Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Turk Psikiyatri Derg ; 31(3): 216-220, 2020.
Article in English, Turkish | MEDLINE | ID: mdl-32978958

ABSTRACT

Lujan-Fryns Syndrome (LFS) is defined as a set of symptoms including mild-moderate mental retardation, marfanoid appearance, hypotonia at birth, hypernasal speech, characteristic craniofacial appearance and normal testis size. The frequency of the syndrome is not known thus the information obtained is solely based on case reports. Hereby, we present a patient with LFS diagnosis. The 29-year old male patient had mental retardation, aggression, and persecutory delusions, characteristic craniofacial and marfanoid features. During his speech pronominal reversal was observed ('the hurt him, he is so upset' when talking abour himself). After examination and genetic analysis, fragile X, Klinefelter, Marfan and Down syndromes and homocystinuria were eliminated as causes of mental retardation. A preliminary diagnoses of LFS done. No mutation was detected in exon 22 of the MED12 gene; but. Whole Exome Sequencing (WES) is ongoing. The patient was started on risperidone (4 mg/day) for psychotic symptoms and carbamazepine (200 mg/day) for impulse control and as an antiepileptic. After a follow up of 8 months, impulse control, psychotic symptoms and aggression improved significantly. Since the specific gene mutation of LFS was not determined in our case, we solely had to depend on clinical evaluation and genetic analysis. Although it is not easy to fully define or classify these syndromes, we believe every reported case will be a step in overcoming these difficulties.


Subject(s)
Autistic Disorder/diagnosis , Craniofacial Abnormalities/diagnosis , Marfan Syndrome/diagnosis , Mental Retardation, X-Linked/diagnosis , Psychotic Disorders/diagnosis , Adult , Autistic Disorder/complications , Autistic Disorder/psychology , Craniofacial Abnormalities/complications , Craniofacial Abnormalities/psychology , Diagnosis, Differential , Humans , Male , Marfan Syndrome/complications , Marfan Syndrome/psychology , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/psychology , Psychotic Disorders/complications , Psychotic Disorders/psychology
2.
Brain Behav ; 9(3): e01221, 2019 03.
Article in English | MEDLINE | ID: mdl-30714330

ABSTRACT

BACKGROUND: Pettigrew syndrome (PGS) is a rare X-linked mental retardation that caused by AP1S2 mutation. The pathogenesis of AP1S2 deficiency has remained elusive. The purpose of this study is to give a comprehensive overview of the phenotypic and genetic spectrum of AP1S2 mutations. METHODS: This study systematically analyzed clinical features and genetic information of a Chinese family with AP1S2 variation, and reviewed previously reported literatures with the same gene variation. RESULTS: We identified a new c.1-1 G>C mutation in AP1S2 gene from a four generation family with seven affected individuals and found the elevated neuron-specific enolase (NSE) in a patient. We summarized the clinical manifestation of 59 patients with AP1S2 mutation. We found that pathogenic point mutations affecting AP1S2 are associated with dysmorphic features and neurodevelopmental problems, which included highly variable mental retardation (MR), delayed in walking, abnormal speech, hypotonia, abnormal brain, abnormal behavior including aggressive behavior, ASD, self-abusive, and abnormal gait. Patients with splice site mutation were more likely to lead to seizures. By contrast, patients with nonsense mutations are more susceptible to microcephaly. CONCLUSION: Our findings suggest AP1S2 mutations contribute to a broad spectrum of neurodevelopmental disorders and are important in the etiological spectrum of PGS.


Subject(s)
Adaptor Protein Complex sigma Subunits/genetics , Basal Ganglia Diseases , Body Dysmorphic Disorders , Dandy-Walker Syndrome , Mental Retardation, X-Linked , Neurodevelopmental Disorders , Seizures , Adult , Basal Ganglia Diseases/complications , Basal Ganglia Diseases/genetics , Basal Ganglia Diseases/psychology , Body Dysmorphic Disorders/etiology , Body Dysmorphic Disorders/genetics , Child , Child, Preschool , Dandy-Walker Syndrome/complications , Dandy-Walker Syndrome/genetics , Dandy-Walker Syndrome/psychology , Female , Humans , Male , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/psychology , Mutation , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/genetics , Pedigree , Phosphopyruvate Hydratase/metabolism , Seizures/complications , Seizures/genetics , Seizures/psychology
3.
ACS Chem Neurosci ; 9(9): 2210-2217, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29227625

ABSTRACT

Rett syndrome and MECP2 Duplication syndrome are neurodevelopmental disorders attributed to loss-of-function mutations in, or duplication of, the gene encoding methyl-CpG-binding protein 2 (MeCP2), respectively. We recently reported decreased expression and function of the metabotropic glutamate receptor 7 (mGlu7) in a mouse model of Rett syndrome. Positive allosteric modulation of mGlu7 activity was sufficient to improve several disease phenotypes including cognition. Here, we tested the hypothesis that mGlu7 expression would be reciprocally regulated in a mouse model of MECP2 Duplication syndrome, such that negative modulation of mGlu7 activity would exert therapeutic benefit. To the contrary, we report that mGlu7 is not functionally increased in mice overexpressing MeCP2 and that neither genetic nor pharmacological reduction of mGlu7 activity impacts phenotypes that are antiparallel to those observed in Rett syndrome model mice. These data expand our understanding of how mGlu7 expression and function is affected by changes in MeCP2 dosage and have important implications for the therapeutic development of mGlu7 modulators.


Subject(s)
Anxiety/metabolism , Fear/physiology , Learning/physiology , Mental Retardation, X-Linked/genetics , Receptors, Metabotropic Glutamate/genetics , Allosteric Regulation , Animals , Anxiety/physiopathology , Anxiety/psychology , Benzoxazoles/pharmacology , Disease Models, Animal , Excitatory Amino Acid Agonists/pharmacology , Fear/drug effects , Fear/psychology , Learning/drug effects , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/psychology , Mice , Phenotype , Phosphinic Acids/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism
4.
Am J Med Genet A ; 173(5): 1358-1363, 2017 May.
Article in English | MEDLINE | ID: mdl-28345801

ABSTRACT

ZC4H2 is involved in human brain development, and, if mutated, can be responsible for a rare X-linked disorder, originally presented in literature as Wieacker-Wolff syndrome and Miles-Carpenter syndrome. In males, severe intellectual disability is associated with variable symptoms of central and peripheral nervous system involvement, such as spasticity, hyperreflexia, muscle weakness, and arthrogryposis. Female carriers are usually described as asymptomatic or only mildly affected. Here, we report on a girl carrying a de novo deletion of ZC4H2 detected by array-CGH analysis. She showed a complex neurodevelopmental disorder resembling the clinical picture commonly observed in male patients. X-inactivation was found to be random. Additionally, she had an unusual appearance of fingers and hand creases, and electromyography showed a peculiar pattern of both neurogenic and myopathic anomalies. The present patient confirms that female carriers can also be severely affected. Systematic clinical investigations of both males and females are needed to define the variety in nature and severity of phenotypes related to ZC4H2 variants.


Subject(s)
Apraxias/genetics , Carrier Proteins/genetics , Contracture/genetics , Genetic Diseases, X-Linked/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/psychology , Muscular Atrophy/genetics , Ophthalmoplegia/genetics , Adolescent , Apraxias/diagnosis , Apraxias/physiopathology , Child , Comparative Genomic Hybridization , Contracture/diagnosis , Contracture/physiopathology , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/physiopathology , Humans , Intellectual Disability/diagnosis , Intellectual Disability/physiopathology , Intracellular Signaling Peptides and Proteins , Male , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/physiopathology , Muscular Atrophy/diagnosis , Muscular Atrophy/physiopathology , Nuclear Proteins , Ophthalmoplegia/diagnosis , Ophthalmoplegia/physiopathology , Sequence Deletion
5.
Pediatr Neurol ; 67: 45-52, 2017 02.
Article in English | MEDLINE | ID: mdl-28065824

ABSTRACT

BACKGROUND: The X-linked creatine transporter deficiency (CRTD) caused by an SLC6A8 mutation represents the second most common cause of X-linked intellectual disability. The clinical phenotype ranges from mild to severe intellectual disability, epilepsy, short stature, poor language skills, and autism spectrum disorders. The objective of this study was to investigate phenotypic variability in the context of genotype, cerebral creatine concentration, and volumetric analysis in a family with CRTD. PATIENTS AND METHODS: The clinical phenotype and manifestations of epilepsy were assessed in a Caucasian family with CRTD. DNA sequencing and creatine metabolism analysis confirmed the diagnosis. Cerebral magnetic resonance imaging (cMRI) with voxel-based morphometry and magnetic resonance spectroscopy was performed in all family members. RESULTS: An SLC6A8 missense mutation (c.1169C>T; p.Pro390Leu, exon 8) was detected in four of five individuals. Both male siblings were hemizygous, the mother and the affected sister heterozygous for the mutation. Structural cMRI was normal, whereas voxel-based morphometry analysis showed reduced white matter volume below the first percentile of the reference population of 290 subjects in the more severely affected boy compared with family members and controls. Normalized creatine concentration differed significantly between the individuals (P < 0.005). CONCLUSIONS: There is a broad phenotypic variability in CRTD even in family members with the same mutation. Differences in mental development could be related to atrophy of the subcortical white matter.


Subject(s)
Brain Diseases, Metabolic, Inborn/diagnostic imaging , Brain Diseases, Metabolic, Inborn/genetics , Creatine/deficiency , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Mental Retardation, X-Linked/diagnostic imaging , Mental Retardation, X-Linked/genetics , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , White Matter/diagnostic imaging , Adolescent , Atrophy/blood , Atrophy/diagnostic imaging , Atrophy/genetics , Atrophy/psychology , Brain Diseases, Metabolic, Inborn/blood , Brain Diseases, Metabolic, Inborn/psychology , Child , Creatine/blood , Creatine/genetics , Female , Genotype , Humans , Intellectual Disability/blood , Intellectual Disability/psychology , Male , Mental Retardation, X-Linked/blood , Mental Retardation, X-Linked/psychology , Middle Aged , Mutation, Missense , Phenotype , Plasma Membrane Neurotransmitter Transport Proteins/blood , Plasma Membrane Neurotransmitter Transport Proteins/genetics , White Matter/metabolism
6.
J Child Neurol ; 31(2): 159-63, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25999300

ABSTRACT

MECP2 duplication syndrome is an X-linked genomic disorder that is characterized by infantile hypotonia, intellectual disability, and recurrent respiratory infections. Regression affects a subset of individuals, and the etiology of regression has yet to be examined. In this study, alterations in the hypothalamus-pituitary-adrenal axis, including diurnal patterns in salivary cortisol, were examined in 4 males with MECP2 duplication syndrome who had regression and 4 males with the same syndrome without regression (aged 3-22 years). Individuals who had experienced regression do not exhibit typical diurnal cortisol rhythms, and their profiles were flatter through the day. In contrast, individuals with MECP2 duplication syndrome who had not experienced regression showed more typical patterns of higher cortisol levels in the morning with linear decreases throughout the day. This study is the first to suggest a link between atypical diurnal cortisol rhythms and regression status in MECP2 duplication syndrome and may have implications for treatment.


Subject(s)
Circadian Rhythm/physiology , Hydrocortisone/metabolism , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/psychology , Saliva/metabolism , Child , Child, Preschool , Humans , Male , Young Adult
7.
Pediatr Neurol ; 53(4): 360-363.e2, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26205312

ABSTRACT

BACKGROUND: Creatine transporter (SLC6A8) deficiency is an X-linked inborn error of metabolism characterized by cerebral creatine deficiency, behavioral problems, seizures, hypotonia, and intellectual developmental disability. A third of patients are amenable to treatment with high-dose oral creatine, glycine, and L-arginine supplementation. METHODS: Given the limited treatment response, we initiated an open-label observational study to evaluate the effect of adjunct S-adenosyl methionine to further enhance intracerebral creatine synthesis. RESULTS: Significant and reproducible issues with sleep and behavior were noted in both male patients on a dose of 50/mg/kg. One of the two patients stopped S-adenosyl methionine and did not come for any follow-up. A safe and tolerable dose (17 mg/kg/day) was identified in the other patient. On magnetic resonance spectroscopy, this 8-year-old male did not show an increase in intracerebral creatine. However, significant improvement in speech/language skills, muscle mass were observed as well as in personal outcomes as defined by the family in activities related to communication and decision making. DISCUSSION: Further research is needed to assess the potential of S-adenosyl methionine as an adjunctive therapy for creatine transporter deficiency patients and to define the optimal dose. Our study also illustrates the importance of pathophysiology-based treatment, individualized outcome assessment, and patient/family participation in rare diseases research.


Subject(s)
Arginine/administration & dosage , Brain Diseases, Metabolic, Inborn/drug therapy , Central Nervous System Agents/administration & dosage , Creatine/administration & dosage , Creatine/deficiency , Glycine/administration & dosage , Mental Retardation, X-Linked/drug therapy , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , S-Adenosylmethionine/administration & dosage , Administration, Oral , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Brain Diseases, Metabolic, Inborn/physiopathology , Brain Diseases, Metabolic, Inborn/psychology , Child , Drug Therapy, Combination , Follow-Up Studies , Humans , Magnetic Resonance Spectroscopy , Male , Medication Adherence , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/psychology , Treatment Outcome
8.
Horm Res Paediatr ; 82(4): 261-71, 2014.
Article in English | MEDLINE | ID: mdl-25247785

ABSTRACT

BACKGROUND/AIMS: Monocarboxylate transporter 8 (MCT8) is essential for thyroid hormone (TH) transport in the brain. Mutations in MCT8 are associated with the Allan-Herndon-Dudley syndrome (AHDS), characterized by severe psychomotor retardation and altered serum thyroid parameters. Here we report two novel mutations in MCT8 and discuss the clinical findings. CASE REPORT AND RESULTS: We describe 4 males with AHDS from two unrelated families varying in age from 1.5 to 11 years. All 4 patients presented with typical clinical signs of AHDS, including severe psychomotor retardation, axial hypotonia, lack of speech, diminished muscle mass, increased muscle tone, hyperreflexia, myopathic facies, high T3, low T4 and rT3, and normal/mildly elevated TSH levels. Comparison of patients at different ages suggests the progressive nature of AHDS. Genetic analyses identified a novel missense MCT8 mutation (p.G495A) in family 1 and a 2.8-kb deletion comprising exons 3 and 4 in family 2. Functional analysis of p.G495A revealed impaired TH transport varying from 20 to 85% depending on the cell context. CONCLUSION: Here we report 4 AHDS patients in unrelated Turkish families harboring novel MCT8 mutations. Despite the widely different mutations, the clinical phenotypes are very similar and findings support the progressive nature of AHDS.


Subject(s)
Mental Retardation, X-Linked/genetics , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/genetics , Muscular Atrophy/genetics , Child , Child, Preschool , DNA Mutational Analysis , Gene Deletion , Humans , Infant , Intellectual Disability/etiology , Intellectual Disability/genetics , Male , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/psychology , Monocarboxylic Acid Transporters/metabolism , Muscle Hypotonia/metabolism , Muscle Hypotonia/psychology , Muscular Atrophy/metabolism , Muscular Atrophy/psychology , Mutation/genetics , Mutation, Missense/genetics , Pedigree , Symporters , Thyroid Hormones/blood
9.
J Autism Dev Disord ; 43(10): 2484-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23456562

ABSTRACT

The aim of this study was to determine the frequency, timing, and associated features of developmental regression in MECP2 duplication syndrome. We also examined whether duplication size was associated with regression. Comprehensive psychological evaluations were used to assess 17 boys with MECP2 duplication syndrome. Information about regression was gathered via parent report. Eight of 17 boys exhibited regression in language skills, while seven of 17 exhibited regression in other skill areas. Regression in "other skill" areas coincided with seizure onset and with a prior autism diagnosis in six of seven participants. Regression was not associated with duplication size. Questions remain as to why some boys regress, and future work is necessary to understand the underlying mechanism(s) that causes regression.


Subject(s)
Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/psychology , Regression, Psychology , Child , Child, Preschool , Humans , Language , Male , Mental Retardation, X-Linked/genetics , Methyl-CpG-Binding Protein 2/genetics , Motor Skills , Seizures/physiopathology , Time Factors
10.
Brain Dev ; 35(2): 172-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22541666

ABSTRACT

Mutations in the SLC9A6 gene cause Christianson syndrome in boys. This X-linked syndrome is characterized by profound mental retardation with autistic behavior, microcephaly, epilepsy, ophthalmoplegia, and ataxia. Progressive cerebellar atrophy with motor regression is a remarkable feature in some patients. We report on a 22year-old male patient with Christianson syndrome carrying the novel p.Gln306X mutation. The infantile phenotype suggested pervasive developmental disorder, then profound mental retardation ensued. In later childhood, progressive cerebellar atrophy was diagnosed on serial brain MRIs and motor regression occurred. Furthermore, ophthalmological evaluations showed a retinitis pigmentosum previously unreported in this condition. We conclude that the natural history of the disease in this patient tends to confirm the degenerative nature of Christianson syndrome, and that retinal degeneration may be part of the condition. Before the onset of degeneration, the syndromic association of severe mental retardation, autistic behavior, external ophthalmoplegia, and facial dysmorphism in male patients is a clue to the diagnosis.


Subject(s)
Mental Retardation, X-Linked/genetics , Mutation/physiology , Retinitis Pigmentosa/genetics , Sodium-Hydrogen Exchangers/genetics , Ataxia/etiology , Atrophy , Cerebellar Diseases/genetics , Codon, Nonsense/genetics , DNA Mutational Analysis , Disease Progression , Electroencephalography , Humans , Magnetic Resonance Imaging , Male , Mental Retardation, X-Linked/psychology , Mutation/genetics , Retinal Degeneration/etiology , Retinal Degeneration/pathology , Retinitis Pigmentosa/psychology , Syndrome , Young Adult
11.
Autism Res ; 6(1): 42-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23169761

ABSTRACT

Alterations in the X-linked gene MECP2 encoding the methyl-CpG-binding protein 2 have been linked to autism spectrum disorders (ASDs). Most recently, data suggest that overexpression of MECP2 may be related to ASD. To better characterize the relevance of MECP2 overexpression to ASD-related behaviors, we compared the core symptoms of ASD in MECP2 duplication syndrome to nonverbal mental age-matched boys with idiopathic ASD. Within the MECP2 duplication group, we further delineated aspects of the behavioral phenotype and also examined how duplication size and gene content corresponded to clinical severity. We compared ten males with MECP2 duplication syndrome (ages 3-10) with a chronological and mental age-matched sample of nine nonverbal males with idiopathic ASD. Our results indicate that boys with MECP2 duplication syndrome share the core behavioral features of ASD (e.g. social affect, restricted/repetitive behaviors). Direct comparisons of ASD profiles revealed that a majority of boys with MECP2 duplication syndrome are similar to idiopathic ASD; they have impairments in social affect (albeit to a lesser degree than idiopathic ASD) and similar severity in restricted/repetitive behaviors. Nonverbal mental age did not correlate with severity of social impairment or repetitive behaviors. Within the MECP2 duplication group, breakpoint size does not predict differences in clinical severity. In addition to social withdrawal and stereotyped behaviors, we also found that hyposensitivity to pain/temperature are part of the behavioral phenotype of MECP2 duplication syndrome. Our results illustrate that overexpression/increased dosage of MECP2 is related to core features of ASD.


Subject(s)
Autistic Disorder/psychology , Child Behavior/psychology , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/psychology , Methyl-CpG-Binding Protein 2/genetics , Autistic Disorder/genetics , Child , Child, Preschool , Gene Duplication/genetics , Genetic Predisposition to Disease/genetics , Humans , Male , Phenotype
12.
J Clin Invest ; 122(8): 2837-46, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22751104

ABSTRACT

The second-largest cause of X-linked mental retardation is a deficiency in creatine transporter (CRT; encoded by SLC6A8), which leads to speech and language disorders with severe cognitive impairment. This syndrome, caused by the absence of creatine in the brain, is currently untreatable because CRT is required for creatine entry into brain cells. Here, we developed a brain-specific Slc6a8 knockout mouse (Slc6a8-/y) as an animal model of human CRT deficiency in order to explore potential therapies for this syndrome. The phenotype of the Slc6a8-/y mouse was comparable to that of human patients. We successfully treated the Slc6a8-/y mice with the creatine analog cyclocreatine. Brain cyclocreatine and cyclocreatine phosphate were detected after 9 weeks of cyclocreatine treatment in Slc6a8-/y mice, in contrast to the same mice treated with creatine or placebo. Cyclocreatine-treated Slc6a8-/y mice also exhibited a profound improvement in cognitive abilities, as seen with novel object recognition as well as spatial learning and memory tests. Thus, cyclocreatine appears promising as a potential therapy for CRT deficiency.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Cognition/drug effects , Creatinine/analogs & derivatives , Membrane Transport Proteins/deficiency , Animals , Base Sequence , Brain/metabolism , Cognition Disorders/genetics , Cognition Disorders/psychology , Creatinine/metabolism , Creatinine/pharmacology , DNA Primers/genetics , Disease Models, Animal , Female , Humans , Imidazolidines/metabolism , Learning/drug effects , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , Memory/drug effects , Mental Retardation, X-Linked/drug therapy , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Phosphocreatine/analogs & derivatives , Phosphocreatine/metabolism
13.
J Inherit Metab Dis ; 33 Suppl 3: S5-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-24137762

ABSTRACT

The urinary creatine:creatinine (Cr:Crn) ratio was measured in males from 49 families with a family history compatible with X-linked mental retardation (XLMR) in order to estimate the prevalence of SLC6A8 deficiency in Estonia. We identified 11 boys from 9 families with an increased urinary Cr:Crn ratio (18%). In three related boys, a hemizygous missense mutation (c.1271G>A; p.Gly424Asp) was identified. Their mother was heterozygous for the same mutation. Although many missense mutations have been described, the p.Gly424Asp mutation has not been previously reported. The clinical expression varied widely among affected males of this family. Patients 1 and 3 had relatively mild clinical expression (mild mental retardation (MR) and attention deficit disorder), but patient 2 had all typical clinical signs of SLC6A8 defect such as moderate MR, autistic features, expressive dysphasia and epilepsy. Among our patients, we saw significant problems in speech and language development combined with attention and behavioural difficulties. The number of false-positive biochemical results with increased urinary Cr:Crn ratio was higher (18%) in our study than in previous reports (1.8­10%). We therefore suggest that repeated biochemical testing should be performed before DNA sequencing analysis. Our study suggests that 2% (95% confidence limits: 0.05­11.1%) of this Estonian XLMR panel are due to mutations in the SLC6A8, which is similar to the prevalence reported in other populations. We therefore conclude that creatine transporter deficiency is a relatively common genetic disorder in males with sporadic or familiar MR and diagnostic screening of them should always include screening for SLC6A8 deficiency.


Subject(s)
Brain Diseases, Metabolic, Inborn/diagnosis , Creatine/deficiency , DNA Mutational Analysis , Genetic Testing/methods , Mental Retardation, X-Linked/diagnosis , Mutation, Missense , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Adolescent , Adult , Biomarkers/urine , Brain Diseases, Metabolic, Inborn/epidemiology , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/psychology , Brain Diseases, Metabolic, Inborn/urine , Child , Creatine/genetics , Creatine/urine , Creatinine/urine , Estonia/epidemiology , Female , Genetic Predisposition to Disease , Heredity , Heterozygote , Humans , Intelligence/genetics , Male , Mental Retardation, X-Linked/epidemiology , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/psychology , Mental Retardation, X-Linked/urine , Middle Aged , Nerve Tissue Proteins/deficiency , Pedigree , Persons with Mental Disabilities , Phenotype , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/urine , Predictive Value of Tests , Prevalence , Severity of Illness Index , Young Adult
14.
Hum Mol Genet ; 18(20): 3894-905, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19617635

ABSTRACT

Long-chain acyl-CoA synthetases (ACSLs) convert long-chain fatty acids to acyl-CoAs, the activated substrates essential in various metabolic and signaling pathways. Mutations in ACSL4 are associated with non-syndromic X-linked mental retardation (MRX). However, the developmental functions of ACSL4 and how it is involved in the pathogenesis of MRX remain largely unknown. The Drosophila ACSL-like protein is highly homologous to human ACSL3 and ACSL4, and we designate it as dAcsl. In this study, we demonstrate that dAcsl and ACSL4 are highly conserved in terms of ACSL4's ability to substitute the functions of dAcsl in organismal viability, lipid storage and the neural wiring in visual center. In neurodevelopment, decapentaplegic (Dpp, a BMP-like molecule) production diminished specifically in the larval brain of dAcsl mutants. Consistent with the Dpp reduction, the number of glial cells and neurons dramatically decreased and the retinal axons mis-targeted in the visual cortex. All these defects in Drosophila brain were rescued by the wild-type ACSL4 but not by the mutant products found in MRX patients. Interestingly, expression of an MRX-associated ACSL4 mutant form in a wild-type background led to the lesions in visual center, suggesting a dominant negative effect. These findings validate Drosophila as a model system to reveal the connection between ACSL4 and BMP pathway in neurodevelopment, and to infer the pathogenesis of ACSL4-related MRX.


Subject(s)
Coenzyme A Ligases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Mental Retardation, X-Linked/enzymology , Visual Perception , Amino Acid Sequence , Animals , Brain/enzymology , Brain/metabolism , Coenzyme A Ligases/chemistry , Coenzyme A Ligases/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/psychology , Molecular Sequence Data , Optic Lobe, Nonmammalian/enzymology , Optic Lobe, Nonmammalian/metabolism , Sequence Alignment
15.
Hum Mol Genet ; 18(1): 105-17, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18829665

ABSTRACT

The GDI1 gene, responsible in human for X-linked non-specific mental retardation, encodes alphaGDI, a regulatory protein common to all GTPases of the Rab family. Its alteration, leading to membrane accumulation of different Rab GTPases, may affect multiple steps in neuronal intracellular traffic. Using electron microscopy and electrophysiology, we now report that lack of alphaGDI impairs several steps in synaptic vesicle (SV) biogenesis and recycling in the hippocampus. Alteration of the SV reserve pool (RP) and a 50% reduction in the total number of SV in adult synapses may be dependent on a defective endosomal-dependent recycling and may lead to the observed alterations in short-term plasticity. As predicted by the synaptic characteristics of the mutant mice, the short-term memory deficit, observed when using fear-conditioning protocols with short intervals between trials, disappeared when the Gdi1 mutants were allowed to have longer intervals between sessions. Likewise, previously observed deficits in radial maze learning could be corrected by providing less challenging pre-training. This implies that an intact RP of SVs is necessary for memory processing under challenging conditions in mice. The possibility to correct the learning deficit in mice may have clinical implication for future studies in human.


Subject(s)
Cognition , Guanine Nucleotide Dissociation Inhibitors/genetics , Learning , Mental Retardation, X-Linked/metabolism , Neuronal Plasticity , Synaptic Vesicles/physiology , Animals , Female , Guanine Nucleotide Dissociation Inhibitors/metabolism , Hippocampus/metabolism , Male , Memory , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Synapses/genetics , Synapses/metabolism , Synaptic Vesicles/genetics
16.
Am J Med Genet A ; 146A(23): 3011-7, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18973276

ABSTRACT

Opitz and Kaveggia [Opitz and Kaveggia (1974); Z Kinderheilk 117:1-18] reported on a family of five affected males with distinctive facial appearance, mental retardation, macrocephaly, imperforate anus and hypotonia. Risheg et al. [Risheg et al. (2007); Nat Genet 39:451-453] identified an identical mutation (p.R961W) in MED12 in six families with Opitz-Kaveggia syndrome, including a surviving affected man from the family reported in 1974. The previously defined behavior phenotype of hyperactivity, affability, and excessive talkativeness is very frequent in young boys with this mutation, along with socially oriented, attention-seeking behaviors. We present case studies of two older males with FG syndrome and the p.R961W mutation to illustrate how their behavior changes with age. We also characterize the behavior of eight additional individuals with FG syndrome and this recurrent mutation in MED12 using the Vineland Adaptive Behavior Scales 2nd edition, the Reiss Profile of Fundamental Goals and Motivation Sensitivities, and the Achenbach Child Behavior Checklist. Males with this MED12 mutation had deficits in communication skills compared to their socialization and daily living skills. In addition, they were at increased risk for maladaptive behavior, with a propensity towards aggression, anxiety, and inattention. Based on the behavior phenotype in 10 males with this recurrent MED12 mutation, we offer specific recommendations and interventional strategies. Our findings reinforce the importance of testing for the p.R961W MED12 mutation in males who are suspected of having developmental and behavioral problems with a clinical phenotype that is consistent with FG syndrome.


Subject(s)
Abnormalities, Multiple/psychology , Child Behavior Disorders/diagnosis , Mental Retardation, X-Linked/psychology , Receptors, Thyroid Hormone/genetics , Social Behavior Disorders/diagnosis , Abnormalities, Multiple/genetics , Adolescent , Adult , Amino Acid Substitution , Arginine/genetics , Arginine/metabolism , Child , Child Behavior Disorders/etiology , Child Behavior Disorders/genetics , Communication , Humans , Male , Mediator Complex , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/genetics , Social Behavior Disorders/etiology , Social Behavior Disorders/genetics , Socialization , Syndrome , Tryptophan/genetics , Tryptophan/metabolism , Young Adult
17.
J Med Genet ; 45(2): 100-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17893116

ABSTRACT

INTRODUCTION: We identified a female patient with mental retardation and sensory hyperarousal. She has a de novo paracentric inversion of one X chromosome with completely skewed inactivation of the normal X chromosome. OBJECTIVE: We aimed to identify whether a single gene or gene region caused her cognitive and behavioural impairment and that of others. RESULTS: Fluorescent in situ hybridisation (FISH) showed that the centromeric breakpoint disrupts a single gene: ARHGEF9 (CDC42 guanine nucleotide exchange factor (GEF) 9). The telomeric break lies in a gene poor region. We also found that the levels of the ARHGEF9 transcript from the patient are 10-fold less than those found in control samples. Consequently, we sequenced the coding exons and intron/exon borders of the ARHGEF9 gene in 99 probands from families with X linked mental retardation (XLMR) and 477 mentally retarded males in whom a diagnosis of Fragile X syndrome had been excluded. We did not identify any pathogenic changes; however, we did identify intronic nucleotide changes that might alter splicing. CONCLUSION: ARHGEF9 encodes a RhoGEF family protein: collybistin (hPEM), which is highly expressed in the developing and adult brain. Collybistin can regulate actin cytoskeletal dynamics and may also modulate GABAergic and glycinergic neurotransmission through binding of a scaffolding protein, gephyrin, at the synapse. This potential dual role may explain both the mental retardation and hyperarousal observed in our patient. While ARHGEF9 appears to be an uncommon cause of mental retardation in males, it should be considered in patients with mental retardation and sensory hyperarousal.


Subject(s)
Arousal/genetics , Guanine Nucleotide Exchange Factors/genetics , Mental Retardation, X-Linked/genetics , Adolescent , Arousal/physiology , Chromosome Breakage , Chromosomes, Human, X/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Mental Retardation, X-Linked/psychology , Rho Guanine Nucleotide Exchange Factors
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...