Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 172(5): 897-909.e21, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474918

RESUMEN

X-linked Dystonia-Parkinsonism (XDP) is a Mendelian neurodegenerative disease that is endemic to the Philippines and is associated with a founder haplotype. We integrated multiple genome and transcriptome assembly technologies to narrow the causal mutation to the TAF1 locus, which included a SINE-VNTR-Alu (SVA) retrotransposition into intron 32 of the gene. Transcriptome analyses identified decreased expression of the canonical cTAF1 transcript among XDP probands, and de novo assembly across multiple pluripotent stem-cell-derived neuronal lineages discovered aberrant TAF1 transcription that involved alternative splicing and intron retention (IR) in proximity to the SVA that was anti-correlated with overall TAF1 expression. CRISPR/Cas9 excision of the SVA rescued this XDP-specific transcriptional signature and normalized TAF1 expression in probands. These data suggest an SVA-mediated aberrant transcriptional mechanism associated with XDP and may provide a roadmap for layered technologies and integrated assembly-based analyses for other unsolved Mendelian disorders.


Asunto(s)
Trastornos Distónicos/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Genoma Humano , Transcriptoma/genética , Empalme Alternativo/genética , Elementos Alu/genética , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Estudios de Cohortes , Familia , Femenino , Sitios Genéticos , Haplotipos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Intrones/genética , Masculino , Repeticiones de Minisatélite/genética , Modelos Genéticos , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elementos de Nucleótido Esparcido Corto , Factores Asociados con la Proteína de Unión a TATA/genética , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/genética , Factor de Transcripción TFIID/metabolismo
2.
Am J Hum Genet ; 109(10): 1789-1813, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36152629

RESUMEN

Chromosome 16p11.2 reciprocal genomic disorder, resulting from recurrent copy-number variants (CNVs), involves intellectual disability, autism spectrum disorder (ASD), and schizophrenia, but the responsible mechanisms are not known. To systemically dissect molecular effects, we performed transcriptome profiling of 350 libraries from six tissues (cortex, cerebellum, striatum, liver, brown fat, and white fat) in mouse models harboring CNVs of the syntenic 7qF3 region, as well as cellular, transcriptional, and single-cell analyses in 54 isogenic neural stem cell, induced neuron, and cerebral organoid models of CRISPR-engineered 16p11.2 CNVs. Transcriptome-wide differentially expressed genes were largely tissue-, cell-type-, and dosage-specific, although more effects were shared between deletion and duplication and across tissue than expected by chance. The broadest effects were observed in the cerebellum (2,163 differentially expressed genes), and the greatest enrichments were associated with synaptic pathways in mouse cerebellum and human induced neurons. Pathway and co-expression analyses identified energy and RNA metabolism as shared processes and enrichment for ASD-associated, loss-of-function constraint, and fragile X messenger ribonucleoprotein target gene sets. Intriguingly, reciprocal 16p11.2 dosage changes resulted in consistent decrements in neurite and electrophysiological features, and single-cell profiling of organoids showed reciprocal alterations to the proportions of excitatory and inhibitory GABAergic neurons. Changes both in neuronal ratios and in gene expression in our organoid analyses point most directly to calretinin GABAergic inhibitory neurons and the excitatory/inhibitory balance as targets of disruption that might contribute to changes in neurodevelopmental and cognitive function in 16p11.2 carriers. Collectively, our data indicate the genomic disorder involves disruption of multiple contributing biological processes and that this disruption has relative impacts that are context specific.


Asunto(s)
Trastorno del Espectro Autista , Trastornos de los Cromosomas , Discapacidad Intelectual , Animales , Trastorno del Espectro Autista/genética , Calbindina 2/genética , Corteza Cerebral , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 16/genética , Variaciones en el Número de Copia de ADN , Genómica , Humanos , Discapacidad Intelectual/genética , Ratones , Neuronas , ARN
4.
Am J Med Genet A ; 173(9): 2478-2484, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28691782

RESUMEN

Genetic alterations of ARID1B have been recently recognized as one of the most common mendelian causes of intellectual disability and are associated with both syndromic and non-syndromic phenotypes. The ARID1B protein, a subunit of the chromatin remodeling complex SWI/SNF-A, is involved in the regulation of transcription and multiple downstream cellular processes. We report here the clinical, genetic, and proteomic phenotypes of an individual with a unique apparent de novo mutation of ARID1B due to an intragenic duplication. His neurodevelopmental phenotype includes a severe speech/language disorder with full scale IQ scores 78-98 and scattered academic skill levels, expanding the phenotypic spectrum of ARID1B mutations. Haploinsufficiency of ARID1B was determined both by RNA sequencing and quantitative RT-PCR. Fluorescence in situ hybridization analysis supported an intragenic localization of the ARID1B copy number gain. Principal component analysis revealed marked differentiation of the subject's lymphoblast proteome from that of controls. Of 3426 proteins quantified, 1014 were significantly up- or down-regulated compared to controls (q < 0.01). Pathway analysis revealed highly significant enrichment for canonical pathways of EIF2 and EIF4 signaling, protein ubiquitination, tRNA charging and chromosomal replication, among others. Network analyses revealed down-regulation of: (1) intracellular components involved in organization of membranes, organelles, and vesicles; (2) aspects of cell cycle control, signal transduction, and nuclear protein export; (3) ubiquitination and proteosomal function; and (4) aspects of mRNA synthesis/splicing. Further studies are needed to determine the detailed molecular and cellular mechanisms by which constitutional haploinsufficiency of ARID1B causes syndromic and non-syndromic developmental disabilities.


Asunto(s)
Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Factores de Transcripción/genética , Anomalías Múltiples , Adolescente , Discapacidades del Desarrollo/fisiopatología , Cara/fisiopatología , Duplicación de Gen/genética , Deformidades Congénitas de la Mano/fisiopatología , Haploinsuficiencia/genética , Humanos , Hibridación Fluorescente in Situ , Discapacidad Intelectual/fisiopatología , Masculino , Mutación , Proteómica
5.
Proc Natl Acad Sci U S A ; 111(42): E4468-77, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25294932

RESUMEN

Truncating mutations of chromodomain helicase DNA-binding protein 8 (CHD8), and of many other genes with diverse functions, are strong-effect risk factors for autism spectrum disorder (ASD), suggesting multiple mechanisms of pathogenesis. We explored the transcriptional networks that CHD8 regulates in neural progenitor cells (NPCs) by reducing its expression and then integrating transcriptome sequencing (RNA sequencing) with genome-wide CHD8 binding (ChIP sequencing). Suppressing CHD8 to levels comparable with the loss of a single allele caused altered expression of 1,756 genes, 64.9% of which were up-regulated. CHD8 showed widespread binding to chromatin, with 7,324 replicated sites that marked 5,658 genes. Integration of these data suggests that a limited array of direct regulatory effects of CHD8 produced a much larger network of secondary expression changes. Genes indirectly down-regulated (i.e., without CHD8-binding sites) reflect pathways involved in brain development, including synapse formation, neuron differentiation, cell adhesion, and axon guidance, whereas CHD8-bound genes are strongly associated with chromatin modification and transcriptional regulation. Genes associated with ASD were strongly enriched among indirectly down-regulated loci (P < 10(-8)) and CHD8-bound genes (P = 0.0043), which align with previously identified coexpression modules during fetal development. We also find an intriguing enrichment of cancer-related gene sets among CHD8-bound genes (P < 10(-10)). In vivo suppression of chd8 in zebrafish produced macrocephaly comparable to that of humans with inactivating mutations. These data indicate that heterozygous disruption of CHD8 precipitates a network of gene-expression changes involved in neurodevelopmental pathways in which many ASD-associated genes may converge on shared mechanisms of pathogenesis.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Axones/metabolismo , Sitios de Unión , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Cromatina/metabolismo , ADN Helicasas/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Genoma , Heterocigoto , Humanos , Megalencefalia/metabolismo , Mutación , Neoplasias/metabolismo , Neuronas/metabolismo , Unión Proteica , Factores de Riesgo , Análisis de Secuencia de ARN , Programas Informáticos , Pez Cebra , Proteínas de Pez Cebra/genética
6.
medRxiv ; 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38234840

RESUMEN

Glioblastoma (GBM) is a primary brain cancer with an abysmal prognosis and few effective therapies. The ability to investigate the tumor microenvironment before and during treatment would greatly enhance both understanding of disease response and progression, as well as the delivery and impact of therapeutics. Stereotactic biopsies are a routine surgical procedure performed primarily for diagnostic histopathologic purposes. The role of investigative biopsies - tissue sampling for the purpose of understanding tumor microenvironmental responses to treatment using integrated multi-modal molecular analyses ('Multi-omics") has yet to be defined. Secondly, it is unknown whether comparatively small tissue samples from brain biopsies can yield sufficient information with such methods. Here we adapt stereotactic needle core biopsy tissue in two separate patients. In the first patient with recurrent GBM we performed highly resolved multi-omics analysis methods including single cell RNA sequencing, spatial-transcriptomics, metabolomics, proteomics, phosphoproteomics, T-cell clonotype analysis, and MHC Class I immunopeptidomics from biopsy tissue that was obtained from a single procedure. In a second patient we analyzed multi-regional core biopsies to decipher spatial and genomic variance. We also investigated the utility of stereotactic biopsies as a method for generating patient derived xenograft models in a separate patient cohort. Dataset integration across modalities showed good correspondence between spatial modalities, highlighted immune cell associated metabolic pathways and revealed poor correlation between RNA expression and the tumor MHC Class I immunopeptidome. In conclusion, stereotactic needle biopsy cores are of sufficient quality to generate multi-omics data, provide data rich insight into a patient's disease process and tumor immune microenvironment and can be of value in evaluating treatment responses. One sentence summary: Integrative multi-omics analysis of stereotactic needle core biopsies in glioblastoma.

7.
J Genet Genomics ; 49(7): 654-665, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34896608

RESUMEN

Familial dysautonomia (FD), a hereditary sensory and autonomic neuropathy, is caused by a mutation in the Elongator complex protein 1 (ELP1) gene that leads to a tissue-specific reduction of ELP1 protein. Our work to generate a phenotypic mouse model for FD headed to the discovery that homozygous deletion of the mouse Elp1 gene leads to embryonic lethality prior to mid-gestation. Given that FD is caused by a reduction, not loss, of ELP1, we generated two new mouse models by introducing different copy numbers of the human FD ELP1 transgene into the Elp1 knockout mouse (Elp1-/-) and observed that human ELP1 expression rescues embryonic development in a dose-dependent manner. We then conducted a comprehensive transcriptome analysis in mouse embryos to identify genes and pathways whose expression correlates with the amount of ELP1. We found that ELP1 is essential for the expression of genes responsible for nervous system development. Further, gene length analysis of the differentially expressed genes showed that the loss of Elp1 mainly impacts the expression of long genes and that by gradually restoring Elongator, their expression is progressively rescued. Finally, through evaluation of co-expression modules, we identified gene sets with unique expression patterns that depended on ELP1 expression.


Asunto(s)
Proteínas Portadoras , Disautonomía Familiar , Animales , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Expresión Génica , Homocigoto , Humanos , Ratones , Eliminación de Secuencia
8.
J Biomol Tech ; 31(1): 7-10, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31920466

RESUMEN

Application of solid-phase reversible immobilization (SPRI) beads for size selection in molecular biology should be expanded, in light of the property of the beads to accommodate to high MW intervals of DNA fragment size selection, depending on composition of bead-suspension buffer. Here we show how the conventional size selection interval of 150-800 bp be shifted to 1.5-7 Kbp with by adjusting the concentration of NaCl in the stock suspension buffer. The MW capacity of SPRI beads also change when NaCl replaced with other cations and when the concentration of polyethylene glycol (PEG) 8000 is decreased. Testing the limits of SPRI beads revealed cuts as high as 10 Kbp are possible for some salt/PEG combinations of modified SPRI beads-suspension buffers.


Asunto(s)
ADN/aislamiento & purificación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Tampones (Química) , Cationes/química , ADN/química , Fragmentación del ADN , Peso Molecular , Polietilenglicoles/química
9.
J Biomol Tech ; 31(4): 151-156, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33100919

RESUMEN

Advances in next-generation sequencing technologies have allowed RNA sequencing to become an increasingly time efficient, cost-effective, and accessible tool for genomic research. We present here an automated and miniaturized workflow for RNA library preparation that minimizes reagent usage and processing time required per sample to generate Illumina compatible libraries for sequencing. The reduced-volume libraries show similar behavior to full-scale libraries with comparable numbers of genes detected and reproducible clustering of samples.


Asunto(s)
Automatización/métodos , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , RNA-Seq/métodos , Genómica , ARN/aislamiento & purificación , Reproducibilidad de los Resultados , Flujo de Trabajo
10.
Elife ; 92020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32990597

RESUMEN

Somatic expansion of the Huntington's disease (HD) CAG repeat drives the rate of a pathogenic process ultimately resulting in neuronal cell death. Although mechanisms of toxicity are poorly delineated, transcriptional dysregulation is a likely contributor. To identify modifiers that act at the level of CAG expansion and/or downstream pathogenic processes, we tested the impact of genetic knockout, in HttQ111 mice, of Hdac2 or Hdac3 in medium-spiny striatal neurons that exhibit extensive CAG expansion and exquisite disease vulnerability. Both knockouts moderately attenuated CAG expansion, with Hdac2 knockout decreasing nuclear huntingtin pathology. Hdac2 knockout resulted in a substantial transcriptional response that included modification of transcriptional dysregulation elicited by the HttQ111 allele, likely via mechanisms unrelated to instability suppression. Our results identify novel modifiers of different aspects of HD pathogenesis in medium-spiny neurons and highlight a complex relationship between the expanded Htt allele and Hdac2 with implications for targeting transcriptional dysregulation in HD.


Asunto(s)
Cuerpo Estriado/fisiopatología , Histona Desacetilasa 2/genética , Histona Desacetilasas/genética , Enfermedad de Huntington/genética , Neuronas/fisiología , Animales , Núcleo Celular , Modelos Animales de Enfermedad , Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/enzimología , Ratones , Ratones Endogámicos C57BL
11.
Mol Autism ; 11(1): 45, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32503625

RESUMEN

BACKGROUND: MBD5, encoding the methyl-CpG-binding domain 5 protein, has been proposed as a necessary and sufficient driver of the 2q23.1 microdeletion syndrome. De novo missense and protein-truncating variants from exome sequencing studies have directly implicated MBD5 in the etiology of autism spectrum disorder (ASD) and related neurodevelopmental disorders (NDDs). However, little is known concerning the specific function(s) of MBD5. METHODS: To gain insight into the complex interactions associated with alteration of MBD5 in individuals with ASD and related NDDs, we explored the transcriptional landscape of MBD5 haploinsufficiency across multiple mouse brain regions of a heterozygous hypomorphic Mbd5+/GT mouse model, and compared these results to CRISPR-mediated mutations of MBD5 in human iPSC-derived neuronal models. RESULTS: Gene expression analyses across three brain regions from Mbd5+/GT mice showed subtle transcriptional changes, with cortex displaying the most widespread changes following Mbd5 reduction, indicating context-dependent effects. Comparison with MBD5 reduction in human neuronal cells reinforced the context-dependence of gene expression changes due to MBD5 deficiency. Gene co-expression network analyses revealed gene clusters that were associated with reduced MBD5 expression and enriched for terms related to ciliary function. LIMITATIONS: These analyses included a limited number of mouse brain regions and neuronal models, and the effects of the gene knockdown are subtle. As such, these results will not reflect the full extent of MBD5 disruption across human brain regions during early neurodevelopment in ASD, or capture the diverse spectrum of cell-type-specific changes associated with MBD5 alterations. CONCLUSIONS: Our study points to modest and context-dependent transcriptional consequences of Mbd5 disruption in the brain. It also suggests a possible link between MBD5 and perturbations in ciliary function, which is an established pathogenic mechanism in developmental disorders and syndromes.


Asunto(s)
Encéfalo/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Mutación , Neuronas/metabolismo , Transcripción Genética , Animales , Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/metabolismo , Sistemas CRISPR-Cas , Diferenciación Celular/genética , Línea Celular , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Transgénicos , Neuronas/citología
12.
Mol Neuropsychiatry ; 3(1): 53-71, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28879201

RESUMEN

Genetic variation within the transcription factor TCF4 locus can cause the intellectual disability and developmental disorder Pitt-Hopkins syndrome (PTHS), whereas single-nucleotide polymorphisms within noncoding regions are associated with schizophrenia. These genetic findings position TCF4 as a link between transcription and cognition; however, the neurobiology of TCF4 remains poorly understood. Here, we quantitated multiple distinct TCF4 transcript levels in human induced pluripotent stem cell-derived neural progenitors and differentiated neurons, and PTHS patient fibroblasts. We identify two classes of pharmacological treatments that regulate TCF4 expression: WNT pathway activation and inhibition of class I histone deacetylases. In PTHS fibroblasts, both of these perturbations upregulate a subset of TCF4 transcripts. Finally, using chromatin immunoprecipitation sequencing in conjunction with genome-wide transcriptome analysis, we identified TCF4 target genes that may mediate the effect of TCF4 loss on neuroplasticity. Our studies identify new pharmacological assays, tools, and targets for the development of therapeutics for cognitive disorders.

13.
Genome Biol ; 18(1): 36, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28260531

RESUMEN

BACKGROUND: Structural variation (SV) influences genome organization and contributes to human disease. However, the complete mutational spectrum of SV has not been routinely captured in disease association studies. RESULTS: We sequenced 689 participants with autism spectrum disorder (ASD) and other developmental abnormalities to construct a genome-wide map of large SV. Using long-insert jumping libraries at 105X mean physical coverage and linked-read whole-genome sequencing from 10X Genomics, we document seven major SV classes at ~5 kb SV resolution. Our results encompass 11,735 distinct large SV sites, 38.1% of which are novel and 16.8% of which are balanced or complex. We characterize 16 recurrent subclasses of complex SV (cxSV), revealing that: (1) cxSV are larger and rarer than canonical SV; (2) each genome harbors 14 large cxSV on average; (3) 84.4% of large cxSVs involve inversion; and (4) most large cxSV (93.8%) have not been delineated in previous studies. Rare SVs are more likely to disrupt coding and regulatory non-coding loci, particularly when truncating constrained and disease-associated genes. We also identify multiple cases of catastrophic chromosomal rearrangements known as chromoanagenesis, including somatic chromoanasynthesis, and extreme balanced germline chromothripsis events involving up to 65 breakpoints and 60.6 Mb across four chromosomes, further defining rare categories of extreme cxSV. CONCLUSIONS: These data provide a foundational map of large SV in the morbid human genome and demonstrate a previously underappreciated abundance and diversity of cxSV that should be considered in genomic studies of human disease.


Asunto(s)
Aberraciones Cromosómicas , Inversión Cromosómica , Cromotripsis , Genoma Humano , Genómica , Trastorno del Espectro Autista/genética , Orden Génico , Reordenamiento Génico , Predisposición Genética a la Enfermedad , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación
14.
Nat Genet ; 49(2): 238-248, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28067909

RESUMEN

Arhinia, or absence of the nose, is a rare malformation of unknown etiology that is often accompanied by ocular and reproductive defects. Sequencing of 40 people with arhinia revealed that 84% of probands harbor a missense mutation localized to a constrained region of SMCHD1 encompassing the ATPase domain. SMCHD1 mutations cause facioscapulohumeral muscular dystrophy type 2 (FSHD2) via a trans-acting loss-of-function epigenetic mechanism. We discovered shared mutations and comparable DNA hypomethylation patterning between these distinct disorders. CRISPR/Cas9-mediated alteration of smchd1 in zebrafish yielded arhinia-relevant phenotypes. Transcriptome and protein analyses in arhinia probands and controls showed no differences in SMCHD1 mRNA or protein abundance but revealed regulatory changes in genes and pathways associated with craniofacial patterning. Mutations in SMCHD1 thus contribute to distinct phenotypic spectra, from craniofacial malformation and reproductive disorders to muscular dystrophy, which we speculate to be consistent with oligogenic mechanisms resulting in pleiotropic outcomes.


Asunto(s)
Atresia de las Coanas/genética , Proteínas Cromosómicas no Histona/genética , Predisposición Genética a la Enfermedad/genética , Microftalmía/genética , Distrofias Musculares/genética , Mutación/genética , Nariz/anomalías , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Fenotipo
15.
Cancer Res ; 62(17): 4985-91, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12208750

RESUMEN

Molecules differentially expressed or overexpressed by malignant cells can serve in detecting and tracking of tumor. Additionally, they potentially can be applied in histologic-specific antitumor therapy. Few breast cancer-associated candidate molecules have been identified. Here we describe the use of combinatorial immunoglobulin [antigen-binding fragment of immunoglobulin molecule (Fab) fragment] phage libraries generated from patients with breast carcinoma to identify cancer-associated gene expression. The libraries were enriched for tumor-binding Fab by 3 logs and yielded a group of antibodies against DNA-binding protein B (DbpB), a 35-kDa thrombin-inducible nuclear factor and member of the Y-box family of proteins, which are known to act both negatively in selective gene suppression and positively as promoters of gene transcription. Sequencing of the anti-DbpB showed a degree of heterogeneity and bp substitutions suggesting that the Fabs selected from the combinatorial library represented a varied anti-DbpB immune response and did not simply arise from in vitro amplification by PCR of a single or limited numbers of immunoglobulin genes. Sequencing of the DbpB molecule expressed in malignant breast cancer showed no evidence of tumor-specific mutations. Evaluation of levels of DbpB gene product expression however showed the molecule to be constitutively expressed in normal nonmalignant breast tissues but to have consistently differentially higher expression in breast cancer. Immunohistological staining revealed DbpB to be present both intracellularly and on the cell surface, which suggests it may be a means whereby malignant cells repair and replicate DNA in a selectively advantageous manner as compared with nonmalignant cells. DbpB expression in breast cancer may advance the basic understanding of the role of Y-box binding proteins as regulatory agents, and in defining malignant cell phenotypes. In addition, DbpB and the antibodies generated against it may have direct application in tumor detection and in molecule-targeted immunotherapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas de Unión al ADN , Inmunoglobulinas/genética , Factores de Transcripción , Secuencia de Aminoácidos , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/inmunología , Técnicas Químicas Combinatorias , ADN Complementario/genética , Genes de Inmunoglobulinas , Biblioteca Genómica , Humanos , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/metabolismo , Inmunoglobulinas/metabolismo , Datos de Secuencia Molecular , Factores de Transcripción NFI , Proteínas Nucleares , Biblioteca de Péptidos , Proteína 1 de Unión a la Caja Y
16.
Nat Neurosci ; 19(3): 517-22, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26829649

RESUMEN

Recurrent, reciprocal genomic disorders resulting from non-allelic homologous recombination (NAHR) between near-identical segmental duplications (SDs) are a major cause of human disease, often producing phenotypically distinct syndromes. The genomic architecture of flanking SDs presents a challenge for modeling these syndromes; however, the capability to efficiently generate reciprocal copy number variants (CNVs) that mimic NAHR would represent a valuable modeling tool. We describe here a CRISPR/Cas9 genome engineering method, single-guide CRISPR/Cas targeting of repetitive elements (SCORE), to model reciprocal genomic disorders and demonstrate its capabilities by generating reciprocal CNVs of 16p11.2 and 15q13.3, including alteration of one copy-equivalent of the SDs that mediate NAHR in vivo. The method is reproducible, and RNA sequencing reliably clusters transcriptional signatures from human subjects with in vivo CNVs and their corresponding in vitro models. This new approach will provide broad applicability for the study of genomic disorders and, with further development, may also permit efficient correction of these defects.


Asunto(s)
Trastorno Autístico/genética , Sistemas CRISPR-Cas/genética , Trastornos de los Cromosomas/genética , Variaciones en el Número de Copia de ADN/genética , Ingeniería Genética/métodos , Discapacidad Intelectual/genética , Duplicaciones Segmentarias en el Genoma/genética , Convulsiones/genética , Eliminación de Secuencia/genética , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Cromosomas Humanos Par 16/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Humanos
17.
Cell Stem Cell ; 15(5): 643-52, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25517468

RESUMEN

Genome editing via CRISPR/Cas9 has rapidly become the tool of choice by virtue of its efficacy and ease of use. However, CRISPR/Cas9-mediated genome editing in clinically relevant human somatic cells remains untested. Here, we report CRISPR/Cas9 targeting of two clinically relevant genes, B2M and CCR5, in primary human CD4+ T cells and CD34+ hematopoietic stem and progenitor cells (HSPCs). Use of single RNA guides led to highly efficient mutagenesis in HSPCs but not in T cells. A dual guide approach improved gene deletion efficacy in both cell types. HSPCs that had undergone genome editing with CRISPR/Cas9 retained multilineage potential. We examined predicted on- and off-target mutations via target capture sequencing in HSPCs and observed low levels of off-target mutagenesis at only one site. These results demonstrate that CRISPR/Cas9 can efficiently ablate genes in HSPCs with minimal off-target mutagenesis, which could have broad applicability for hematopoietic cell-based therapy.


Asunto(s)
Proteínas Asociadas a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Animales , Antígenos CD34/metabolismo , Linaje de la Célula , Células Cultivadas , Marcación de Gen , Sitios Genéticos , Genoma Humano/genética , Células Madre Hematopoyéticas/citología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Edición de ARN/genética , ARN Guía de Kinetoplastida/metabolismo , Receptores CCR5/metabolismo
18.
PLoS One ; 8(5): e62481, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23650516

RESUMEN

Genomic instability is a known precursor to cancer and aging. The RecQ helicases are a highly conserved family of DNA-unwinding enzymes that play key roles in maintaining genome stability in all living organisms. Human RecQ homologs include RECQ1, BLM, WRN, RECQ4, and RECQ5ß, three of which have been linked to diseases with elevated risk of cancer and growth defects (Bloom Syndrome and Rothmund-Thomson Syndrome) or premature aging (Werner Syndrome). RECQ1, the first RecQ helicase discovered and the most abundant in human cells, is the least well understood of the five human RecQ homologs. We have previously described that knockout of RECQ1 in mice or knockdown of its expression in human cells results in elevated frequency of spontaneous sister chromatid exchanges, chromosomal instability, increased load of DNA damage and heightened sensitivity to ionizing radiation. We have now obtained evidence implicating RECQ1 in the nonhomologous end-joining pathway of DNA double-strand break repair. We show that RECQ1 interacts directly with the Ku70/80 subunit of the DNA-PK complex, and depletion of RECQ1 results in reduced end-joining in cell free extracts. In vitro, RECQ1 binds and unwinds the Ku70/80-bound partial duplex DNA substrate efficiently. Linear DNA is co-bound by RECQ1 and Ku70/80, and DNA binding by Ku70/80 is modulated by RECQ1. Collectively, these results provide the first evidence for an interaction of RECQ1 with Ku70/80 and a role of the human RecQ helicase in double-strand break repair through nonhomologous end-joining.


Asunto(s)
Antígenos Nucleares/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , RecQ Helicasas/metabolismo , Antígenos Nucleares/química , ADN/química , Proteínas de Unión al ADN/química , Ensayo de Cambio de Movilidad Electroforética , Células HeLa , Humanos , Autoantígeno Ku , Conformación de Ácido Nucleico , Oligonucleótidos/química , Unión Proteica , Mapas de Interacción de Proteínas , RecQ Helicasas/química
19.
DNA Repair (Amst) ; 11(6): 537-49, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22542292

RESUMEN

RECQ1 is the most abundant RecQ homolog in humans but its functions have remained mostly elusive. Biochemically, RECQ1 displays distinct substrate specificities from WRN and BLM, indicating that these RecQ helicases likely perform non-overlapping functions. Our earlier work demonstrated that RECQ1-deficient cells display spontaneous genomic instability. We have obtained key evidence suggesting a unique role of RECQ1 in repair of oxidative DNA damage. We show that similar to WRN, RECQ1 associates with PARP-1 in nuclear extracts and exhibits direct protein interaction in vitro. Deficiency in WRN or BLM helicases have been shown to result in reduced homologous recombination and hyperactivation of PARP under basal condition. However, RECQ1-deficiency did not lead to PARP activation in undamaged cells and nor did it result in reduction in homologous recombination repair. In stark contrast to what is seen in WRN-deficiency, RECQ1-deficient cells hyperactivate PARP in a specific response to H2O2treatment. RECQ1-deficient cells are more sensitive to oxidative DNA damage and exposure to oxidative stress results in a rapid and reversible recruitment of RECQ1 to chromatin. Chromatin localization of RECQ1 precedes WRN helicase, which has been shown to function in oxidative DNA damage repair. However, oxidative DNA damage-induced chromatin recruitment of these RecQ helicases is independent of PARP activity. As other RecQ helicases are known to interact with PARP-1, this study provides a paradigm to delineate specialized and redundant functions of RecQ homologs in repair of oxidative DNA damage.


Asunto(s)
Daño del ADN , Reparación del ADN , Poli(ADP-Ribosa) Polimerasas/metabolismo , RecQ Helicasas/metabolismo , Western Blotting , Línea Celular Tumoral , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Roturas del ADN de Cadena Simple/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células HeLa , Recombinación Homóloga , Humanos , Peróxido de Hidrógeno/farmacología , Inmunoprecipitación , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/genética , Unión Proteica , Interferencia de ARN , RecQ Helicasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA