Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 497
Filtrar
1.
Med Sci (Paris) ; 40(8-9): 643-652, 2024.
Artículo en Francés | MEDLINE | ID: mdl-39303116

RESUMEN

Rare genetic diseases with neurodevelopmental disorders (NDDs) encompass several heterogeneous conditions (autism spectrum disorder (ASD), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), specific learning disorder (SLD), among others). Currently, few treatments are available for these patients. The difficulty in accessing human brain samples and the discrepancies between human and animal models highlight the need for new research approaches. One promising approach is the use of the cerebral organoids. These 3D, self-organized structures, generated from induced pluripotent stem cells (iPSCs), enable the reproduction of the stages of human brain development, from the proliferation of neural stem cells to their differentiation into neurons, oligodentrocytes, and astrocytes. Cerebral organoids hold great promise in understanding brain development and in the search for treatments.


Title: Des organoïdes cérébraux pour la compréhension et la thérapie des maladies génétiques rares avec troubles neurodéveloppementaux. Abstract: Les maladies génétiques associées à des troubles neurodéveloppementaux (TND) regroupent plusieurs maladies pour lesquelles peu de traitements sont proposés. L'impossibilité d'accéder à des échantillons de cerveaux humains pour des études ex vivo, et les divergences entre l'homme et les modèles animaux rendent nécessaires de nouvelles approches de recherche. L'organoïde cérébral, une structure en trois dimensions, auto-organisée, et générée à partir de cellules souches pluripotentes induites, permet de reproduire les étapes de développement du cerveau humain, de la prolifération des cellules souches neurales à leur différenciation en neurones, en oligodendrocytes, ou en astrocytes. L'intérêt de ce modèle est désormais prouvé pour la compréhension du développement cérébral et pour la recherche de traitements. Après une présentation des cellules souches pluripotentes induites et des organoïdes, nous exposerons comment cette technique est actuellement déployée, en particulier pour étudier les mécanismes physiopathologiques résultant de variations génétiques pathogènes de gènes candidats de TND.


Asunto(s)
Trastornos del Neurodesarrollo , Organoides , Enfermedades Raras , Humanos , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/terapia , Trastornos del Neurodesarrollo/patología , Enfermedades Raras/genética , Enfermedades Raras/terapia , Animales , Células Madre Pluripotentes Inducidas/trasplante , Encéfalo/patología , Enfermedades Genéticas Congénitas/terapia , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Diferenciación Celular/genética
2.
Hum Brain Mapp ; 45(13): e26815, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39254138

RESUMEN

With brain structure and function undergoing complex changes throughout childhood and adolescence, age is a critical consideration in neuroimaging studies, particularly for those of individuals with neurodevelopmental conditions. However, despite the increasing use of large, consortium-based datasets to examine brain structure and function in neurotypical and neurodivergent populations, it is unclear whether age-related changes are consistent between datasets and whether inconsistencies related to differences in sample characteristics, such as demographics and phenotypic features, exist. To address this, we built models of age-related changes of brain structure (regional cortical thickness and regional surface area; N = 1218) and function (resting-state functional connectivity strength; N = 1254) in two neurodiverse datasets: the Province of Ontario Neurodevelopmental Network and the Healthy Brain Network. We examined whether deviations from these models differed between the datasets, and explored whether these deviations were associated with demographic and clinical variables. We found significant differences between the two datasets for measures of cortical surface area and functional connectivity strength throughout the brain. For regional measures of cortical surface area, the patterns of differences were associated with race/ethnicity, while for functional connectivity strength, positive associations were observed with head motion. Our findings highlight that patterns of age-related changes in the brain may be influenced by demographic and phenotypic characteristics, and thus future studies should consider these when examining or controlling for age effects in analyses.


Asunto(s)
Conjuntos de Datos como Asunto , Imagen por Resonancia Magnética , Humanos , Femenino , Masculino , Niño , Adolescente , Adulto Joven , Adulto , Trastornos del Neurodesarrollo/diagnóstico por imagen , Trastornos del Neurodesarrollo/fisiopatología , Trastornos del Neurodesarrollo/patología , Conectoma , Encéfalo/diagnóstico por imagen , Encéfalo/crecimiento & desarrollo , Encéfalo/anatomía & histología , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/anatomía & histología , Envejecimiento/fisiología
3.
J Cell Mol Med ; 28(17): e18560, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39258535

RESUMEN

Neurodevelopmental disorders are mostly studied using mice as models. However, the mouse brain lacks similar cell types and structures as those of the human brain. In recent years, emergence of three-dimensional brain organoids derived from human embryonic stem cells or induced pluripotent stem cells allows for controlled monitoring and evaluation of early neurodevelopmental processes and has opened a window for studying various aspects of human brain development. However, such organoids lack original anatomical structure of the brain during maturation, and neurodevelopmental maturation processes that rely on unique cellular interactions and neural network connections are limited. Consequently, organoids are difficult to be used extensively and effectively while modelling later stages of human brain development and disease progression. To address this problem, several methods and technologies have emerged that aim to enhance the sophisticated regulation of brain organoids developmental processes through bioengineering approaches, which may alleviate some of the current limitations. This review discusses recent advances and application areas of human brain organoid culture methods, aiming to generalize optimization strategies for organoid systems, improve the ability to mimic human brain development, and enhance the application value of organoids.


Asunto(s)
Encéfalo , Trastornos del Neurodesarrollo , Organoides , Humanos , Trastornos del Neurodesarrollo/patología , Encéfalo/patología , Encéfalo/crecimiento & desarrollo , Encéfalo/citología , Animales , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos
4.
Eur J Med Genet ; 71: 104965, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39094681

RESUMEN

Neurodevelopmental disorders have been linked to numerous genes, particularly pathogenic variants in genes encoding postsynaptic scaffolding proteins, like SHANK3. This study aims to provide insights into the cardiovascular profile of patients with pathogenic SHANK3 variants, expanding beyond the well-established associations with neurodevelopmental disorders and epilepsy. We conducted a prospective study involving patients affected by neurodevelopmental disorders with pathogenic SHANK3 variants. Comprehensive cardiovascular assessments were performed and molecular genetic testing included chromosomal microarray followed by clinical exome sequencing. We identified five patients with de novo SHANK3 variants, all of whom exhibited cardiac involvement, including myocardial dysfunction, congenital heart disease (patent ductus arteriosus), and a case of postictal atrial fibrillation. Our findings emphasize an elevated risk of cardiovascular abnormalities in patients with SHANK3 pathogenic variants compared to prior reports. Despite their young age, these patients displayed significant cardiac abnormalities. The study highlights the necessity of integrating cardiac evaluation and ongoing cardiovascular monitoring into multidisciplinary care, facilitating early detection of heart failure and assessment of the risk of sudden unexpected death in epilepsy (SUDEP). Further research is needed to elucidate the underlying mechanisms of cardiac manifestations in SHANK3 mutation carriers.


Asunto(s)
Epilepsia , Proteínas del Tejido Nervioso , Trastornos del Neurodesarrollo , Humanos , Proteínas del Tejido Nervioso/genética , Femenino , Masculino , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Epilepsia/genética , Epilepsia/patología , Niño , Preescolar , Mutación , Adulto , Adolescente , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/patología , Lactante , Secuenciación del Exoma
5.
Am J Hum Genet ; 111(9): 1994-2011, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39168120

RESUMEN

Zinc and RING finger 3 (ZNRF3) is a negative-feedback regulator of Wnt/ß-catenin signaling, which plays an important role in human brain development. Although somatically frequently mutated in cancer, germline variants in ZNRF3 have not been established as causative for neurodevelopmental disorders (NDDs). We identified 12 individuals with ZNRF3 variants and various phenotypes via GeneMatcher/Decipher and evaluated genotype-phenotype correlation. We performed structural modeling and representative deleterious and control variants were assessed using in vitro transcriptional reporter assays with and without Wnt-ligand Wnt3a and/or Wnt-potentiator R-spondin (RSPO). Eight individuals harbored de novo missense variants and presented with NDD. We found missense variants associated with macrocephalic NDD to cluster in the RING ligase domain. Structural modeling predicted disruption of the ubiquitin ligase function likely compromising Wnt receptor turnover. Accordingly, the functional assays showed enhanced Wnt/ß-catenin signaling for these variants in a dominant negative manner. Contrarily, an individual with microcephalic NDD harbored a missense variant in the RSPO-binding domain predicted to disrupt binding affinity to RSPO and showed attenuated Wnt/ß-catenin signaling in the same assays. Additionally, four individuals harbored de novo truncating or de novo or inherited large in-frame deletion variants with non-NDD phenotypes, including heart, adrenal, or nephrotic problems. In contrast to NDD-associated missense variants, the effects on Wnt/ß-catenin signaling were comparable between the truncating variant and the empty vector and between benign variants and the wild type. In summary, we provide evidence for mirror brain size phenotypes caused by distinct pathomechanisms in Wnt/ß-catenin signaling through protein domain-specific deleterious ZNRF3 germline missense variants.


Asunto(s)
Encéfalo , Mutación de Línea Germinal , Trastornos del Neurodesarrollo , Fenotipo , Ubiquitina-Proteína Ligasas , Vía de Señalización Wnt , Humanos , Vía de Señalización Wnt/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Femenino , Masculino , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Niño , Preescolar , beta Catenina/genética , beta Catenina/metabolismo , Adolescente , Mutación Missense , Estudios de Asociación Genética , Dominios Proteicos
6.
Genes (Basel) ; 15(8)2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39202413

RESUMEN

The human 16p11.2 chromosomal region is rich in segmental duplications which mediate the formation of recurrent CNVs. CNVs affecting the 16p11.2 region are associated with an increased risk for developing neuropsychiatric disorders, including autism spectrum disorder (ASD), schizophrenia, and intellectual disability (ID), as well as abnormal body weight and head circumference and dysmorphic features, with marked phenotypic variability and reduced penetrance. CNVs affecting the 16p11.2 region mainly affect a distal interval of ~220 Kb, between Breakpoints 2 and 3 (BP2-BP3), and a proximal interval of ~593 Kb (BP4-BP5). Here, we report on 15 patients with recurrent 16p11.2 rearrangements that were identified among a cohort of 1600 patients (0.9%) with neurodevelopmental disorders. A total of 13 deletions and two duplications were identified, of which eight deletions included the proximal 16p11.2 region (BP4-BP5) and five included the distal 16p11.2 region (BP2-BP3). Of the two duplications that were identified, one affected the proximal and one the distal 16p11.2 region; however, both patients had additional CNVs contributing to phenotypic severity. The features observed and their severity varied greatly, even between patients within the same family. This article aims to further delineate the clinical spectrum of patients with 16p11.2 recurrent rearrangements in order to aid the counselling of patients and their families.


Asunto(s)
Cromosomas Humanos Par 16 , Discapacidad Intelectual , Fenotipo , Humanos , Cromosomas Humanos Par 16/genética , Masculino , Femenino , Niño , Adolescente , Preescolar , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Variaciones en el Número de Copia de ADN , Deleción Cromosómica , Adulto , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Aberraciones Cromosómicas , Adulto Joven
7.
Adv Neurobiol ; 37: 457-495, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39207708

RESUMEN

This chapter will focus on microglial involvement in neurodevelopmental and neuropsychiatric disorders, particularly autism spectrum disorder (ASD), schizophrenia and major depressive disorder (MDD). We will describe the neuroimmune risk factors that contribute to the etiopathology of these disorders across the lifespan, including both in early life and adulthood. Microglia, being the resident immune cells of the central nervous system, could play a key role in triggering and determining the outcome of these disorders. This chapter will review preclinical and clinical findings where microglial morphology and function were examined in the contexts of ASD, schizophrenia and MDD. Clinical evidence points out to altered microglial morphology and reactivity, as well as increased expression of pro-inflammatory cytokines, supporting the idea that microglial abnormalities are involved in these disorders. Indeed, animal models for these disorders found altered microglial morphology and homeostatic functions which resulted in behaviours related to these disorders. Additionally, as microglia have emerged as promising therapeutic targets, we will also address in this chapter therapies involving microglial mechanisms for the treatment of neurodevelopmental and neuropsychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Depresivo Mayor , Microglía , Trastornos del Neurodesarrollo , Esquizofrenia , Humanos , Microglía/metabolismo , Microglía/patología , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Esquizofrenia/metabolismo , Esquizofrenia/patología , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/metabolismo , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/patología , Animales , Trastornos Mentales/metabolismo , Trastornos Mentales/patología
8.
Hum Mol Genet ; 33(19): 1671-1687, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-38981622

RESUMEN

De novo variants in the Cytoplasmic FMR1-interacting protein 2 (CYFIP2) have been repeatedly associated with neurodevelopmental disorders and epilepsy, underscoring its critical role in brain development and function. While CYFIP2's role in regulating actin polymerization as part of the WAVE regulatory complex (WRC) is well-established, its additional molecular functions remain relatively unexplored. In this study, we performed unbiased quantitative proteomic analysis, revealing 278 differentially expressed proteins (DEPs) in the forebrain of Cyfip2 knock-out embryonic mice compared to wild-type mice. Unexpectedly, these DEPs, in conjunction with previously identified CYFIP2 brain interactors, included not only other WRC components but also numerous proteins associated with membraneless organelles (MLOs) involved in mRNA processing and translation within cells, including the nucleolus, stress granules, and processing bodies. Additionally, single-cell transcriptomic analysis of the Cyfip2 knock-out forebrain revealed gene expression changes linked to cellular stress responses and MLOs. We also observed morphological changes in MLOs in Cyfip2 knock-out brains and CYFIP2 knock-down cells under basal and stress conditions. Lastly, we demonstrated that CYFIP2 knock-down in cells, potentially through WRC-dependent actin regulation, suppressed the phosphorylation levels of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2α), thereby enhancing protein synthesis. These results suggest a physical and functional connection between CYFIP2 and various MLO proteins and also extend CYFIP2's role within the WRC from actin regulation to influencing eIF2α phosphorylation and protein synthesis. With these dual functions, CYFIP2 may fine-tune the balance between MLO formation/dynamics and protein synthesis, a crucial aspect of proper mRNA processing and translation.


Asunto(s)
Citoesqueleto de Actina , Proteínas Adaptadoras Transductoras de Señales , Factor 2 Eucariótico de Iniciación , Ratones Noqueados , Trastornos del Neurodesarrollo , Animales , Ratones , Fosforilación , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Humanos , Proteómica/métodos , Prosencéfalo/metabolismo , Encéfalo/metabolismo
9.
Stem Cell Res ; 79: 103487, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38972232

RESUMEN

CTNNB1 encodes beta-catenin, which plays a crucial role in Wnt signaling pathway. Mutations in CTNNB1 involve in tumor developing, Primary Aldosteronism, Neurodevelopmental disorders (NDDs), etc. NDDs is a class of disorders that impact brain development and function, manifesting symptom including autism spectrum disorder (ASD), intellectual disability (ID), schizophrenia (SCZ), and epilepsy. Here, we generated an iPSC line (CTUi005-A) from a patient diagnosed with NDDs, carrying a heterozygous mutation of the CTNNB1 gene. CTUi005-A exhibits typical iPSC characteristics, and holds potential as a cellular tool for investigating the pathogenic mechanisms underlying NDDs.


Asunto(s)
Heterocigoto , Células Madre Pluripotentes Inducidas , Mutación , beta Catenina , Humanos , Masculino , beta Catenina/genética , beta Catenina/metabolismo , Línea Celular , Células Madre Pluripotentes Inducidas/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología
10.
Stem Cell Reports ; 19(8): 1074-1091, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39059378

RESUMEN

Although microglia are macrophages of the central nervous system, their involvement is not limited to immune functions. The roles of microglia during development in humans remain poorly understood due to limited access to fetal tissue. To understand how microglia can impact human neurodevelopment, the methyl-CpG binding protein 2 (MECP2) gene was knocked out in human microglia-like cells (MGLs). Disruption of the MECP2 in MGLs led to transcriptional and functional perturbations, including impaired phagocytosis. The co-culture of healthy MGLs with MECP2-knockout (KO) neurons rescued synaptogenesis defects, suggesting a microglial role in synapse formation. A targeted drug screening identified ADH-503, a CD11b agonist, restored phagocytosis and synapse formation in spheroid-MGL co-cultures, significantly improved disease progression, and increased survival in MeCP2-null mice. These results unveil a MECP2-specific regulation of human microglial phagocytosis and identify a novel therapeutic treatment for MECP2-related conditions.


Asunto(s)
Proteína 2 de Unión a Metil-CpG , Microglía , Trastornos del Neurodesarrollo , Fagocitosis , Microglía/metabolismo , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Ratones , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ratones Noqueados , Sinapsis/metabolismo , Neuronas/metabolismo
11.
Eur J Neurosci ; 60(5): 4877-4892, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39054743

RESUMEN

The prevalence of pyramidal cells (PCs) in the mammalian cerebral cortex underscore their value as they play a crucial role in various brain functions, ranging from cognition, sensory processing, to motor output. PC morphology significantly influences brain connectivity and plays a critical role in maintaining normal brain function. Pathological alterations to PC morphology are thought to contribute to the aetiology of neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. This review explores the relationship between abnormalities in PC morphology in key cortical areas and the clinical manifestations in schizophrenia and ASD. We focus largely on human postmortem studies and provide evidence that dendritic segment length, complexity and spine density are differentially affected in these disorders. These morphological alterations can lead to disruptions in cortical connectivity, potentially contributing to the cognitive and behavioural deficits observed in these disorders. Furthermore, we highlight the importance of investigating the functional and structural characteristics of PCs in these disorders to illuminate the underlying pathogenesis and stimulate further research in this area.


Asunto(s)
Trastornos del Neurodesarrollo , Células Piramidales , Humanos , Células Piramidales/patología , Animales , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/fisiopatología , Trastorno del Espectro Autista/patología , Trastorno del Espectro Autista/fisiopatología , Esquizofrenia/patología , Esquizofrenia/fisiopatología , Corteza Cerebral/patología
12.
Neurobiol Dis ; 199: 106604, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39002810

RESUMEN

Mitochondria are essential regulators of cellular energy metabolism and play a crucial role in the maintenance and function of neuronal cells. Studies in the last decade have highlighted the importance of mitochondrial dynamics and bioenergetics in adult neurogenesis, a process that significantly influences cognitive function and brain plasticity. In this review, we examine the mechanisms by which mitochondria regulate adult neurogenesis, focusing on the impact of mitochondrial function on the behavior of neural stem/progenitor cells and the maturation and plasticity of newborn neurons in the adult mouse hippocampus. In addition, we explore the link between mitochondrial dysfunction, adult hippocampal neurogenesis and genes associated with cognitive deficits in neurodevelopmental disorders. In particular, we provide insights into how alterations in the transcriptional regulator NR2F1 affect mitochondrial dynamics and may contribute to the pathophysiology of the emerging neurodevelopmental disorder Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS). Understanding how genes involved in embryonic and adult neurogenesis affect mitochondrial function in neurological diseases might open new directions for therapeutic interventions aimed at boosting mitochondrial function during postnatal life.


Asunto(s)
Hipocampo , Mitocondrias , Trastornos del Neurodesarrollo , Neurogénesis , Neurogénesis/fisiología , Animales , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/fisiopatología , Hipocampo/metabolismo , Mitocondrias/metabolismo , Humanos , Células-Madre Neurales/metabolismo
13.
Neurobiol Dis ; 199: 106607, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39029564

RESUMEN

Cell metabolism is a key regulator of human neocortex development and evolution. Several lines of evidence indicate that alterations in neural stem/progenitor cell (NPC) metabolism lead to abnormal brain development, particularly brain size-associated neurodevelopmental disorders, such as microcephaly. Abnormal NPC metabolism causes impaired cell proliferation and thus insufficient expansion of NPCs for neurogenesis. Therefore, the production of neurons, which is a major determinant of brain size, is decreased and the size of the brain, especially the size of the neocortex, is significantly reduced. This review discusses recent progress understanding NPC metabolism, focusing in particular on glucose metabolism, fatty acid metabolism and amino acid metabolism (e.g., glutaminolysis and serine metabolism). We provide an overview of the contributions of these metabolic pathways to brain development and evolution, as well as to the etiology of neurodevelopmental disorders. Furthermore, we discuss the advantages and disadvantages of various experimental models to study cell metabolism in the developing brain.


Asunto(s)
Encéfalo , Células-Madre Neurales , Trastornos del Neurodesarrollo , Humanos , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/fisiopatología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/crecimiento & desarrollo , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neurogénesis/fisiología , Tamaño de los Órganos/fisiología
14.
Genes (Basel) ; 15(7)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-39062680

RESUMEN

BACKGROUND: Duplications on the short arm of chromosome X, including the gene NR0B1, have been associated with gonadal dysgenesis and with male to female sex reversal. Additional clinical manifestations can be observed in the affected patients, depending on the duplicated genomic region. Here we report one of the largest duplications on chromosome X, in a Lebanese patient, and we provide the first comprehensive review of duplications in this genomic region. CASE PRESENTATION: A 2-year-old female patient born to non-consanguineous Lebanese parents, with a family history of one miscarriage, is included in this study. The patient presents with sex reversal, dysmorphic features, optic atrophy, epilepsy, psychomotor and neurodevelopmental delay. Single nucleotide variants and copy number variants analysis were carried out on the patient through exome sequencing (ES). This showed an increased coverage of a genomic region of around 23.6 Mb on chromosome Xp22.31-p21.2 (g.7137718-30739112) in the patient, suggestive of a large duplication encompassing more than 60 genes, including the NR0B1 gene involved in sex reversal. A karyotype analysis confirmed sex reversal in the proband presenting with the duplication, and revealed a balanced translocation between the short arms of chromosomes X and 14:46, X, t(X;14) (p11;p11) in her/his mother. CONCLUSIONS: This case highlights the added value of CNV analysis from ES data in the genetic diagnosis of patients. It also underscores the challenges encountered in announcing unsolicited incidental findings to the family.


Asunto(s)
Variaciones en el Número de Copia de ADN , Epilepsia , Secuenciación del Exoma , Trastornos del Neurodesarrollo , Atrofia Óptica , Preescolar , Humanos , Duplicación Cromosómica/genética , Cromosomas Humanos X/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Epilepsia/genética , Epilepsia/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Atrofia Óptica/genética , Atrofia Óptica/patología
15.
Mol Genet Genomic Med ; 12(7): e2473, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39038237

RESUMEN

BACKGROUND: Neuron navigator 3 (NAV3) is characterized as one of the neuron navigator family (NAV1, NAV2, NAV3) proteins predominantly expressed in the nervous system. The NAV3-encoded protein comprises a conserved AAA and coiled-coil domains characteristic of ATPases, which are associated with different cellular activities. METHODS: We describe a Saudi proband presenting a complex recessive neurodevelopmental disorder (NDD). Whole exome sequencing (WES) followed by Sanger sequencing, 3D protein modeling and RT-qPCR was performed. RESULTS: WES revealed a bi-allelic frameshift variant (c.2604_2605delAG; p.Val870SerfsTer12) in exon 12 of the NAV3 gene. Furthermore, RT-qPCR revealed a significant decrease in the NAV3 mRNA expression in the patient sample, and 3D protein modeling revealed disruption of the overall secondary structure. CONCLUSION: For the time, we associate a bi-allelic variant in the NAV3 gene causing NDD in humans.


Asunto(s)
Mutación del Sistema de Lectura , Trastornos del Neurodesarrollo , Femenino , Humanos , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Linaje
16.
Sci Adv ; 10(28): eadk5462, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38985877

RESUMEN

Adherens junction-associated protein 1 (AJAP1) has been implicated in brain diseases; however, a pathogenic mechanism has not been identified. AJAP1 is widely expressed in neurons and binds to γ-aminobutyric acid type B receptors (GBRs), which inhibit neurotransmitter release at most synapses in the brain. Here, we show that AJAP1 is selectively expressed in dendrites and trans-synaptically recruits GBRs to presynaptic sites of neurons expressing AJAP1. We have identified several monoallelic AJAP1 variants in individuals with epilepsy and/or neurodevelopmental disorders. Specifically, we show that the variant p.(W183C) lacks binding to GBRs, resulting in the inability to recruit them. Ultrastructural analysis revealed significantly decreased presynaptic GBR levels in Ajap1-/- and Ajap1W183C/+ mice. Consequently, these mice exhibited reduced GBR-mediated presynaptic inhibition at excitatory and inhibitory synapses, along with impaired synaptic plasticity. Our study reveals that AJAP1 enables the postsynaptic neuron to regulate the level of presynaptic GBR-mediated inhibition, supporting the clinical relevance of loss-of-function AJAP1 variants.


Asunto(s)
Neurotransmisores , Sinapsis , Transmisión Sináptica , Animales , Femenino , Humanos , Masculino , Ratones , Alelos , Epilepsia/metabolismo , Epilepsia/genética , Epilepsia/patología , Mutación con Pérdida de Función , Ratones Noqueados , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Plasticidad Neuronal , Neuronas/metabolismo , Neurotransmisores/metabolismo , Sinapsis/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo
17.
Neurobiol Dis ; 199: 106597, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38992777

RESUMEN

Pediatric low grade brain tumors and neurodevelopmental disorders share proteins, signaling pathways, and networks. They also share germline mutations and an impaired prenatal differentiation origin. They may differ in the timing of the events and proliferation. We suggest that their pivotal distinct, albeit partially overlapping, outcomes relate to the cell states, which depend on their spatial location, and timing of gene expression during brain development. These attributes are crucial as the brain develops sequentially, and single-cell spatial organization influences cell state, thus function. Our underlying premise is that the root cause in neurodevelopmental disorders and pediatric tumors is impaired prenatal differentiation. Data related to pediatric brain tumors, neurodevelopmental disorders, brain cell (sub)types, locations, and timing of expression in the developing brain are scant. However, emerging single cell technologies, including transcriptomic, spatial biology, spatial high-resolution imaging performed over the brain developmental time, could be transformational in deciphering brain pathologies thereby pharmacology.


Asunto(s)
Encéfalo , Análisis de la Célula Individual , Humanos , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Encéfalo/metabolismo , Niño , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Animales
18.
Neuroimage ; 297: 120721, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38968977

RESUMEN

Individuals with congenital heart disease (CHD) have an increased risk of neurodevelopmental impairments. Given the hypothesized complexity linking genomics, atypical brain structure, cardiac diagnoses and their management, and neurodevelopmental outcomes, unsupervised methods may provide unique insight into neurodevelopmental variability in CHD. Using data from the Pediatric Cardiac Genomics Consortium Brain and Genes study, we identified data-driven subgroups of individuals with CHD from measures of brain structure. Using structural magnetic resonance imaging (MRI; N = 93; cortical thickness, cortical volume, and subcortical volume), we identified subgroups that differed primarily on cardiac anatomic lesion and language ability. In contrast, using diffusion MRI (N = 88; white matter connectivity strength), we identified subgroups that were characterized by differences in associations with rare genetic variants and visual-motor function. This work provides insight into the differential impacts of cardiac lesions and genomic variation on brain growth and architecture in patients with CHD, with potentially distinct effects on neurodevelopmental outcomes.


Asunto(s)
Encéfalo , Cardiopatías Congénitas , Imagen por Resonancia Magnética , Humanos , Cardiopatías Congénitas/patología , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/genética , Femenino , Masculino , Niño , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Adolescente , Adulto Joven , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/patología , Adulto , Preescolar , Imagen de Difusión por Resonancia Magnética , Trastornos del Neurodesarrollo/diagnóstico por imagen , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/genética
19.
J Med Genet ; 61(9): 833-838, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-38876772

RESUMEN

Homozygous VPS50 variants have been previously described in two unrelated patients with a neurodevelopmental disorder with microcephaly, seizures and neonatal cholestasis. VPS50 encodes a subunit that is unique to the heterotetrameric endosome-associated recycling protein (EARP) complex. The other subunits of the EARP complex, such as VPS51, VPS52 and VPS53, are also shared by the Golgi-associated retrograde protein complex. We report on an 18-month-old female patient with biallelic VPS50 variants. She carried a paternally inherited heterozygous nonsense c.13A>T; p.(Lys5*) variant. By long-read genome sequencing, we characterised a structural variant with a 4.3 Mb inversion flanked by deletions at both breakpoints on the maternal allele. The ~428 kb deletion at the telomeric inversion breakpoint encompasses the entire VPS50 gene. We demonstrated a deficiency of VPS50 in patient-derived fibroblasts, confirming the loss-of-function nature of both VPS50 variants. VPS53 and VPS52 protein levels were significantly reduced and absent, respectively, in fibroblasts of the patient. These data show that VPS50 and/or EARP deficiency and the associated functional defects underlie the phenotype in patients with VPS50 pathogenic variants. The VPS50-related core phenotype comprises severe developmental delay, postnatal microcephaly, hypoplastic corpus callosum, neonatal low gamma-glutamyl transpeptidase cholestasis and failure to thrive. The disease is potentially fatal in early childhood.


Asunto(s)
Codón sin Sentido , Proteínas de Transporte Vesicular , Humanos , Femenino , Lactante , Codón sin Sentido/genética , Proteínas de Transporte Vesicular/genética , Microcefalia/genética , Microcefalia/patología , Fenotipo , Colestasis/genética , Colestasis/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología
20.
Curr Opin Genet Dev ; 87: 102210, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38833893

RESUMEN

Pseudouridine (Ψ), the most abundant RNA modification, plays a role in pre-mRNA splicing, RNA stability, protein translation efficiency, and cellular responses to environmental stress. Dysregulation of pseudouridylation is linked to human diseases. This review explores recent insights into the role of RNA pseudouridylation alterations in human disorders and the therapeutic potential of Ψ. We discuss the impact of the reduction of Ψ levels in ribosomal, messenger, and transfer RNA in RNA processing, protein translation, and consequently its role in neurodevelopmental diseases and cancer. Furthermore, we review the success of N1-methyl-Ψ messenger RNA vaccines against COVID-19 and the development of RNA-guided pseudouridylation enzymes for treating genetic diseases caused by premature stop codons.


Asunto(s)
COVID-19 , Seudouridina , Humanos , Seudouridina/metabolismo , Seudouridina/genética , COVID-19/genética , Neoplasias/genética , Neoplasias/terapia , Neoplasias/metabolismo , Neoplasias/patología , SARS-CoV-2/genética , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Animales , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/terapia , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Vacunas contra la COVID-19 , Biosíntesis de Proteínas , Empalme del ARN/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA