Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 42(1): e27-e43, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34670408

RESUMEN

OBJECTIVE: Capillary malformation (CM) occurs sporadically and is associated with Sturge-Weber syndrome. The somatic mosaic mutation in GNAQ (c.548G>A, p.R183Q) is enriched in endothelial cells (ECs) in skin CM and Sturge-Weber syndrome brain CM. Our goal was to investigate how the mutant Gαq (G-protein αq subunit) alters EC signaling and disrupts capillary morphogenesis. Approach and Results: We used lentiviral constructs to express p.R183Q or wild-type GNAQ in normal human endothelial colony forming cells (EC-R183Q and EC-WT, respectively). EC-R183Q constitutively activated PLC (phospholipase C) ß3, a downstream effector of Gαq. Activated PLCß3 was also detected in human CM tissue sections. Bulk RNA sequencing analyses of mutant versus wild-type EC indicated constitutive activation of PKC (protein kinase C), NF-κB (nuclear factor kappa B) and calcineurin signaling in EC-R183Q. Increased expression of downstream targets in these pathways, ANGPT2 (angiopoietin-2) and DSCR (Down syndrome critical region protein) 1.4 were confirmed by quantitative PCR and immunostaining of human CM tissue sections. The Gαq inhibitor YM-254890 as well as siRNA targeted to PLCß3 reduced mRNA expression levels of these targets in EC-R183Q while the pan-PKC inhibitor AEB071 reduced ANGPT2 but not DSCR1.4. EC-R183Q formed enlarged blood vessels in mice, reminiscent of those found in human CM. shRNA knockdown of ANGPT2 in EC-R183Q normalized the enlarged vessels to sizes comparable those formed by EC-WT. CONCLUSIONS: Gαq-R183Q, when expressed in ECs, establishes constitutively active PLCß3 signaling that leads to increased ANGPT2 and a proangiogenic, proinflammatory phenotype. EC-R183Q are sufficient to form enlarged CM-like vessels in mice, and suppression of ANGPT2 prevents the enlargement. Our study provides the first evidence that endothelial Gαq-R183Q is causative for CM and identifies ANGPT2 as a contributor to CM vascular phenotype.


Asunto(s)
Angiopoyetina 2/metabolismo , Capilares/metabolismo , Células Progenitoras Endoteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Neovascularización Patológica , Síndrome de Sturge-Weber/metabolismo , Adolescente , Adulto , Anciano , Angiopoyetina 2/genética , Animales , Capilares/anomalías , Células Cultivadas , Niño , Preescolar , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Lactante , Recién Nacido , Masculino , Ratones Desnudos , Mutación , Fenotipo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Síndrome de Sturge-Weber/genética , Síndrome de Sturge-Weber/patología , Regulación hacia Arriba
2.
Genome Biol ; 22(1): 92, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33781308

RESUMEN

BACKGROUND: Post-zygotic mutations incurred during DNA replication, DNA repair, and other cellular processes lead to somatic mosaicism. Somatic mosaicism is an established cause of various diseases, including cancers. However, detecting mosaic variants in DNA from non-cancerous somatic tissues poses significant challenges, particularly if the variants only are present in a small fraction of cells. RESULTS: Here, the Brain Somatic Mosaicism Network conducts a coordinated, multi-institutional study to examine the ability of existing methods to detect simulated somatic single-nucleotide variants (SNVs) in DNA mixing experiments, generate multiple replicates of whole-genome sequencing data from the dorsolateral prefrontal cortex, other brain regions, dura mater, and dural fibroblasts of a single neurotypical individual, devise strategies to discover somatic SNVs, and apply various approaches to validate somatic SNVs. These efforts lead to the identification of 43 bona fide somatic SNVs that range in variant allele fractions from ~ 0.005 to ~ 0.28. Guided by these results, we devise best practices for calling mosaic SNVs from 250× whole-genome sequencing data in the accessible portion of the human genome that achieve 90% specificity and sensitivity. Finally, we demonstrate that analysis of multiple bulk DNA samples from a single individual allows the reconstruction of early developmental cell lineage trees. CONCLUSIONS: This study provides a unified set of best practices to detect somatic SNVs in non-cancerous tissues. The data and methods are freely available to the scientific community and should serve as a guide to assess the contributions of somatic SNVs to neuropsychiatric diseases.


Asunto(s)
Encéfalo/metabolismo , Estudios de Asociación Genética , Variación Genética , Alelos , Mapeo Cromosómico , Biología Computacional/métodos , Estudios de Asociación Genética/métodos , Genómica/métodos , Células Germinativas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple
4.
Artículo en Inglés | MEDLINE | ID: mdl-33335013

RESUMEN

Chromosomal structural variation can cause severe neurodevelopmental and neuropsychiatric phenotypes. Here we present a nonverbal female adolescent with severe stereotypic movement disorder with severe problem behavior (e.g., self-injurious behavior, aggression, and disruptive and destructive behaviors), autism spectrum disorder, severe intellectual disability, attention deficit hyperactivity disorder, and global developmental delay. Previous cytogenetic analysis revealed balanced translocations present in the patient's apparently normal mother. We hypothesized the presence of unbalanced translocations in the patient due to maternal history of spontaneous abortions. Whole-genome sequencing and whole-genome optical mapping, complementary next-generation genomic technologies capable of the accurate and robust detection of structural variants, identified t(3;10), t(10;14), and t(3;14) three-way balanced translocations in the mother and der(10)t(3;14;10) and der(14)t(3;14;10) translocations in the patient. Instead of a t(3;10), she inherited a normal maternal copy of Chromosome 3, resulting in an unbalanced state of a 3q28qter duplication and 10q26.2qter deletion. Copy-imbalanced genes in one or both of these regions, such as DLG1, DOCK1, and EBF3, may contribute to the patient's phenotype that spans neurodevelopmental, musculoskeletal, and psychiatric domains, with the possible contribution of a maternally inherited 15q13.2q13.3 deletion.


Asunto(s)
Deleción Cromosómica , Malformaciones del Sistema Nervioso/genética , Conducta Autodestructiva , Translocación Genética , Adolescente , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Homólogo 1 de la Proteína Discs Large , Femenino , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/terapia , Fenotipo , Trastorno Específico del Lenguaje/genética , Factores de Transcripción , Secuenciación Completa del Genoma , Proteínas de Unión al GTP rac
5.
Annu Rev Genet ; 54: 487-510, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32916079

RESUMEN

Mosaicism refers to the occurrence of two or more genomes in an individual derived from a single zygote. Germline mosaicism is a mutation that is limited to the gonads and can be transmitted to offspring. Somatic mosaicism is a postzygotic mutation that occurs in the soma, and it may occur at any developmental stage or in adult tissues. Mosaic variation may be classified in six ways: (a) germline or somatic origin, (b) class of DNA mutation (ranging in scale from single base pairs to multiple chromosomes), (c) developmental context, (d) body location(s), (e) functional consequence (including deleterious, neutral, or advantageous), and (f) additional sources of mosaicism, including mitochondrial heteroplasmy, exogenous DNA sources such as vectors, and epigenetic changes such as imprinting and X-chromosome inactivation. Technological advances, including single-cell and other next-generation sequencing, have facilitated improved sensitivity and specificity to detect mosaicism in a variety of biological contexts.


Asunto(s)
Genoma/genética , Mutación/genética , Animales , Cromosomas/genética , ADN/genética , Células Germinativas/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mitocondrias/genética , Mosaicismo
6.
Science ; 356(6336)2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28450582

RESUMEN

Neuropsychiatric disorders have a complex genetic architecture. Human genetic population-based studies have identified numerous heritable sequence and structural genomic variants associated with susceptibility to neuropsychiatric disease. However, these germline variants do not fully account for disease risk. During brain development, progenitor cells undergo billions of cell divisions to generate the ~80 billion neurons in the brain. The failure to accurately repair DNA damage arising during replication, transcription, and cellular metabolism amid this dramatic cellular expansion can lead to somatic mutations. Somatic mutations that alter subsets of neuronal transcriptomes and proteomes can, in turn, affect cell proliferation and survival and lead to neurodevelopmental disorders. The long life span of individual neurons and the direct relationship between neural circuits and behavior suggest that somatic mutations in small populations of neurons can significantly affect individual neurodevelopment. The Brain Somatic Mosaicism Network has been founded to study somatic mosaicism both in neurotypical human brains and in the context of complex neuropsychiatric disorders.


Asunto(s)
Encéfalo/anomalías , Trastornos Mentales/genética , Mosaicismo , Enfermedades del Sistema Nervioso/genética , Células-Madre Neurales/fisiología , Neuronas/fisiología , Encéfalo/metabolismo , División Celular/genética , Daño del ADN , Análisis Mutacional de ADN/métodos , Reparación del ADN/genética , Replicación del ADN , Genoma Humano , Células Germinativas/metabolismo , Humanos , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo
7.
Nat Commun ; 7: 13387, 2016 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-27841353

RESUMEN

The KDM4 histone demethylases are conserved epigenetic regulators linked to development, spermatogenesis and tumorigenesis. However, how the KDM4 family targets specific chromatin regions is largely unknown. Here, an extensive histone peptide microarray analysis uncovers trimethyl-lysine histone-binding preferences among the closely related KDM4 double tudor domains (DTDs). KDM4A/B DTDs bind strongly to H3K23me3, a poorly understood histone modification recently shown to be enriched in meiotic chromatin of ciliates and nematodes. The 2.28 Å co-crystal structure of KDM4A-DTD in complex with H3K23me3 peptide reveals key intermolecular interactions for H3K23me3 recognition. Furthermore, analysis of the 2.56 Å KDM4B-DTD crystal structure pinpoints the underlying residues required for exclusive H3K23me3 specificity, an interaction supported by in vivo co-localization of KDM4B and H3K23me3 at heterochromatin in mammalian meiotic and newly postmeiotic spermatocytes. In vitro demethylation assays suggest H3K23me3 binding by KDM4B stimulates H3K36 demethylation. Together, these results provide a possible mechanism whereby H3K23me3-binding by KDM4B directs localized H3K36 demethylation during meiosis and spermatogenesis.


Asunto(s)
Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Lisina/metabolismo , Animales , Sitios de Unión/genética , Cristalografía por Rayos X , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/genética , Masculino , Metilación , Ratones Endogámicos C57BL , Modelos Moleculares , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Ratas Sprague-Dawley
8.
Cell Rep ; 11(5): 808-20, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25921540

RESUMEN

The MLL gene is a common target of chromosomal translocations found in human leukemia. MLL-fusion leukemia has a consistently poor outcome. One of the most common translocation partners is AF9 (MLLT3). MLL-AF9 recruits DOT1L, a histone 3 lysine 79 methyltransferase (H3K79me1/me2/me3), leading to aberrant gene transcription. We show that DOT1L has three AF9 binding sites and present the nuclear magnetic resonance (NMR) solution structure of a DOT1L-AF9 complex. We generate structure-guided point mutations and find that they have graded effects on recruitment of DOT1L to MLL-AF9. Chromatin immunoprecipitation sequencing (ChIP-seq) analyses of H3K79me2 and H3K79me3 show that graded reduction of the DOT1L interaction with MLL-AF9 results in differential loss of H3K79me2 and me3 at MLL-AF9 target genes. Furthermore, the degree of DOT1L recruitment is linked to the level of MLL-AF9 hematopoietic transformation.


Asunto(s)
Histonas/metabolismo , Metiltransferasas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Inmunoprecipitación de Cromatina , N-Metiltransferasa de Histona-Lisina , Humanos , Espectroscopía de Resonancia Magnética , Metilación , Metiltransferasas/química , Metiltransferasas/genética , Datos de Secuencia Molecular , Proteína de la Leucemia Mieloide-Linfoide/química , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Mutación Puntual , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Análisis de Secuencia de ADN
9.
Nat Biotechnol ; 32(7): 677-83, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24837660

RESUMEN

RNA-guided genome editing with the CRISPR-Cas9 system has great potential for basic and clinical research, but the determinants of targeting specificity and the extent of off-target cleavage remain insufficiently understood. Using chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq), we mapped genome-wide binding sites of catalytically inactive Cas9 (dCas9) in HEK293T cells, in combination with 12 different single guide RNAs (sgRNAs). The number of off-target sites bound by dCas9 varied from ∼10 to >1,000 depending on the sgRNA. Analysis of off-target binding sites showed the importance of the PAM-proximal region of the sgRNA guiding sequence and that dCas9 binding sites are enriched in open chromatin regions. When targeted with catalytically active Cas9, some off-target binding sites had indels above background levels in a region around the ChIP-seq peak, but generally at lower rates than the on-target sites. Our results elucidate major determinants of Cas9 targeting, and we show that ChIP-seq allows unbiased detection of Cas9 binding sites genome-wide.


Asunto(s)
Sistemas CRISPR-Cas/genética , Mapeo Cromosómico , Desoxirribonucleasa I/genética , Células Madre Embrionarias/fisiología , Marcación de Gen/métodos , Genoma/genética , Modelos Genéticos , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Células HEK293 , Humanos , Datos de Secuencia Molecular
10.
Indian J Ophthalmol ; 60(5): 475-80, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22944763

RESUMEN

AIMS: To complete an initial estimate of the global cost of eliminating avoidable blindness, including the investment required to build ongoing primary and secondary health care systems, as well as to eliminate the 'backlog' of avoidable blindness. This analysis also seeks to understand and articulate where key data limitations lie. MATERIALS AND METHODS: Data were collected in line with a global estimation approach, including separate costing frameworks for the primary and secondary care sectors, and the treatment of backlog. RESULTS: The global direct health cost to eliminate avoidable blindness over a 10-year period from 2011 to 2020 is estimated at $632 billion per year (2009 US$). As countries already spend $592 billion per annum on eye health, this represents additional investment of $397.8 billion over 10 years, which is $40 billion per year or $5.80 per person for each year between 2010 and 2020. This is concentrated in high-income nations, which require 68% of the investment but comprise 16% of the world's inhabitants. For all other regions, the additional investment required is $127 billion. CONCLUSIONS: This costing estimate has identified that low- and middle-income countries require less than half the additional investment compared with high-income nations. Low- and middle-income countries comprise the greater investment proportion in secondary care whereas high-income countries require the majority of investment into the primary sector. However, there is a need to improve sector data. Investment in better data will have positive flow-on effects for the eye health sector.


Asunto(s)
Ceguera/economía , Costo de Enfermedad , Atención a la Salud/economía , Gastos en Salud/tendencias , Ceguera/prevención & control , Salud Global , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA