Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
PLoS Genet ; 20(10): e1011428, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39405291

ABSTRACT

Börjeson-Forssman-Lehmann syndrome (BFLS) is an X-linked intellectual disability and endocrine disorder caused by pathogenic variants of plant homeodomain finger gene 6 (PHF6). An understanding of the role of PHF6 in vivo in the development of the mammalian nervous system is required to advance our knowledge of how PHF6 mutations cause BFLS. Here, we show that PHF6 protein levels are greatly reduced in cells derived from a subset of patients with BFLS. We report the phenotypic, anatomical, cellular and molecular characterization of the brain in males and females in two mouse models of BFLS, namely loss of Phf6 in the germline and nervous system-specific deletion of Phf6. We show that loss of PHF6 resulted in spontaneous seizures occurring via a neural intrinsic mechanism. Histological and morphological analysis revealed a significant enlargement of the lateral ventricles in adult Phf6-deficient mice, while other brain structures and cortical lamination were normal. Phf6 deficient neural precursor cells showed a reduced capacity for self-renewal and increased differentiation into neurons. Phf6 deficient cortical neurons commenced spontaneous neuronal activity prematurely suggesting precocious neuronal maturation. We show that loss of PHF6 in the foetal cortex and isolated cortical neurons predominantly caused upregulation of genes, including Reln, Nr4a2, Slc12a5, Phip and ZIC family transcription factor genes, involved in neural development and function, providing insight into the molecular effects of loss of PHF6 in the developing brain.


Subject(s)
Disease Models, Animal , Mental Retardation, X-Linked , Repressor Proteins , Seizures , Animals , Mice , Seizures/genetics , Seizures/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Humans , Repressor Proteins/genetics , Repressor Proteins/metabolism , Female , Male , Face/abnormalities , Intellectual Disability/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hypogonadism/genetics , Hypogonadism/pathology , Hypogonadism/metabolism , Vestibular Diseases/genetics , Vestibular Diseases/pathology , Calcinosis/genetics , Calcinosis/pathology , Calcinosis/metabolism , Neural Stem Cells/metabolism , Mice, Knockout , Transcription, Genetic , Epilepsy , Fingers/abnormalities , Growth Disorders , Obesity
2.
Neurobiol Dis ; 200: 106621, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39097035

ABSTRACT

Allan-Herndon-Dudley syndrome (AHDS) is a rare X-linked disorder that causes severe neurological damage, for which there is no effective treatment. AHDS is due to inactivating mutations in the thyroid hormone transporter MCT8 that impair the entry of thyroid hormones into the brain, resulting in cerebral hypothyroidism. However, the pathophysiology of AHDS is still not fully understood and this is essential to develop therapeutic strategies. Based on evidence suggesting that thyroid hormone deficit leads to alterations in astroglial cells, including gliosis, in this work, we have evaluated astroglial impairments in MCT8 deficiency by means of magnetic resonance imaging, histological, ultrastructural, and immunohistochemical techniques, and by mining available RNA sequencing outputs. Apparent diffusion coefficient (ADC) imaging values obtained from magnetic resonance imaging showed changes indicative of alterations in brain cytoarchitecture in MCT8-deficient patients (n = 11) compared to control subjects (n = 11). Astroglial alterations were confirmed by immunohistochemistry against astroglial markers in autopsy brain samples of an 11-year-old and a 30th gestational week MCT8-deficient subjects in comparison to brain samples from control subjects at similar ages. These findings were validated and further explored in a mouse model of AHDS. Our findings confirm changes in all the astroglial populations of the cerebral cortex in MCT8 deficiency that impact astrocytic metabolic and mitochondrial cellular respiration functions. These impairments arise early in brain development and persist at adult stages, revealing an abnormal distribution, density, morphology of cortical astrocytes, along with altered transcriptome, compatible with an astrogliosis-like phenotype at adult stages. We conclude that astrocytes are potential novel therapeutic targets in AHDS, and we propose ADC imaging as a tool to monitor the progression of neurological impairments and potential effects of treatments in MCT8 deficiency.


Subject(s)
Astrocytes , Brain , Monocarboxylic Acid Transporters , Muscle Hypotonia , Symporters , Thyroid Hormones , Astrocytes/metabolism , Astrocytes/pathology , Animals , Mice , Humans , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Male , Brain/metabolism , Brain/pathology , Thyroid Hormones/metabolism , Child , Symporters/metabolism , Symporters/genetics , Muscle Hypotonia/metabolism , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Female , Adult , Magnetic Resonance Imaging/methods , Muscular Atrophy
3.
Dis Model Mech ; 17(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38881329

ABSTRACT

MECP2 duplication syndrome (MDS) is a neurodevelopmental disorder caused by tandem duplication of the MECP2 locus and its surrounding genes, including IRAK1. Current MDS mouse models involve transgenic expression of MECP2 only, limiting their applicability to the study of the disease. Herein, we show that an efficient and precise CRISPR/Cas9 fusion proximity-based approach can be utilized to generate an Irak1-Mecp2 tandem duplication mouse model ('Mecp2 Dup'). The Mecp2 Dup mouse model recapitulates the genomic landscape of human MDS by harboring a 160 kb tandem duplication encompassing Mecp2 and Irak1, representing the minimal disease-causing duplication, and the neighboring genes Opn1mw and Tex28. The Mecp2 Dup model exhibits neuro-behavioral abnormalities, and an abnormal immune response to infection not previously observed in other mouse models, possibly owing to Irak1 overexpression. The Mecp2 Dup model thus provides a tool to investigate MDS disease mechanisms and develop potential therapies applicable to patients.


Subject(s)
Disease Models, Animal , Gene Duplication , Interleukin-1 Receptor-Associated Kinases , Mental Retardation, X-Linked , Methyl-CpG-Binding Protein 2 , Animals , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Humans , Mice, Inbred C57BL , Mice , CRISPR-Cas Systems/genetics , Behavior, Animal , Male
4.
Epilepsia ; 65(8): 2483-2496, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38819633

ABSTRACT

OBJECTIVE: Methyl CpG-binding protein 2 (MECP2) duplication syndrome is a rare X-linked genomic disorder affecting predominantly males, which is usually manifested as epilepsy and autism spectrum disorder (ASD) comorbidity. The transgenic line MeCP2Tg1 was used for mimicking MECP2 duplication syndrome and showed autism-epilepsy co-occurrence. Previous works suggested that the excitatory/inhibitory (E/I) imbalance is a potential common mechanism for both epilepsy and ASD. The projection neurons and parvalbumin (PV) interneurons account for the majority of E/I balance in the hippocampus. Therefore, we explored how structural changes of projection and PV+ neurons occur in the hippocampus of MeCP2Tg1 mice and whether these morphological changes contribute to epilepsy susceptibility. METHODS: We used the interneuron Designer receptors exclusively activated by designer drugs mouse model to inhibit inhibitory neurons in the hippocampus to verify the epilepsy susceptibility of MeCP2Tg1 (FVB, an inbred strain named as sensitivity to Friend leukemia virus) mice. Electroencephalograms were recorded for the definition of seizure. We performed retro-orbital injection of virus in MeCP2Tg1 (FVB):CaMKIIα-Cre (C57BL/6) mice or MeCP2Tg1:PV-Cre (C57BL/6) mice and their littermate controls to specifically label projection and PV+ neurons for structural analysis. RESULTS: Epilepsy susceptibility was increased in MeCP2Tg1 mice. There was a reduced number of PV neurons and reduced dendritic complexity in the hippocampus of MeCP2Tg1 mice. The dendritic complexity in MeCP2Tg1 mice was increased compared to wild-type mice, and total dendritic spine density in dentate gyrus of MeCP2Tg1 mice was also increased. Total dendritic spine density was increased in CA1 of MeCP2Tg1 mice. SIGNIFICANCE: Overexpression of MeCP2 may disrupt crucial signaling pathways, resulting in decreased dendritic complexity of PV interneurons and increased dendritic spine density of projection neurons. This reciprocal modulation of excitatory and inhibitory neuronal structures associated with MeCP2 implies its significance as a potential target in the development of epilepsy and offers a novel perspective on the co-occurrence of autism and epilepsy.


Subject(s)
Epilepsy , Hippocampus , Interneurons , Parvalbumins , Animals , Male , Mice , Disease Models, Animal , Electroencephalography , Epilepsy/pathology , Epilepsy/genetics , Hippocampus/pathology , Hippocampus/metabolism , Interneurons/pathology , Interneurons/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Methyl-CpG-Binding Protein 2/genetics , Mice, Transgenic , Parvalbumins/metabolism
5.
Eur J Med Genet ; 69: 104949, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38797245

ABSTRACT

Variation in the non-coding genome is being increasingly recognized to be involved in monogenic disease etiology. However, the interpretation of non-coding variation is complicated by a lack of understanding of how non-coding genetic elements function. Additional lines of evidence are therefore needed to recognize non-coding variants as pathogenic. We here present a case where a collective body of evidence resulted in the identification and conclusive classification of a pathogenic deep intronic variant in ATRX. This report demonstrates the utility of a multi-platform approach in aiding the identification of pathogenic variants outside coding regions. Furthermore, it marks the first reported instance of a deep intronic pathogenic variant in ATRX.


Subject(s)
Introns , X-linked Nuclear Protein , Humans , X-linked Nuclear Protein/genetics , Male , Mutation , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Mental Retardation, X-Linked/diagnosis
6.
Am J Med Genet A ; 194(9): e63639, 2024 09.
Article in English | MEDLINE | ID: mdl-38682877

ABSTRACT

Pettigrew syndrome (PGS), an X-linked intellectual disability (XLID), is caused by mutations in the AP1S2 gene. Herein, we described a Thai family with six patients who had severe-to-profound intellectual impairment, limited verbal communication, and varying degrees of limb spasticity. One patient had a unilateral cataract. We demonstrated facial evolution over time, namely coarse facies, long faces, and thick lip vermilions. We identified a novel AP1S2 variant, c.1-2A>G. The mRNA analysis revealed that the variant resulted in splicing defects with leaky splicing, yielding two distinct aberrant transcripts, one of which likely resulting in the mutant protein lacking the first 44 amino acids whereas the other possibly leading to no production of the protein. By performing a literature review, we found 51 patients and 11 AP1S2 pathogenic alleles described and that all the variants were loss-of-function alleles. The severity of ID in Pettigrew syndrome is mostly severe-to-profound (54.8%), followed by moderate (26.2%) and mild. Progressive spasticity was noted in multiple patients. In summary, leaky splicing found in the present family was likely related to the intrafamilial clinical variability. Our data also support the previous notion of variable expression and neuroprogressive nature of the disorder.


Subject(s)
Adaptor Protein Complex sigma Subunits , Mental Retardation, X-Linked , RNA Splicing , Adult , Child, Preschool , Female , Humans , Male , Adaptor Protein Complex sigma Subunits/genetics , Alleles , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Mutation/genetics , Pedigree , Phenotype , RNA Splicing/genetics
7.
Dis Model Mech ; 17(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38463005

ABSTRACT

Snyder-Robinson syndrome (SRS) is a rare X-linked recessive disorder caused by a mutation in the SMS gene, which encodes spermine synthase, and aberrant polyamine metabolism. SRS is characterized by intellectual disability, thin habitus, seizure, low muscle tone/hypotonia and osteoporosis. Progress towards understanding and treating SRS requires a model that recapitulates human gene variants and disease presentations. Here, we evaluated molecular and neurological presentations in the G56S mouse model, which carries a missense mutation in the Sms gene. The lack of SMS protein in the G56S mice resulted in increased spermidine/spermine ratio, failure to thrive, short stature and reduced bone density. They showed impaired learning capacity, increased anxiety, reduced mobility and heightened fear responses, accompanied by reduced total and regional brain volumes. Furthermore, impaired mitochondrial oxidative phosphorylation was evident in G56S cerebral cortex, G56S fibroblasts and Sms-null hippocampal cells, indicating that SMS may serve as a future therapeutic target. Collectively, our study establishes the suitability of the G56S mice as a preclinical model for SRS and provides a set of molecular and functional outcome measures that can be used to evaluate therapeutic interventions for SRS.


Subject(s)
Behavior, Animal , Disease Models, Animal , Mental Retardation, X-Linked , Polyamines , Spermine Synthase , Animals , Mental Retardation, X-Linked/pathology , Mental Retardation, X-Linked/genetics , Spermine Synthase/metabolism , Spermine Synthase/genetics , Polyamines/metabolism , Mitochondria/metabolism , Male , Mice , Fibroblasts/metabolism , Fibroblasts/pathology , Oxidative Phosphorylation , Hippocampus/pathology , Hippocampus/metabolism , Anxiety/pathology , Bone Density , Brain/pathology , Brain/metabolism , Fear , Humans , Organ Size
8.
J Pediatr Endocrinol Metab ; 37(4): 371-374, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38345890

ABSTRACT

OBJECTIVES: To report an unusual case of MCT8 deficiency (Allan-Herndon-Dudley syndrome), an X-linked condition caused by pathogenic variants in the SLC16A2 gene. Defective transport of thyroid hormones (THs) in this condition leads to severe neurodevelopmental impairment in males, while heterozygous females are usually asymptomatic or have mild TH abnormalities. CASE PRESENTATION: A girl with profound developmental delay, epilepsy, primary amenorrhea, elevated T3, low T4 and free T4 levels was diagnosed with MCT8-deficiency at age 17 years, during evaluation for primary ovarian insufficiency (POI). Cytogenetic analysis demonstrated balanced t(X;16)(q13.2;q12.1) translocation with a breakpoint disrupting SLC16A2. X-chromosome inactivation studies revealed a skewed inactivation of the normal X chromosome. CONCLUSIONS: MCT8-deficiency can manifest clinically and phenotypically in women with SLC16A2 aberrations when nonrandom X inactivation occurs, while lack of X chromosome integrity due to translocation can cause POI.


Subject(s)
Mental Retardation, X-Linked , Primary Ovarian Insufficiency , Symporters , Male , Adolescent , Humans , Female , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Muscle Hypotonia/diagnosis , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Delayed Diagnosis , Primary Ovarian Insufficiency/genetics , Monocarboxylic Acid Transporters/genetics , Translocation, Genetic , Symporters/genetics
9.
Fluids Barriers CNS ; 20(1): 79, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37924081

ABSTRACT

BACKGROUND: The monocarboxylate transporter 8 (MCT8) plays a vital role in maintaining brain thyroid hormone homeostasis. This transmembrane transporter is expressed at the brain barriers, as the blood-brain barrier (BBB), and in neural cells, being the sole known thyroid hormone-specific transporter to date. Inactivating mutations in the MCT8 gene (SLC16A2) cause the Allan-Herndon-Dudley Syndrome (AHDS) or MCT8 deficiency, a rare X-linked disease characterized by delayed neurodevelopment and severe psychomotor disorders. The underlying pathophysiological mechanisms of AHDS remain unclear, and no effective treatments are available for the neurological symptoms of the disease. METHODS: Neurovascular unit ultrastructure was studied by means of transmission electron microscopy. BBB permeability and integrity were evaluated by immunohistochemistry, non-permeable dye infiltration assays and histological staining techniques. Brain blood-vessel density was evaluated by immunofluorescence and magnetic resonance angiography. Finally, angiogenic-related factors expression was evaluated by qRT-PCR. The studies were carried out both in an MCT8 deficient subject and Mct8/Dio2KO mice, an AHDS murine model, and their respective controls. RESULTS: Ultrastructural analysis of the BBB of Mct8/Dio2KO mice revealed significant alterations in neurovascular unit integrity and increased transcytotic flux. We also found functional alterations in the BBB permeability, as shown by an increased presence of peripheral IgG, Sodium Fluorescein and Evans Blue, along with increased brain microhemorrhages. We also observed alterations in the angiogenic process, with reduced blood vessel density in adult mice brain and altered expression of angiogenesis-related factors during brain development. Similarly, AHDS human brain samples showed increased BBB permeability to IgG and decreased blood vessel density. CONCLUSIONS: These findings identify for the first time neurovascular alterations in the MCT8-deficient brain, including a disruption of the integrity of the BBB and alterations in the neurovascular unit ultrastructure as a new pathophysiological mechanism for AHDS. These results open a new field for potential therapeutic targets for the neurological symptoms of these patients and unveils magnetic resonance angiography as a new non-invasive in vivo technique for evaluating the progression of the disease.


Subject(s)
Mental Retardation, X-Linked , Symporters , Animals , Humans , Mice , Blood-Brain Barrier/metabolism , Immunoglobulin G , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Hypotonia/diagnosis , Muscle Hypotonia/genetics , Muscle Hypotonia/metabolism , Muscular Atrophy/diagnosis , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Symporters/genetics , Symporters/metabolism , Symporters/therapeutic use , Thyroid Hormones/metabolism , Thyroid Hormones/therapeutic use
10.
Cells ; 12(8)2023 04 18.
Article in English | MEDLINE | ID: mdl-37190086

ABSTRACT

Microcephaly with pontine and cerebellar hypoplasia (MICPCH) syndrome is a neurodevelopmental disorder caused by the deficiency of the X-chromosomal gene CASK. However, the molecular mechanisms by which CASK deficiency causes cerebellar hypoplasia in this syndrome remain elusive. In this study, we used CASK knockout (KO) mice as models for MICPCH syndrome and investigated the effect of CASK mutants. Female CASK heterozygote KO mice replicate the progressive cerebellar hypoplasia observed in MICPCH syndrome. CASK KO cultured cerebellar granule (CG) cells show progressive cell death that can be rescued by co-infection with lentivirus expressing wild-type CASK. Rescue experiments with CASK deletion mutants identify that the CaMK, PDZ, and SH3, but not L27 and guanylate kinase domains of CASK are required for the survival of CG cells. We identify missense mutations in the CaMK domain of CASK derived from human patients that fail to rescue the cell death of cultured CASK KO CG cells. Machine learning-based structural analysis using AlphaFold 2.2 predicts that these mutations disrupt the structure of the binding interface with Liprin-α2. These results suggest that the interaction with Liprin-α2 via the CaMK domain of CASK may be involved in the pathophysiology of cerebellar hypoplasia in MICPCH syndrome.


Subject(s)
Adaptor Proteins, Signal Transducing , Cerebellum , Guanylate Kinases , Membrane Proteins , Mental Retardation, X-Linked , Microcephaly , Cerebellum/metabolism , Cerebellum/pathology , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Guanylate Kinases/chemistry , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Humans , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Mice, Knockout , Animals , Mice , Female , Cells, Cultured , Mutation , Protein Domains , Machine Learning , Software , Apoptosis
11.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(2): 294-301, 2023 Feb 28.
Article in English, Chinese | MEDLINE | ID: mdl-36999477

ABSTRACT

Börjeson-Forssman-Lehmann syndrome (BFLS) is a rare X-linked intellectual disability. The main features of the patients include intellectual disability/global developmental delay, characteristic face, anomalies of fingers and toes, hypogonadism, linear skin hyperpigmentation, and tooth abnormalities in female patients, and obesity in male patients. A case of BFLS caused by a novel mutation of PHF6 gene who was treated in the Department of Pediatrics, Xiangya Hospital, Central South University was reported. The 11 months old girl presented the following symptons: Global developmental delay, characteristic face, sparse hair, ocular hypertelorism, flat nasal bridge, hairy anterior to the tragus, thin upper lip, dental anomalies, ankyloglossia, simian line, tapering fingers, camptodactylia, and linear skin hyperpigmentation. The gene results of the second-generation sequencing technology showed that there was a novel heterozygous mutation site c.346C>T (p.Arg116*) of the PHF6 (NM032458.3), variation rating as pathogenic variation. During the follow-up, the patient developed astigmatism, strabismus, awake bruxism, and stereotyped behavior, and the linear skin hyperpigmentation became gradually more evident. The disease is lack of effective therapy so far.


Subject(s)
Hypogonadism , Intellectual Disability , Mental Retardation, X-Linked , Humans , Male , Female , Child , Infant , Intellectual Disability/genetics , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Obesity/complications , Hypogonadism/genetics , Hypogonadism/pathology
12.
Biochem Soc Trans ; 51(1): 363-372, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36815699

ABSTRACT

Mutations in the polyglutamine tract-binding protein 1 (PQBP1) gene are associated with Renpenning syndrome, which is characterized by microcephaly, intellectual deficiency, short stature, small testes, and distinct facial dysmorphism. Studies using different models have revealed that PQBP1 plays essential roles in neural development and function. In this mini-review, we summarize recent findings relating to the roles of PQBP1 in these processes, including in the regulation of neural progenitor proliferation, neural projection, synaptic growth, neuronal survival, and cognitive function via mRNA transcription and splicing-dependent or -independent processes. The novel findings provide insights into the mechanisms underlying the pathogenesis of Renpenning syndrome and may advance drug discovery and treatment for this condition.


Subject(s)
Cerebral Palsy , Intellectual Disability , Mental Retardation, X-Linked , Humans , Carrier Proteins/chemistry , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Mutation , Cerebral Palsy/genetics , Cerebral Palsy/pathology , Intellectual Disability/genetics , DNA-Binding Proteins/genetics
13.
Eur J Med Genet ; 65(2): 104421, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34999262

ABSTRACT

EIF2S3 pathogenic variants have been shown to cause MEHMO syndrome - a rare X-linked intellectual disability syndrome. In most cases, DNA diagnostics of MEHMO syndrome is performed using exome sequencing. We describe two cousins with profound intellectual disability, severe microcephaly, microgenitalism, hypoglycemia, epileptic seizures, and hypertrichosis, whose clinical symptoms allowed us to suspect MEHMO syndrome. To confirm this diagnosis, we designed an mRNA analysis for the EIF2S3 gene. It is a cost-effective method to detect coding sequence variants in multi-exonic genes, as well as splicing defects and allelic imbalance. Our mRNA sequence analysis revealed a novel EIF2S3 variant c.820C>G in both cousins. We also found the same variant in female family members in the heterozygous state. To investigate the pathogenicity of the c.820C>G variant, we performed expression analysis, which showed that the DDIT3 transcript level was significantly increased in the patient relative to the controls. We, thus, demonstrate that mRNA analysis is an efficient tool for performing genetic testing in patients with distinct phenotypic features.


Subject(s)
Epilepsy/genetics , Eukaryotic Initiation Factor-2/genetics , Genitalia/abnormalities , Hypogonadism/genetics , Mental Retardation, X-Linked/genetics , Microcephaly/genetics , Obesity/genetics , Allelic Imbalance , Cells, Cultured , Child, Preschool , Epilepsy/pathology , Eukaryotic Initiation Factor-2/metabolism , Genitalia/pathology , Heterozygote , Humans , Hypogonadism/pathology , Infant , Male , Mental Retardation, X-Linked/pathology , Microcephaly/pathology , Mutation , Obesity/pathology , Pedigree , RNA Splicing
14.
Genes (Basel) ; 12(12)2021 11 28.
Article in English | MEDLINE | ID: mdl-34946860

ABSTRACT

Investigating novel genetic variants involved in intellectual disability (ID) development is essential. X-linked intellectual disability (XLID) accounts for over 10% of all cases of ID in males. XLID genes are involved in many cellular pathways and processes. Some of them are not specific to the development and functioning of the neural system. The implementation of exome sequencing simplifies the search for novel variants, especially those less expected. Here, we describe a nonsense variant of the XLID gene, WDR13. The mutation c.757C>T (p.Arg253Ter) was uncovered by X-chromosome exome sequencing in males with a familial form of intellectual disability. Quantitative PCR (qPCR) analysis showed that variant c.757C>T caused a significant decrease in WDR13 expression in the patient's fibroblast. Moreover, it dysregulated other genes linked to intellectual disability, such as FMR1, SYN1, CAMK2A, and THOC2. The obtained results indicate the pathogenic nature of the detected variant and suggest that the WDR13 gene interacts with other genes essential for the functioning of the nervous system, especially the synaptic plasticity process.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression Regulation , Genes, X-Linked , Intellectual Disability/pathology , Mental Retardation, X-Linked/pathology , Mutation , Adult , Female , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Pedigree , Exome Sequencing/methods , Young Adult
15.
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
16.
PLoS One ; 16(8): e0256181, 2021.
Article in English | MEDLINE | ID: mdl-34388204

ABSTRACT

Identifying causative variants in cis-regulatory elements (CRE) in neurodevelopmental disorders has proven challenging. We have used in vivo functional analyses to categorize rigorously filtered CRE variants in a clinical cohort that is plausibly enriched for causative CRE mutations: 48 unrelated males with a family history consistent with X-linked intellectual disability (XLID) in whom no detectable cause could be identified in the coding regions of the X chromosome (chrX). Targeted sequencing of all chrX CRE identified six rare variants in five affected individuals that altered conserved bases in CRE targeting known XLID genes and segregated appropriately in families. Two of these variants, FMR1CRE and TENM1CRE, showed consistent site- and stage-specific differences of enhancer function in the developing zebrafish brain using dual-color fluorescent reporter assay. Mouse models were created for both variants. In male mice Fmr1CRE induced alterations in neurodevelopmental Fmr1 expression, olfactory behavior and neurophysiological indicators of FMRP function. The absence of another likely causative variant on whole genome sequencing further supported FMR1CRE as the likely basis of the XLID in this family. Tenm1CRE mice showed no phenotypic anomalies. Following the release of gnomAD 2.1, reanalysis showed that TENM1CRE exceeded the maximum plausible population frequency of a XLID causative allele. Assigning causative status to any ultra-rare CRE variant remains problematic and requires disease-relevant in vivo functional data from multiple sources. The sequential and bespoke nature of such analyses renders them time-consuming and challenging to scale for routine clinical use.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Genes, X-Linked , Genome, Human , Mental Retardation, X-Linked/genetics , Nerve Tissue Proteins/genetics , Regulatory Elements, Transcriptional , Tenascin/genetics , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/pathology , Chromosome Mapping , Cohort Studies , Disease Models, Animal , Embryo, Nonmammalian , Exome , Fragile X Mental Retardation Protein/metabolism , Gene Frequency , Genotype , Humans , Male , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Mice , Nerve Tissue Proteins/deficiency , Pedigree , Phenotype , Tenascin/deficiency , Zebrafish
17.
Hum Genomics ; 15(1): 49, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34348791

ABSTRACT

BACKGROUND: The diagnostic process for uncommon disorders with similar manifestations is complicated and requires newer technology, like gene sequencing for a correct diagnosis. MAIN BODY: We described two brothers clinically diagnosed with Carpenter syndrome, which is a condition characterized by the premature fusion of certain skull bones (craniosynostosis), abnormalities of the fingers and toes, and other developmental problems, for which they underwent craniotomies. However, whole exome sequencing analysis concluded a novel pathological variation in the ATRX chromatin remodeler gene and protein remodeling demonstrated structural variations that decreased the function, giving a completely different diagnosis to these patients. CONCLUSION: Our study focuses on the importance of using newer technologies, such as whole exome sequencing analysis, in patients with ambiguous phenotypes.


Subject(s)
Acrocephalosyndactylia/genetics , Mental Retardation, X-Linked/genetics , Nuclear Proteins/genetics , X-linked Nuclear Protein/genetics , alpha-Thalassemia/genetics , Acrocephalosyndactylia/pathology , DNA Helicases/genetics , Exome/genetics , Humans , Mental Retardation, X-Linked/pathology , Mutation/genetics , Phenotype , Exome Sequencing , alpha-Thalassemia/pathology
18.
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
19.
Cell Rep ; 36(2): 109337, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260915

ABSTRACT

RNA-binding proteins play important roles in X-linked intellectual disability (XLID). In this study, we investigate the contribution of the XLID-associated RBMX in neuronal differentiation. We show that RBMX-depleted cells exhibit aberrant activation of the p53 pathway. Moreover, we identify that the RBMX RGG/RG motif is methylated by protein arginine methyltransferase 5 (PRMT5), and this regulates assembly with the SRSF1 splicing factor into higher-order complexes. Depletion of RBMX or disruption of the RBMX/SRSF1 complex in PRMT5-depleted cells reduces SRSF1 binding to the MDM4 precursor (pre-)mRNA, leading to exon 6 exclusion and lower MDM4 protein levels. Transcriptomic analysis of isogenic Shashi-XLID human-induced pluripotent stem cells (hiPSCs) generated using CRISPR-Cas9 reveals a dysregulation of MDM4 splicing and aberrant p53 upregulation. Shashi-XLID neural progenitor cells (NPCs) display differentiation and morphological abnormalities accompanied with excessive apoptosis. Our findings identify RBMX as a regulator of SRSF1 and the p53 pathway, suggesting that the loss of function of the RBMX RGG/RG motif is the cause of Shashi-XLID syndrome.


Subject(s)
Cell Differentiation , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Mental Retardation, X-Linked/pathology , Neurons/metabolism , Neurons/pathology , Sequence Deletion , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Alternative Splicing/genetics , Amino Acid Motifs , Arginine/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Methylation , Neural Stem Cells/metabolism , Neurogenesis , Protein Binding , Protein Stability , Protein-Arginine N-Methyltransferases/metabolism , Proto-Oncogene Proteins/genetics , RNA/metabolism , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism
20.
Genes (Basel) ; 12(6)2021 06 04.
Article in English | MEDLINE | ID: mdl-34199727

ABSTRACT

The int22h1/int22h2-mediated Xq28 duplication syndrome is a rare X-linked intellectual disability syndrome (XLIDS) arising from a duplication of the segment between intron 22 homologous regions 1 and 2, on the q28 subregion of the X chromosome. The main clinical features of the syndrome include intellectual disability, neurobehavioral abnormalities, and dysmorphic facial features. Due to the X-linked nature of the syndrome, affected males exhibit more severe phenotypes compared with heterozygous females. A unique distinguishing feature of the syndrome across the sexes, however, is a peculiar combination of recurrent sinopulmonary infections and atopy exclusively seen in a subset of affected males. In addition to the 'typical' 0.5 Mb duplication detected in most cases reported to date with the syndrome, a shortened centromeric version, and another 0.2 Mb telomerically shifted one, have been recently identified, with most detected duplications being maternally inherited, except for three recent cases found to have de novo duplications. Interestingly, a recently reported case of an affected male suggests a possible association of the syndrome with multiple malignancies, an observation that has been recently replicated in two pediatric patients. As a result, a better understanding of the pathogenesis of int22h1/int22h2-mediated Xq28 duplication syndrome may grant us a better understanding of the sex-specific differences in immunological responses, as well as the potential role of the genes involved by the duplication, in oncogenesis.


Subject(s)
Chromosome Duplication , Mental Retardation, X-Linked/genetics , Phenotype , Genetic Loci , Humans , Mental Retardation, X-Linked/immunology , Mental Retardation, X-Linked/pathology
SELECTION OF CITATIONS
SEARCH DETAIL