Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.729
Filtrer
1.
Nat Commun ; 15(1): 5524, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951485

RÉSUMÉ

The three-dimensional genome structure organized by CTCF is required for development. Clinically identified mutations in CTCF have been linked to adverse developmental outcomes. Nevertheless, the underlying mechanism remains elusive. In this investigation, we explore the regulatory roles of a clinically relevant R567W point mutation, located within the 11th zinc finger of CTCF, by introducing this mutation into both murine models and human embryonic stem cell-derived cortical organoid models. Mice with homozygous CTCFR567W mutation exhibit growth impediments, resulting in postnatal mortality, and deviations in brain, heart, and lung development at the pathological and single-cell transcriptome levels. This mutation induces premature stem-like cell exhaustion, accelerates the maturation of GABAergic neurons, and disrupts neurodevelopmental and synaptic pathways. Additionally, it specifically hinders CTCF binding to peripheral motifs upstream to the core consensus site, causing alterations in local chromatin structure and gene expression, particularly at the clustered protocadherin locus. Comparative analysis using human cortical organoids mirrors the consequences induced by this mutation. In summary, this study elucidates the influence of the CTCFR567W mutation on human neurodevelopmental disorders, paving the way for potential therapeutic interventions.


Sujet(s)
Facteur de liaison à la séquence CCCTC , Troubles du développement neurologique , Organoïdes , Facteur de liaison à la séquence CCCTC/métabolisme , Facteur de liaison à la séquence CCCTC/génétique , Humains , Animaux , Souris , Troubles du développement neurologique/génétique , Organoïdes/métabolisme , Mutation , Neurones GABAergiques/métabolisme , Neurones GABAergiques/anatomopathologie , Mâle , Chromatine/métabolisme , Chromatine/génétique , Femelle , Encéphale/métabolisme , Encéphale/anatomopathologie , Mutation ponctuelle , Cellules souches embryonnaires humaines/métabolisme
2.
J Neurodev Disord ; 16(1): 37, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970057

RÉSUMÉ

BACKGROUND: A sizeable proportion of pathogenic genetic variants identified in young children tested for congenital differences are associated with neurodevelopmental psychiatric disorders (NPD). In this growing group, a genetic diagnosis often precedes the emergence of diagnosable developmental concerns. Here, we describe DAGSY (Developmental Assessment of Genetically Susceptible Youth), a novel interdisciplinary 'genetic-diagnosis-first' clinic integrating psychiatric, psychological and genetic expertise, and report our first observations and feedback from families and referring clinicians. METHODS: We retrieved data on referral sources and indications, genetic and NPD diagnoses and recommendations for children seen at DAGSY between 2018 and 2022. Through a survey, we obtained feedback from twenty families and eleven referring clinicians. RESULTS: 159 children (mean age 10.2 years, 57.2% males) completed an interdisciplinary (psychiatry, psychology, genetic counselling) DAGSY assessment during this period. Of these, 69.8% had a pathogenic microdeletion or microduplication, 21.5% a sequence-level variant, 4.4% a chromosomal disorder, and 4.4% a variant of unknown significance with emerging evidence of pathogenicity. One in four children did not have a prior NPD diagnosis, and referral to DAGSY was motivated by their genetic vulnerability alone. Following assessment, 76.7% received at least one new NPD diagnosis, most frequently intellectual disability (24.5%), anxiety (20.7%), autism spectrum (18.9%) and specific learning (16.4%) disorder. Both families and clinicians responding to our survey expressed satisfaction, but also highlighted some areas for potential improvement. CONCLUSIONS: DAGSY addresses an unmet clinical need for children identified with genetic variants that confer increased vulnerability for NPD and provides a crucial platform for research in this area. DAGSY can serve as a model for interdisciplinary clinics integrating child psychiatry, psychology and genetics, addressing both clinical and research needs for this emerging population.


Sujet(s)
Troubles mentaux , Troubles du développement neurologique , Humains , Enfant , Troubles du développement neurologique/génétique , Femelle , Mâle , Troubles mentaux/génétique , Prédisposition génétique à une maladie , Adolescent
3.
Transl Psychiatry ; 14(1): 259, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890284

RÉSUMÉ

A range of rare mutations involving micro-deletion or -duplication of genetic material (copy number variants (CNVs)) have been associated with high neurodevelopmental and psychiatric risk (ND-CNVs). Irritability is frequently observed in childhood neurodevelopmental conditions, yet its aetiology is largely unknown. Genetic variation may play a role, but there is a sparsity of studies investigating the presentation of irritability in young people with ND-CNVs. This study aimed to investigate whether there is a difference in irritability in young people with rare ND-CNVs compared to those without ND-CNVs, and to what extent irritability is associated with psychiatric diagnoses and cognitive ability (IQ). Irritability and broader psychopathology were assessed in 485 young people with ND-CNVs and 164 sibling controls, using the child and adolescent psychiatric assessment. Autism was assessed using the social communication questionnaire, and intelligence quotient (IQ) by the Wechsler abbreviated scale of intelligence. Fifty four percent of young people with ND-CNVs met the threshold for irritability; significantly more than controls (OR = 3.77, CI = 3.07-7.90, p = 5.31 × 10-11). When controlling for the presence of other psychiatric comorbidities, ND-CNV status was still associated with irritability. There was no evidence for a relationship between irritability and IQ. Irritability is an important aspect of the clinical picture in young people with ND-CNVs. This work shows that genetic variation is associated with irritability in young people with ND-CNVs, independent of psychiatric comorbidities or IQ impairment. Clinicians should be aware of this increased risk to inform management and interventions.


Sujet(s)
Variations de nombre de copies de segment d'ADN , Humeur irritable , Troubles du développement neurologique , Humains , Mâle , Femelle , Adolescent , Troubles du développement neurologique/génétique , Enfant , Intelligence/génétique , Études cas-témoins , Fratrie
4.
Learn Mem ; 31(5)2024 May.
Article de Anglais | MEDLINE | ID: mdl-38876485

RÉSUMÉ

The common fruit fly Drosophila melanogaster provides a powerful platform to investigate the genetic, molecular, cellular, and neural circuit mechanisms of behavior. Research in this model system has shed light on multiple aspects of brain physiology and behavior, from fundamental neuronal function to complex behaviors. A major anatomical region that modulates complex behaviors is the mushroom body (MB). The MB integrates multimodal sensory information and is involved in behaviors ranging from sensory processing/responses to learning and memory. Many genes that underlie brain disorders are conserved, from flies to humans, and studies in Drosophila have contributed significantly to our understanding of the mechanisms of brain disorders. Genetic mutations that mimic human diseases-such as Fragile X syndrome, neurofibromatosis type 1, Parkinson's disease, and Alzheimer's disease-affect MB structure and function, altering behavior. Studies dissecting the effects of disease-causing mutations in the MB have identified key pathological mechanisms, and the development of a complete connectome promises to add a comprehensive anatomical framework for disease modeling. Here, we review Drosophila models of human neurodevelopmental and neurodegenerative disorders via the effects of their underlying mutations on MB structure, function, and the resulting behavioral alterations.


Sujet(s)
Modèles animaux de maladie humaine , Corps pédonculés , Maladies neurodégénératives , Troubles du développement neurologique , Animaux , Corps pédonculés/physiologie , Maladies neurodégénératives/physiopathologie , Maladies neurodégénératives/génétique , Maladies neurodégénératives/anatomopathologie , Troubles du développement neurologique/génétique , Troubles du développement neurologique/physiopathologie , Drosophila melanogaster , Humains , Drosophila
5.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891827

RÉSUMÉ

In this Special Issue, we focus on the complex mechanisms underlying neurodevelopmental disorders (as delineated in the DSM-5), which are a group of neurological disorders that begin in childhood but significantly impact adult life [...].


Sujet(s)
Épigenèse génétique , Troubles du développement neurologique , Humains , Troubles du développement neurologique/génétique , Troubles du développement neurologique/thérapie
6.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891897

RÉSUMÉ

Heterozygous mutations in the FOXP1 gene (OMIM#605515) are responsible for a well-characterized neurodevelopmental syndrome known as "intellectual developmental disorder with language impairment with or without autistic features" (OMIM#613670) or FOXP1 syndrome for short. The main features of the condition are global developmental delay/intellectual disability; speech impairment in all individuals, regardless of their level of cognitive abilities; behavioral abnormalities; congenital anomalies, including subtle dysmorphic features; and strabismus, brain, cardiac, and urogenital abnormalities. Here, we present two siblings with a de novo heterozygous FOXP1 variant, namely, a four-year-old boy and 14-month-old girl. Both children have significantly delayed early psychomotor development, hypotonia, and very similar, slightly dysmorphic facial features. A lack of expressive speech was the leading symptom in the case of the four-year-old boy. We performed whole-exome sequencing on the male patient, which identified a pathogenic heterozygous c.1541G>A (p.Arg514His) FOXP1 mutation. His sister's targeted mutation analysis also showed the same heterozygous FOXP1 variant. Segregation analysis revealed the de novo origin of the mutation, suggesting the presence of parental gonadal mosaicism. To the best of our knowledge, this is the first report of gonadal mosaicism in FOXP1-related neurodevelopmental disorders in the medical literature.


Sujet(s)
Facteurs de transcription Forkhead , Mosaïcisme , Troubles du développement neurologique , Protéines de répression , Humains , Facteurs de transcription Forkhead/génétique , Mâle , Femelle , Enfant d'âge préscolaire , Troubles du développement neurologique/génétique , Troubles du développement neurologique/diagnostic , Nourrisson , Protéines de répression/génétique , Mutation , , Hétérozygote
7.
Mol Autism ; 15(1): 25, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849897

RÉSUMÉ

BACKGROUND: Autism and different neurodevelopmental conditions frequently co-occur, as do their symptoms at sub-diagnostic threshold levels. Overlapping traits and shared genetic liability are potential explanations. METHODS: In the population-based Norwegian Mother, Father, and Child Cohort study (MoBa), we leverage item-level data to explore the phenotypic factor structure and genetic architecture underlying neurodevelopmental traits at age 3 years (N = 41,708-58,630) using maternal reports on 76 items assessing children's motor and language development, social functioning, communication, attention, activity regulation, and flexibility of behaviors and interests. RESULTS: We identified 11 latent factors at the phenotypic level. These factors showed associations with diagnoses of autism and other neurodevelopmental conditions. Most shared genetic liabilities with autism, ADHD, and/or schizophrenia. Item-level GWAS revealed trait-specific genetic correlations with autism (items rg range = - 0.27-0.78), ADHD (items rg range = - 0.40-1), and schizophrenia (items rg range = - 0.24-0.34). We find little evidence of common genetic liability across all neurodevelopmental traits but more so for several genetic factors across more specific areas of neurodevelopment, particularly social and communication traits. Some of these factors, such as one capturing prosocial behavior, overlap with factors found in the phenotypic analyses. Other areas, such as motor development, seemed to have more heterogenous etiology, with specific traits showing a less consistent pattern of genetic correlations with each other. CONCLUSIONS: These exploratory findings emphasize the etiological complexity of neurodevelopmental traits at this early age. In particular, diverse associations with neurodevelopmental conditions and genetic heterogeneity could inform follow-up work to identify shared and differentiating factors in the early manifestations of neurodevelopmental traits and their relation to autism and other neurodevelopmental conditions. This in turn could have implications for clinical screening tools and programs.


Sujet(s)
Phénotype , Humains , Norvège , Femelle , Mâle , Enfant d'âge préscolaire , Études de cohortes , Troubles du développement neurologique/génétique , Troubles du développement neurologique/diagnostic , Mères , Trouble autistique/génétique , Prédisposition génétique à une maladie , Adulte , Pères , Étude d'association pangénomique , Trouble déficitaire de l'attention avec hyperactivité/génétique , Trouble déficitaire de l'attention avec hyperactivité/diagnostic , Schizophrénie/génétique , Hétérogénéité génétique
8.
Am J Psychiatry ; 181(6): 482-492, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38822584

RÉSUMÉ

Schizophrenia is routinely referred to as a neurodevelopmental disorder, but the role of brain development in a disorder typically diagnosed during early adult life is enigmatic. The authors revisit the neurodevelopmental model of schizophrenia with genomic insights from the most recent schizophrenia clinical genetic association studies, transcriptomic and epigenomic analyses from human postmortem brain studies, and analyses from cellular models that recapitulate neurodevelopment. Emerging insights into schizophrenia genetic risk continue to converge on brain development, particularly stages of early brain development, that may be perturbed to deviate from a typical, normative course, resulting in schizophrenia clinical symptomatology. As the authors explicate, schizophrenia genetic risk is likely dynamic and context dependent, with effects of genetic risk varying spatiotemporally, across the neurodevelopmental continuum. Optimizing therapeutic strategies for the heterogeneous collective of individuals with schizophrenia may likely be guided by leveraging markers of genetic risk and derivative functional insights, well before the emergence of psychosis. Ultimately, rather than a focus on therapeutic intervention during adolescence or adulthood, principles of prediction and prophylaxis in the pre- and perinatal and neonatal stages may best comport with the biology of schizophrenia to address the early-stage perturbations that alter the normative neurodevelopmental trajectory.


Sujet(s)
Prédisposition génétique à une maladie , Schizophrénie , Humains , Schizophrénie/génétique , Schizophrénie/étiologie , Prédisposition génétique à une maladie/génétique , Encéphale/anatomopathologie , Troubles du développement neurologique/génétique , Troubles du développement neurologique/étiologie
9.
Methods Mol Biol ; 2799: 1-11, 2024.
Article de Anglais | MEDLINE | ID: mdl-38727899

RÉSUMÉ

N-methyl-D-aspartate receptors (NMDAR) are ligand-gated ion channels mediating excitatory neurotransmission and are important for normal brain development, cognitive abilities, and motor functions. Pathogenic variants in the Glutamate receptor Ionotropic N-methyl-D-aspartate (GRIN) genes (GRIN1, GRIN2A-D) encoding NMDAR subunits have been associated with a wide spectrum of neurodevelopmental disorders and epilepsies ranging from treatable focal epilepsies to devastating early-onset developmental and epileptic encephalopathies. Genetic variants in NMDA receptor genes can cause a range of complex alterations to receptor properties resulting in various degrees of loss-of-function, gain-of-function, or mixtures thereof. Understanding how genetic variants affect the function of the receptors, therefore, represents an important first step in the ongoing development towards targeted therapies. Currently, targeted treatment options for GRIN-related diseases are limited. However, treatment with memantine has been reported to significantly reduce seizure frequency in a few individuals with developmental and epileptic encephalopathies harboring de novo gain-of-function GRIN2A missense variants, and supplementary treatment with L-serine has been associated with improved motor and cognitive performance as well as reduced seizure frequency in patients with GRIN2B loss-of-function missense variants as well as GRIN2A and GRIN2B null variants.


Sujet(s)
Épilepsie , Troubles du développement neurologique , Récepteurs du N-méthyl-D-aspartate , Récepteurs du N-méthyl-D-aspartate/génétique , Récepteurs du N-méthyl-D-aspartate/métabolisme , Humains , Troubles du développement neurologique/génétique , Épilepsie/génétique , Épilepsie/traitement médicamenteux , Prédisposition génétique à une maladie , Variation génétique , Mémantine/usage thérapeutique , Mémantine/pharmacologie
10.
Arch Pediatr ; 31(5): 320-325, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38719651

RÉSUMÉ

INTRODUCTION: KBG syndrome is an autosomal dominant, polymalformative genetic syndrome that is mainly associated with neurodevelopmental and learning disorders, intellectual disability, behavioral disorders, and epilepsy as well as characteristic dysmorphic features, short stature, and ENT (ear, nose, and throat) abnormalities. However, the diagnostic pathway of these individuals is an element that has not been broadly evaluated. The main aim of this study was therefore to characterize the diagnostic pathway for these individuals, by assessing the different healthcare professionals involved and the main referral elements. METHOD: This was a multicenter, retrospective, descriptive study. A cohort of 30 individuals with KBG syndrome who were followed up at Poitiers University Hospital and Bordeaux University Hospital we recruited. RESULTS: Pediatricians were the main healthcare professionals who referred individuals for genetic consultation, and the main reason for referral was an assessment of learning delays or intellectual disability, in association with other abnormalities. CONCLUSION: Pediatricians play a crucial role in the diagnostic guidance of individuals with KBG syndrome, and the main reason for referral remains the assessment of a learning delay or intellectual disability. Healthcare professionals must therefore remain attentive to the child's development and the various anomalies associated with it, in particular characteristic dysmorphic features, behavioral disorders, and statural growth.


Sujet(s)
Troubles du développement neurologique , Humains , Études rétrospectives , Femelle , Mâle , Enfant , Troubles du développement neurologique/diagnostic , Troubles du développement neurologique/génétique , Enfant d'âge préscolaire , Orientation vers un spécialiste/statistiques et données numériques , Déficience intellectuelle/génétique , Déficience intellectuelle/diagnostic , Adolescent , Nourrisson , Pédiatres/psychologie , Malformations multiples/génétique , Malformations multiples/diagnostic , Malformations multiples/psychologie
11.
Eur J Hum Genet ; 32(7): 879-883, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38702431

RÉSUMÉ

Numerous large scale genomic studies have uncovered rare but recurrent pathogenetic variants in a significant number of genes encoding epigenetic machinery in cases with neurodevelopmental disorders (NDD) especially autism spectrum disorder (ASD). These findings provide strong support for the functional importance of epigenetic regulators in neurodevelopment. After the clinical genomics evaluation of the patients using exome sequencing, we have identified, three novel protein-truncating variants (PTVs) in the MSL2 gene (OMIM: 614802) which encodes a chromatin modifying enzyme. MSL2 modifies chromatin through both mono-ubiquitination of histone 2B on lysine 34 (K34) and acetylation of histone H4 on lysine 16 (K16). We reported first time the detailed clinical features associated with 3 MSL2 PTVs. There are 15 PTVs (13 de novo) reported from the large genomics studies (12 cases) or ClinVar (3 cases) of NDD, ASD, and developmental disorders (DD) but the specific clinical features for these cases are not described. Taken together, our descriptions of dysmorphic face and other features support the causal role of MSL2 in a likely syndromic neurodevelopmental disorder and add MSL2 to a growing list of epigenetic genes implicated in ASD.


Sujet(s)
Trouble du spectre autistique , Humains , Mâle , Trouble du spectre autistique/génétique , Femelle , Enfant , Troubles du développement neurologique/génétique , Troubles du développement neurologique/anatomopathologie , Chromatine/génétique , Chromatine/métabolisme , Enfant d'âge préscolaire , Syndrome , Mutation
12.
Neurobiol Dis ; 197: 106525, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38729272

RÉSUMÉ

RNA-binding proteins (RBPs) bind to RNAs and are crucial for regulating RNA splicing, stability, translation, and transport. Among these proteins, the CUGBP Elav-like family (CELF) is a highly conserved group crucial for posttranscriptional regulation by binding to CUG repeats. Comprising CELF1-6, this family exhibits diverse expression patterns and functions. Dysregulation of CELF has been implicated in various neural disorders, encompassing both neurodegenerative and neurodevelopmental conditions, such as Alzheimer's disease and autism. This article aims to provide a comprehensive summary of the CELF family's role in neurodevelopment and neurodevelopmental disorders. Understanding CELF's mechanisms may offer clues for potential therapeutic strategies by regulating their targets in neurodevelopmental disorders.


Sujet(s)
Protéines CELF , Troubles du développement neurologique , Humains , Troubles du développement neurologique/génétique , Animaux , Protéines CELF/métabolisme , Protéines CELF/génétique
14.
Hum Genet ; 143(6): 761-773, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38787418

RÉSUMÉ

Chung-Jansen syndrome is a neurodevelopmental disorder characterized by intellectual disability, behavioral problems, obesity and dysmorphic features. It is caused by pathogenic variants in the PHIP gene that encodes for the Pleckstrin homology domain-interacting protein, which is part of an epigenetic modifier protein complex. Therefore, we hypothesized that PHIP haploinsufficiency may impact genome-wide DNA methylation (DNAm). We assessed the DNAm profiles of affected individuals with pathogenic and likely pathogenic PHIP variants with Infinium Methylation EPIC arrays and report a specific and sensitive DNAm episignature biomarker for Chung-Jansen syndrome. In addition, we observed similarities between the methylation profile of Chung-Jansen syndrome and that of functionally related and clinically partially overlapping genetic disorders, White-Kernohan syndrome (caused by variants in DDB1 gene) and Börjeson-Forssman-Lehmann syndrome (caused by variants in PHF6 gene). Based on these observations we also proceeded to develop a common episignature biomarker for these disorders. These newly defined episignatures can be used as part of a multiclass episignature classifier for screening of affected individuals with rare disorders and interpretation of genetic variants of unknown clinical significance, and provide further insights into the common molecular pathophysiology of the clinically-related Chung-Jansen, Börjeson-Forssman-Lehmann and White-Kernohan syndromes.


Sujet(s)
Méthylation de l'ADN , Déficience intellectuelle , Humains , Déficience intellectuelle/génétique , Déficience intellectuelle/diagnostic , Mâle , Femelle , Haploinsuffisance/génétique , Troubles du développement neurologique/génétique , Troubles du développement neurologique/diagnostic , Enfant
15.
Neurology ; 102(11): e209413, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38759134

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Knowledge of young-onset Alzheimer disease in adults with Down syndrome has greatly improved clinical care. However, little is known about dementia in rare genetic neurodevelopmental disorders (RGNDs). In this review, a comprehensive overview is provided of reports on dementia and cognitive/adaptive trajectories in adults with RGNDs. METHODS: A systematic literature review was conducted in Embase, Medline ALL, and PsycINFO on December 6, 2022. The protocol was registered in PROSPERO (CRD42021223041). Search terms for dementia, cognitive and adaptive functioning, and RGNDs were combined using generic terms and the Orphanet database. Study characteristics and descriptive data on genetic diagnosis, clinical and neuropathologic features, comorbidities, and diagnostic methods were extracted using a modified version of the Cochrane Data Extraction Template. RESULTS: The literature search yielded 40 publications (17 cohorts, 23 case studies) describing dementia and/or cognitive or adaptive trajectories in adults with 14 different RGNDs. Dementia was reported in 49 individuals (5 cohorts, 20 cases) with a mean age at onset of 44.4 years. Diagnostics were not disclosed for half of the reported individuals (n = 25/49, 51.0%). A total of 44 different psychodiagnostic instruments were used. MRI was the most reported additional investigation (n = 12/49, 24.5%). Comorbid disorders most frequently associated with cognitive/adaptive decline were epilepsy, psychotic disorders, and movement disorders. DISCUSSION: Currently available literature shows limited information on aging in RGNDs, with relatively many reports of young-onset dementia. Longitudinal data may provide insights into converging neurodevelopmental degenerative pathways. We provide recommendations to optimize dementia screening, diagnosis, and research.


Sujet(s)
Démence , Troubles du développement neurologique , Humains , Démence/génétique , Démence/épidémiologie , Démence/diagnostic , Troubles du développement neurologique/génétique , Troubles du développement neurologique/diagnostic , Maladies rares/génétique , Adulte
16.
Am J Hum Genet ; 111(6): 1206-1221, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38772379

RÉSUMÉ

Utilizing trio whole-exome sequencing and a gene matching approach, we identified a cohort of 18 male individuals from 17 families with hemizygous variants in KCND1, including two de novo missense variants, three maternally inherited protein-truncating variants, and 12 maternally inherited missense variants. Affected subjects present with a neurodevelopmental disorder characterized by diverse neurological abnormalities, mostly delays in different developmental domains, but also distinct neuropsychiatric signs and epilepsy. Heterozygous carrier mothers are clinically unaffected. KCND1 encodes the α-subunit of Kv4.1 voltage-gated potassium channels. All variant-associated amino acid substitutions affect either the cytoplasmic N- or C-terminus of the channel protein except for two occurring in transmembrane segments 1 and 4. Kv4.1 channels were functionally characterized in the absence and presence of auxiliary ß subunits. Variant-specific alterations of biophysical channel properties were diverse and varied in magnitude. Genetic data analysis in combination with our functional assessment shows that Kv4.1 channel dysfunction is involved in the pathogenesis of an X-linked neurodevelopmental disorder frequently associated with a variable neuropsychiatric clinical phenotype.


Sujet(s)
Troubles du développement neurologique , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Mâle , Épilepsie/génétique , , Maladies génétiques liées au chromosome X/génétique , Hétérozygote , Mutation faux-sens/génétique , Troubles du développement neurologique/génétique , Pedigree , Phénotype , Canaux potassiques Shal/génétique
17.
Am J Hum Genet ; 111(6): 1222-1238, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38781976

RÉSUMÉ

Heterozygous variants in SLC6A1, encoding the GAT-1 GABA transporter, are associated with seizures, developmental delay, and autism. The majority of affected individuals carry missense variants, many of which are recurrent germline de novo mutations, raising the possibility of gain-of-function or dominant-negative effects. To understand the functional consequences, we performed an in vitro GABA uptake assay for 213 unique variants, including 24 control variants. De novo variants consistently resulted in a decrease in GABA uptake, in keeping with haploinsufficiency underlying all neurodevelopmental phenotypes. Where present, ClinVar pathogenicity reports correlated well with GABA uptake data; the functional data can inform future reports for the remaining 72% of unscored variants. Surface localization was assessed for 86 variants; two-thirds of loss-of-function missense variants prevented GAT-1 from being present on the membrane while GAT-1 was on the surface but with reduced activity for the remaining third. Surprisingly, recurrent de novo missense variants showed moderate loss-of-function effects that reduced GABA uptake with no evidence for dominant-negative or gain-of-function effects. Using linear regression across multiple missense severity scores to extrapolate the functional data to all potential SLC6A1 missense variants, we observe an abundance of GAT-1 residues that are sensitive to substitution. The extent of this missense vulnerability accounts for the clinically observed missense enrichment; overlap with hypermutable CpG sites accounts for the recurrent missense variants. Strategies to increase the expression of the wild-type SLC6A1 allele are likely to be beneficial across neurodevelopmental disorders, though the developmental stage and extent of required rescue remain unknown.


Sujet(s)
Transporteurs de GABA , Haploinsuffisance , Mutation faux-sens , Humains , Transporteurs de GABA/génétique , Haploinsuffisance/génétique , Acide gamma-amino-butyrique/métabolisme , Troubles du développement neurologique/génétique , Incapacités de développement/génétique , Trouble autistique/génétique , Cellules HEK293
19.
Sci Rep ; 14(1): 11239, 2024 05 16.
Article de Anglais | MEDLINE | ID: mdl-38755281

RÉSUMÉ

While short-read sequencing currently dominates genetic research and diagnostics, it frequently falls short of capturing certain structural variants (SVs), which are often implicated in the etiology of neurodevelopmental disorders (NDDs). Optical genome mapping (OGM) is an innovative technique capable of capturing SVs that are undetectable or challenging-to-detect via short-read methods. This study aimed to investigate NDDs using OGM, specifically focusing on cases that remained unsolved after standard exome sequencing. OGM was performed in 47 families using ultra-high molecular weight DNA. Single-molecule maps were assembled de novo, followed by SV and copy number variant calling. We identified 7 variants of interest, of which 5 (10.6%) were classified as likely pathogenic or pathogenic, located in BCL11A, OPHN1, PHF8, SON, and NFIA. We also identified an inversion disrupting NAALADL2, a gene which previously was found to harbor complex rearrangements in two NDD cases. Variants in known NDD genes or candidate variants of interest missed by exome sequencing mainly consisted of larger insertions (> 1kbp), inversions, and deletions/duplications of a low number of exons (1-4 exons). In conclusion, in addition to improving molecular diagnosis in NDDs, this technique may also reveal novel NDD genes which may harbor complex SVs often missed by standard sequencing techniques.


Sujet(s)
Cartographie chromosomique , Variations de nombre de copies de segment d'ADN , Troubles du développement neurologique , Humains , Troubles du développement neurologique/génétique , Troubles du développement neurologique/diagnostic , Femelle , Mâle , Cartographie chromosomique/méthodes , /méthodes , Enfant , Variation structurale du génome , Enfant d'âge préscolaire
20.
Psychiatr Genet ; 34(3): 74-80, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38690959

RÉSUMÉ

BACKGROUND: Biallelic loss-of-function variants in SMPD4 cause a rare and severe neurodevelopmental disorder. These variants have been identified in a group of children with neurodevelopmental disorders with microcephaly, arthrogryposis, and structural brain anomalies. SMPD4 encodes a sphingomyelinase that hydrolyzes sphingomyelin into ceramide at neutral pH and can thereby affect membrane lipid homeostasis. SMPD4 localizes to the membranes of the endoplasmic reticulum and nuclear envelope and interacts with nuclear pore complexes. MATERIALS AND METHODS: For the efficient prenatal diagnosis of rare and undiagnosed diseases, the parallel detection of copy number variants (CNVs) and single nucleotide variants using whole-exome analysis is required. A physical examination of the parents was performed. Karyotype and whole-exome analysis were performed for the fetus and the parents. RESULTS: A fetus with microcephaly and arthrogryposis; biallelic null variants (c.387-1G>A; Chr2[GRCh38]: g.130142742_130202459del) were detected by whole-exome sequencing (WES). We have reported for the first time the biallelic loss-of-function mutations in SMPD4 in patients born to unrelated parents in China. CONCLUSION: WES could replace chromosomal microarray analysis and copy number variation sequencing as a more cost-effective genetic test for detecting CNVs and diagnosing highly heterogeneous conditions.


Sujet(s)
Variations de nombre de copies de segment d'ADN , , Microcéphalie , Polymorphisme de nucléotide simple , Diagnostic prénatal , Sphingomyeline phosphodiesterase , Humains , Variations de nombre de copies de segment d'ADN/génétique , /méthodes , Femelle , Diagnostic prénatal/méthodes , Sphingomyeline phosphodiesterase/génétique , Polymorphisme de nucléotide simple/génétique , Grossesse , Microcéphalie/génétique , Hétérozygote , Arthrogrypose/génétique , Arthrogrypose/diagnostic , Mâle , Exome/génétique , Mutation/génétique , Troubles du développement neurologique/génétique , Troubles du développement neurologique/diagnostic
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...