Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Thyroid ; 32(7): 849-859, 2022 07.
Article in English | MEDLINE | ID: mdl-35350867

ABSTRACT

Background: Allan-Herndon-Dudley syndrome (AHDS) is a severe psychomotor disability disorder that also manifests characteristic abnormal thyroid hormone (TH) levels. AHDS is caused by inactivating mutations in monocarboxylate transporter 8 (MCT8), a specific TH plasma membrane transporter widely expressed in the central nervous system (CNS). MCT8 mutations cause impaired transport of TH across brain barriers, leading to insufficient neural TH supply. There is currently no successful therapy for the neurological symptoms. Earlier work has shown that intravenous (IV), but not intracerebroventricular adeno-associated virus serotype 9 (AAV9) -based gene therapy given to newborn Mct8 knockout (Mct8-/y) male mice increased triiodothyronine (T3) brain content and partially rescued TH-dependent gene expression, suggesting a promising approach to treat this neurological disorder. Methods: The potential of IV delivery of AAV9 carrying human MCT8 was tested in the well-established Mct8-/y/Organic anion-transporting polypeptide 1c1 (Oatp1c1)-/ - double knockout (dKO) mouse model of AHDS, which, unlike Mct8-/y mice, displays both neurological and TH phenotype. Further, as the condition is usually diagnosed during childhood, treatment was given intravenously to P30 mice and psychomotor tests were carried out blindly at P120-P140 after which tissues were collected and analyzed. Results: Systemic IV delivery of AAV9-MCT8 at a juvenile stage led to improved locomotor and cognitive functions at P120-P140, which was accompanied by a near normalization of T3 content and an increased response of positively regulated TH-dependent gene expression in different brain regions examined (thalamus, hippocampus, and parietal cortex). The effects on serum TH concentrations and peripheral tissues were less pronounced, showing only improvement in the serum T3/reverse T3 (rT3) ratio and in liver deiodinase 1 expression. Conclusion: IV administration of AAV9, carrying the human MCT8, to juvenile dKO mice manifesting AHDS has long-term beneficial effects, predominantly on the CNS. This preclinical study indicates that this gene therapy has the potential to ameliorate the devastating neurological symptoms in patients with AHDS.


Subject(s)
Mental Retardation, X-Linked , Monocarboxylic Acid Transporters , Symporters , Animals , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/therapy , Mice , Monocarboxylic Acid Transporters/administration & dosage , Monocarboxylic Acid Transporters/deficiency , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Hypotonia , Muscular Atrophy , Mutation , Serogroup , Symporters/administration & dosage , Symporters/deficiency , Symporters/genetics , Symporters/metabolism , Triiodothyronine/metabolism
2.
Front Endocrinol (Lausanne) ; 12: 723750, 2021.
Article in English | MEDLINE | ID: mdl-34539576

ABSTRACT

Genetic defects in the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) result in MCT8 deficiency. This disorder is characterized by a combination of severe intellectual and motor disability, caused by decreased cerebral thyroid hormone signalling, and a chronic thyrotoxic state in peripheral tissues, caused by exposure to elevated serum T3 concentrations. In particular, MCT8 plays a crucial role in the transport of thyroid hormone across the blood-brain-barrier. The life expectancy of patients with MCT8 deficiency is strongly impaired. Absence of head control and being underweight at a young age, which are considered proxies of the severity of the neurocognitive and peripheral phenotype, respectively, are associated with higher mortality rate. The thyroid hormone analogue triiodothyroacetic acid is able to effectively and safely ameliorate the peripheral thyrotoxicosis; its effect on the neurocognitive phenotype is currently under investigation. Other possible therapies are at a pre-clinical stage. This review provides an overview of the current understanding of the physiological role of MCT8 and the pathophysiology, key clinical characteristics and developing treatment options for MCT8 deficiency.


Subject(s)
Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/therapy , Muscle Hypotonia/genetics , Muscle Hypotonia/therapy , Muscular Atrophy/genetics , Muscular Atrophy/therapy , Humans , Mental Retardation, X-Linked/mortality , Mental Retardation, X-Linked/pathology , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/mortality , Muscle Hypotonia/pathology , Muscular Atrophy/mortality , Muscular Atrophy/pathology , Phenotype , Signal Transduction/genetics , Symporters/genetics , Therapies, Investigational/methods , Therapies, Investigational/trends
3.
Hum Genet ; 140(12): 1625-1634, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34524523

ABSTRACT

ATR-X, an acronym for alpha thalassemia and mental retardation X-linked, syndrome is a congenital condition predominantly affecting males, characterized by mild to severe intellectual disability, facial, skeletal, urogenital, and hematopoietic anomalies. Less common are heart defects, eye anomalies, renal abnormalities, and gastrointestinal dysfunction. ATR-X syndrome is caused by germline variants in the ATRX gene. Until recently, the diagnosis of the ATR-X syndrome had been guided by the classical clinical manifestations and confirmed by molecular techniques. However, our new systematic analysis shows that the only clinical sign shared by all affected individuals is intellectual disability, with the other manifestations varying even within the same family. More than 190 different germline ATRX mutations in some 200 patients have been analyzed. With improved and more frequent analysis by molecular technologies, more subtle deletions and insertions have been detected recently. Moreover, emerging technologies reveal non-classic phenotypes of ATR-X syndrome as well as the description of a new clinical feature, the development of osteosarcoma which suggests an increased cancer risk in ATR-X syndrome. This review will focus on the different types of inherited ATRX mutations and their relation to clinical features in the ATR-X syndrome. We will provide an update of the frequency of clinical manifestations, the affected organs, and the genotype-phenotype correlations. Finally, we propose a shift in the diagnosis of ATR-X patients, from a clinical diagnosis to a molecular-based approach. This may assist clinicians in patient management, risk assessment and genetic counseling.


Subject(s)
Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/therapy , alpha-Thalassemia/genetics , alpha-Thalassemia/physiopathology , alpha-Thalassemia/therapy , Animals , Humans , Intellectual Disability/genetics , Mental Retardation, X-Linked/diagnosis , Molecular Diagnostic Techniques , Mutation , alpha-Thalassemia/diagnosis
4.
Genes (Basel) ; 12(8)2021 07 24.
Article in English | MEDLINE | ID: mdl-34440297

ABSTRACT

Creatine (Cr) Transporter Deficiency (CTD) is an X-linked metabolic disorder, mostly caused by missense mutations in the SLC6A8 gene and presenting with intellectual disability, autistic behavior, and epilepsy. There is no effective treatment for CTD and patients need lifelong assistance. Thus, the research of novel intervention strategies is a major scientific challenge. Animal models are an excellent tool to dissect the disease pathogenetic mechanisms and drive the preclinical development of therapeutics. This review illustrates the current knowledge about Cr metabolism and CTD clinical aspects, with a focus on mainstay diagnostic and therapeutic options. Then, we discuss the rodent models of CTD characterized in the last decade, comparing the phenotypes expressed within clinically relevant domains and the timeline of symptom development. This analysis highlights that animals with the ubiquitous deletion/mutation of SLC6A8 genes well recapitulate the early onset and the complex pathological phenotype of the human condition. Thus, they should represent the preferred model for preclinical efficacy studies. On the other hand, brain- and cell-specific conditional mutants are ideal for understanding the basis of CTD at a cellular and molecular level. Finally, we explain how CTD models might provide novel insight about the pathogenesis of other disorders, including cancer.


Subject(s)
Brain Diseases, Metabolic, Inborn/pathology , Brain Diseases, Metabolic, Inborn/therapy , Central Nervous System/pathology , Creatine/deficiency , Disease Models, Animal , Mental Retardation, X-Linked/pathology , Mental Retardation, X-Linked/therapy , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Animals , Biomarkers/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Creatine/metabolism , Humans , Mental Retardation, X-Linked/metabolism , Mice , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Rats
5.
Sci Transl Med ; 13(583)2021 03 03.
Article in English | MEDLINE | ID: mdl-33658357

ABSTRACT

Many intellectual disability disorders are due to copy number variations, and, to date, there have been no treatment options tested for this class of diseases. MECP2 duplication syndrome (MDS) is one of the most common genomic rearrangements in males and results from duplications spanning the methyl-CpG binding protein 2 (MECP2) gene locus. We previously showed that antisense oligonucleotide (ASO) therapy can reduce MeCP2 protein amount in an MDS mouse model and reverse its disease features. This MDS mouse model, however, carried one transgenic human allele and one mouse allele, with the latter being protected from human-specific MECP2-ASO targeting. Because MeCP2 is a dosage-sensitive protein, the ASO must be titrated such that the amount of MeCP2 is not reduced too far, which would cause Rett syndrome. Therefore, we generated an "MECP2 humanized" MDS model that carries two human MECP2 alleles and no mouse endogenous allele. Intracerebroventricular injection of the MECP2-ASO efficiently down-regulated MeCP2 expression throughout the brain in these mice. Moreover, MECP2-ASO mitigated several behavioral deficits and restored expression of selected MeCP2-regulated genes in a dose-dependent manner without any toxicity. Central nervous system administration of MECP2-ASO is therefore well tolerated and beneficial in this mouse model and provides a translatable approach that could be feasible for treating MDS.


Subject(s)
Mental Retardation, X-Linked , Methyl-CpG-Binding Protein 2 , Oligonucleotides, Antisense/therapeutic use , Animals , DNA Copy Number Variations , Mental Retardation, X-Linked/therapy , Methyl-CpG-Binding Protein 2/genetics , Mice
6.
Lancet Neurol ; 19(8): 689-698, 2020 08.
Article in English | MEDLINE | ID: mdl-32702338

ABSTRACT

The X-linked gene encoding MECP2 is involved in two severe and complex neurodevelopmental disorders. Loss of function of the MeCP2 protein underlies Rett syndrome, whereas duplications of the MECP2 locus cause MECP2 duplication syndrome. Research on the mechanisms by which MeCP2 exerts effects on gene expression in neurons, studies of animal models bearing different disease-causing mutations, and more in-depth observations of clinical presentations have clarified some issues even as they have raised further questions. Yet there is enough evidence so far to suggest possible approaches to therapy for these two diseases that could go beyond attempting to address specific signs and symptoms (of which there are many) and instead target the pathophysiology underlying MECP2 disorders. Further work could bring antisense oligonucleotides, deep brain stimulation, and gene therapy into the clinic within the next decade or so.


Subject(s)
Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/therapy , Methyl-CpG-Binding Protein 2/genetics , Mutation/genetics , Rett Syndrome/genetics , Rett Syndrome/therapy , Humans
7.
Endocr Rev ; 41(2)2020 04 01.
Article in English | MEDLINE | ID: mdl-31754699

ABSTRACT

Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).


Subject(s)
Biological Transport/physiology , Membrane Transport Proteins/physiology , Mental Retardation, X-Linked , Monocarboxylic Acid Transporters/physiology , Muscle Hypotonia , Muscular Atrophy , Organic Anion Transporters/physiology , Symporters/physiology , Thyroid Hormones/metabolism , Animals , Humans , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/therapy , Monocarboxylic Acid Transporters/deficiency , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/genetics , Muscle Hypotonia/metabolism , Muscle Hypotonia/physiopathology , Muscle Hypotonia/therapy , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Muscular Atrophy/therapy , Organic Anion Transporters/deficiency , Organic Anion Transporters/genetics , Symporters/deficiency , Symporters/genetics , Thyroid Hormones/therapeutic use
8.
Mov Disord ; 34(5): 625-636, 2019 05.
Article in English | MEDLINE | ID: mdl-30913345

ABSTRACT

The term "cerebral palsy mimic" is used to describe a number of neurogenetic disorders that may present with motor symptoms in early childhood, resulting in a misdiagnosis of cerebral palsy. Cerebral palsy describes a heterogeneous group of neurodevelopmental disorders characterized by onset in infancy or early childhood of motor symptoms (including hypotonia, spasticity, dystonia, and chorea), often accompanied by developmental delay. The primary etiology of a cerebral palsy syndrome should always be identified if possible. This is particularly important in the case of genetic or metabolic disorders that have specific disease-modifying treatment. In this article, we discuss clinical features that should alert the clinician to the possibility of a cerebral palsy mimic, provide a practical framework for selecting and interpreting neuroimaging, biochemical, and genetic investigations, and highlight selected conditions that may present with predominant spasticity, dystonia/chorea, and ataxia. Making a precise diagnosis of a genetic disorder has important implications for treatment, and for advising the family regarding prognosis and genetic counseling. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Cerebral Palsy/diagnosis , Diagnosis, Differential , Movement Disorders/diagnosis , Adenylyl Cyclases/genetics , Ataxia/physiopathology , Ataxia Telangiectasia/diagnosis , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/physiopathology , Ataxia Telangiectasia/therapy , Brain/diagnostic imaging , Brain Diseases, Metabolic, Inborn/diagnosis , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/physiopathology , Brain Diseases, Metabolic, Inborn/therapy , Carbohydrate Metabolism, Inborn Errors/diagnosis , Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/physiopathology , Carbohydrate Metabolism, Inborn Errors/therapy , Cerebral Palsy/physiopathology , Chorea/physiopathology , Creatine/deficiency , Creatine/genetics , Dyskinesias/diagnosis , Dyskinesias/genetics , Dyskinesias/physiopathology , Dyskinesias/therapy , Dystonia/physiopathology , Folic Acid Deficiency/diagnosis , Folic Acid Deficiency/genetics , Folic Acid Deficiency/physiopathology , Folic Acid Deficiency/therapy , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Humans , Hyperargininemia/diagnosis , Hyperargininemia/genetics , Hyperargininemia/physiopathology , Hyperargininemia/therapy , Lesch-Nyhan Syndrome/diagnosis , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/physiopathology , Lesch-Nyhan Syndrome/therapy , Magnetic Resonance Imaging , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/therapy , Monosaccharide Transport Proteins/deficiency , Monosaccharide Transport Proteins/genetics , Movement Disorders/genetics , Movement Disorders/physiopathology , Movement Disorders/therapy , Multiple Carboxylase Deficiency/diagnosis , Multiple Carboxylase Deficiency/genetics
9.
Nat Med ; 24(6): 802-813, 2018 06.
Article in English | MEDLINE | ID: mdl-29785027

ABSTRACT

Alpha-thalassemia X-linked intellectual disability (ATR-X) syndrome is caused by mutations in ATRX, which encodes a chromatin-remodeling protein. Genome-wide analyses in mouse and human cells indicate that ATRX tends to bind to G-rich sequences with a high potential to form G-quadruplexes. Here, we report that Atrx mutation induces aberrant upregulation of Xlr3b expression in the mouse brain, an outcome associated with neuronal pathogenesis displayed by ATR-X model mice. We show that ATRX normally binds to G-quadruplexes in CpG islands of the imprinted Xlr3b gene, regulating its expression by recruiting DNA methyltransferases. Xlr3b binds to dendritic mRNAs, and its overexpression inhibits dendritic transport of the mRNA encoding CaMKII-α, promoting synaptic dysfunction. Notably, treatment with 5-ALA, which is converted into G-quadruplex-binding metabolites, reduces RNA polymerase II recruitment and represses Xlr3b transcription in ATR-X model mice. 5-ALA treatment also rescues decreased synaptic plasticity and cognitive deficits seen in ATR-X model mice. Our findings suggest a potential therapeutic strategy to target G-quadruplexes and decrease cognitive impairment associated with ATR-X syndrome.


Subject(s)
Cognition , DNA/metabolism , G-Quadruplexes , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/therapy , alpha-Thalassemia/physiopathology , alpha-Thalassemia/therapy , 3' Untranslated Regions/genetics , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cytoplasmic Granules/metabolism , Cytoskeletal Proteins/metabolism , Dendrites/metabolism , Ligands , Male , Mental Retardation, X-Linked/genetics , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , RNA Polymerase II/metabolism , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , alpha-Thalassemia/genetics
10.
BMC Res Notes ; 10(1): 743, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29258560

ABSTRACT

OBJECTIVES: Children with microcephaly face lifelong psychomotor, cognitive, and communications skills disabilities. Etiology of microcephaly is heterogeneous but presentation often includes seizures, hypotonia, ataxia, stereotypic movements, attention deficits, excitability, cognitive delays, and poor communication skills. Molecular diagnostics have outpaced available interventions and most children receive generic physical, speech, and occupational therapies with little attention to the efficacy of such treatments. Mutations in the X-linked intellectual disability gene (XLID) CASK is one etiology associated with microcephaly which produces mental retardation and microcephaly with pontine and cerebellar hypoplasia (MICPCH; OMIM# 300749). We pilot-tested an intensive therapy in three girls with heterozygous mutation in the gene CASK and MICPCH. Child A = 54 months; Child B = 89 months; and Child C = 24 months received a targeted treatment to improve gross/fine motor skills, visual-motor coordination, social interaction, and communication. Treatment was 4 h each weekday for 10 treatment days. Operant training promoted/refined goal-directed activities. The Peabody Developmental Motor Scales 2 was administered pre- and post-treatment. RESULTS: Child A gained 14 developmental months; Child B gained 20 developmental months; and Child C gained 39 developmental months. This case series suggests that children with MICPCH are responsive to intensive therapy aimed at increasing functional skills/independence. Trial Registration ClinicalTrials.gov Registration Number: NCT03325946; Release Date: October 30, 2017.


Subject(s)
Cognition/physiology , Guanylate Kinases/genetics , Motor Skills/physiology , Mutation , Neurological Rehabilitation/methods , Child , Child, Preschool , Female , Humans , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/therapy , Microcephaly/genetics , Microcephaly/physiopathology , Microcephaly/therapy , Pilot Projects , Treatment Outcome
11.
Hum Mutat ; 38(3): 260-264, 2017 03.
Article in English | MEDLINE | ID: mdl-27805744

ABSTRACT

Mutations in the thyroid hormone transporter SLC16A2 (MCT8) cause the Allan-Herndon-Dudley Syndrome (AHDS), characterized by severe psychomotor retardation and peripheral thyrotoxicosis. Here, we report three newly identified AHDS patients. Previously documented mutations were identified in probands 1 (p.R271H) and 2 (p.G564R), resulting in a severe clinical phenotype. A novel mutation (p.G564E) was identified in proband 3, affecting the same Gly564 residue, but resulting in a relatively mild clinical phenotype. Functional analysis in transiently transfected COS-1 and JEG-3 cells showed a near-complete inactivation of TH transport for p.G564R, whereas considerable cell-type-dependent residual transport activity was observed for p.G564E. Both mutants showed a strong decrease in protein expression levels, but differentially affected Vmax and Km values of T3 transport. Our findings illustrate that different mutations affecting the same residue may have a differential impact on SLC16A2 transporter function, which translates into differences in severity of the clinical phenotype.


Subject(s)
Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/diagnosis , Muscle Hypotonia/genetics , Muscular Atrophy/diagnosis , Muscular Atrophy/genetics , Mutation , Phenotype , Biomarkers , Child , Child, Preschool , Humans , Magnetic Resonance Imaging , Male , Mental Retardation, X-Linked/therapy , Muscle Hypotonia/therapy , Muscular Atrophy/therapy , Pedigree , Symporters
12.
Clin Genet ; 91(4): 557-563, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27247049

ABSTRACT

Individuals with two or more copies of the MECP2 gene, located at Xq28, share clinical features and a distinct facial phenotype known as MECP2 Duplication syndrome. We have examined perinatal characteristics, early childhood development and medical co-morbidities in this disorder. The International Rett Syndrome Phenotype Database (InterRett), which collects information from caregivers and clinicians on individuals with Rett syndrome and MECP2 associated disorders, was used as the data source. Data were available on 56 cases (49 males and 7 females) with MECP2 Duplication syndrome. Median age at ascertainment was 7.9 years (range: 1.2-37.6 years) and at diagnosis 3.0 years (range: 3 weeks-37 years). Less than a third (29%) learned to walk. Speech deterioration was reported in 34% and only 20% used word approximations or better at ascertainment. Over half (55%) had been hospitalised for respiratory infections in the first 2 years of life. Just under half (44%) had seizures, occurring daily in nearly half of this group. The majority (89%) had gastrointestinal problems and a third had a gastrostomy. Following the recent demonstration of phenotype reversal in a mouse model of MECP2 Duplication, a clear understanding of the natural history is crucial to the design and implementation of future therapeutic strategies.


Subject(s)
Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Methyl-CpG-Binding Protein 2/genetics , Adolescent , Adult , Animals , Child , Child, Preschool , Databases, Genetic , Female , Gastrointestinal Tract/physiopathology , Gastrointestinal Tract/surgery , Humans , Infant , Intellectual Disability/physiopathology , Kaplan-Meier Estimate , Language Development Disorders/genetics , Language Development Disorders/physiopathology , Male , Mental Retardation, X-Linked/epidemiology , Mental Retardation, X-Linked/therapy , Mice , Phenotype , Rett Syndrome/epidemiology , Rett Syndrome/genetics , Rett Syndrome/physiopathology , Young Adult
13.
Blood Cells Mol Dis ; 55(1): 27-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25976463

ABSTRACT

α-Thalassemia X-linked mental retardation syndrome is a rare inherited intellectual disability disorder due to mutations in the ATRX gene. In our previous study of the prevalence of ß-thalassemia mutations in the Eastern Province of Saudi Arabia, we confirmed the widespread coinheritance of α-thalassemia mutation. Some of these subjects have a family history of mental retardation, the cause of which is unknown. Therefore, we investigated the presence or absence of mutations in the ATRX gene in these patients. Three exons of the ATRX gene and their flanking regions were directly sequenced. Only four female transfusion dependent ß-thalassemia patients were found to be carriers of a novel mutation in the ATRX gene. Two of the ATRX gene mutations, c.623delA and c.848T>C were present in patients homozygous for IVS I-5(G→C) and homozygous for Cd39(C → T) ß-thalassemia mutation, respectively. While the other two that were located in the intronic region (flanking regions), were present in patients homozygous for Cd39(C → T) ß-thalassemia mutation. The two subjects with the mutations in the coding region had family members with mental retardation, which suggests that the novel frame shift mutation and the missense mutation at coding region of ATRX gene are involved in ATRX syndrome.


Subject(s)
DNA Helicases/genetics , Mental Retardation, X-Linked/genetics , Mutation , Nuclear Proteins/genetics , alpha-Globins/genetics , alpha-Thalassemia/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , Adolescent , Adult , Base Sequence , Blood Transfusion , Exons , Female , Heterozygote , Homozygote , Humans , Introns , Male , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/pathology , Mental Retardation, X-Linked/therapy , Molecular Sequence Data , Pedigree , Saudi Arabia , X-linked Nuclear Protein , alpha-Thalassemia/complications , alpha-Thalassemia/pathology , alpha-Thalassemia/therapy , beta-Thalassemia/complications , beta-Thalassemia/pathology , beta-Thalassemia/therapy
14.
J Child Neurol ; 30(12): 1664-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25900139

ABSTRACT

X-linked monocarboxylate transporter 8 (MCT8) deficiency results from a loss-of-function mutation in the monocarboxylate transporter 8 gene, located on chromosome Xq13.2 (Allan-Herndon-Dudley syndrome). Affected boys present early in life with neurodevelopment delays but have pleasant dispositions and commonly have elevated serum triiodothyronine. They also have marked axial hypotonia and quadriparesis but surprisingly little spasticity early in their disease course. They do, however, have subtle involuntary movements, most often dystonia. The combination of hypotonia and dystonia presents a neurorehabilitation challenge and explains why spasticity-directed therapies have commonly produced suboptimal responses. Our aim was to better define the spectrum of motor disability and to elucidate the neuroanatomic basis of the motor impairments seen in MCT8 deficiency using clinical observation and brain magnetic resonance imaging (MRI) in a cohort of 6 affected pediatric patients. Our findings identified potential imaging biomarkers and suggest that rehabilitation efforts targeting dystonia may be more beneficial than those targeting spasticity in the prepubertal pediatric MCT8 deficiency population.


Subject(s)
Brain/pathology , Mental Retardation, X-Linked/pathology , Mental Retardation, X-Linked/physiopathology , Muscle Hypotonia/pathology , Muscle Hypotonia/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Child , Child, Preschool , Cohort Studies , Diffusion Tensor Imaging , Dystonia/pathology , Dystonia/physiopathology , Humans , Infant , Magnetic Resonance Imaging , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/therapy , Muscle Hypotonia/diagnosis , Muscle Hypotonia/therapy , Muscular Atrophy/diagnosis , Muscular Atrophy/therapy , Phenotype
15.
Handb Clin Neurol ; 111: 297-306, 2013.
Article in English | MEDLINE | ID: mdl-23622180

ABSTRACT

Ten percent of cases of intellectual deficiency in boys are caused by genes located on the X chromosome. X-linked mental retardation (XLMR) includes more than 200 syndromes and 80 genes identified to date. The fragile X syndrome is the most frequent syndrome, due to a dynamic mutation with a CGG triplet amplification. Mental retardation is virtually always present. Phonological and syntactic impairments are often combined with pragmatic language impairment and visuospatial reasoning difficulties. A minority fulfill the criteria for autism. In girls, the clinical expression of the complete mutation varies according to the X chromosome inactivation profile. Several XLMR occur as severe early onset encephalopathies: Lowe oculocerebrorenal syndrome, ATR-X syndrome (alpha thalassemia/mental retardation X-linked), Allan-Herdon-Dudley syndrome (MCT8 gene). Two genes, ARX (X-LAG; Partington syndrome) and MECP2 (Rett syndrome in females; mild MR with spastic diplegia/psychotic problems in males) are associated with various phenotypes, according to the mutation involved. Oligophrenine 1 (OPHN-1) gene mutations lead to vermal dysplasia. PQBP1 gene mutations (Renpenning syndrome) are responsible for moderate to severe mental deficiency, microcephaly, and small stature. Although some forms of XLMR are not very specific and the phenotype for each given gene is somewhat heterogeneous, a clinical diagnostic strategy is emerging.


Subject(s)
Ataxia , Fragile X Syndrome , Mental Retardation, X-Linked , Seizures , alpha-Thalassemia , Ataxia/genetics , Ataxia/therapy , Female , Fragile X Syndrome/genetics , Fragile X Syndrome/therapy , Humans , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/therapy , Phenotype , Seizures/genetics , Seizures/therapy , alpha-Thalassemia/genetics , alpha-Thalassemia/therapy
16.
J Am Med Inform Assoc ; 20(4): 643-51, 2013.
Article in English | MEDLINE | ID: mdl-23408511

ABSTRACT

BACKGROUND AND SIGNIFICANCE: Intellectual disability is a condition characterized by significant limitations in cognitive abilities and social/behavioral adaptive skills and is an important reason for pediatric, neurologic, and genetic referrals. Approximately 10% of protein-encoding genes on the X chromosome are implicated in intellectual disability, and the corresponding intellectual disability is termed X-linked ID (XLID). Although few mutations and a small number of families have been identified and XLID is rare, collectively the impact of XLID is significant because patients usually are unable to fully participate in society. OBJECTIVE: To reveal the molecular mechanisms of various intellectual disabilities and to suggest small molecules which by binding to the malfunctioning protein can reduce unwanted effects. METHODS: Using various in silico methods we reveal the molecular mechanism of XLID in cases involving proteins with known 3D structure. The 3D structures were used to predict the effect of disease-causing missense mutations on the folding free energy, conformational dynamics, hydrogen bond network and, if appropriate, protein-protein binding free energy. RESULTS: It is shown that the vast majority of XLID mutation sites are outside the active pocket and are accessible from the water phase, thus providing the opportunity to alter their effect by binding appropriate small molecules in the vicinity of the mutation site. CONCLUSIONS: This observation is used to demonstrate, computationally and experimentally, that a particular condition, Snyder-Robinson syndrome caused by the G56S spermine synthase mutation, might be ameliorated by small molecule binding.


Subject(s)
Mental Retardation, X-Linked/genetics , Mutation, Missense , Spermine Synthase/genetics , Binding Sites , Brain Diseases, Metabolic, Inborn/genetics , Chloride Channels/genetics , Computational Biology , Creatine/deficiency , Creatine/genetics , Genes, X-Linked , Humans , Mental Retardation, X-Linked/therapy , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Protein Binding/physiology , Protein Structure, Quaternary , Spermine Synthase/metabolism , Translational Research, Biomedical
17.
Am J Med Genet C Semin Med Genet ; 157C(1): 72-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21308988

ABSTRACT

Creatine is a nitrogen containing compound that serves as an energy shuttle between the mitochondrial sites of ATP production and the cytosol where ATP is utilized. There are two known disorders of creatine synthesis (both transmitted as autosomal recessive traits: arginine: glycine amidinotransferase (AGAT) deficiency; OMIM 602360; and guanidinoacetate methyltransferase (GAMT) deficiency (OMIM 601240)) and one disorder of creatine transport (X-linked recessive SLC6A8 creatine transporter deficiency (OMIM 300036)). All these disorders are characterized by brain creatine deficiency, detectable by magnetic resonance spectroscopy. Affected patients can have mental retardation, hypotonia, autism or behavioral problems and seizures. The diagnosis of these conditions relies on the measurement of plasma and urine creatine and guanidinoacetate. Creatine levels in plasma are reduced in both creatine synthesis defects and guanidinoacetate is increased in GAMT deficiency. The urine creatine/creatinine ratio is elevated in creatine transporter deficiency with normal plasma levels of creatine and guanidinoacetate. The diagnosis is confirmed in all cases by DNA testing or functional studies. Defects of creatine biosynthesis are treated with creatine supplements and, in GAMT deficiency, with ornithine and dietary restriction of arginine through limitation of protein intake. No causal therapy is yet available for creatine transporter deficiency and supplementation with the guanidinoacetate precursors arginine and glycine is being explored. The excellent response to therapy of early identified patients with GAMT or AGAT deficiency candidates these condition for inclusion in newborn screening programs.


Subject(s)
Brain Diseases, Metabolic, Inborn , Mental Retardation, X-Linked , Amidinotransferases/genetics , Brain Diseases, Metabolic, Inborn/diagnosis , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/therapy , Creatine/deficiency , Creatine/genetics , Creatinine/blood , Creatinine/urine , Glycine/analogs & derivatives , Glycine/blood , Glycine/urine , Guanidinoacetate N-Methyltransferase/genetics , Humans , Infant, Newborn , Magnetic Resonance Spectroscopy , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/therapy , Neonatal Screening , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Plasma Membrane Neurotransmitter Transport Proteins/genetics
19.
Wiad Lek ; 61(4-6): 146-53, 2008.
Article in Polish | MEDLINE | ID: mdl-18939366

ABSTRACT

Mental retardation is a serious medical and social problem. The prevalence of mental retardation is estimated at 2-3%. Establishing the cause of mental retardation is extremely important for prognosis, management, and genetic counseling. It is postulated that 25-35% of mental retardation cases may be of genetic background. Among the genetic causes 25-30% are probably result of mutations located in the X chromosome (X-linked mental retardation--XLMR). X-linked mental retardation is a heterogeneous set of conditions responsible for a large proportion of inherited mental retardation. More than 200 XLMR conditions and 45 cloned genes are listed in catalogue available on the Internet. Traditionally, based on clinical presentation, XLMR conditions were divided into specific and nonspecific forms or syndromic and nonsyndromic. The distinction between specific and non-specific forms of XLMR is gradually becoming less clear and spectrum of phenotypic variability is very large as both syndromic and nonsyndromic forms have been described for several of the XLMR genes. Mutations in patients suffering from X-linked mental retardation genes have been found only in a relatively limited number of cases. Up to 50% of the patients from XLMR families might have mutations in one of the known genes implicated in XLMR so far. However, current methods are generally too expensive or too unreliable to justify mutation screening of all known XLMR genes in diagnostic testing. Thus it is necessary to use empirical data of recurrence risk in genetic counseling of the family with mental retardation.


Subject(s)
Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/therapy , Genetic Counseling , Humans , Mental Retardation, X-Linked/diagnosis , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...