and suggested that their case resembled 2 brothers previously thought to have an atypical form of the Coffin-Lowry syndrome (303600) (illustrated in Smith's Recognizable Patterns of Human Malformation, Jones, 1988). However, Wilkie et al. (1991) found that there were hematologic signs of the nondeletion ATR syndrome in the patient reported by Porteous and Burn (1990). In addition, Wilkie et al. (1991) reported that hematologic evaluation of 1 of the brothers reported in Smith's book showed that he had nondeletion ATR and that a male first-cousin through the maternal line had the same condition. Wilkie et al. (1991) suggested that this condition be called 'X-linked alpha-thalassemia/mental retardation' (ATR-X) to distinguish it from the deletion form.

In a review, Gibbons et al. (1991) noted that some patients with ATR-X syndrome have normal or only mildly abnormal hematologic indices; thus normal hemoglobin levels and red cell indices do not necessarily exclude the condition.

Cole et al. (1991) described an affected boy whose maternal uncle was also affected. The boy had right-sided renal agenesis with left-sided hydronephrosis and hydroureter. He had recurrent hypochromic, microcytic anemia. His otherwise unaffected sister had had recurrent urinary tract infections and persistent renal impairment in the absence of any identifiable renal tract anomaly. Kurosawa et al. (1996) described a boy with self-induced vomiting followed by rumination and noted that Cole et al. (1991) made the same observation in a man and his nephew.

Donnai et al. (1991) described 4 brothers with this syndrome in whom the diagnosis was first suspected because of their characteristic clinical features and was confirmed in each case by the demonstration of Hb H inclusions in a proportion of their red blood cells. Very rare Hb H inclusions were found in the red blood cells of the mother and one sister who both shared some facial features with the affected boys; they were presumed to be carriers of the disorder.

Gorlin (1993) examined patients with typical features of the ATR-X syndrome, but without hemoglobin H. The facies were identical and mapping studies in several families suggested location of the mutation in the site on the X chromosome involved in ATR-X. The facies of this syndrome, which is often confused with that of Coffin-Lowry syndrome, were marked by telecanthus, epicanthic folds, flat nasal bridge, midface hypoplasia, a carp-shaped mouth with full lips, and small triangular nose with anteverted nostrils. Gorlin (1993) noted that the alae of the nose extended lower than the columella and septum. All developmental milestones, especially walking, were delayed and speech was almost absent. On further investigation, Gibbons (1994) found that the patients of Gorlin (1993) did have alpha-thalassemia, as indicated by the presence of hemoglobin H inclusions after use of 1% brilliant cresyl blue staining overnight in buffered solution at room temperature. With the staining, the Hb H inclusions give the erythrocytes the appearance of golf balls.

Logie et al. (1994) reported a pedigree with 6 affected males in 4 sibships spanning 2 generations. Two affected cousins were described in detail, one of whom had an unusually mild hematologic phenotype. Hb H inclusions, the hallmark of the disorder, were detected in the peripheral red blood cells only after repeated observations. The cousins had strikingly similar facies with telecanthus, anteverted nares, carp-shaped mouth, and large tongue. Gibbons et al. (1995) showed that the hematologic findings in ATR-X may vary widely; indeed, in some cases, the manifestation of alpha-thalassemia may be subtle and missed without repeated examinations. McPherson et al. (1995) described a kindred with 4 affected members. The hematologic abnormality was not detected on routine hematologic studies, including hemoglobin electrophoresis, but the patients were found to have hemoglobin H inclusions on brilliant cresyl blue staining of peripheral smears.

Reardon et al. (1995) reported 2 phenotypic females with a 46,XY karyotype who had abnormalities of the external genitalia resulting in male pseudohermaphroditism. They pointed out that 1 of the 5 original patients described in defining the ATR-X syndrome was a phenotypic female with a 46,XY karyotype (Wilkie et al., 1990). McPherson et al. (1995) described genital anomalies that led to a female sex of rearing in 3 of 4 affected members of a family. Gibbons et al. (1995) emphasized the progressive coarsening of the facial appearance. Kuno et al. (1997) described a 5-year-old Japanese boy with this condition. He had an abnormal hemoglobin which was found to consist exclusively of a beta subunit. Severe mental retardation and hypoplastic penis and testes were present. Anemia was only mild (hematocrit 35.8%). The family history was unremarkable.

Martinez et al. (1998) reported 2 brothers and 1 maternal cousin with severe mental retardation, microcephaly, short stature, cryptorchidism, and spastic diplegia. Some facial dysmorphic features were present. Martinez et al. (1998) pointed out the similarity in phenotype between their family and that described by Sutherland et al. (1988) (see 309500). They suggested that the greater phenotypic severity in their family was due to allelic heterogeneity. X-inactivation analysis of 1 potential and 3 obligate carriers showed nonrandom inactivation of the disease-linked variant. On further analysis of this family, Lossi et al. (1999) found that 3% of the patients' erythrocytes showed Hb H inclusions, consistent with ATR-X. Lossi et al. (1999) also reported dysmorphic facial features, including 'carp-like' triangular mouth, hypertelorism, small triangular nose, and broad nasal root. The hypertonia and spasticity were unusual findings in this family. A mutation was found in the ATRX gene in affected individuals (300032.0016).

Gibbons and Higgs (2000) provided a review of the clinical spectrum of syndromes caused by mutation in the XH2 gene.

Martucciello et al. (2006) described male 3-year-old dizygotic twins with ATRX who exhibited gastrointestinal problems including severe regurgitation of food, vomiting, dysphagia, irritability, respiratory disorders, meteorism, and chronic constipation. Barium studies in both twins showed gastric pseudovolvulus, and 24-hour pH monitoring showed severe gastroesophageal reflux. Enzymo-histochemical studies of full-thickness colonic biopsies revealed a complex dysganglionosis: ultrashort Hirschsprung disease (see 142623) associated with hypoganglionosis. Martucciello et al. (2006) reviewed the gastrointestinal phenotype of 128 confirmed cases of ATRX and found that drooling was reported in 36% of cases, gastroesophageal reflux was present in 72%, and constipation in 30%. Fundoplication was performed in 10% of cases, and 9% were fed by gastrostomy. Upper GI bleeding was reported in 10% of cases. Fatal aspiration of vomitus occurred in 3 patients; volvulus was seen in 4 patients, 2 of whom died after intestinal infarction; and 4 patients had recurrent hospitalizations for ileus or pseudoobstruction. Martucciello et al. (2006) also noted that there were numerous anecdotal reports from parents describing prolonged episodes of patient distress with refusal to eat or drink.

Jezela-Stanek et al. (2009) reported a patient with ATRX confirmed by genetic analysis. He had hypertelorism, epicanthal folds, strabismus, short nose with flat bridge and triangular upturned tip, and tented upper lip with everted lower lip. Other features included hypotonia, psychomotor retardation, and hemoglobin H inclusions. The patient also had undescended testes and ambiguous genitalia, which the authors referred to as male pseudohermaphroditism. Laboratory studies showed increased FSH and decreased testosterone. A deceased sib was believed to have been affected and reportedly had ambiguous external genitalia. Jezela-Stanek et al. (2009) postulated that the distinctive facial features in ATRX result from facial hypotonia and can be confused with Coffin-Lowry syndrome (CLS; 303600) or SLO syndromes (SLOS; 270400).

Based on a literature review, Leon and Harley (2021) reported the frequency of clinical features in individuals with ATR-X syndrome. The clinical features were highly variable, even within the same family. The most common feature was intellectual disability, identified in 100% of patients, followed by hematologic abnormalities in 70%, genital abnormalities in 62%, characteristic facies in 55%, skeletal abnormalities in 43%, and microcephaly in 42%. Less frequent features included hypotonia (34%), gastrointestinal problems (30%), seizures (17%), abnormal behavior (10%), heart defects (8%), and osteosarcoma (3%).

Carrier Females

Studying 7 pedigrees that included individuals with the ATR-X syndrome, Gibbons et al. (1992) concluded that intellectually normal female carriers could be identified by the presence of rare cells containing Hb H inclusions in their peripheral blood and by an extremely skewed pattern of X inactivation in cells from a variety of tissues. McPherson et al. (1995) used a combination of skewed X inactivation and haplotype analysis at Xq12-q21.3 to establish carrier status.

Wada et al. (2005) found skewed X-inactivation patterns (greater than 90:10) in 6 of 7 unaffected Japanese female ATR-X carriers; the 1 carrier with nonskewed X inactivation (72:28) demonstrated moderate mental retardation. The woman did not have dysmorphic features or hemoglobin inclusions. Wada et al. (2005) concluded that mutations in the ATRX gene may cause mental retardation in females if the chromosome carrying the mutation is not properly inactivated.

Badens et al. (2006) reported a 4-year-old girl with typical features of the ATR-X syndrome. Molecular studies showed a totally skewed X-inactivation pattern, with the active chromosome carrying a heterozygous mutation in the ATRX gene (300032.0018). Neither parent had the mutation in peripheral blood leukocytes, but SNP analysis indicated that the mutation occurred on the maternal chromosome. The child was conceived with assisted reproduction technologies (ART) due to micropolycystic ovaries and endometriosis in the mother. Badens et al. (2006) suggested that some aspect of ART may have disturbed imprinting in this patient.


Mapping

By linkage analysis of 7 affected pedigrees, Gibbons et al. (1992) mapped the ATR-X locus to an 11-cM interval on chromosome Xq12-q21.31 between markers DXS106 and DXYS1X (peak lod score of 5.4 at theta = 0 at DXS72).

In a 3-generation ATR-X family with 3 affected males, Houdayer et al. (1993) demonstrated a maximum lod score of 2.09 at a recombination fraction of zero for linkage with DXS453 located at the boundary Xq12-q13.1. The nearest flanking loci demonstrating recombination with the disease locus were the androgen receptor (AR; 313700) at Xq11.2-q12 on the centromeric side and DXS72 at Xq21.1 on the telomeric side. Houdayer et al. (1993) interpreted their results as compatible with a distal boundary at Xq21.1 instead of Xq21.31 as previously held.

Gibbons et al. (1995) performed linkage analysis in 9 families with ATR-X syndrome and identified key recombinants that reduced the area of interest to 1.4 cM (estimated to be 15 Mb) between DXS454 and DXS72 within Xq13.1-q21.1 (Wang et al., 1994).


Diagnosis

Leon and Harley (2021) recommended that targeted sequencing of the ATRX gene should be the first diagnostic test in patients with more than one hallmark clinical feature of ATR-X syndrome. If a mutation is not identified by this method, other techniques to identify deletions or duplications in the ATRX gene, such as chromosome microarray or MLPA, should be considered.


Molecular Genetics

In patients with the ATR-X syndrome, Gibbons et al. (1995) identified mutations in the ATRX gene (300032.0001-300032.0009).

In affected members of a family with ATR-X syndrome, Villard et al. (1996) identified a splice site mutation in the ATRX gene (300032.0010). In 2 first cousins presenting the classic ATR-X phenotype with alpha-thalassemia and Hb H inclusions, only the abnormal transcript was expressed. In a distant cousin presenting with a similar dysmorphic mental retardation phenotype, but without thalassemia, they found that approximately 30% of the ATRX transcripts were normal. These data suggested that the mode of action of the ATRX gene product on globin expression is distinct from its mode of action in brain development and facial morphogenesis, and that the mutated splice site could be used with varying efficiency in different individuals.

Hendrich and Bickmore (2001) reviewed human disorders that share in common defects of chromatin structure or modification, including the ATR-X spectrum of disorders, ICF syndrome (242860), Rett syndrome (312750), Rubinstein-Taybi syndrome (180849), and Coffin-Lowry syndrome.

Partial Duplication of the ATRX Gene

Thienpont et al. (2007) reported 3 patients, including 2 sibs, with the ATRX syndrome due to partial duplications of the ATRX gene. In 1 family, the duplication included exons 2 to 35; in the other family, exons 2 to 29. Further analysis showed that both mothers carried the duplication and both had skewed X inactivation. In 1 patient, ATRX mRNA levels were about 3% of normal values. Thienpont et al. (2007) noted that the duplications were not identified by sequence analysis and suggested that quantitative analysis to detect copy numbers of the ATRX gene may be required in some cases.

Cohn et al. (2009) reported a family in which 3 males had ATRX syndrome due to a partial intragenic duplication of the ATRX gene that spanned exons 2 to 31. Northern blot analysis failed to identify a full-length transcript, but cDNA sequencing was consistent with some level of expression. The authors noted that complete loss of ATRX is most likely lethal, suggesting that the mutation was likely hypomorphic and associated with some residual protein function. Unaffected obligate carrier females in the family had highly skewed X inactivation. The phenotype was typical for the disorder, although the facial features were not as readily apparent in the 2 older affected individuals. The proband was identified from 2 larger cohorts comprising 300 males with mental retardation. Cohn et al. (2009) did not find ATRX duplications in 29 additional males with ATRX syndrome who were negative on sequence analysis, suggesting that duplications are a rare cause of the disorder.


Genotype/Phenotype Correlations

In a review article, Gibbons and Higgs (2000) noted that mutations in the ATRX gene resulting in the loss of the C terminal domain are associated with the most severe urogenital abnormalities. However, at other sites, there is no obvious link between genotype and phenotype, and there is considerable variation in the degree of abnormalities seen in individuals with the same mutation.

Among 22 ATRX patients from 16 families, Badens et al. (2006) found that those with mutations in the PHD-like domain of the ATRX protein had significantly more severe and permanent psychomotor retardation and significantly more severe urogenital anomalies compared to those with mutations in the helicase domain.

Based on a review of the literature, Leon and Harley (2021) noted that the ATRX mutations in individuals with ATR-X syndrome who developed osteosarcoma were located in the C-terminal region of the gene.


Animal Model

Medina et al. (2009) surveyed ATR-X syndrome clinical findings and noted that ocular defects were present in 47 (23%) of 202 patients. They showed that Atrx was expressed in the neuroprogenitor pool in embryonic mouse retina and in all cell types of adult mouse retina except rod photoreceptors. Conditional inactivation of Atrx in mouse retina during embryogenesis resulted in loss of only 2 types of neurons, amacrine and horizontal cells. This defect did not arise from a failure to specify these cells, but rather a defect in interneuron differentiation and survival postnatally. The timing of cell loss was concomitant with light-dependent changes in synaptic organization in mouse retina and with a change in Atrx subnuclear localization within these interneurons. The interneuron defects were associated with functional deficits as demonstrated by reduced b-wave amplitudes upon electroretinogram analysis. Medina et al. (2009) proposed a role for Atrx in interneuron survival and differentiation.


REFERENCES

  1. Badens, C., Lacoste, C., Philip, N., Martini, N., Courrier, S., Giuliano, F., Verloes, A., Munnich, A., Leheup, B., Burglen, L., Odent, S., Van Esch, H., Levy, N. Mutations in PHD-like domain of the ATRX gene correlate with severe psychomotor impairment and severe urogenital abnormalities in patients with ATRX syndrome. Clin. Genet. 70: 57-62, 2006. [PubMed: 16813605, related citations] [Full Text]

  2. Badens, C., Martini, N., Courrier, S., DesPortes, V., Touraine, R., Levy, N., Edery, P. ATRX syndrome in a girl with a heterozygous mutation in the ATRX Zn finger domain and a totally skewed X-inactivation pattern. Am. J. Med. Genet. 140A: 2212-2215, 2006. [PubMed: 16955409, related citations] [Full Text]

  3. Cohn, D. M., Pagon, R. A., Hudgins, L., Schwartz, C. E., Stevenson, R. E., Friez, M. J. Partial ATRX gene duplication causes ATR-X syndrome. (Letter) Am. J. Med. Genet. 149A: 2317-2320, 2009. [PubMed: 19764021, related citations] [Full Text]

  4. Cole, T. R. P., May, A., Hughes, H. E. Alpha-thalassaemia/mental retardation syndrome (non-deletional type): report of a family supporting X linked inheritance. J. Med. Genet. 28: 734-737, 1991. [PubMed: 1770528, related citations] [Full Text]

  5. Donnai, D., Clayton-Smith, J., Gibbons, R. J., Higgs, D. R. The non-deletion alpha-thalassaemia/mental retardation syndrome: further support for X linkage. J. Med. Genet. 28: 742-745, 1991. [PubMed: 1770530, related citations] [Full Text]

  6. Gibbons, R. J., Brueton, L., Buckle, V. J., Burn, J., Clayton-Smith, J., Davison, B. C. C., Gardner, R. J. M., Homfray, T., Kearney, L., Kingston, H. M., Newbury-Ecob, R., Porteous, M. E. P., Wilkie, A. O. M., Higgs, D. R. Clinical and hematologic aspects of the X-linked alpha-thalassemia/mental retardation syndrome (ATR-X). Am. J. Med. Genet. 55: 288-299, 1995. [PubMed: 7726225, related citations] [Full Text]

  7. Gibbons, R. J., Higgs, D. R. Molecular-clinical spectrum of the ATR-X syndrome. Am. J. Med. Genet. 97: 204-212, 2000. [PubMed: 11449489, related citations] [Full Text]

  8. Gibbons, R. J., Picketts, D. J., Villard, L., Higgs, D. R. Mutations in a putative global transcriptional regulator cause X-linked mental retardation with alpha-thalassemia (ATR-X syndrome). Cell 80: 837-845, 1995. [PubMed: 7697714, related citations] [Full Text]

  9. Gibbons, R. J., Suthers, G. K., Wilkie, A. O. M., Buckle, V. J., Higgs, D. R. X-linked alpha-thalassemia/mental retardation (ATR-X) syndrome: localization to Xq12-q21.31 by X inactivation and linkage analysis. Am. J. Hum. Genet. 51: 1136-1149, 1992. [PubMed: 1415255, related citations]

  10. Gibbons, R. J., Wilkie, A. O. M., Weatherall, D. J., Higgs, D. R. A newly defined X linked mental retardation syndrome associated with alpha-thalassaemia. J. Med. Genet. 28: 729-733, 1991. [PubMed: 1770527, related citations] [Full Text]

  11. Gibbons, R. Personal Communication. Oxford, England 11/7/1994.

  12. Gorlin, R. J. Personal Communication. Minneapolis, Minn. 5/29/1993.

  13. Harvey, M. P., Kearney, A., Smith, A., Trent, R. J. Occurrence of the alpha-thalassaemia-mental retardation syndrome (non-deletional type) in an Australian male. J. Med. Genet. 27: 577-581, 1990. [PubMed: 2231651, related citations] [Full Text]

  14. Hendrich, B., Bickmore, W. Human diseases with underlying defects in chromatin structure and modification. Hum. Molec. Genet. 10: 2233-2242, 2001. [PubMed: 11673406, related citations] [Full Text]

  15. Houdayer, C., Toutain, A., Ronce, N., Lefort, G., Sarda, P., Taib, J., Briault, S., Lambert, J. C., Moraine, C. X-linked alpha-thalassemia/mental retardation syndrome: linkage analysis in a new family further supports localization in proximal Xq. Ann. Genet. 36: 194-199, 1993. [PubMed: 8166423, related citations]

  16. Jezela-Stanek, A., Fisher, C., Szarras-Czapnik, M., Olczak-Kowalczyk, D., Gibbons, R. J., Slowikowska-Hilczer, J., Krajewska-Walasek, M. X-linked alpha thalassaemia/mental retardation syndrome: a case with gonadal dysgenesis, caused by a novel mutation in ATRX gene. Clin. Dysmorph. 18: 168-171, 2009. [PubMed: 19444090, related citations] [Full Text]

  17. Jones, K. L. Smith's Recognizable Patterns of Human Malformation. (4th ed.) Philadelphia: W. B. Saunders (pub.) 1988. P. 237.

  18. Kuno, T., Ideguchi, H., Yoshida, N., Masuyama, T., Ohta, M., Nishimura, S., Tasaki, H., Miyazaki, S., Hara, H., Matsumoto, K. A case of X-linked alpha-thalassemia/mental retardation syndrome: analysis of hemoglobin by an automated glycated hemoglobin analyzer. Acta Paediat. Jpn. 39: 615-618, 1997. [PubMed: 9363663, related citations] [Full Text]

  19. Kurosawa, K., Akatsuka, A., Ochiai, Y., Ikeda, J., Maekawa, K. Self-induced vomiting in X-linked alpha-thalassemia/mental retardation syndrome. (Letter) Am. J. Med. Genet. 63: 505-506, 1996. [PubMed: 8967323, related citations] [Full Text]

  20. Leon, N. Y., Harley, V. R. ATR-X syndrome: genetics, clinical spectrum, and management. Hum. Genet. 140: 1625-1634, 2021. [PubMed: 34524523, related citations] [Full Text]

  21. Logie, L. J., Gibbons, R. J., Higgs, D. R., Brown, J. K., Porteous, M. E. M. Alpha thalassaemia mental retardation (ATR-X): an atypical family. Arch. Dis. Child. 70: 439-440, 1994. [PubMed: 8017970, related citations] [Full Text]

  22. Lossi, A. M., Millan, J. M., Villard, L., Orellana, C., Cardoso, C., Prieto, F., Fontes, M., Martinez, F. Mutation of the XNP/ATR-X gene in a family with severe mental retardation, spastic paraplegia and skewed pattern of X inactivation: demonstration that the mutation is involved in the inactivation bias. (Letter) Am. J. Hum. Genet. 65: 558-562, 1999. [PubMed: 10417298, related citations] [Full Text]

  23. Martinez, F., Tomas, M., Millan, J. M., Fernandez, A., Palau, F., Prieto, F. Genetic localisation of mental retardation with spastic diplegia to the pericentromeric region of the X chromosome: X inactivation in female carriers. J. Med. Genet. 35: 284-287, 1998. [PubMed: 9598720, related citations] [Full Text]

  24. Martucciello, G., Lombardi, L., Savasta, S., Gibbons, R. J. Gastrointestinal phenotype of ATR-X syndrome. Am. J. Med. Genet. 140A: 1172-1176, 2006. [PubMed: 16688741, related citations] [Full Text]

  25. McPherson, E. W., Clemens, M. M., Gibbons, R. J., Higgs, D. R. X-linked alpha-thalassemia/mental retardation (ATR-X) syndrome: a new kindred with severe genital anomalies and mild hematologic expression. Am. J. Med. Genet. 55: 302-306, 1995. [PubMed: 7726227, related citations] [Full Text]

  26. Medina, C. F., Mazerolle, C., Wang, Y., Berube, N. G., Coupland, S., Gibbons, R. J., Wallace, V. A., Picketts, D. J. Altered visual function and interneuron survival in Atrx knockout mice: inference for the human syndrome. Hum. Molec. Genet. 18: 966-977, 2009. [PubMed: 19088125, related citations] [Full Text]

  27. Porteous, M. E. M., Burn, J. Unknown syndrome. A possible new X linked retardation syndrome: dysmorphic facies, microcephaly, hypotonia, and small genitalia. J. Med. Genet. 27: 339-340, 1990. [PubMed: 2352265, related citations] [Full Text]

  28. Reardon, W., Gibbons, R. J., Winter, R. M., Baraitser, M. Male pseudohermaphroditism in sibs with the alpha-thalassemia/mental retardation (ATR-X) syndrome. Am. J. Med. Genet. 55: 285-287, 1995. [PubMed: 7726224, related citations] [Full Text]

  29. Sutherland, G. R., Gedeon, A. K., Haan, E. A., Woodroffe, P., Mulley, J. C. Linkage studies with the gene for an X-linked syndrome of mental retardation, microcephaly and spastic diplegia. Am. J. Med. Genet. 30: 493-508, 1988. [PubMed: 3177467, related citations] [Full Text]

  30. Thienpont, B., de Ravel, T., Van Esch, H., Van Schoubroeck, D., Moerman, P., Vermeesch, J. R., Fryns, J.-P., Froyen, G., Lacoste, C., Badens, C., Devriendt, K. Partial duplications of the ATRX gene cause the ATR-X syndrome. Europ. J. Hum. Genet. 15: 1094-1097, 2007. [PubMed: 17579672, related citations] [Full Text]

  31. Villard, L., Toutain, A., Lossi, A.-M., Gecz, J., Houdayer, C., Moraine, C., Fontes, M. Splicing mutation in the ATR-X gene can lead to a dysmorphic mental retardation phenotype without alpha-thalassemia. Am. J. Hum. Genet. 58: 499-505, 1996. [PubMed: 8644709, related citations]

  32. Wada, T., Sugie, H., Fukushima, Y., Saitoh, S. Non-skewed X-inactivation may cause mental retardation in a female carrier of X-linked alpha-thalassemia/mental retardation syndrome (ATR-X): X-inactivation study of nine female carriers of ATR-X. Am. J. Med. Genet. 138A: 18-20, 2005. [PubMed: 16100724, related citations] [Full Text]

  33. Wang, L. H., Collins, A., Lawrence, S., Keats, B. J., Morton, N. E. Integration of gene maps: chromosome X. Genomics 22: 590-604, 1994. [PubMed: 8001970, related citations] [Full Text]

  34. Weatherall, D. J., Higgs, D. R., Bunch, C., Old, J. M., Hunt, D. M., Pressley, L., Clegg, J. B., Bethlenfalvay, N. C., Sjolin, S., Koler, R. D., Magenis, E., Francis, J. L., Bebbington, D. Hemoglobin H disease and mental retardation: a new syndrome or a remarkable coincidence? New Eng. J. Med. 305: 607-612, 1981. [PubMed: 6267462, related citations] [Full Text]

  35. Wilkie, A. O. M., Pembrey, M. E., Gibbons, R. J., Higgs, D. R., Porteous, M. E. M., Burn, J., Winter, R. M. The non-deletion type of alpha thalassaemia/mental retardation: a recognisable dysmorphic syndrome with X linked inheritance. (Letter) J. Med. Genet. 28: 724 only, 1991. [PubMed: 1941971, related citations] [Full Text]

  36. Wilkie, A. O. M., Zeitlin, H. C., Lindenbaum, R. H., Buckle, V. J., Fischel-Ghodsian, N., Chui, D. H. K., Gardner-Medwin, D., MacGillivray, M. H., Weatherall, D. J., Higgs, D. R. Clinical features and molecular analysis of the alpha-thalassemia/mental retardation syndromes. II. Cases without detectable abnormality of the alpha-globin complex. Am. J. Hum. Genet. 46: 1127-1140, 1990. [PubMed: 2339705, related citations]


Hilary J. Vernon - updated : 07/19/2022
Cassandra L. Kniffin - updated : 10/13/2010
George E. Tiller - updated : 8/24/2009
Cassandra L. Kniffin - updated : 7/31/2009
Cassandra L. Kniffin - updated : 12/18/2006
Marla J. F. O'Neill - updated : 10/11/2006
Cassandra L. Kniffin - updated : 8/24/2006
Cassandra L. Kniffin - updated : 9/19/2005
Cassandra L. Kniffin - reorganized : 7/14/2005
Cassandra L. Kniffin - updated : 6/23/2005
Victor A. McKusick - updated : 1/22/2004
George E. Tiller - updated : 2/12/2002
Victor A. McKusick - updated : 10/3/2000
Creation Date:
Victor A. McKusick : 11/7/1991
carol : 05/22/2023
carol : 12/18/2022
carol : 07/20/2022
carol : 07/19/2022
carol : 07/09/2016
alopez : 7/25/2011
wwang : 10/20/2010
ckniffin : 10/13/2010
terry : 5/12/2010
wwang : 8/24/2009
wwang : 8/5/2009
ckniffin : 7/31/2009
wwang : 12/21/2006
ckniffin : 12/18/2006
wwang : 10/12/2006
terry : 10/11/2006
wwang : 9/1/2006
ckniffin : 8/24/2006
wwang : 11/23/2005
wwang : 10/3/2005
wwang : 9/30/2005
ckniffin : 9/19/2005
carol : 7/14/2005
ckniffin : 6/23/2005
ckniffin : 6/15/2005
ckniffin : 5/12/2005
terry : 1/22/2004
cwells : 2/18/2002
cwells : 2/12/2002
mcapotos : 10/6/2000
mcapotos : 10/5/2000
terry : 10/3/2000
carol : 8/4/1999
carol : 8/4/1999
mark : 2/12/1998
mark : 3/27/1997
mark : 6/25/1996
mark : 6/25/1996
mark : 6/25/1996
mark : 6/25/1996
mark : 6/25/1996
terry : 6/14/1996
mark : 3/7/1996
terry : 3/4/1996
mark : 4/8/1995
terry : 1/20/1995
carol : 10/6/1994
davew : 8/22/1994
warfield : 4/19/1994

# 301040

ALPHA-THALASSEMIA/IMPAIRED INTELLECTUAL DEVELOPMENT SYNDROME, X-LINKED; ATRX


Alternative titles; symbols

ALPHA-THALASSEMIA/MENTAL RETARDATION SYNDROME, X-LINKED
ALPHA-THALASSEMIA/MENTAL RETARDATION SYNDROME, NONDELETION TYPE
ATR-X SYNDROME
ATR, NONDELETION TYPE


SNOMEDCT: 715342005;   ORPHA: 847;   DO: 0110030;  


Phenotype-Gene Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Gene/Locus Gene/Locus
MIM number
Xq21.1 Alpha-thalassemia/impaired intellectual development syndrome 301040 X-linked dominant 3 ATRX 300032

TEXT

A number sign (#) is used with this entry because X-linked alpha-thalassemia/impaired intellectual development syndrome is caused by mutation in the ATRX gene (300032) on Xq21.

The 'deletion' type of alpha-thalassemia/impaired intellectual development syndrome (141750) is a contiguous gene syndrome due to a deletion in chromosome 16p that involves the hemoglobin alpha-1 (HBA1; 141800) and alpha-2 (HBA2; 141850) genes.

The X-linked mental retardation-hypotonic facies syndrome (309580) is also caused by mutation in the ATRX gene.


Clinical Features

Weatherall et al. (1981) reported the association of hemoglobin H disease (Hb H; see alpha-thalassemias, 141800) and mental retardation in 3 unrelated patients of northern European descent.

Wilkie et al. (1990) reported 5 unrelated patients, 2 of whom were reported by Weatherall et al. (1981), with mental retardation and alpha-thalassemia without molecular abnormalities of the alpha-globin gene complex on chromosome 16p. The patients showed a strikingly uniform phenotype comprising severe mental handicap, characteristic dysmorphic facies, genital abnormalities, and an unusual, mild form of hemoglobin H disease. Facial features included microcephaly, hypertelorism, epicanthus, a small triangular upturned nose, and flat face. The degree of red blood cell hypochromia and Hb H levels, which varied from 0.7 to 6.7%, were milder than usually found in alpha-thalassemia. Although several approaches failed to find a defect in the alpha-globin genes, 3 patients tested had markedly reduced total mRNA levels of both HBA1 and HBA2. The authors suggested that the responsible locus encoded a trans-acting factor involved in the normal regulation of alpha-globin expression.

Harvey et al. (1990) described the syndrome in a 21-year-old male and his brother who had died earlier, suggesting X-linked inheritance. DNA analysis showed no deletions within the alpha-globin gene cluster. Hb H bodies were present at a low level (1.6%).

Porteous and Burn (1990) described a 6-year-old boy who had a maternal uncle with an X-linked mental retardation syndrome, and suggested that their case resembled 2 brothers previously thought to have an atypical form of the Coffin-Lowry syndrome (303600) (illustrated in Smith's Recognizable Patterns of Human Malformation, Jones, 1988). However, Wilkie et al. (1991) found that there were hematologic signs of the nondeletion ATR syndrome in the patient reported by Porteous and Burn (1990). In addition, Wilkie et al. (1991) reported that hematologic evaluation of 1 of the brothers reported in Smith's book showed that he had nondeletion ATR and that a male first-cousin through the maternal line had the same condition. Wilkie et al. (1991) suggested that this condition be called 'X-linked alpha-thalassemia/mental retardation' (ATR-X) to distinguish it from the deletion form.

In a review, Gibbons et al. (1991) noted that some patients with ATR-X syndrome have normal or only mildly abnormal hematologic indices; thus normal hemoglobin levels and red cell indices do not necessarily exclude the condition.

Cole et al. (1991) described an affected boy whose maternal uncle was also affected. The boy had right-sided renal agenesis with left-sided hydronephrosis and hydroureter. He had recurrent hypochromic, microcytic anemia. His otherwise unaffected sister had had recurrent urinary tract infections and persistent renal impairment in the absence of any identifiable renal tract anomaly. Kurosawa et al. (1996) described a boy with self-induced vomiting followed by rumination and noted that Cole et al. (1991) made the same observation in a man and his nephew.

Donnai et al. (1991) described 4 brothers with this syndrome in whom the diagnosis was first suspected because of their characteristic clinical features and was confirmed in each case by the demonstration of Hb H inclusions in a proportion of their red blood cells. Very rare Hb H inclusions were found in the red blood cells of the mother and one sister who both shared some facial features with the affected boys; they were presumed to be carriers of the disorder.

Gorlin (1993) examined patients with typical features of the ATR-X syndrome, but without hemoglobin H. The facies were identical and mapping studies in several families suggested location of the mutation in the site on the X chromosome involved in ATR-X. The facies of this syndrome, which is often confused with that of Coffin-Lowry syndrome, were marked by telecanthus, epicanthic folds, flat nasal bridge, midface hypoplasia, a carp-shaped mouth with full lips, and small triangular nose with anteverted nostrils. Gorlin (1993) noted that the alae of the nose extended lower than the columella and septum. All developmental milestones, especially walking, were delayed and speech was almost absent. On further investigation, Gibbons (1994) found that the patients of Gorlin (1993) did have alpha-thalassemia, as indicated by the presence of hemoglobin H inclusions after use of 1% brilliant cresyl blue staining overnight in buffered solution at room temperature. With the staining, the Hb H inclusions give the erythrocytes the appearance of golf balls.

Logie et al. (1994) reported a pedigree with 6 affected males in 4 sibships spanning 2 generations. Two affected cousins were described in detail, one of whom had an unusually mild hematologic phenotype. Hb H inclusions, the hallmark of the disorder, were detected in the peripheral red blood cells only after repeated observations. The cousins had strikingly similar facies with telecanthus, anteverted nares, carp-shaped mouth, and large tongue. Gibbons et al. (1995) showed that the hematologic findings in ATR-X may vary widely; indeed, in some cases, the manifestation of alpha-thalassemia may be subtle and missed without repeated examinations. McPherson et al. (1995) described a kindred with 4 affected members. The hematologic abnormality was not detected on routine hematologic studies, including hemoglobin electrophoresis, but the patients were found to have hemoglobin H inclusions on brilliant cresyl blue staining of peripheral smears.

Reardon et al. (1995) reported 2 phenotypic females with a 46,XY karyotype who had abnormalities of the external genitalia resulting in male pseudohermaphroditism. They pointed out that 1 of the 5 original patients described in defining the ATR-X syndrome was a phenotypic female with a 46,XY karyotype (Wilkie et al., 1990). McPherson et al. (1995) described genital anomalies that led to a female sex of rearing in 3 of 4 affected members of a family. Gibbons et al. (1995) emphasized the progressive coarsening of the facial appearance. Kuno et al. (1997) described a 5-year-old Japanese boy with this condition. He had an abnormal hemoglobin which was found to consist exclusively of a beta subunit. Severe mental retardation and hypoplastic penis and testes were present. Anemia was only mild (hematocrit 35.8%). The family history was unremarkable.

Martinez et al. (1998) reported 2 brothers and 1 maternal cousin with severe mental retardation, microcephaly, short stature, cryptorchidism, and spastic diplegia. Some facial dysmorphic features were present. Martinez et al. (1998) pointed out the similarity in phenotype between their family and that described by Sutherland et al. (1988) (see 309500). They suggested that the greater phenotypic severity in their family was due to allelic heterogeneity. X-inactivation analysis of 1 potential and 3 obligate carriers showed nonrandom inactivation of the disease-linked variant. On further analysis of this family, Lossi et al. (1999) found that 3% of the patients' erythrocytes showed Hb H inclusions, consistent with ATR-X. Lossi et al. (1999) also reported dysmorphic facial features, including 'carp-like' triangular mouth, hypertelorism, small triangular nose, and broad nasal root. The hypertonia and spasticity were unusual findings in this family. A mutation was found in the ATRX gene in affected individuals (300032.0016).

Gibbons and Higgs (2000) provided a review of the clinical spectrum of syndromes caused by mutation in the XH2 gene.

Martucciello et al. (2006) described male 3-year-old dizygotic twins with ATRX who exhibited gastrointestinal problems including severe regurgitation of food, vomiting, dysphagia, irritability, respiratory disorders, meteorism, and chronic constipation. Barium studies in both twins showed gastric pseudovolvulus, and 24-hour pH monitoring showed severe gastroesophageal reflux. Enzymo-histochemical studies of full-thickness colonic biopsies revealed a complex dysganglionosis: ultrashort Hirschsprung disease (see 142623) associated with hypoganglionosis. Martucciello et al. (2006) reviewed the gastrointestinal phenotype of 128 confirmed cases of ATRX and found that drooling was reported in 36% of cases, gastroesophageal reflux was present in 72%, and constipation in 30%. Fundoplication was performed in 10% of cases, and 9% were fed by gastrostomy. Upper GI bleeding was reported in 10% of cases. Fatal aspiration of vomitus occurred in 3 patients; volvulus was seen in 4 patients, 2 of whom died after intestinal infarction; and 4 patients had recurrent hospitalizations for ileus or pseudoobstruction. Martucciello et al. (2006) also noted that there were numerous anecdotal reports from parents describing prolonged episodes of patient distress with refusal to eat or drink.

Jezela-Stanek et al. (2009) reported a patient with ATRX confirmed by genetic analysis. He had hypertelorism, epicanthal folds, strabismus, short nose with flat bridge and triangular upturned tip, and tented upper lip with everted lower lip. Other features included hypotonia, psychomotor retardation, and hemoglobin H inclusions. The patient also had undescended testes and ambiguous genitalia, which the authors referred to as male pseudohermaphroditism. Laboratory studies showed increased FSH and decreased testosterone. A deceased sib was believed to have been affected and reportedly had ambiguous external genitalia. Jezela-Stanek et al. (2009) postulated that the distinctive facial features in ATRX result from facial hypotonia and can be confused with Coffin-Lowry syndrome (CLS; 303600) or SLO syndromes (SLOS; 270400).

Based on a literature review, Leon and Harley (2021) reported the frequency of clinical features in individuals with ATR-X syndrome. The clinical features were highly variable, even within the same family. The most common feature was intellectual disability, identified in 100% of patients, followed by hematologic abnormalities in 70%, genital abnormalities in 62%, characteristic facies in 55%, skeletal abnormalities in 43%, and microcephaly in 42%. Less frequent features included hypotonia (34%), gastrointestinal problems (30%), seizures (17%), abnormal behavior (10%), heart defects (8%), and osteosarcoma (3%).

Carrier Females

Studying 7 pedigrees that included individuals with the ATR-X syndrome, Gibbons et al. (1992) concluded that intellectually normal female carriers could be identified by the presence of rare cells containing Hb H inclusions in their peripheral blood and by an extremely skewed pattern of X inactivation in cells from a variety of tissues. McPherson et al. (1995) used a combination of skewed X inactivation and haplotype analysis at Xq12-q21.3 to establish carrier status.

Wada et al. (2005) found skewed X-inactivation patterns (greater than 90:10) in 6 of 7 unaffected Japanese female ATR-X carriers; the 1 carrier with nonskewed X inactivation (72:28) demonstrated moderate mental retardation. The woman did not have dysmorphic features or hemoglobin inclusions. Wada et al. (2005) concluded that mutations in the ATRX gene may cause mental retardation in females if the chromosome carrying the mutation is not properly inactivated.

Badens et al. (2006) reported a 4-year-old girl with typical features of the ATR-X syndrome. Molecular studies showed a totally skewed X-inactivation pattern, with the active chromosome carrying a heterozygous mutation in the ATRX gene (300032.0018). Neither parent had the mutation in peripheral blood leukocytes, but SNP analysis indicated that the mutation occurred on the maternal chromosome. The child was conceived with assisted reproduction technologies (ART) due to micropolycystic ovaries and endometriosis in the mother. Badens et al. (2006) suggested that some aspect of ART may have disturbed imprinting in this patient.


Mapping

By linkage analysis of 7 affected pedigrees, Gibbons et al. (1992) mapped the ATR-X locus to an 11-cM interval on chromosome Xq12-q21.31 between markers DXS106 and DXYS1X (peak lod score of 5.4 at theta = 0 at DXS72).

In a 3-generation ATR-X family with 3 affected males, Houdayer et al. (1993) demonstrated a maximum lod score of 2.09 at a recombination fraction of zero for linkage with DXS453 located at the boundary Xq12-q13.1. The nearest flanking loci demonstrating recombination with the disease locus were the androgen receptor (AR; 313700) at Xq11.2-q12 on the centromeric side and DXS72 at Xq21.1 on the telomeric side. Houdayer et al. (1993) interpreted their results as compatible with a distal boundary at Xq21.1 instead of Xq21.31 as previously held.

Gibbons et al. (1995) performed linkage analysis in 9 families with ATR-X syndrome and identified key recombinants that reduced the area of interest to 1.4 cM (estimated to be 15 Mb) between DXS454 and DXS72 within Xq13.1-q21.1 (Wang et al., 1994).


Diagnosis

Leon and Harley (2021) recommended that targeted sequencing of the ATRX gene should be the first diagnostic test in patients with more than one hallmark clinical feature of ATR-X syndrome. If a mutation is not identified by this method, other techniques to identify deletions or duplications in the ATRX gene, such as chromosome microarray or MLPA, should be considered.


Molecular Genetics

In patients with the ATR-X syndrome, Gibbons et al. (1995) identified mutations in the ATRX gene (300032.0001-300032.0009).

In affected members of a family with ATR-X syndrome, Villard et al. (1996) identified a splice site mutation in the ATRX gene (300032.0010). In 2 first cousins presenting the classic ATR-X phenotype with alpha-thalassemia and Hb H inclusions, only the abnormal transcript was expressed. In a distant cousin presenting with a similar dysmorphic mental retardation phenotype, but without thalassemia, they found that approximately 30% of the ATRX transcripts were normal. These data suggested that the mode of action of the ATRX gene product on globin expression is distinct from its mode of action in brain development and facial morphogenesis, and that the mutated splice site could be used with varying efficiency in different individuals.

Hendrich and Bickmore (2001) reviewed human disorders that share in common defects of chromatin structure or modification, including the ATR-X spectrum of disorders, ICF syndrome (242860), Rett syndrome (312750), Rubinstein-Taybi syndrome (180849), and Coffin-Lowry syndrome.

Partial Duplication of the ATRX Gene

Thienpont et al. (2007) reported 3 patients, including 2 sibs, with the ATRX syndrome due to partial duplications of the ATRX gene. In 1 family, the duplication included exons 2 to 35; in the other family, exons 2 to 29. Further analysis showed that both mothers carried the duplication and both had skewed X inactivation. In 1 patient, ATRX mRNA levels were about 3% of normal values. Thienpont et al. (2007) noted that the duplications were not identified by sequence analysis and suggested that quantitative analysis to detect copy numbers of the ATRX gene may be required in some cases.

Cohn et al. (2009) reported a family in which 3 males had ATRX syndrome due to a partial intragenic duplication of the ATRX gene that spanned exons 2 to 31. Northern blot analysis failed to identify a full-length transcript, but cDNA sequencing was consistent with some level of expression. The authors noted that complete loss of ATRX is most likely lethal, suggesting that the mutation was likely hypomorphic and associated with some residual protein function. Unaffected obligate carrier females in the family had highly skewed X inactivation. The phenotype was typical for the disorder, although the facial features were not as readily apparent in the 2 older affected individuals. The proband was identified from 2 larger cohorts comprising 300 males with mental retardation. Cohn et al. (2009) did not find ATRX duplications in 29 additional males with ATRX syndrome who were negative on sequence analysis, suggesting that duplications are a rare cause of the disorder.


Genotype/Phenotype Correlations

In a review article, Gibbons and Higgs (2000) noted that mutations in the ATRX gene resulting in the loss of the C terminal domain are associated with the most severe urogenital abnormalities. However, at other sites, there is no obvious link between genotype and phenotype, and there is considerable variation in the degree of abnormalities seen in individuals with the same mutation.

Among 22 ATRX patients from 16 families, Badens et al. (2006) found that those with mutations in the PHD-like domain of the ATRX protein had significantly more severe and permanent psychomotor retardation and significantly more severe urogenital anomalies compared to those with mutations in the helicase domain.

Based on a review of the literature, Leon and Harley (2021) noted that the ATRX mutations in individuals with ATR-X syndrome who developed osteosarcoma were located in the C-terminal region of the gene.


Animal Model

Medina et al. (2009) surveyed ATR-X syndrome clinical findings and noted that ocular defects were present in 47 (23%) of 202 patients. They showed that Atrx was expressed in the neuroprogenitor pool in embryonic mouse retina and in all cell types of adult mouse retina except rod photoreceptors. Conditional inactivation of Atrx in mouse retina during embryogenesis resulted in loss of only 2 types of neurons, amacrine and horizontal cells. This defect did not arise from a failure to specify these cells, but rather a defect in interneuron differentiation and survival postnatally. The timing of cell loss was concomitant with light-dependent changes in synaptic organization in mouse retina and with a change in Atrx subnuclear localization within these interneurons. The interneuron defects were associated with functional deficits as demonstrated by reduced b-wave amplitudes upon electroretinogram analysis. Medina et al. (2009) proposed a role for Atrx in interneuron survival and differentiation.


REFERENCES

  1. Badens, C., Lacoste, C., Philip, N., Martini, N., Courrier, S., Giuliano, F., Verloes, A., Munnich, A., Leheup, B., Burglen, L., Odent, S., Van Esch, H., Levy, N. Mutations in PHD-like domain of the ATRX gene correlate with severe psychomotor impairment and severe urogenital abnormalities in patients with ATRX syndrome. Clin. Genet. 70: 57-62, 2006. [PubMed: 16813605] [Full Text: https://doi.org/10.1111/j.1399-0004.2006.00641.x]

  2. Badens, C., Martini, N., Courrier, S., DesPortes, V., Touraine, R., Levy, N., Edery, P. ATRX syndrome in a girl with a heterozygous mutation in the ATRX Zn finger domain and a totally skewed X-inactivation pattern. Am. J. Med. Genet. 140A: 2212-2215, 2006. [PubMed: 16955409] [Full Text: https://doi.org/10.1002/ajmg.a.31400]

  3. Cohn, D. M., Pagon, R. A., Hudgins, L., Schwartz, C. E., Stevenson, R. E., Friez, M. J. Partial ATRX gene duplication causes ATR-X syndrome. (Letter) Am. J. Med. Genet. 149A: 2317-2320, 2009. [PubMed: 19764021] [Full Text: https://doi.org/10.1002/ajmg.a.33006]

  4. Cole, T. R. P., May, A., Hughes, H. E. Alpha-thalassaemia/mental retardation syndrome (non-deletional type): report of a family supporting X linked inheritance. J. Med. Genet. 28: 734-737, 1991. [PubMed: 1770528] [Full Text: https://doi.org/10.1136/jmg.28.11.734]

  5. Donnai, D., Clayton-Smith, J., Gibbons, R. J., Higgs, D. R. The non-deletion alpha-thalassaemia/mental retardation syndrome: further support for X linkage. J. Med. Genet. 28: 742-745, 1991. [PubMed: 1770530] [Full Text: https://doi.org/10.1136/jmg.28.11.742]

  6. Gibbons, R. J., Brueton, L., Buckle, V. J., Burn, J., Clayton-Smith, J., Davison, B. C. C., Gardner, R. J. M., Homfray, T., Kearney, L., Kingston, H. M., Newbury-Ecob, R., Porteous, M. E. P., Wilkie, A. O. M., Higgs, D. R. Clinical and hematologic aspects of the X-linked alpha-thalassemia/mental retardation syndrome (ATR-X). Am. J. Med. Genet. 55: 288-299, 1995. [PubMed: 7726225] [Full Text: https://doi.org/10.1002/ajmg.1320550309]

  7. Gibbons, R. J., Higgs, D. R. Molecular-clinical spectrum of the ATR-X syndrome. Am. J. Med. Genet. 97: 204-212, 2000. [PubMed: 11449489] [Full Text: https://doi.org/10.1002/1096-8628(200023)97:3<204::AID-AJMG1038>3.0.CO;2-X]

  8. Gibbons, R. J., Picketts, D. J., Villard, L., Higgs, D. R. Mutations in a putative global transcriptional regulator cause X-linked mental retardation with alpha-thalassemia (ATR-X syndrome). Cell 80: 837-845, 1995. [PubMed: 7697714] [Full Text: https://doi.org/10.1016/0092-8674(95)90287-2]

  9. Gibbons, R. J., Suthers, G. K., Wilkie, A. O. M., Buckle, V. J., Higgs, D. R. X-linked alpha-thalassemia/mental retardation (ATR-X) syndrome: localization to Xq12-q21.31 by X inactivation and linkage analysis. Am. J. Hum. Genet. 51: 1136-1149, 1992. [PubMed: 1415255]

  10. Gibbons, R. J., Wilkie, A. O. M., Weatherall, D. J., Higgs, D. R. A newly defined X linked mental retardation syndrome associated with alpha-thalassaemia. J. Med. Genet. 28: 729-733, 1991. [PubMed: 1770527] [Full Text: https://doi.org/10.1136/jmg.28.11.729]

  11. Gibbons, R. Personal Communication. Oxford, England 11/7/1994.

  12. Gorlin, R. J. Personal Communication. Minneapolis, Minn. 5/29/1993.

  13. Harvey, M. P., Kearney, A., Smith, A., Trent, R. J. Occurrence of the alpha-thalassaemia-mental retardation syndrome (non-deletional type) in an Australian male. J. Med. Genet. 27: 577-581, 1990. [PubMed: 2231651] [Full Text: https://doi.org/10.1136/jmg.27.9.577]

  14. Hendrich, B., Bickmore, W. Human diseases with underlying defects in chromatin structure and modification. Hum. Molec. Genet. 10: 2233-2242, 2001. [PubMed: 11673406] [Full Text: https://doi.org/10.1093/hmg/10.20.2233]

  15. Houdayer, C., Toutain, A., Ronce, N., Lefort, G., Sarda, P., Taib, J., Briault, S., Lambert, J. C., Moraine, C. X-linked alpha-thalassemia/mental retardation syndrome: linkage analysis in a new family further supports localization in proximal Xq. Ann. Genet. 36: 194-199, 1993. [PubMed: 8166423]

  16. Jezela-Stanek, A., Fisher, C., Szarras-Czapnik, M., Olczak-Kowalczyk, D., Gibbons, R. J., Slowikowska-Hilczer, J., Krajewska-Walasek, M. X-linked alpha thalassaemia/mental retardation syndrome: a case with gonadal dysgenesis, caused by a novel mutation in ATRX gene. Clin. Dysmorph. 18: 168-171, 2009. [PubMed: 19444090] [Full Text: https://doi.org/10.1097/MCD.0b013e32832a9ea5]

  17. Jones, K. L. Smith's Recognizable Patterns of Human Malformation. (4th ed.) Philadelphia: W. B. Saunders (pub.) 1988. P. 237.

  18. Kuno, T., Ideguchi, H., Yoshida, N., Masuyama, T., Ohta, M., Nishimura, S., Tasaki, H., Miyazaki, S., Hara, H., Matsumoto, K. A case of X-linked alpha-thalassemia/mental retardation syndrome: analysis of hemoglobin by an automated glycated hemoglobin analyzer. Acta Paediat. Jpn. 39: 615-618, 1997. [PubMed: 9363663] [Full Text: https://doi.org/10.1111/j.1442-200x.1997.tb03651.x]

  19. Kurosawa, K., Akatsuka, A., Ochiai, Y., Ikeda, J., Maekawa, K. Self-induced vomiting in X-linked alpha-thalassemia/mental retardation syndrome. (Letter) Am. J. Med. Genet. 63: 505-506, 1996. [PubMed: 8967323] [Full Text: https://doi.org/10.1002/ajmg.1320630304]

  20. Leon, N. Y., Harley, V. R. ATR-X syndrome: genetics, clinical spectrum, and management. Hum. Genet. 140: 1625-1634, 2021. [PubMed: 34524523] [Full Text: https://doi.org/10.1007/s00439-021-02361-5]

  21. Logie, L. J., Gibbons, R. J., Higgs, D. R., Brown, J. K., Porteous, M. E. M. Alpha thalassaemia mental retardation (ATR-X): an atypical family. Arch. Dis. Child. 70: 439-440, 1994. [PubMed: 8017970] [Full Text: https://doi.org/10.1136/adc.70.5.439]

  22. Lossi, A. M., Millan, J. M., Villard, L., Orellana, C., Cardoso, C., Prieto, F., Fontes, M., Martinez, F. Mutation of the XNP/ATR-X gene in a family with severe mental retardation, spastic paraplegia and skewed pattern of X inactivation: demonstration that the mutation is involved in the inactivation bias. (Letter) Am. J. Hum. Genet. 65: 558-562, 1999. [PubMed: 10417298] [Full Text: https://doi.org/10.1086/302499]

  23. Martinez, F., Tomas, M., Millan, J. M., Fernandez, A., Palau, F., Prieto, F. Genetic localisation of mental retardation with spastic diplegia to the pericentromeric region of the X chromosome: X inactivation in female carriers. J. Med. Genet. 35: 284-287, 1998. [PubMed: 9598720] [Full Text: https://doi.org/10.1136/jmg.35.4.284]

  24. Martucciello, G., Lombardi, L., Savasta, S., Gibbons, R. J. Gastrointestinal phenotype of ATR-X syndrome. Am. J. Med. Genet. 140A: 1172-1176, 2006. [PubMed: 16688741] [Full Text: https://doi.org/10.1002/ajmg.a.31248]

  25. McPherson, E. W., Clemens, M. M., Gibbons, R. J., Higgs, D. R. X-linked alpha-thalassemia/mental retardation (ATR-X) syndrome: a new kindred with severe genital anomalies and mild hematologic expression. Am. J. Med. Genet. 55: 302-306, 1995. [PubMed: 7726227] [Full Text: https://doi.org/10.1002/ajmg.1320550311]

  26. Medina, C. F., Mazerolle, C., Wang, Y., Berube, N. G., Coupland, S., Gibbons, R. J., Wallace, V. A., Picketts, D. J. Altered visual function and interneuron survival in Atrx knockout mice: inference for the human syndrome. Hum. Molec. Genet. 18: 966-977, 2009. [PubMed: 19088125] [Full Text: https://doi.org/10.1093/hmg/ddn424]

  27. Porteous, M. E. M., Burn, J. Unknown syndrome. A possible new X linked retardation syndrome: dysmorphic facies, microcephaly, hypotonia, and small genitalia. J. Med. Genet. 27: 339-340, 1990. [PubMed: 2352265] [Full Text: https://doi.org/10.1136/jmg.27.5.339]

  28. Reardon, W., Gibbons, R. J., Winter, R. M., Baraitser, M. Male pseudohermaphroditism in sibs with the alpha-thalassemia/mental retardation (ATR-X) syndrome. Am. J. Med. Genet. 55: 285-287, 1995. [PubMed: 7726224] [Full Text: https://doi.org/10.1002/ajmg.1320550308]

  29. Sutherland, G. R., Gedeon, A. K., Haan, E. A., Woodroffe, P., Mulley, J. C. Linkage studies with the gene for an X-linked syndrome of mental retardation, microcephaly and spastic diplegia. Am. J. Med. Genet. 30: 493-508, 1988. [PubMed: 3177467] [Full Text: https://doi.org/10.1002/ajmg.1320300152]

  30. Thienpont, B., de Ravel, T., Van Esch, H., Van Schoubroeck, D., Moerman, P., Vermeesch, J. R., Fryns, J.-P., Froyen, G., Lacoste, C., Badens, C., Devriendt, K. Partial duplications of the ATRX gene cause the ATR-X syndrome. Europ. J. Hum. Genet. 15: 1094-1097, 2007. [PubMed: 17579672] [Full Text: https://doi.org/10.1038/sj.ejhg.5201878]

  31. Villard, L., Toutain, A., Lossi, A.-M., Gecz, J., Houdayer, C., Moraine, C., Fontes, M. Splicing mutation in the ATR-X gene can lead to a dysmorphic mental retardation phenotype without alpha-thalassemia. Am. J. Hum. Genet. 58: 499-505, 1996. [PubMed: 8644709]

  32. Wada, T., Sugie, H., Fukushima, Y., Saitoh, S. Non-skewed X-inactivation may cause mental retardation in a female carrier of X-linked alpha-thalassemia/mental retardation syndrome (ATR-X): X-inactivation study of nine female carriers of ATR-X. Am. J. Med. Genet. 138A: 18-20, 2005. [PubMed: 16100724] [Full Text: https://doi.org/10.1002/ajmg.a.30901]

  33. Wang, L. H., Collins, A., Lawrence, S., Keats, B. J., Morton, N. E. Integration of gene maps: chromosome X. Genomics 22: 590-604, 1994. [PubMed: 8001970] [Full Text: https://doi.org/10.1006/geno.1994.1432]

  34. Weatherall, D. J., Higgs, D. R., Bunch, C., Old, J. M., Hunt, D. M., Pressley, L., Clegg, J. B., Bethlenfalvay, N. C., Sjolin, S., Koler, R. D., Magenis, E., Francis, J. L., Bebbington, D. Hemoglobin H disease and mental retardation: a new syndrome or a remarkable coincidence? New Eng. J. Med. 305: 607-612, 1981. [PubMed: 6267462] [Full Text: https://doi.org/10.1056/NEJM198109103051103]

  35. Wilkie, A. O. M., Pembrey, M. E., Gibbons, R. J., Higgs, D. R., Porteous, M. E. M., Burn, J., Winter, R. M. The non-deletion type of alpha thalassaemia/mental retardation: a recognisable dysmorphic syndrome with X linked inheritance. (Letter) J. Med. Genet. 28: 724 only, 1991. [PubMed: 1941971] [Full Text: https://doi.org/10.1136/jmg.28.10.724]

  36. Wilkie, A. O. M., Zeitlin, H. C., Lindenbaum, R. H., Buckle, V. J., Fischel-Ghodsian, N., Chui, D. H. K., Gardner-Medwin, D., MacGillivray, M. H., Weatherall, D. J., Higgs, D. R. Clinical features and molecular analysis of the alpha-thalassemia/mental retardation syndromes. II. Cases without detectable abnormality of the alpha-globin complex. Am. J. Hum. Genet. 46: 1127-1140, 1990. [PubMed: 2339705]


Contributors:
Hilary J. Vernon - updated : 07/19/2022
Cassandra L. Kniffin - updated : 10/13/2010
George E. Tiller - updated : 8/24/2009
Cassandra L. Kniffin - updated : 7/31/2009
Cassandra L. Kniffin - updated : 12/18/2006
Marla J. F. O'Neill - updated : 10/11/2006
Cassandra L. Kniffin - updated : 8/24/2006
Cassandra L. Kniffin - updated : 9/19/2005
Cassandra L. Kniffin - reorganized : 7/14/2005
Cassandra L. Kniffin - updated : 6/23/2005
Victor A. McKusick - updated : 1/22/2004
George E. Tiller - updated : 2/12/2002
Victor A. McKusick - updated : 10/3/2000

Creation Date:
Victor A. McKusick : 11/7/1991

Edit History:
carol : 05/22/2023
carol : 12/18/2022
carol : 07/20/2022
carol : 07/19/2022
carol : 07/09/2016
alopez : 7/25/2011
wwang : 10/20/2010
ckniffin : 10/13/2010
terry : 5/12/2010
wwang : 8/24/2009
wwang : 8/5/2009
ckniffin : 7/31/2009
wwang : 12/21/2006
ckniffin : 12/18/2006
wwang : 10/12/2006
terry : 10/11/2006
wwang : 9/1/2006
ckniffin : 8/24/2006
wwang : 11/23/2005
wwang : 10/3/2005
wwang : 9/30/2005
ckniffin : 9/19/2005
carol : 7/14/2005
ckniffin : 6/23/2005
ckniffin : 6/15/2005
ckniffin : 5/12/2005
terry : 1/22/2004
cwells : 2/18/2002
cwells : 2/12/2002
mcapotos : 10/6/2000
mcapotos : 10/5/2000
terry : 10/3/2000
carol : 8/4/1999
carol : 8/4/1999
mark : 2/12/1998
mark : 3/27/1997
mark : 6/25/1996
mark : 6/25/1996
mark : 6/25/1996
mark : 6/25/1996
mark : 6/25/1996
terry : 6/14/1996
mark : 3/7/1996
terry : 3/4/1996
mark : 4/8/1995
terry : 1/20/1995
carol : 10/6/1994
davew : 8/22/1994
warfield : 4/19/1994