Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Mol Sci ; 22(3)2021 Jan 23.
Article in English | MEDLINE | ID: mdl-33498634

ABSTRACT

A growing number of genetic neurodevelopmental disorders are known to be associated with unique genomic DNA methylation patterns, called episignatures, which are detectable in peripheral blood. The intellectual developmental disorder, X-linked, syndromic, Armfield type (MRXSA) is caused by missense variants in FAM50A. Functional studies revealed the pathogenesis to be a spliceosomopathy that is characterized by atypical mRNA processing during development. In this study, we assessed the peripheral blood specimens in a cohort of individuals with MRXSA and detected a unique and highly specific DNA methylation episignature associated with this disorder. We used this episignature to construct a support vector machine model capable of sensitive and specific identification of individuals with pathogenic variants in FAM50A. This study contributes to the expanding number of genetic neurodevelopmental disorders with defined DNA methylation episignatures, provides an additional understanding of the associated molecular mechanisms, and further enhances our ability to diagnose patients with rare disorders.


Subject(s)
DNA Methylation , Mental Retardation, X-Linked/genetics , Adult , Case-Control Studies , Child , DNA-Binding Proteins/genetics , Epigenome , Humans , Male , Mental Retardation, X-Linked/etiology , Middle Aged , Models, Genetic , Neurodevelopmental Disorders/genetics , RNA-Binding Proteins/genetics
2.
Am J Med Genet A ; 182(4): 823-828, 2020 04.
Article in English | MEDLINE | ID: mdl-31943778

ABSTRACT

Missense variants in HNRNPH2 cause Bain type syndromic X-linked intellectual disability (XLID). To date, only six affected females and three affected males have been reported in the literature, and the phenotype has yet to be delineated in detail. Here, we report on a 35-year-old female with a novel de novo variant in HNRNPH2, providing further evidence that missense changes in the nuclear localization sequence cause Bain type XLID and that aminoacid 206 likely represents a mutational hotspot. We expand the phenotype of Bain type XLID to include breathing, sleep and movement disorders, cerebellar vermis hypoplasia, stereotypies, and hypersensitivity to noise. Our data indicate that the phenotype may be broader and more variable than initially reported, and suggest Rett syndrome as a possible differential diagnosis.


Subject(s)
Abnormalities, Multiple/etiology , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Intellectual Disability/etiology , Mental Retardation, X-Linked/etiology , Mutation, Missense , Abnormalities, Multiple/pathology , Adult , Exome , Female , Humans , Intellectual Disability/pathology , Mental Retardation, X-Linked/pathology , Phenotype
3.
Eur J Med Genet ; 63(4): 103777, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31580924

ABSTRACT

Snyder-Robinson syndrome (SRS) is an X-linked syndromic intellectual disability condition caused by variants in the spermine synthase gene (SMS). The syndrome is characterized by facial dysmorphism, thin body build, kyphoscoliosis, osteoporosis, hypotonia, developmental delay and associated neurological features (seizures, unsteady gait, abnormal speech). Until now, only missense variants with a functionally characterized partial loss of function (LoF) have been described. Here we describe the first complete LoF variant, Met303Lysfs*, in a male patient with a severe form of Snyder-Robinson syndrome. He presented with multiple malformations and severly delayed development, and died at 4 months of age. Functional in vitro assays showed a complete absence of functional SMS protein. Taken together, our findings and those of previously reported patients confirm that pathogenic variants of SMS are indeed LoF and that there might exist a genotype-phenotype correlation between the type of variant and the severity of the syndrome.


Subject(s)
Loss of Function Mutation , Mental Retardation, X-Linked/etiology , Spermine Synthase/genetics , Humans , Infant , Male , Mental Retardation, X-Linked/pathology , Prognosis
4.
Yakugaku Zasshi ; 139(4): 497-503, 2019.
Article in Japanese | MEDLINE | ID: mdl-30930376

ABSTRACT

Tissue barriers contribute to the maintenance of homeostasis in the body, and tissue barrier dysfunction presents a risk factor for a variety of diseases. The blood-brain barrier (BBB) is a major tissue barrier acting as a static barrier and dynamic interface that plays an important role in the maintenance of central nervous system homeostasis. We show the functional characterization of the brain-to-blood efflux transport system of amyloid-ß peptide (Aß) across the BBB. We found that activated vitamin D3 may be a candidate agent for modulating the Aß clearance across the BBB. Cerebral creatine deficiency syndromes are caused by loss-of-function mutations in the creatine transporter (CRT; SLC6A8), which transports creatine at the BBB. We found that functional impairment of CRT due to a G561R mutation resulted in incomplete N-linked glycosylation due to misfolding during protein maturation, leading to impaired creatine transport activity at the BBB. To develop a delivery system for biomedicine across the tissue barrier, we established a screening system to identify cell-penetrating peptides by a combination of in vitro cell permeability screening assays and phage display technology. Using this system, we identified cyclic hepta-peptides that are able to facilitate intestinal absorption of phages in vitro and in vivo, which are promising candidates as a carrier for macromolecular biomedicines. In conclusion, these studies focusing on the dynamic interface of tissue barriers will contribute to knowledge on disease pathogenesis as well as the development of a targeted biomedicine delivery system.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems , Drug Discovery , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Animals , Brain Diseases, Metabolic, Inborn/etiology , Brain Diseases, Metabolic, Inborn/genetics , Cholecalciferol/pharmacology , Creatine/deficiency , Creatine/genetics , Glycosylation , Humans , Loss of Function Mutation , Membrane Transport Proteins/genetics , Mental Retardation, X-Linked/etiology , Mental Retardation, X-Linked/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Plasma Membrane Neurotransmitter Transport Proteins/genetics
5.
BMC Med Genet ; 16: 112, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26672597

ABSTRACT

BACKGROUND: Methyl-CpG-binding protein 2 (MeCP2) is a key transcriptional regulator of gene expression in the maintenance and development of the central nervous system. Loss- or gain-function of this gene may contribute to neurodevelopmental disorders. The aim of this study is to delineate the clinical characteristics of MECP2 duplication syndrome and the hereditary mechanism in a Chinese family. CASE PRESENTATION: We identified a Chinese family with three persons carry MECP2 gene duplication: a boy, his mother and his grandmother. The duplication segment which was detected by multiplex ligation-dependent probe amplification (MLPA) included gene MECP2, interleukin-1 receptor-associated kinase 1 (IRAK1), filamin A (FLNA), and L1 cell adhesion molecule (L1CAM). Furthermore, array comparative genomic hybridization (aCGH) was performed on the mother, showed that MECP2 containing duplication was 510 Kb (153,113,885-153,624,154), including 16 other genes except MECP2. The boy showed most symptoms of MECP2 duplication syndrome. His mother and maternal grandmother were asymptomatic. Both female carriers had a skewed X chromosome inactivation (XCI), which were 80:20 and 74:26 respectively. CONCLUSION: To our knowledge, this is the second reported Chinese Han family with MECP2-containing duplications. And this patient had recurrent respiratory infections which was different from the first two Chinese-brother cases. MECP2 is the core gene responsible for MECP2 duplication syndrome. XCI may play an important role in modulating the clinical manifestation.


Subject(s)
Mental Retardation, X-Linked/genetics , Methyl-CpG-Binding Protein 2/genetics , Asian People/genetics , Child, Preschool , Female , Gene Duplication , Humans , In Situ Hybridization, Fluorescence , Male , Mental Retardation, X-Linked/etiology , Pedigree , X Chromosome Inactivation
6.
PLoS Genet ; 10(9): e1004615, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25255244

ABSTRACT

The mechanisms and treatment of psychomotor retardation, which includes motor and cognitive impairment, are indefinite. The Allan-Herndon-Dudley syndrome (AHDS) is an X-linked psychomotor retardation characterized by delayed development, severe intellectual disability, muscle hypotonia, and spastic paraplegia, in combination with disturbed thyroid hormone (TH) parameters. AHDS has been associated with mutations in the monocarboxylate transporter 8 (mct8/slc16a2) gene, which is a TH transporter. In order to determine the pathophysiological mechanisms of AHDS, MCT8 knockout mice were intensively studied. Although these mice faithfully replicated the abnormal serum TH levels, they failed to exhibit the neurological and behavioral symptoms of AHDS patients. Here, we generated an mct8 mutant (mct8-/-) zebrafish using zinc-finger nuclease (ZFN)-mediated targeted gene editing system. The elimination of MCT8 decreased the expression levels of TH receptors; however, it did not affect the expression of other TH-related genes. Similar to human patients, mct8-/- larvae exhibited neurological and behavioral deficiencies. High-throughput behavioral assays demonstrated that mct8-/- larvae exhibited reduced locomotor activity, altered response to external light and dark transitions and an increase in sleep time. These deficiencies in behavioral performance were associated with altered expression of myelin-related genes and neuron-specific deficiencies in circuit formation. Time-lapse imaging of single-axon arbors and synapses in live mct8-/- larvae revealed a reduction in filopodia dynamics and axon branching in sensory neurons and decreased synaptic density in motor neurons. These phenotypes enable assessment of the therapeutic potential of three TH analogs that can enter the cells in the absence of MCT8. The TH analogs restored the myelin and axon outgrowth deficiencies in mct8-/- larvae. These findings suggest a mechanism by which MCT8 regulates neural circuit assembly, ultimately mediating sensory and motor control of behavioral performance. We also propose that the administration of TH analogs early during embryo development can specifically reduce neurological damage in AHDS patients.


Subject(s)
Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/etiology , Muscle Hypotonia/diagnosis , Muscle Hypotonia/etiology , Muscular Atrophy/diagnosis , Muscular Atrophy/etiology , Animals , Animals, Genetically Modified , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Gene Order , Gene Targeting , Hypothalamo-Hypophyseal System , Kruppel-Like Transcription Factors/genetics , Mental Retardation, X-Linked/drug therapy , Monocarboxylic Acid Transporters/genetics , Motor Activity/genetics , Muscle Hypotonia/drug therapy , Muscular Atrophy/drug therapy , Mutation , Myelin Sheath/metabolism , Neurogranin/genetics , Neurons/metabolism , Phenotype , Photoperiod , Pseudopodia/genetics , Pseudopodia/metabolism , Thyroid Gland , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormones/pharmacology , Zebrafish , Zinc Fingers
7.
Am J Med Genet C Semin Med Genet ; 166C(3): 290-301, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25099957

ABSTRACT

Recently, de novo aberrations in PHF6 were identified in females with intellectual disability and with a distinct phenotype including a characteristic facial gestalt with bitemporal narrowing, prominent supraorbital ridges, synophrys, a short nose and dental anomalies, tapering fingers with brachytelephalangy, clinodactyly and hypoplastic nails, short toes with hypoplastic nails, and linear skin hyperpigmentation. In adolescent or older patients, this phenotype overlaps but is not identical with Borjeson-Forssman-Lehmann syndrome in males, caused by X-linked recessive mutations in PHF6. In younger girls there seems to be a striking phenotypic overlap with Coffin-Siris syndrome, which is characterized by intellectual disability, sparse hair and hypoplastic nails. This review will summarize and characterize the female phenotype caused by de novo aberrations in PHF6 and will discuss the overlapping and distinguishing features with Coffin-Siris syndrome.


Subject(s)
Abnormalities, Multiple/etiology , Carrier Proteins/genetics , Epilepsy/etiology , Face/abnormalities , Fingers/abnormalities , Growth Disorders/etiology , Hand Deformities, Congenital/etiology , Hypogonadism/etiology , Intellectual Disability/etiology , Mental Retardation, X-Linked/etiology , Micrognathism/etiology , Neck/abnormalities , Obesity/etiology , Abnormalities, Multiple/genetics , Child , Epilepsy/genetics , Female , Genetic Association Studies , Growth Disorders/genetics , Hand Deformities, Congenital/genetics , Humans , Hypogonadism/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Micrognathism/genetics , Nails, Malformed/genetics , Obesity/genetics , Pedigree , Repressor Proteins , Young Adult
8.
Curr Opin Genet Dev ; 23(3): 289-94, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23453691

ABSTRACT

Understanding the underlying molecular basis for disease can often be a prolonged and tortuous process with many false leads and blind alleys. Relating the cause of ATR-X syndrome to the function of the protein ATRX is a case in point. In this review we attempt to bring together the diverse biological phenomena associated with ATRX dysfunction with what has recently been discovered concerning the chromatin remodelling activity of this protein. This potentially casts light on how defective DNA replication/histone replacement can impact on transcription, telomere maintenance and also possibly chromosome segregation.


Subject(s)
DNA Helicases/genetics , DNA Replication/genetics , Mental Retardation, X-Linked/genetics , Nuclear Proteins/genetics , Telomere Homeostasis/genetics , alpha-Thalassemia/genetics , Chromatin Assembly and Disassembly/genetics , Chromosome Segregation , Histones/genetics , Histones/metabolism , Humans , Mental Retardation, X-Linked/etiology , Mental Retardation, X-Linked/pathology , Mutation , Transcription, Genetic , X-linked Nuclear Protein , alpha-Thalassemia/etiology , alpha-Thalassemia/pathology
9.
Cell Res ; 20(8): 908-18, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20548336

ABSTRACT

Recent studies have identified mutations in PHF8, an X-linked gene encoding a JmjC domain-containing protein, as a causal factor for X-linked mental retardation (XLMR) and cleft lip/cleft palate. However, the underlying mechanism is unknown. Here we show that PHF8 is a histone demethylase and coactivator for retinoic acid receptor (RAR). Although activities for both H3K4me3/2/1 and H3K9me2/1 demethylation were detected in cellular-based assays, recombinant PHF8 exhibited only H3K9me2/1 demethylase activity in vitro, suggesting that PHF8 is an H3K9me2/1 demethylase whose specificity may be modulated in vivo. Importantly, a mutant PHF8 (phenylalanine at position 279 to serine) identified in the XLMR patients is defective in enzymatic activity, indicating that the loss of histone demethylase activity is causally linked with the onset of disease. In addition, we show that PHF8 binds specifically to H3K4me3/2 peptides via an N-terminal PHD finger domain. Consistent with a role for PHF8 in neuronal differentiation, knockdown of PHF8 in mouse embryonic carcinoma P19 cells impairs RA-induced neuronal differentiation, whereas overexpression of the wild-type but not the F279S mutant PHF8 drives P19 cells toward neuronal differentiation. Furthermore, we show that PHF8 interacts with RARalpha and functions as a coactivator for RARalpha. Taken together, our results suggest that histone methylation modulated by PHF8 plays a critical role in neuronal differentiation.


Subject(s)
Histone Demethylases/metabolism , Mental Retardation, X-Linked/genetics , Neurons/enzymology , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Amino Acid Substitution , Animals , Cell Differentiation , Gene Knockdown Techniques , Histone Demethylases/genetics , Histones/metabolism , Humans , Mental Retardation, X-Linked/etiology , Mice , Mutagenesis, Site-Directed , Neurons/cytology , Neurons/metabolism , Protein Binding , Protein Structure, Tertiary , Transcription Factors/genetics
10.
Rev Neurol ; 47(12): 634-7, 2008.
Article in Spanish | MEDLINE | ID: mdl-19085879

ABSTRACT

INTRODUCTION: Mental retardation has an approximated prevalence of 2% in the general population and its most frequent cause is X-fragile syndrome. This genetic disorder predominantly affects males and it is mainly caused by the expansion of CGG in FMR1 gene. Recently has been demonstrated that mutations in a new called ARX gene (aristaless-related homeobox) can also cause a similar form of X linked mental retardation, as well as other neurological disorders (autism, Partington or West syndrome). The most frequent mutation that has been reported is the c.428_451 dup24, which comprises almost 60% of all described. It causes an expansion of a polyalanine tract of ARX protein. CASE REPORTS: We report three cases of mental retardation in two different families where the mutation in ARX gene c.428_451 dup24 was found while X-fragile syndrome screening was made. Personal and familiar history, phenotype and evolution are described. CONCLUSION: The molecular analysis of this mutation should be considered as a routine for the genetic diagnosis of mental retardation in males of nondrafted cause.


Subject(s)
Homeodomain Proteins/genetics , Mental Retardation, X-Linked/etiology , Mental Retardation, X-Linked/genetics , Mutation , Transcription Factors/genetics , Child, Preschool , Female , Fragile X Syndrome/genetics , Humans , Infant , Male , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/epidemiology , Pedigree , Spain/epidemiology
11.
Rev. neurol. (Ed. impr.) ; 47(12): 634-637, 16 dic., 2008. ilus
Article in Es | IBECS | ID: ibc-71829

ABSTRACT

Introducción. El retraso mental tiene una prevalencia aproximada del 2% en la población general, y la causa hereditaria más frecuente es el síndrome X frágil. Esta entidad afecta predominantemente a varones y está fundamentalmente causada por la expansión del triplete CGG en el gen FMR1. Recientemente, se ha demostrado que mutaciones en un nuevo gen llamado ARX (aristaless related homeobox) pueden ocasionar también una forma similar de retraso mental ligado al X, entreun amplio espectro de trastornos neurológicos relacionados (autismo, síndrome de Partington o síndrome de West, entre otros). La mutación más frecuentemente descrita, aproximadamente un 60% del total, es la duplicación de 24 pares de bases en el exón 2 (c.428_451 dup24), que produce una expansión de un tramo de polialanina en la proteína ARX. Casos clínicos.Se comunican tres casos de retraso mental no filiado, pertenecientes a dos familias distintas, en los que se halló la mutación en el gen ARX c.428_451 dup24 al realizar un estudio genético adicional al cribado de síndrome X frágil. Se describen los antecedentes personales y familiares, características fenotípicas y evolución de cada uno de ellos. Conclusión. El análisis molecular de dicha mutación debería considerarse de rutina para el diagnóstico genético de retraso mental en varones de causa no filiada


Introduction. Mental retardation has an approximated prevalence of 2% in the general population and its mostfrequent cause is X-fragile syndrome. This genetic disorder predominantly affects males and it is mainly caused by the expansion of CGG in FMR1 gene. Recently has been demonstrated that mutations in a new called ARX gene (aristalessrelated homeobox) can also cause a similar form of X linked mental retardation, as well as other neurological disorders (autism, Partington or West syndrome). The most frequent mutation that has been reported is the c.428_451 dup24, which comprises almost 60% of all described. It causes an expansion of a polyalanine tract of ARX protein. Case reports. We reportthree cases of mental retardation in two different families where the mutation in ARX gene c.428_451 dup24 was found while X-fragile syndrome screening was made. Personal and familiar history, phenotype and evolution are described. Conclusion. The molecular analysis of this mutation should be considered as a routine for the genetic diagnosis of mental retardation inmales of nondrafted cause


Subject(s)
Humans , Genes, Homeobox/genetics , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/etiology , Mutation/genetics , Autistic Disorder/genetics , Dystonia/genetics , Epilepsy/genetics , Fragile X Syndrome/genetics
12.
Clin Neurol Neurosurg ; 110(6): 631-4, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18462864

ABSTRACT

We present four patients with ARX mutations and widely variant clinical presentations. Case 1, a female with a known ARX mutation has refractory infantile spasms and severe mental retardation. Case 2, a male presented with a neurodegenerative disorder and has a known ARX mutation likely de novo as mother is not a carrier. Cases 3 and 4, two siblings with a novel variant in ARX, which is not clearly pathogenic, have developmental delay. One of the siblings had a diagnosis of autistic spectrum disorder, failure to thrive with severe feeding difficulties, intracranial hemorrhage, and seizures. There are very few affected females with ARX related infantile spasms. These cases expand the known phenotype of this emerging condition.


Subject(s)
Homeodomain Proteins/genetics , Mental Retardation, X-Linked/genetics , Spasms, Infantile/genetics , Transcription Factors/genetics , Autistic Disorder/etiology , Autistic Disorder/genetics , Child, Preschool , DNA/biosynthesis , DNA/genetics , Failure to Thrive/etiology , Failure to Thrive/genetics , Female , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Humans , Infant , Infant, Newborn , Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/genetics , Mental Retardation, X-Linked/etiology , Mutation/physiology , Seizures/etiology , Seizures/genetics , Spasms, Infantile/etiology
13.
Mol Genet Metab ; 92(1-2): 36-42, 2007.
Article in English | MEDLINE | ID: mdl-17618155

ABSTRACT

The HSD17B10 gene maps on chromosome Xp11.2, a region highly associated with X-linked mental retardation. This gene encodes HSD10, a mitochondrial multifunctional enzyme that plays a significant part in the metabolism of neuroactive steroids and the degradation of isoleucine. The HSD17B10 gene is composed of six exons and five introns. Its exon 5 is an alternative exon such that there are several HSD17B10 mRNA isoforms in brain. A silent mutation (c.605C-->A) and three missense mutations (c.395C-->G; c.419C-->T; c.771A-->G), respectively, cause the X-linked mental retardation, choreoathetosis, and abnormal behavior (MRXS10) and the hydroxyacyl-CoA dehydrogenase II deficiency. The latter condition seems to be a multifactorial disease due to the disturbance of more than one metabolic pathway by the HSD10 deficiency. HSD10 inactivates the positive modulators of GABAA receptors, and plays a role in the maintenance of GABAergic neuronal function. This working model may account for the mental retardation of these patients. The dehydrogenase activity is slightly inhibited by the binding of amyloid-beta peptide to the loop D of HSD10. Elevated levels of HSD10 were observed in hippocampi of Alzheimer disease patients so this multifunctional enzyme may be related to Alzheimer disease pathogenesis; however, the molecular mechanism of its involvement remains to be ascertained.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/physiology , Cognition Disorders/enzymology , Isoleucine/metabolism , Neurotransmitter Agents/metabolism , Steroids/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/genetics , Amino Acid Sequence , Humans , Mental Retardation, X-Linked/enzymology , Mental Retardation, X-Linked/etiology , Molecular Sequence Data , Sequence Homology, Amino Acid
15.
Am J Hum Genet ; 79(6): 1119-24, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17186471

ABSTRACT

In a systematic sequencing screen of the coding exons of the X chromosome in 250 families with X-linked mental retardation (XLMR), we identified two nonsense mutations and one consensus splice-site mutation in the AP1S2 gene on Xp22 in three families. Affected individuals in these families showed mild-to-profound mental retardation. Other features included hypotonia early in life and delay in walking. AP1S2 encodes an adaptin protein that constitutes part of the adaptor protein complex found at the cytoplasmic face of coated vesicles located at the Golgi complex. The complex mediates the recruitment of clathrin to the vesicle membrane. Aberrant endocytic processing through disruption of adaptor protein complexes is likely to result from the AP1S2 mutations identified in the three XLMR-affected families, and such defects may plausibly cause abnormal synaptic development and function. AP1S2 is the first reported XLMR gene that encodes a protein directly involved in the assembly of endocytic vesicles.


Subject(s)
Adaptor Protein Complex sigma Subunits/genetics , Mental Retardation, X-Linked/genetics , Mutation , Adaptor Protein Complex sigma Subunits/metabolism , Adult , Child , Endosomes/metabolism , Female , Humans , Male , Mental Retardation, X-Linked/etiology , Mental Retardation, X-Linked/psychology , Pedigree
17.
Nat Genet ; 35(4): 313-5, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14634649

ABSTRACT

We found mutations in the gene PQBP1 in 5 of 29 families with nonsyndromic (MRX) and syndromic (MRXS) forms of X-linked mental retardation (XLMR). Clinical features in affected males include mental retardation, microcephaly, short stature, spastic paraplegia and midline defects. PQBP1 has previously been implicated in the pathogenesis of polyglutamine expansion diseases. Our findings link this gene to XLMR and shed more light on the pathogenesis of this common disorder.


Subject(s)
Mental Retardation, X-Linked/genetics , Mutation/genetics , Oligopeptides/genetics , Carrier Proteins/genetics , DNA-Binding Proteins , Female , Genetic Linkage , Humans , Male , Mental Retardation, X-Linked/classification , Mental Retardation, X-Linked/etiology , Molecular Sequence Data , Nuclear Proteins/genetics , Pedigree , Syndrome
18.
J Med Genet ; 40(1): 11-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12525535

ABSTRACT

BACKGROUND: The gene encoding fatty acid CoA ligase 4 (FACL4) is mutated in families with non-specific X linked mental retardation (MRX) and is responsible for cognitive impairment in the contiguous gene syndrome ATS-MR (Alport syndrome and mental retardation), mapped to Xq22.3. This finding makes this gene a good candidate for other mental retardation disorders mapping in this region. METHODS: We have screened the FACL4 gene in eight families, two MRX and six syndromic X linked mental retardation (MRXS), mapping in a large interval encompassing Xq22.3. RESULTS: We have found a missense mutation in MRX68. The mutation (c.1001C>T in the brain isoform) cosegregates with the disease and changes a highly conserved proline into a leucine (p.P375L) in the first luciferase domain, which markedly reduces the enzymatic activity. Furthermore, all heterozygous females showed completely skewed X inactivation in blood leucocytes, as happens in all reported females with other FACL4 point mutations or deletions. CONCLUSIONS: Since the FACL4 gene is highly expressed in brain, where it encodes a brain specific isoform, and is located in hippocampal and cerebellar neurones, a role for this gene in cognitive processes can be expected. Here we report the third MRX family with a FACL4 mutation and describe the development of a rapid enzymatic assay on peripheral blood that we propose as a sensitive, robust, and efficient diagnostic tool in mentally retarded males.


Subject(s)
Coenzyme A Ligases/genetics , Genetic Testing/methods , Mental Retardation, X-Linked/enzymology , Mental Retardation, X-Linked/genetics , Mutation, Missense/genetics , Repressor Proteins , Saccharomyces cerevisiae Proteins , Adolescent , Adult , Amino Acid Substitution/genetics , Cell Extracts/chemistry , Cell Line , Child , Chromosomes, Human, X/genetics , Coenzyme A Ligases/blood , Female , Genetic Carrier Screening/methods , Humans , Infant , Leucine/genetics , Lymphocytes/chemistry , Male , Mental Retardation, X-Linked/blood , Mental Retardation, X-Linked/etiology , Middle Aged , Molecular Diagnostic Techniques/methods , Pedigree , Proline/genetics , Sex Chromosome Aberrations
19.
Clin Genet ; 62(6): 423-32, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12485186

ABSTRACT

This review covers the history and nosology of X-linked mental retardation (XLMR) in which the following, largely clinically based, subclassification was used: fragile X syndrome (FRAXA), syndromic forms (MRXS) and non-specific forms (MRX). After the discovery of the FMR2 gene at the FRAXE site, 10 MRX genes have been identified in the last 6 years. A short description is given of the strategies used to identify the genes that cause mental retardation (MR). Furthermore, their potential functions and the association with MR will be discussed. It is emphasized that mutations in several of these MR genes can result in non-specific, as well as in syndromic forms of XLMR. Present findings stress the importance of accurate clinical evaluation. Most considerably, genotype-phenotype correlation studies of affected individuals in XLMR families with MRX gene mutations are necessary to define the criteria of MRX vs MRXS subclassification.


Subject(s)
Mental Retardation, X-Linked/classification , Mental Retardation, X-Linked/genetics , Animals , Family Health , Humans , Mental Retardation, X-Linked/etiology , Mutation , Syndrome
20.
Rev. cuba. pediatr ; 45(3): 333-336, may.-jun.1973. tab
Article in Spanish | CUMED | ID: cum-25621

ABSTRACT

Se estudia el rendimiento intelectual en 26 pacientes no hospitalizados portadores del síndrome de Turner, atendidos en el Instituto de Endocrinología y Enfermedades Metabólicas, encontrándose una alta incidencia de deficiencia mental (50 por ciento). Se relaciona el índice de rendimiento intelectual con el cariotipo de los pacientes y se comentan los resultados(AU)


Subject(s)
Humans , Female , Child , Turner Syndrome/complications , Mental Retardation, X-Linked/epidemiology , Mental Retardation, X-Linked/etiology , Chromosome Aberrations , Underachievement
SELECTION OF CITATIONS
SEARCH DETAIL
...