Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 452
Filtrar
1.
Genes (Basel) ; 15(4)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38674358

RESUMEN

Pathogenic ASH1L variants have been reported in probands with broad phenotypic presentations, including intellectual disability, autism spectrum disorder, attention deficit hyperactivity disorder, seizures, congenital anomalies, and other skeletal, muscular, and sleep differences. Here, we review previously published individuals with pathogenic ASH1L variants and report three further probands with novel ASH1L variants and previously unreported phenotypic features, including mixed receptive language disorder and gait disturbances. These novel data from the Brain Gene Registry, an accessible repository of clinically derived genotypic and phenotypic data, have allowed for the expansion of the phenotypic and genotypic spectrum of this condition.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Trastornos del Neurodesarrollo , Fenotipo , Humanos , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Masculino , N-Metiltransferasa de Histona-Lisina/genética , Femenino , Niño , Genotipo , Proteínas de Unión al ADN/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Factores de Transcripción/genética , Preescolar , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Mutación , Adolescente
2.
Elife ; 132024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38655849

RESUMEN

Mutations in the human PURA gene cause the neurodevelopmental PURA syndrome. In contrast to several other monogenetic disorders, almost all reported mutations in this nucleic acid-binding protein result in the full disease penetrance. In this study, we observed that patient mutations across PURA impair its previously reported co-localization with processing bodies. These mutations either destroyed the folding integrity, RNA binding, or dimerization of PURA. We also solved the crystal structures of the N- and C-terminal PUR domains of human PURA and combined them with molecular dynamics simulations and nuclear magnetic resonance measurements. The observed unusually high dynamics and structural promiscuity of PURA indicated that this protein is particularly susceptible to mutations impairing its structural integrity. It offers an explanation why even conservative mutations across PURA result in the full penetrance of symptoms in patients with PURA syndrome.


PURA syndrome is a neurodevelopmental disorder that affects about 650 patients worldwide, resulting in a range of symptoms including neurodevelopmental delays, intellectual disability, muscle weakness, seizures, and eating difficulties. The condition is caused by a mutated gene that codes for a protein called PURA. PURA binds RNA ­ the molecule that carries genetic information so it can be translated into proteins ­ and has roles in regulating the production of new proteins. Contrary to other conditions that result from mutations in a single gene, PURA syndrome patients show 'high penetrance', meaning almost every reported mutation in the gene leads to symptoms. Proske, Janowski et al. wanted to understand the molecular basis for this high penetrance. To find out more, the researchers first examined how patient mutations affected the location of the PURA in the cell, using human cells grown in the laboratory. Normally, PURA travels to P-bodies, which are groupings of RNA and proteins involved in regulating which genes get translated into proteins. The researchers found that in cells carrying PURA syndrome mutations, PURA failed to move adequately to P-bodies. To find out how this 'mislocalization' might happen, Proske, Janowski et al. tested how different mutations affected the three-dimensional folding of PURA. These analyses showed that the mutations impair the protein's folding and thereby disrupt PURA's ability to bind RNA, which may explain why mutant PURA cannot localize correctly. Proske, Janowski et al. describe the molecular abnormalities of PURA underlying this disorder and show how molecular analysis of patient mutations can reveal the mechanisms of a disease at the cell level. The results show that the impact of mutations on the structural integrity of the protein, which affects its ability to bind RNA, are likely key to the symptoms of the syndrome. Additionally, their approach used establishes a way to predict and test mutations that will cause PURA syndrome. This may help to develop diagnostic tools for this condition.


Asunto(s)
Trastornos del Neurodesarrollo , Cuerpos de Procesamiento , Humanos , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Cuerpos de Procesamiento/metabolismo , Cuerpos de Procesamiento/patología , Gránulos de Estrés/metabolismo , Cristalografía por Rayos X , Dimerización , Dominios Proteicos , Dicroismo Circular , Proteínas Recombinantes , Pliegue de Proteína , Penetrancia , Sustitución de Aminoácidos , Mutación Puntual , Células HeLa
3.
Dis Model Mech ; 17(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38566589

RESUMEN

The addition of O-linked ß-N-acetylglucosamine (O-GlcNAc) to proteins (referred to as O-GlcNAcylation) is a modification that is crucial for vertebrate development. O-GlcNAcylation is catalyzed by O-GlcNAc transferase (OGT) and reversed by O-GlcNAcase (OGA). Missense variants of OGT have recently been shown to segregate with an X-linked syndromic form of intellectual disability, OGT-linked congenital disorder of glycosylation (OGT-CDG). Although the existence of OGT-CDG suggests that O-GlcNAcylation is crucial for neurodevelopment and/or cognitive function, the underlying pathophysiologic mechanisms remain unknown. Here we report a mouse line that carries a catalytically impaired OGT-CDG variant. These mice show altered O-GlcNAc homeostasis with decreased global O-GlcNAcylation and reduced levels of OGT and OGA in the brain. Phenotypic characterization of the mice revealed lower body weight associated with reduced body fat mass, short stature and microcephaly. This mouse model will serve as an important tool to study genotype-phenotype correlations in OGT-CDG in vivo and for the development of possible treatment avenues for this disorder.


Asunto(s)
Modelos Animales de Enfermedad , Discapacidad Intelectual , N-Acetilglucosaminiltransferasas , Animales , N-Acetilglucosaminiltransferasas/metabolismo , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/deficiencia , Discapacidad Intelectual/genética , Encéfalo/patología , Encéfalo/metabolismo , Fenotipo , Ratones , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/enzimología , beta-N-Acetilhexosaminidasas/metabolismo , Glicosilación , Peso Corporal
4.
J Biol Chem ; 300(4): 107124, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432637

RESUMEN

Rab35 (Ras-associated binding protein) is a small GTPase that regulates endosomal membrane trafficking and functions in cell polarity, cytokinesis, and growth factor signaling. Altered Rab35 function contributes to progression of glioblastoma, defects in primary cilia formation, and altered cytokinesis. Here, we report a pediatric patient with global developmental delay, hydrocephalus, a Dandy-Walker malformation, axial hypotonia with peripheral hypertonia, visual problems, and conductive hearing impairment. Exome sequencing identified a homozygous missense variant in the GTPase fold of RAB35 (c.80G>A; p.R27H) as the most likely candidate. Functional analysis of the R27H-Rab35 variant protein revealed enhanced interaction with its guanine-nucleotide exchange factor, DENND1A and decreased interaction with a known effector, MICAL1, indicating that the protein is in an inactive conformation. Cellular expression of the variant drives the activation of Arf6, a small GTPase under negative regulatory control of Rab35. Importantly, variant expression leads to delayed cytokinesis and altered length, number, and Arl13b composition of primary cilia, known factors in neurodevelopmental disease. Our findings provide evidence of altered Rab35 function as a causative factor of a neurodevelopmental disorder.


Asunto(s)
Mutación Missense , Trastornos del Neurodesarrollo , Proteínas de Unión al GTP rab , Femenino , Humanos , Masculino , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Línea Celular , Cilios/metabolismo , Cilios/genética , Cilios/patología , Citocinesis/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Mutación con Pérdida de Función , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Linaje , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Modelos Moleculares , Estructura Terciaria de Proteína
5.
Am J Med Genet A ; 194(6): e63555, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38326731

RESUMEN

Heterozygous pathogenic variants in KDM6B have recently been associated to a rare neurodevelopmental disorder referred to as "Neurodevelopmental disorder with coarse facies and mild distal skeletal abnormalities" and characterized by non-pathognomonic facial and body dysmorphisms, a wide range of neurodevelopmental and behavioral disorders and nonspecific neuroradiological findings. KDM6B encodes a histone demethylase, expressed in different tissues during development, which regulates gene expression through the modulation of chromatin accessibility by RNA polymerase. We herein describe a 11-year-old male patient carrying a novel de novo pathogenic variant in KDM6B exhibiting facial dysmorphisms, dysgraphia, behavioral traits relatable to oppositional defiant, autism spectrum, and attention deficit hyperactivity disorders, a single seizure episode, and a neuroimaging finding of a single cerebellar heterotopic nodule, never described to date in this genetic condition. These findings expand the phenotypic spectrum of this syndrome, highlighting the potential role for KDM6B in cerebellar development and providing valuable insights for genetic counseling.


Asunto(s)
Cerebelo , Histona Demetilasas con Dominio de Jumonji , Trastornos del Neurodesarrollo , Humanos , Masculino , Niño , Histona Demetilasas con Dominio de Jumonji/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Cerebelo/anomalías , Cerebelo/patología , Cerebelo/diagnóstico por imagen , Fenotipo , Mutación/genética
6.
Genet Med ; 26(5): 101097, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38334070

RESUMEN

PURPOSE: Pathogenic variants of FIG4 generate enlarged lysosomes and neurological and developmental disorders. To identify additional genes regulating lysosomal volume, we carried out a genome-wide activation screen to detect suppression of enlarged lysosomes in FIG4-/- cells. METHODS: The CRISPR-a gene activation screen utilized sgRNAs from the promoters of protein-coding genes. Fluorescence-activated cell sorting separated cells with correction of the enlarged lysosomes from uncorrected cells. Patient variants of SLC12A9 were identified by exome or genome sequencing and studied by segregation analysis and clinical characterization. RESULTS: Overexpression of SLC12A9, a solute co-transporter, corrected lysosomal swelling in FIG4-/- cells. SLC12A9 (NP_064631.2) colocalized with LAMP2 at the lysosome membrane. Biallelic variants of SLC12A9 were identified in 3 unrelated probands with neurodevelopmental disorders. Common features included intellectual disability, skeletal and brain structural abnormalities, congenital heart defects, and hypopigmented hair. Patient 1 was homozygous for nonsense variant p.(Arg615∗), patient 2 was compound heterozygous for p.(Ser109Lysfs∗20) and a large deletion, and proband 3 was compound heterozygous for p.(Glu290Glyfs∗36) and p.(Asn552Lys). Fibroblasts from proband 1 contained enlarged lysosomes that were corrected by wild-type SLC12A9 cDNA. Patient variant p.(Asn552Lys) failed to correct the lysosomal defect. CONCLUSION: Impaired function of SLC12A9 results in enlarged lysosomes and a recessive disorder with a recognizable neurodevelopmental phenotype.


Asunto(s)
Mutación con Pérdida de Función , Lisosomas , Trastornos del Neurodesarrollo , Humanos , Lisosomas/metabolismo , Lisosomas/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Masculino , Mutación con Pérdida de Función/genética , Femenino , Alelos , Niño , Preescolar , Lactante , Fenotipo , Linaje
7.
Clin Genet ; 105(6): 620-629, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38356149

RESUMEN

PPP1R21 encodes for a conserved protein that is involved in endosomal maturation. Biallelic pathogenic variants in PPP1R21 have been associated with a syndromic neurodevelopmental disorder from studying 13 affected individuals. In this report, we present 11 additional individuals from nine unrelated families and their clinical, radiological, and molecular findings. We identified eight different variants in PPP1R21, of which six were novel variants. Global developmental delay and hypotonia are neurological features that were observed in all individuals. There is also a similar pattern of dysmorphic features with coarse faces as a gestalt observed in several individuals. Common findings in 75% of individuals with available brain imaging include delays in myelination, wavy outline of the bodies of the lateral ventricles, and slight prominence of the bodies of the lateral ventricles. PPP1R21-related neurodevelopmental disorder is associated with a consistent phenotype and should be considered in highly consanguineous individuals presenting with developmental delay/intellectual disability along with coarse facial features.


Asunto(s)
Trastornos del Neurodesarrollo , Fenotipo , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Mutación , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Linaje
8.
Clin Genet ; 105(6): 655-660, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38384171

RESUMEN

Precise regulation of gene expression is important for correct neurodevelopment. 9q34.3 deletions affecting the EHMT1 gene result in a syndromic neurodevelopmental disorder named Kleefstra syndrome. In contrast, duplications of the 9q34.3 locus encompassing EHMT1 have been suggested to cause developmental disorders, but only limited information has been available. We have identified 15 individuals from 10 unrelated families, with 9q34.3 duplications <1.5 Mb in size, encompassing EHMT1 entirely. Clinical features included mild developmental delay, mild intellectual disability or learning problems, autism spectrum disorder, and behavior problems. The individuals did not consistently display dysmorphic features, congenital anomalies, or growth abnormalities. DNA methylation analysis revealed a weak DNAm profile for the cases with 9q34.3 duplication encompassing EHMT1, which could segregate the majority of the affected cases from controls. This study shows that individuals with 9q34.3 duplications including EHMT1 gene present with mild non-syndromic neurodevelopmental disorders and DNA methylation changes different from Kleefstra syndrome.


Asunto(s)
Deleción Cromosómica , Duplicación Cromosómica , Cromosomas Humanos Par 9 , Metilación de ADN , Cardiopatías Congénitas , N-Metiltransferasa de Histona-Lisina , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Metilación de ADN/genética , Cromosomas Humanos Par 9/genética , Masculino , Femenino , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Duplicación Cromosómica/genética , Niño , Preescolar , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Adolescente , Fenotipo
9.
Am J Med Genet A ; 194(6): e63528, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38169111

RESUMEN

Somatic variants in the NOTCH pathway regulator FBXW7 are frequently seen in a variety of malignancies. Heterozygous loss-of-function germline variants in FBXW7 have recently been described as causative for a neurodevelopmental syndrome. Independently, FBXW7 was also considered as a susceptibility gene for Wilms tumor due to a few observations of heterozygous germline variants in patients with Wilms tumor. Whether the same FBXW7 variants are implicated in both, neurodevelopmental delay and Wilms tumor formation, remained unclear. By clinical testing, we now observed a patient with neurodevelopmental delay due to a de novo constitutional mosaic FBXW7 splice site pathogenic variant who developed Wilms tumor. In the tumor, we identified a second hit frameshift variant in FBXW7. Immunohistochemical staining was consistent with mosaic loss of FBXW7 protein expression in the tumor. Our data support the role of constitutional FBXW7 pathogenic variants in both, neurodevelopmental disorder and the etiology of Wilms tumor. Therefore, Wilms tumor screening should be considered in individuals with constitutional or germline pathogenic variants in FBXW7 and associated neurodevelopmental syndrome.


Asunto(s)
Proteína 7 que Contiene Repeticiones F-Box-WD , Predisposición Genética a la Enfermedad , Tumor de Wilms , Humanos , Masculino , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Mutación del Sistema de Lectura/genética , Mutación de Línea Germinal/genética , Neoplasias Renales/genética , Neoplasias Renales/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Tumor de Wilms/genética , Tumor de Wilms/patología , Niño
10.
Genet Med ; 26(5): 101087, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38288683

RESUMEN

PURPOSE: Interneuronopathies are a group of neurodevelopmental disorders characterized by deficient migration and differentiation of gamma-aminobutyric acidergic interneurons resulting in a broad clinical spectrum, including autism spectrum disorders, early-onset epileptic encephalopathy, intellectual disability, and schizophrenic disorders. SP9 is a transcription factor belonging to the Krüppel-like factor and specificity protein family, the members of which harbor highly conserved DNA-binding domains. SP9 plays a central role in interneuron development and tangential migration, but it has not yet been implicated in a human neurodevelopmental disorder. METHODS: Cases with SP9 variants were collected through international data-sharing networks. To address the specific impact of SP9 variants, in silico and in vitro assays were carried out. RESULTS: De novo heterozygous variants in SP9 cause a novel form of interneuronopathy. SP9 missense variants affecting the glutamate 378 amino acid result in severe epileptic encephalopathy because of hypomorphic and neomorphic DNA-binding effects, whereas SP9 loss-of-function variants result in a milder phenotype with epilepsy, developmental delay, and autism spectrum disorder. CONCLUSION: De novo heterozygous SP9 variants are responsible for a neurodevelopmental disease. Interestingly, variants located in conserved DNA-binding domains of KLF/SP family transcription factors may lead to neomorphic DNA-binding functions resulting in a combination of loss- and gain-of-function effects.


Asunto(s)
Trastorno del Espectro Autista , Epilepsia , Discapacidad Intelectual , Interneuronas , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Epilepsia/genética , Epilepsia/patología , Masculino , Femenino , Interneuronas/metabolismo , Interneuronas/patología , Niño , Preescolar , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Fenotipo , Mutación Missense/genética , Heterocigoto , Adolescente , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología
11.
Am J Med Genet A ; 194(6): e63548, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38264805

RESUMEN

Pathogenic PHF21A variation causes PHF21A-related neurodevelopmental disorders (NDDs). Although amorphic alleles, including haploinsufficiency, have been established as a disease mechanism, increasing evidence suggests that missense variants as well as frameshift variants extending the BHC80 carboxyl terminus also cause disease. Expanding on these, we report a proposita with intellectual disability and overgrowth and a novel de novo heterozygous PHF21A splice variant (NM_001352027.3:c.[153+1G>C];[=]) causing skipping of exon 6, which encodes an in-frame BHC80 deletion (p.(Asn30_Gln51del)). This deletion disrupts a predicted leucine zipper domain and implicates this domain in BHC80 function and as a target of variation causing PHF21A-related NDDs. This extension of understanding emphasizes the application of RNA analysis in precision genomic medicine practice.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Empalme del ARN , Femenino , Humanos , Alelos , Exones/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Empalme del ARN/genética , Análisis de Secuencia de ARN , Niño
12.
J Neurosci ; 43(45): 7554-7564, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37940582

RESUMEN

The cerebellum, traditionally associated with motor coordination and balance, also plays a crucial role in various aspects of higher-order function and dysfunction. Emerging research has shed light on the cerebellum's broader contributions to cognitive, emotional, and reward processes. The cerebellum's influence on autonomic function further highlights its significance in regulating motivational and emotional states. Perturbations in cerebellar development and function have been implicated in various neurodevelopmental disorders, including autism spectrum disorder and attention deficit hyperactivity disorder. An increasing appreciation for neuropsychiatric symptoms that arise from cerebellar dysfunction underscores the importance of elucidating the circuit mechanisms that underlie complex interactions between the cerebellum and other brain regions for a comprehensive understanding of complex behavior. By briefly discussing new advances in mapping cerebellar function in affective, cognitive, autonomic, and social processing and reviewing the role of the cerebellum in neuropathology beyond the motor domain, this Mini-Symposium review aims to provide a broad perspective of cerebellar intersections with the limbic brain in health and disease.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno del Espectro Autista , Trastornos del Neurodesarrollo , Humanos , Cognición/fisiología , Cerebelo/fisiología , Trastornos del Neurodesarrollo/patología
13.
Nature ; 622(7982): 359-366, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37758944

RESUMEN

The assembly of cortical circuits involves the generation and migration of interneurons from the ventral to the dorsal forebrain1-3, which has been challenging to study at inaccessible stages of late gestation and early postnatal human development4. Autism spectrum disorder and other neurodevelopmental disorders (NDDs) have been associated with abnormal cortical interneuron development5, but which of these NDD genes affect interneuron generation and migration, and how they mediate these effects remains unknown. We previously developed a platform to study interneuron development and migration in subpallial organoids and forebrain assembloids6. Here we integrate assembloids with CRISPR screening to investigate the involvement of 425 NDD genes in human interneuron development. The first screen aimed at interneuron generation revealed 13 candidate genes, including CSDE1 and SMAD4. We subsequently conducted an interneuron migration screen in more than 1,000 forebrain assembloids that identified 33 candidate genes, including cytoskeleton-related genes and the endoplasmic reticulum-related gene LNPK. We discovered that, during interneuron migration, the endoplasmic reticulum is displaced along the leading neuronal branch before nuclear translocation. LNPK deletion interfered with this endoplasmic reticulum displacement and resulted in abnormal migration. These results highlight the power of this CRISPR-assembloid platform to systematically map NDD genes onto human development and reveal disease mechanisms.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Trastornos del Neurodesarrollo , Femenino , Humanos , Recién Nacido , Embarazo , Movimiento Celular/genética , Sistemas CRISPR-Cas/genética , Interneuronas/citología , Interneuronas/metabolismo , Interneuronas/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Organoides/citología , Organoides/embriología , Organoides/crecimiento & desarrollo , Organoides/metabolismo , Organoides/patología , Retículo Endoplásmico/metabolismo , Prosencéfalo/citología , Prosencéfalo/embriología , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Prosencéfalo/patología , Transporte Activo de Núcleo Celular
14.
Acta Neuropathol ; 146(5): 663-683, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37656188

RESUMEN

Microglia are the brain's resident macrophages, which guide various developmental processes crucial for brain maturation, activity, and plasticity. Microglial progenitors enter the telencephalic wall by the 4th postconceptional week and colonise the fetal brain in a manner that spatiotemporally tracks key neurodevelopmental processes in humans. However, much of what we know about how microglia shape neurodevelopment comes from rodent studies. Multiple differences exist between human and rodent microglia warranting further focus on the human condition, particularly as microglia are emerging as critically involved in the pathological signature of various cognitive and neurodevelopmental disorders. In this article, we review the evidence supporting microglial involvement in basic neurodevelopmental processes by focusing on the human species. We next concur on the neuropathological evidence demonstrating whether and how microglia contribute to the aetiology of two neurodevelopmental disorders: autism spectrum conditions and schizophrenia. Next, we highlight how recent technologies have revolutionised our understanding of microglial biology with a focus on how these tools can help us elucidate at unprecedented resolution the links between microglia and neurodevelopmental disorders. We conclude by reviewing which current treatment approaches have shown most promise towards targeting microglia in neurodevelopmental disorders and suggest novel avenues for future consideration.


Asunto(s)
Trastorno del Espectro Autista , Trastornos del Neurodesarrollo , Humanos , Microglía/patología , Trastornos del Neurodesarrollo/patología , Macrófagos/patología , Neuropatología , Encéfalo/patología
15.
Eur J Hum Genet ; 31(11): 1251-1260, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37644171

RESUMEN

Heterozygous, pathogenic CUX1 variants are associated with global developmental delay or intellectual disability. This study delineates the clinical presentation in an extended cohort and investigates the molecular mechanism underlying the disorder in a Cux1+/- mouse model. Through international collaboration, we assembled the phenotypic and molecular information for 34 individuals (23 unpublished individuals). We analyze brain CUX1 expression and susceptibility to epilepsy in Cux1+/- mice. We describe 34 individuals, from which 30 were unrelated, with 26 different null and four missense variants. The leading symptoms were mild to moderate delayed speech and motor development and borderline to moderate intellectual disability. Additional symptoms were muscular hypotonia, seizures, joint laxity, and abnormalities of the forehead. In Cux1+/- mice, we found delayed growth, histologically normal brains, and increased susceptibility to seizures. In Cux1+/- brains, the expression of Cux1 transcripts was half of WT animals. Expression of CUX1 proteins was reduced, although in early postnatal animals significantly more than in adults. In summary, disease-causing CUX1 variants result in a non-syndromic phenotype of developmental delay and intellectual disability. In some individuals, this phenotype ameliorates with age, resulting in a clinical catch-up and normal IQ in adulthood. The post-transcriptional balance of CUX1 expression in the heterozygous brain at late developmental stages appears important for this favorable clinical course.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Adulto , Animales , Humanos , Ratones , Heterocigoto , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Proteínas Represoras/genética , Convulsiones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Methods Mol Biol ; 2695: 337-349, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37450130

RESUMEN

Neurodevelopmental disorders in children have an important impact on the quality of life in the whole life cycle. Severe neurodevelopmental disorders will become a serious social and family burden and an important social and economic problem. The early and middle childhood is the critical period of children's neurodevelopment. Early diagnosis of neurological disorders plays an important role in guiding children's neurological development. Existing monitoring tools lack prenatal and even early assessment of children's neurodevelopment, so reliable biomarkers are conducive to personalized care at an earlier stage. In this review, we will discuss different methods of neurodevelopmental monitoring at different times and the role and evaluation of liquid biopsy in neurodevelopmental monitoring.


Asunto(s)
Biopsia Líquida , Trastornos del Neurodesarrollo , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/patología , Biopsia Líquida/métodos , Humanos , Pruebas Prenatales no Invasivas/métodos , Ácidos Nucleicos Libres de Células/análisis , Encéfalo/crecimiento & desarrollo , Encéfalo/patología
17.
Artículo en Inglés | MEDLINE | ID: mdl-37263801

RESUMEN

Ankyrins are a family of proteins that link integral membrane proteins to the underlying spectrin-actin cytoskeleton and play a key role in activities such as cell motility, activation, proliferation, cell-cell contact, and the maintenance of specialized membrane domains. Ankyrin 3 (ANK3) is one of the three major subtypes of the ankyrin protein family. Ankryin genes are ubiquitously expressed, but their expression is highest in the brain. In the central nervous system, ankyrins have critical roles at the axonal initial segment, the nodes of Ranvier, and at synapses. To date, pathogenic variants in ANK3 have been reported in individuals with neuropsychiatric, cognitive, and neurodevelopmental disorders. The clinical severity is variable in these individuals with both autosomal recessive and autosomal dominant patterns of inheritance observed. These findings have suggested genotype-phenotype correlations and even isoform-specific implications for individuals with ANK3 pathogenic variants. Here, we report a patient with speech delay, autism spectrum disorder, and a language disorder in which a de novo nonsense ANK3 alteration was discovered by exome sequencing. Interestingly, the next-generation sequencing data suggested the change was mosaic in the affected child, and it was confirmed by digital polymerase chain reaction (dPCR) at 22% allelic fraction. To our knowledge, this is the first case of an individual with a pathogenic mosaic ANK3 variant. This finding expands upon the existing genotype-phenotype information available for the ANK3 gene while also highlighting potential gene expression correlations with phenotype.


Asunto(s)
Trastorno del Espectro Autista , Trastornos del Neurodesarrollo , Humanos , Trastorno del Espectro Autista/genética , Ancirinas/genética , Isoformas de Proteínas/genética , Encéfalo/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología
18.
Am J Hum Genet ; 110(6): 963-978, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37196654

RESUMEN

De novo variants are a leading cause of neurodevelopmental disorders (NDDs), but because every monogenic NDD is different and usually extremely rare, it remains a major challenge to understand the complete phenotype and genotype spectrum of any morbid gene. According to OMIM, heterozygous variants in KDM6B cause "neurodevelopmental disorder with coarse facies and mild distal skeletal abnormalities." Here, by examining the molecular and clinical spectrum of 85 reported individuals with mostly de novo (likely) pathogenic KDM6B variants, we demonstrate that this description is inaccurate and potentially misleading. Cognitive deficits are seen consistently in all individuals, but the overall phenotype is highly variable. Notably, coarse facies and distal skeletal anomalies, as defined by OMIM, are rare in this expanded cohort while other features are unexpectedly common (e.g., hypotonia, psychosis, etc.). Using 3D protein structure analysis and an innovative dual Drosophila gain-of-function assay, we demonstrated a disruptive effect of 11 missense/in-frame indels located in or near the enzymatic JmJC or Zn-containing domain of KDM6B. Consistent with the role of KDM6B in human cognition, we demonstrated a role for the Drosophila KDM6B ortholog in memory and behavior. Taken together, we accurately define the broad clinical spectrum of the KDM6B-related NDD, introduce an innovative functional testing paradigm for the assessment of KDM6B variants, and demonstrate a conserved role for KDM6B in cognition and behavior. Our study demonstrates the critical importance of international collaboration, sharing of clinical data, and rigorous functional analysis of genetic variants to ensure correct disease diagnosis for rare disorders.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Animales , Facies , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Drosophila , Discapacidad Intelectual/patología , Histona Demetilasas con Dominio de Jumonji/genética
19.
Genet Med ; 25(7): 100859, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37092538

RESUMEN

PURPOSE: The study aimed to clinically and molecularly characterize the neurodevelopmental disorder associated with heterozygous de novo variants in CNOT9. METHODS: Individuals were clinically examined. Variants were identified using exome or genome sequencing. These variants were evaluated using in silico predictions, and their functional relevance was further assessed by molecular models and research in the literature. The variants have been classified according to the criteria of the American College of Medical Genetics. RESULTS: We report on 7 individuals carrying de novo missense variants in CNOT9, p.(Arg46Gly), p.(Pro131Leu), and p.(Arg227His), and, recurrent in 4 unrelated individuals, p.(Arg292Trp). All affected persons have developmental delay/intellectual disability, with 5 of them showing seizures. Other symptoms include muscular hypotonia, facial dysmorphism, and behavioral abnormalities. Molecular modeling predicted that the variants are damaging and would lead to reduced protein stability or impaired recognition of interaction partners. Functional analyses in previous studies showed a pathogenic effect of p.(Pro131Leu) and p.(Arg227His). CONCLUSION: We propose CNOT9 as a novel gene for neurodevelopmental disorder and epilepsy.


Asunto(s)
Epilepsia , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Epilepsia/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Mutación Missense/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Convulsiones/genética
20.
Genet Med ; 25(7): 100839, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37057675

RESUMEN

PURPOSE: LHX2 encodes the LIM homeobox 2 transcription factor (LHX2), which is highly expressed in brain and well conserved across species, but it has not been clearly linked to neurodevelopmental disorders (NDDs) to date. METHODS: Through international collaboration, we identified 19 individuals from 18 families with variable neurodevelopmental phenotypes, carrying a small chromosomal deletion, likely gene-disrupting or missense variants in LHX2. Functional consequences of missense variants were investigated in cellular systems. RESULTS: Affected individuals presented with developmental and/or behavioral abnormalities, autism spectrum disorder, variable intellectual disability, and microcephaly. We observed nucleolar accumulation for 2 missense variants located within the DNA-binding HOX domain, impaired interaction with co-factor LDB1 for another variant located in the protein-protein interaction-mediating LIM domain, and impaired transcriptional activation by luciferase assay for 4 missense variants. CONCLUSION: We implicate LHX2 haploinsufficiency by deletion and likely gene-disrupting variants as causative for a variable NDD. Our findings suggest a loss-of-function mechanism also for likely pathogenic LHX2 missense variants. Together, our observations underscore the importance of LHX2 in the nervous system and for variable neurodevelopmental phenotypes.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Proteínas con Homeodominio LIM/genética , Trastorno del Espectro Autista/genética , Haploinsuficiencia/genética , Trastornos del Neurodesarrollo/patología , Factores de Transcripción/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA