Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 13(2)2024 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-38275824

RESUMEN

PTCHD1 has been implicated in Autism Spectrum Disorders (ASDs) and/or intellectual disability, where copy-number-variant losses or loss-of-function coding mutations segregate with disease in an X-linked recessive fashion. Missense variants of PTCHD1 have also been reported in patients. However, the significance of these mutations remains undetermined since the activities, subcellular localization, and regulation of the PTCHD1 protein are currently unknown. This paucity of data concerning PTCHD1 prevents the effective evaluation of sequence variants identified during diagnostic screening. Here, we characterize PTCHD1 protein binding partners, extending previously reported interactions with postsynaptic scaffolding protein, SAP102. Six rare missense variants of PTCHD1 were also identified from patients with neurodevelopmental disorders. After modelling these variants on a hypothetical three-dimensional structure of PTCHD1, based on the solved structure of NPC1, PTCHD1 variants harboring these mutations were assessed for protein stability, post-translational processing, and protein trafficking. We show here that the wild-type PTCHD1 post-translational modification includes complex N-glycosylation and that specific mutant proteins disrupt normal N-link glycosylation processing. However, regardless of their processing, these mutants still localized to PSD95-containing dendritic processes and remained competent for complexing SAP102.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Humanos , Discapacidad Intelectual/genética , Trastorno del Espectro Autista/genética , Glicosilación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación/genética , Estabilidad Proteica
2.
Sci Rep ; 13(1): 20391, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37990104

RESUMEN

Patched domain-containing 1 (PTCHD1) is a well-established susceptibility gene for autism spectrum disorder (ASD) and intellectual disability (ID). Previous studies have suggested that alterations in the dosage of PTCHD1 may contribute to the etiology of both ASD and ID. However, there has not yet been a thorough investigation regarding mechanisms that regulate PTCHD1 expression. We sought to characterize the Ptchd1 promoter in a mouse neuronal model, as well as to identify and validate cis regulatory elements. We defined specific regions of the Ptchd1 promoter essential for robust expression in P19-induced neurons. Evolutionarily-conserved putative transcription factor binding sites within these regions were subsequently identified. Using a pairwise comparison of chromatin accessibility between mouse forebrain and liver tissues, a candidate regulatory region, ~ 9.1 kbp downstream of the Ptchd1 stop codon was defined. This region harbours two ENCODE-predicted enhancer cis-regulatory elements. Further, using DNase footprint analysis, a putative YY1-binding motif was also identified. Genomic deletion of the entire 8 kbp downstream open chromatin region attenuated Ptchd1 transcription by over 60% in our neuronal model, corroborating its predicted regulatory function. This study provides mechanistic insights related to the expression of PTCHD1, and provides important context to interpret genetic and genomic variation at this locus which may influence neurodevelopment.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Animales , Ratones , Trastorno Autístico/genética , Trastorno del Espectro Autista/genética , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Secuencia Conservada , Elementos de Facilitación Genéticos , Cromatina/genética
3.
Psychiatr Genet ; 33(6): 213-232, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37851134

RESUMEN

Chromatin, a protein-DNA complex, is a dynamic structure that stores genetic information within the nucleus and responds to molecular/cellular changes in its structure, providing conditional access to the genetic machinery. ATP-dependent chromatin modifiers regulate access of transcription factors and RNA polymerases to DNA by either "opening" or "closing" the structure of chromatin, and its aberrant regulation leads to a variety of neurodevelopmental disorders. The chromodomain helicase DNA-binding (CHD) proteins are ATP-dependent chromatin modifiers involved in the organization of chromatin structure, act as gatekeepers of genomic access, and deposit histone variants required for gene regulation. In this review, we first discuss the structural and functional domains of the CHD proteins, and their binding sites, and phosphorylation, acetylation, and methylation sites. The conservation of important amino acids in SWItch/sucrose non-fermenting (SWI/SNF) domains, and their protein and mRNA tissue expression profiles are discussed. Next, we convey the important binding partners of CHD proteins, their protein complexes and activities, and their involvements in epigenetic regulation. We also show the ChIP-seq binding dynamics for CHD1, CHD2, CHD4, and CHD7 proteins at promoter regions of histone genes, as well as several genes that are critical for neurodevelopment. The role of CHD proteins in development is also discussed. Finally, this review provides information about CHD protein mutations reported in autism and neurodevelopmental disorders, and their pathogenicity. Overall, this review provides information on the progress of research into CHD proteins, their structural and functional domains, epigenetics, and their role in stem cell, development, and neurological disorders.


Asunto(s)
Trastorno Autístico , Enfermedades del Sistema Nervioso , Humanos , Cromatina/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histonas/genética , Histonas/metabolismo , Epigénesis Genética , Trastorno Autístico/genética , Ensamble y Desensamble de Cromatina/genética , ADN , ADN Helicasas/genética , ADN Helicasas/química , ADN Helicasas/metabolismo , Adenosina Trifosfato/metabolismo , Enfermedades del Sistema Nervioso/genética
4.
Genes (Basel) ; 13(3)2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35328080

RESUMEN

Over the last one and a half decades, copy number variation and whole-genome sequencing studies have illuminated the considerable genetic heterogeneity that underlies the etiologies of autism spectrum disorder (ASD) and intellectual disability (ID). These investigations support the idea that ASD may result from complex interactions between susceptibility-related genetic variants (single nucleotide variants or copy number variants) and the environment. This review outlines the identification and neurobiological characterization of two such genes located in Xp22.11, Patched domain-containing 1 (PTCHD1), and its antisense lncRNA PTCHD1-AS. Animal models of Ptchd1 disruption have recapitulated a subset of clinical symptoms related to ASD as well as to ID. Furthermore, these Ptchd1 mouse knockout studies implicate the expression of Ptchd1 in both the thalamic and the hippocampal brain regions as being crucial for proper neurodevelopment and cognitive function. Altered kynurenine metabolic signalling has been postulated as a disease mechanism in one of these animal studies. Additionally, ASD patient-derived induced pluripotent stem cells (iPSCs) carrying a copy number loss impacting the antisense non-coding RNA PTCHD1-AS have been used to generate 2D neuronal cultures. While copy number loss of PTCHD1-AS does not affect the transcription of PTCHD1, the neurons exhibit diminished miniature excitatory postsynaptic current frequency, supporting its role in ASD etiology. A more thorough understanding of risk factor genes, such as PTCHD1 and PTCHD1-AS, will help to clarify the intricate genetic and biological mechanisms that underlie ASD and ID, providing a foundation for meaningful therapeutic interventions to enhance the quality of life of individuals who experience these conditions.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Animales , Trastorno del Espectro Autista/genética , Variaciones en el Número de Copia de ADN , Humanos , Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Ratones , Calidad de Vida
5.
Sci Rep ; 11(1): 23113, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34848785

RESUMEN

In a multi-branch family from Pakistan, individuals presenting with palmoplantar keratoderma segregate in autosomal dominant fashion, and individuals with intellectual disability (ID) segregate in apparent autosomal recessive fashion. Initial attempts to identify the ID locus using homozygosity-by-descent (HBD) mapping were unsuccessful. However, following an assumption of locus heterogeneity, a reiterative HBD approach in concert with whole exome sequencing (WES) was employed. We identified a known disease-linked mutation in the polymicrogyria gene, ADGRG1, in two affected members. In the remaining two (living) affected members, HBD mapping cross-referenced with WES data identified a single biallelic frameshifting variant in the gene encoding retinol dehydrogenase 14 (RDH14). Transcription data indicate that RDH14 is expressed in brain, but not in retina. Magnetic resonance imaging for the individuals with this RDH14 mutation show no signs of polymicrogyria, however cerebellar atrophy was a notable feature. RDH14 in HEK293 cells localized mainly in the nucleoplasm. Co-immunoprecipitation studies confirmed binding to the proton-activated chloride channel 1 (PACC1/TMEM206), which is greatly diminished by the mutation. Our studies suggest RDH14 as a candidate for autosomal recessive ID and cerebellar atrophy, implicating either disrupted retinoic acid signaling, or, through PACC1, disrupted chloride ion homeostasis in the brain as a putative disease mechanism.


Asunto(s)
Oxidorreductasas de Alcohol , Discapacidad Intelectual , Receptores Acoplados a Proteínas G , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Oxidorreductasas de Alcohol/genética , Alelos , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Núcleo Celular/metabolismo , Cerebelo/patología , Cloruros , Mapeo Cromosómico , Citoplasma/metabolismo , Mutación del Sistema de Lectura , Variación Genética , Genotipo , Células HEK293 , Homocigoto , Discapacidad Intelectual/genética , Iones , Imagen por Resonancia Magnética , Mutagénesis Sitio-Dirigida , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Pakistán , Linaje , Receptores Acoplados a Proteínas G/genética , Retina/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Secuenciación del Exoma
6.
Hum Genet ; 138(10): 1183-1200, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31471722

RESUMEN

The glutamate pyruvate transaminase 2 (GPT2) gene produces a nuclear-encoded mitochondrial enzyme that catalyzes the reversible transfer of an amino group from glutamate to pyruvate, generating alanine and alpha-ketoglutarate. Recessive mutations in GPT2 have been recently identified in a new syndrome involving intellectual and developmental disability (IDD), postnatal microcephaly, and spastic paraplegia. We have identified additional families with recessive GPT2 mutations and expanded the phenotype to include small stature. GPT2 loss-of-function mutations were identified in four families, nine patients total, including: a homozygous mutation in one child [c.775T>C (p.C259R)]; compound heterozygous mutations in two siblings [c.812A>C (p.N271T)/c.1432_1433delGT (p.V478Rfs*73)]; a novel homozygous, putative splicing mutation [c.1035C>T (p.G345=)]; and finally, a recurrent mutation, previously identified in a distinct family [c.1210C>T (p.R404*)]. All patients were diagnosed with IDD. A majority of patients had remarkably small stature throughout development, many < 1st percentile for height and weight. Given the potential biological function of GPT2 in cellular growth, this phenotype is strongly suggestive of a newly identified clinical susceptibility. Further, homozygous GPT2 mutations manifested in at least 2 of 176 families with IDD (approximately 1.1%) in a Pakistani cohort, thereby representing a relatively common cause of recessive IDD in this population, with recurrence of the p.R404* mutation in this population. Based on variants in the ExAC database, we estimated that approximately 1 in 248 individuals are carriers of moderately or severely deleterious variants in GPT2.


Asunto(s)
Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Genes Recesivos , Predisposición Genética a la Enfermedad , Mutación , Fenotipo , Transaminasas/genética , Adolescente , Alelos , Sustitución de Aminoácidos , Discapacidades del Desarrollo/metabolismo , Activación Enzimática , Exones , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Genética de Población , Genotipo , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Imagen por Resonancia Magnética , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , Modelos Moleculares , Linaje , Conformación Proteica , Sitios de Empalme de ARN , Análisis de Secuencia de ADN , Relación Estructura-Actividad , Transaminasas/química , Transaminasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...