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










Base de datos
Intervalo de año de publicación
1.
PLoS Genet ; 19(6): e1010759, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37289818

RESUMEN

Gene regulation is highly cell type-specific and understanding the function of non-coding genetic variants associated with complex traits requires molecular phenotyping at cell type resolution. In this study we performed single nucleus ATAC-seq (snATAC-seq) and genotyping in peripheral blood mononuclear cells from 13 individuals. Clustering chromatin accessibility profiles of 96,002 total nuclei identified 17 immune cell types and sub-types. We mapped chromatin accessibility QTLs (caQTLs) in each immune cell type and sub-type using individuals of European ancestry which identified 6,901 caQTLs at FDR < .10 and 4,220 caQTLs at FDR < .05, including those obscured from assays of bulk tissue such as with divergent effects on different cell types. For 3,941 caQTLs we further annotated putative target genes of variant activity using single cell co-accessibility, and caQTL variants were significantly correlated with the accessibility level of linked gene promoters. We fine-mapped loci associated with 16 complex immune traits and identified immune cell caQTLs at 622 candidate causal variants, including those with cell type-specific effects. At the 6q15 locus associated with type 1 diabetes, in line with previous reports, variant rs72928038 was a naïve CD4+ T cell caQTL linked to BACH2 and we validated the allelic effects of this variant on regulatory activity in Jurkat T cells. These results highlight the utility of snATAC-seq for mapping genetic effects on accessible chromatin in specific cell types.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Cromatina , Humanos , Cromatina/genética , Herencia Multifactorial , Leucocitos Mononucleares , Sitios de Carácter Cuantitativo/genética
2.
Proc Natl Acad Sci U S A ; 120(20): e2210991120, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37155843

RESUMEN

In 2021, the World Health Organization reclassified glioblastoma, the most common form of adult brain cancer, into isocitrate dehydrogenase (IDH)-wild-type glioblastomas and grade IV IDH mutant (G4 IDHm) astrocytomas. For both tumor types, intratumoral heterogeneity is a key contributor to therapeutic failure. To better define this heterogeneity, genome-wide chromatin accessibility and transcription profiles of clinical samples of glioblastomas and G4 IDHm astrocytomas were analyzed at single-cell resolution. These profiles afforded resolution of intratumoral genetic heterogeneity, including delineation of cell-to-cell variations in distinct cell states, focal gene amplifications, as well as extrachromosomal circular DNAs. Despite differences in IDH mutation status and significant intratumoral heterogeneity, the profiled tumor cells shared a common chromatin structure defined by open regions enriched for nuclear factor 1 transcription factors (NFIA and NFIB). Silencing of NFIA or NFIB suppressed in vitro and in vivo growths of patient-derived glioblastomas and G4 IDHm astrocytoma models. These findings suggest that despite distinct genotypes and cell states, glioblastoma/G4 astrocytoma cells share dependency on core transcriptional programs, yielding an attractive platform for addressing therapeutic challenges associated with intratumoral heterogeneity.


Asunto(s)
Astrocitoma , Neoplasias Encefálicas , Glioblastoma , Adulto , Humanos , Glioblastoma/genética , Glioblastoma/patología , Cromatina/genética , Transcriptoma , Astrocitoma/genética , Astrocitoma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Mutación , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo
3.
Nat Genet ; 55(6): 984-994, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37231096

RESUMEN

Dysfunctional pancreatic islet beta cells are a hallmark of type 2 diabetes (T2D), but a comprehensive understanding of the underlying mechanisms, including gene dysregulation, is lacking. Here we integrate information from measurements of chromatin accessibility, gene expression and function in single beta cells with genetic association data to nominate disease-causal gene regulatory changes in T2D. Using machine learning on chromatin accessibility data from 34 nondiabetic, pre-T2D and T2D donors, we identify two transcriptionally and functionally distinct beta cell subtypes that undergo an abundance shift during T2D progression. Subtype-defining accessible chromatin is enriched for T2D risk variants, suggesting a causal contribution of subtype identity to T2D. Both beta cell subtypes exhibit activation of a stress-response transcriptional program and functional impairment in T2D, which is probably induced by the T2D-associated metabolic environment. Our findings demonstrate the power of multimodal single-cell measurements combined with machine learning for characterizing mechanisms of complex diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Diabetes Mellitus Tipo 2/genética , Multiómica , Células Secretoras de Insulina/metabolismo , Regulación de la Expresión Génica , Cromatina/metabolismo
4.
bioRxiv ; 2023 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-36711922

RESUMEN

Altered function and gene regulation of pancreatic islet beta cells is a hallmark of type 2 diabetes (T2D), but a comprehensive understanding of mechanisms driving T2D is still missing. Here we integrate information from measurements of chromatin activity, gene expression and function in single beta cells with genetic association data to identify disease-causal gene regulatory changes in T2D. Using machine learning on chromatin accessibility data from 34 non-diabetic, pre-T2D and T2D donors, we robustly identify two transcriptionally and functionally distinct beta cell subtypes that undergo an abundance shift in T2D. Subtype-defining active chromatin is enriched for T2D risk variants, suggesting a causal contribution of subtype identity to T2D. Both subtypes exhibit activation of a stress-response transcriptional program and functional impairment in T2D, which is likely induced by the T2D-associated metabolic environment. Our findings demonstrate the power of multimodal single-cell measurements combined with machine learning for identifying mechanisms of complex diseases.

5.
Nature ; 598(7879): 129-136, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616068

RESUMEN

The mammalian cerebrum performs high-level sensory perception, motor control and cognitive functions through highly specialized cortical and subcortical structures1. Recent surveys of mouse and human brains with single-cell transcriptomics2-6 and high-throughput imaging technologies7,8 have uncovered hundreds of neural cell types distributed in different brain regions, but the transcriptional regulatory programs that are responsible for the unique identity and function of each cell type remain unknown. Here we probe the accessible chromatin in more than 800,000 individual nuclei from 45 regions that span the adult mouse isocortex, olfactory bulb, hippocampus and cerebral nuclei, and use the resulting data to map the state of 491,818 candidate cis-regulatory DNA elements in 160 distinct cell types. We find high specificity of spatial distribution for not only excitatory neurons, but also most classes of inhibitory neurons and a subset of glial cell types. We characterize the gene regulatory sequences associated with the regional specificity within these cell types. We further link a considerable fraction of the cis-regulatory elements to putative target genes expressed in diverse cerebral cell types and predict transcriptional regulators that are involved in a broad spectrum of molecular and cellular pathways in different neuronal and glial cell populations. Our results provide a foundation for comprehensive analysis of gene regulatory programs of the mammalian brain and assist in the interpretation of noncoding risk variants associated with various neurological diseases and traits in humans.


Asunto(s)
Cerebro/citología , Cerebro/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Atlas como Asunto , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/genética , Neuroglía/clasificación , Neuroglía/metabolismo , Neuronas/clasificación , Neuronas/metabolismo , Análisis de Secuencia de ADN , Análisis de la Célula Individual
6.
Nature ; 594(7863): 398-402, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34012112

RESUMEN

Genetic risk variants that have been identified in genome-wide association studies of complex diseases are primarily non-coding1. Translating these risk variants into mechanistic insights requires detailed maps of gene regulation in disease-relevant cell types2. Here we combined two approaches: a genome-wide association study of type 1 diabetes (T1D) using 520,580 samples, and the identification of candidate cis-regulatory elements (cCREs) in pancreas and peripheral blood mononuclear cells using single-nucleus assay for transposase-accessible chromatin with sequencing (snATAC-seq) of 131,554 nuclei. Risk variants for T1D were enriched in cCREs that were active in T cells and other cell types, including acinar and ductal cells of the exocrine pancreas. Risk variants at multiple T1D signals overlapped with exocrine-specific cCREs that were linked to genes with exocrine-specific expression. At the CFTR locus, the T1D risk variant rs7795896 mapped to a ductal-specific cCRE that regulated CFTR; the risk allele reduced transcription factor binding, enhancer activity and CFTR expression in ductal cells. These findings support a role for the exocrine pancreas in the pathogenesis of T1D and highlight the power of large-scale genome-wide association studies and single-cell epigenomics for understanding the cellular origins of complex disease.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Epigenómica , Predisposición Genética a la Enfermedad , Análisis de la Célula Individual , Cromatina/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Femenino , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Inmunidad/genética , Masculino , Conductos Pancreáticos/metabolismo , Conductos Pancreáticos/patología
7.
Nat Genet ; 53(4): 455-466, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33795864

RESUMEN

Single-nucleus assay for transposase-accessible chromatin using sequencing (snATAC-seq) creates new opportunities to dissect cell type-specific mechanisms of complex diseases. Since pancreatic islets are central to type 2 diabetes (T2D), we profiled 15,298 islet cells by using combinatorial barcoding snATAC-seq and identified 12 clusters, including multiple alpha, beta and delta cell states. We cataloged 228,873 accessible chromatin sites and identified transcription factors underlying lineage- and state-specific regulation. We observed state-specific enrichment of fasting glucose and T2D genome-wide association studies for beta cells and enrichment for other endocrine cell types. At T2D signals localized to islet-accessible chromatin, we prioritized variants with predicted regulatory function and co-accessibility with target genes. A causal T2D variant rs231361 at the KCNQ1 locus had predicted effects on a beta cell enhancer co-accessible with INS and genome editing in embryonic stem cell-derived beta cells affected INS levels. Together our findings demonstrate the power of single-cell epigenomics for interpreting complex disease genetics.


Asunto(s)
Cromatina/química , Diabetes Mellitus Tipo 2/genética , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Canal de Potasio KCNQ1/genética , Células Secretoras de Polipéptido Pancreático/metabolismo , Células Secretoras de Somatostatina/metabolismo , Glucemia/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Epigenómica , Ayuno , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Células Secretoras de Glucagón/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Células Madre Embrionarias Humanas/citología , Humanos , Células Secretoras de Insulina/patología , Canal de Potasio KCNQ1/metabolismo , Familia de Multigenes , Células Secretoras de Polipéptido Pancreático/patología , Polimorfismo Genético , Análisis de la Célula Individual , Células Secretoras de Somatostatina/patología , Factores de Transcripción/clasificación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Nature ; 583(7818): 744-751, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728240

RESUMEN

The Encyclopedia of DNA Elements (ENCODE) project has established a genomic resource for mammalian development, profiling a diverse panel of mouse tissues at 8 developmental stages from 10.5 days after conception until birth, including transcriptomes, methylomes and chromatin states. Here we systematically examined the state and accessibility of chromatin in the developing mouse fetus. In total we performed 1,128 chromatin immunoprecipitation with sequencing (ChIP-seq) assays for histone modifications and 132 assay for transposase-accessible chromatin using sequencing (ATAC-seq) assays for chromatin accessibility across 72 distinct tissue-stages. We used integrative analysis to develop a unified set of chromatin state annotations, infer the identities of dynamic enhancers and key transcriptional regulators, and characterize the relationship between chromatin state and accessibility during developmental gene regulation. We also leveraged these data to link enhancers to putative target genes and demonstrate tissue-specific enrichments of sequence variants associated with disease in humans. The mouse ENCODE data sets provide a compendium of resources for biomedical researchers and achieve, to our knowledge, the most comprehensive view of chromatin dynamics during mammalian fetal development to date.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Conjuntos de Datos como Asunto , Desarrollo Fetal/genética , Histonas/metabolismo , Anotación de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Cromatina/química , Secuenciación de Inmunoprecipitación de Cromatina , Enfermedad/genética , Elementos de Facilitación Genéticos/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Variación Genética , Histonas/química , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/genética , Reproducibilidad de los Resultados , Transposasas/metabolismo
11.
FASEB J ; 34(5): 6449-6465, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32196731

RESUMEN

The steroid hormone ecdysone is the central regulator of insect metamorphosis, during which a growing, immature larva is remodeled, through pupal stages, to a reproductive adult. However, the underlying mechanisms of ecdysone-mediated metamorphosis remain to be fully elucidated. Here, we identified metamorphosis-associated microRNAs (miRNAs) and their potential targets by cross-linking immunoprecipitation coupled with deep sequencing of endogenous Argonaute 1 protein in Drosophila. Interestingly, miR-8-3p targeted five Vha genes encoding distinct subunits of vacuolar H+ -ATPase (V-ATPase), which has a vital role in the organellar acidification. The expression of ecdysone-responsive miR-8-3p is normally downregulated during Drosophila metamorphosis, but temporary overexpression of miR-8-3p in the whole body at the end of larval development led to defects in metamorphosis and survival, hallmarks of aberrant ecdysone signaling. In addition, miR-8-3p was expressed in the prothoracic gland (PG), which produces and releases ecdysone in response to prothoracicotropic hormone (PTTH). Notably, overexpression of miR-8-3p or knockdown of its Vha targets in the PG resulted in larger than normal, ecdysone-deficient larvae that failed to develop into the pupal stage but could be rescued by ecdysone feeding. Moreover, these animals showed defective PTTH signaling with a concomitant decrease in the expression of ecdysone biosynthetic genes. We also demonstrated that the regulatory network between the conserved miR-8-3p/miR-200 family and V-ATPase was functional in human cells. Consequently, our data indicate that the coordinated regulation of V-ATPase subunits by miR-8-3p is involved in Drosophila metamorphosis by controlling the ecdysone biosynthesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Ecdisona/biosíntesis , Metamorfosis Biológica , MicroARNs/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteínas de Drosophila/genética , ATPasas de Translocación de Protón Vacuolares/genética
12.
Mol Cancer Res ; 17(11): 2208-2220, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31444232

RESUMEN

The epidermal growth factor receptor (EGFR) is overexpressed in numerous solid tumors and is the subject of extensive therapeutic efforts. Much of the research on EGFR is focused on protein dynamics and downstream signaling; however, few studies have explored its transcriptional regulation. Here, we identified two enhancers (CE1 and CE2) present within the first intron of the EGFR gene in models of glioblastoma (GBM) and head and neck squamous cell carcinoma (HNSCC). CE1 and CE2 contain open chromatin and H3K27Ac histone marks, enhance transcription in reporter assays, and interact with the EGFR promoter. Enhancer genetic deletion by CRISPR/Cas9 significantly reduces EGFR transcript levels, with double deletion exercising an additive effect. Targeted repression of CE1 and CE2 by dCas9-KRAB demonstrates repression of transcription similar to that of genomic deletion. We identify AP-1 transcription factor family members in concert with BET bromodomain proteins as modulators of CE1 and CE2 activity in HNSCC and GBM through de novo motif identification and validate their presence. Genetic inhibition of AP-1 or pharmacologic disruption of BET/AP-1 binding results in downregulated EGFR protein and transcript levels, confirming a role for these factors in CE1 and CE2. Our results identify and characterize these novel enhancers, shedding light on the role that epigenetic mechanisms play in regulating EGFR transcription in EGFR-dependent cancers. IMPLICATIONS: We identify critical constituent enhancers present in the first intron of the EGFR gene, and provide a rationale for therapeutic targeting of EGFR intron 1 enhancers through perturbation of AP-1 and BET in EGFR-positive malignancies.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Proteínas/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Factor de Transcripción AP-1/metabolismo , Línea Celular Tumoral , Cromatina/genética , Epigénesis Genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Genes Reporteros , Glioblastoma/patología , Humanos , Intrones/genética , Regiones Promotoras Genéticas/genética , Proteínas/genética , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Factor de Transcripción AP-1/genética
13.
Nat Commun ; 10(1): 2078, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31064983

RESUMEN

Genetic variants affecting pancreatic islet enhancers are central to T2D risk, but the gene targets of islet enhancer activity are largely unknown. We generate a high-resolution map of islet chromatin loops using Hi-C assays in three islet samples and use loops to annotate target genes of islet enhancers defined using ATAC-seq and published ChIP-seq data. We identify candidate target genes for thousands of islet enhancers, and find that enhancer looping is correlated with islet-specific gene expression. We fine-map T2D risk variants affecting islet enhancers, and find that candidate target genes of these variants defined using chromatin looping and eQTL mapping are enriched in protein transport and secretion pathways. At IGF2BP2, a fine-mapped T2D variant reduces islet enhancer activity and IGF2BP2 expression, and conditional inactivation of IGF2BP2 in mouse islets impairs glucose-stimulated insulin secretion. Our findings provide a resource for studying islet enhancer function and identifying genes involved in T2D risk.


Asunto(s)
Cromatina/metabolismo , Diabetes Mellitus Tipo 2/genética , Redes Reguladoras de Genes/genética , Islotes Pancreáticos/metabolismo , Proteínas de Unión al ARN/genética , Adulto , Animales , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina/genética , Diabetes Mellitus Tipo 2/patología , Elementos de Facilitación Genéticos/genética , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Conformación Molecular , Sitios de Carácter Cuantitativo/genética , Proteínas de Unión al ARN/metabolismo
14.
FASEB J ; 32(8): 4519-4533, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29543534

RESUMEN

The steroid hormone ecdysone has a central role in the developmental transitions of insects through its control of responsive protein-coding and microRNA (miRNA) gene expression. However, the complete regulatory network controlling the expression of these genes remains to be elucidated. In this study, we performed cross-linking immunoprecipitation coupled with deep sequencing of endogenous Argonaute 1 (Ago1) protein, the core effector of the miRNA pathway, in Drosophila S2 cells. We found that regulatory interactions between miRNAs and their cognate targets were substantially altered by Ago1 in response to ecdysone signaling. Additionally, during the larva-to-adult metamorphosis, miR-252-5p was up-regulated via the canonical ecdysone-signaling pathway. Moreover, we provide evidence that miR-252-5p targets Abelson interacting protein ( Abi) to decrease the protein levels of cyclins A and B, controlling the cell cycle. Overall, our data suggest a potential role for the ecdysone/miR-252-5p/Abi regulatory axis partly in cell-cycle control during metamorphosis in Drosophila.-Lim, D.-H., Lee, S., Han, J. Y., Choi, M.-S., Hong, J.-S., Seong, Y., Kwon, Y.-S., Lee, Y. S. Ecdysone-responsive microR-252-5p controls the cell cycle by targeting Abi in Drosophila.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclo Celular/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Ecdisona/metabolismo , MicroARNs/metabolismo , Animales , Proteínas Argonautas/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Larva/metabolismo , Transporte de Proteínas/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
15.
Mol Cancer Ther ; 16(10): 2178-2190, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28611106

RESUMEN

Colorectal cancer is the third most commonly diagnosed cancer in the world, and exhibits heterogeneous characteristics in terms of genomic alterations, expression signature, and drug responsiveness. Although there have been considerable efforts to classify this disease based on high-throughput sequencing techniques, targeted treatments for specific subgroups have been limited. KRAS and BRAF mutations are prevalent genetic alterations in colorectal cancers, and patients with mutations in either of these genes have a worse prognosis and are resistant to anti-EGFR treatments. In this study, we have found that a subgroup of colorectal cancers, defined by having either KRAS or BRAF (KRAS/BRAF) mutations and BCL2L1 (encoding BCL-XL) amplification, can be effectively targeted by simultaneous inhibition of BCL-XL (with ABT-263) and MCL1 (with YM-155). This combination treatment of ABT-263 and YM-155 was shown to have a synergistic effect in vitro as well as in in vivo patient-derived xenograft models. Our data suggest that combined inhibition of BCL-XL and MCL1 provides a promising treatment strategy for this genomically defined colorectal cancer subgroup. Mol Cancer Ther; 16(10); 2178-90. ©2017 AACR.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína bcl-X/genética , Anciano , Compuestos de Anilina/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Humanos , Imidazoles/administración & dosificación , Ratones , Mutación , Naftoquinonas/administración & dosificación , Sulfonamidas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Exp Mol Med ; 49(4): e317, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28408749

RESUMEN

Gliosarcoma (GS) is a rare variant (2%) of glioblastoma (GBM) that poses clinical genomic challenges because of its poor prognosis and limited genomic information. To gain a comprehensive view of the genomic alterations in GS and to understand the molecular etiology of GS, we applied whole-exome sequencing analyses for 28 GS cases (6 blood-matched fresh-frozen tissues for the discovery set, 22 formalin-fixed paraffin-embedded tissues for the validation set) and copy-number variation microarrays for 5 blood-matched fresh-frozen tissues. TP53 mutations were more prevalent in the GS cases (20/28, 70%) compared to the GBM cases (29/90, 32%), and the GS patients with TP53 mutations showed a significantly shorter survival (multivariate Cox analysis, hazard ratio=23.9, 95% confidence interval, 2.87-199.63, P=0.003). A pathway analysis showed recurrent alterations in MAPK signaling (EGFR, RASGRF2 and TP53), phosphatidylinositol/calcium signaling (CACNA1s, PLCs and ITPRs) and focal adhesion/tight junction (PTEN and PAK3) pathways. Genomic profiling of the matched recurrent GS cases detected the occurrence of TP53 mutations in two recurrent GS cases, which suggests that TP53 mutations play a role in treatment resistance. Functionally, we found that TP53 mutations are associated with the epithelial-mesenchymal transition (EMT) process of sarcomatous components of GS. We provide the first comprehensive genome-wide genetic alternation profiling of GS, which suggests novel prognostic subgroups in GS patients based on their TP53 mutation status and provides new insight in the pathogenesis and targeted treatment of GS.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Transición Epitelial-Mesenquimal/genética , Gliosarcoma/genética , Mutación , Proteína p53 Supresora de Tumor/genética , Neoplasias Encefálicas/patología , Señalización del Calcio , Línea Celular Tumoral , Femenino , Gliosarcoma/patología , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Uniones Estrechas/genética , Uniones Estrechas/metabolismo
17.
Mol Cells ; 39(2): 77-86, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26831452

RESUMEN

Cancer is a heterogeneous disease caused by diverse genomic alterations in oncogenes and tumor suppressor genes. Despite recent advances in high-throughput sequencing technologies and development of targeted therapies, novel cancer drug development is limited due to the high attrition rate from clinical studies. Patient-derived xenografts (PDX), which are established by the transfer of patient tumors into immunodeficient mice, serve as a platform for co-clinical trials by enabling the integration of clinical data, genomic profiles, and drug responsiveness data to determine precisely targeted therapies. PDX models retain many of the key characteristics of patients' tumors including histology, genomic signature, cellular heterogeneity, and drug responsiveness. These models can also be applied to the development of biomarkers for drug responsiveness and personalized drug selection. This review summarizes our current knowledge of this field, including methodologic aspects, applications in drug development, challenges and limitations, and utilization for precision cancer medicine.


Asunto(s)
Supervivencia de Injerto , Terapia Molecular Dirigida/métodos , Neoplasias Gástricas/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Antineoplásicos/farmacología , Biomarcadores Farmacológicos/metabolismo , Compuestos de Bifenilo/farmacología , Camptotecina/análogos & derivados , Camptotecina/farmacología , Sinergismo Farmacológico , Humanos , Irinotecán , Ratones , Ratones Desnudos , Ratones SCID , Nitrofenoles/farmacología , Piperazinas/farmacología , Medicina de Precisión , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Sulfonamidas/farmacología , Carga Tumoral/efectos de los fármacos
18.
Proc Natl Acad Sci U S A ; 112(40): 12492-7, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26401016

RESUMEN

Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide. Recent high-throughput analyses of genomic alterations revealed several driver genes and altered pathways in GC. However, therapeutic applications from genomic data are limited, largely as a result of the lack of druggable molecular targets and preclinical models for drug selection. To identify new therapeutic targets for GC, we performed array comparative genomic hybridization (aCGH) of DNA from 103 patients with GC for copy number alteration (CNA) analysis, and whole-exome sequencing from 55 GCs from the same patients for mutation profiling. Pathway analysis showed recurrent alterations in the Wnt signaling [APC, CTNNB1, and DLC1 (deleted in liver cancer 1)], ErbB signaling (ERBB2, PIK3CA, and KRAS), and p53 signaling/apoptosis [TP53 and BCL2L1 (BCL2-like 1)] pathways. In 18.4% of GC cases (19/103), amplification of the antiapoptotic gene BCL2L1 was observed, and subsequently a BCL2L1 inhibitor was shown to markedly decrease cell viability in BCL2L1-amplified cell lines and in similarly altered patient-derived GC xenografts, especially when combined with other chemotherapeutic agents. In 10.9% of cases (6/55), mutations in DLC1 were found and were also shown to confer a growth advantage for these cells via activation of Rho-ROCK signaling, rendering these cells more susceptible to a ROCK inhibitor. Taken together, our study implicates BCL2L1 and DLC1 as potential druggable targets for specific subsets of GC cases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteínas Activadoras de GTPasa/genética , Estudio de Asociación del Genoma Completo/métodos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Proteínas Supresoras de Tumor/genética , Proteína bcl-X/genética , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Supervivencia Celular/genética , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Exoma/genética , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Mutación , Interferencia de ARN , Análisis de Secuencia de ADN/métodos , Neoplasias Gástricas/patología , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína bcl-X/metabolismo
19.
Biochem Biophys Res Commun ; 420(1): 130-5, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22405767

RESUMEN

Methionine sulfoxide reductase B3A (MsrB3A), which catalyzes the stereospecific reduction of methionine-R-sulfoxide to methionine, is localized to the endoplasmic reticulum (ER). Here, we report a critical role of the ER-targeted MsrB3 in protection against ER stress in Drosophila and in mammalian cells. Flies overexpressing human MsrB3A exhibited significantly increased resistance to ER stress induced by dithiothreitol. These flies also showed slightly enhanced resistance to tunicamycin-induced ER stress. In addition, overexpression of MsrB3A in mammalian cells increased resistance to dithiothreitol- and thapsigargin-induced ER stresses. However, MsrB3A overexpression had no effect on the resistance to tunicamycin-induced ER stress. Knockdown of MsrB3A in mammalian cells led to a significant decrease in the resistance to thapsigargin-induced ER stress, but had no effects on the resistance to either dithiothreitol- or tunicamycin-induced ER stress. Collectively, our data provide evidence that the ER-type of MsrB3 plays an important role in protection against ER stress, suggesting that MsrB3 may be involved in the regulation of ER homeostasis.


Asunto(s)
Drosophila melanogaster/fisiología , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/enzimología , Metionina Sulfóxido Reductasas/fisiología , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Animales , Animales Modificados Genéticamente , Línea Celular , Regulación hacia Abajo , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Metionina Sulfóxido Reductasas/genética , Estrés Oxidativo/efectos de los fármacos , Tapsigargina/farmacología
20.
Biochem Biophys Res Commun ; 419(1): 20-6, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22310715

RESUMEN

Methionine sulfoxide reductase B (MsrB) is an enzyme that repairs oxidatively damaged proteins by specifically reducing methionine-R-sulfoxide back to methionine. Three MsrBs, localized in different cellular compartments, are expressed in mammals. However, the physiological roles of each MsrB with regard to its location remain poorly understood. Here, we expressed endoplasmic reticulum (ER)-targeted human MsrB3A (hMsrB3A) in Drosophila and examined its effects on various phenotypes. In two independent transgenic lines, both ubiquitous and neuronal expression of hMsrB3A rendered flies resistant to oxidative stress. Interestingly, these flies also showed significantly enhanced cold and heat tolerance. More strikingly, expression of hMsrB3A in the whole body and nervous system extended the lifespan of fruit flies at 29 °C by 43-50% and 12-37%, respectively, suggesting that the targeted expression of MsrB in the ER regulates Drosophila lifespan. A significant increase in lifespan was also observed at 25 °C only when hMsrB3A was expressed in neurons. Additionally, hMsrB3A overexpression significantly delayed the age-related decline in locomotor activity and fecundity. Taken together, our data provide evidence that the ER type of MsrB, MsrB3A, plays an important role in protection mechanisms against oxidative, cold and heat stresses and, moreover, in the regulation of fruit fly aging.


Asunto(s)
Envejecimiento/metabolismo , Respuesta al Choque por Frío , Drosophila melanogaster/fisiología , Retículo Endoplásmico/enzimología , Respuesta al Choque Térmico , Metionina Sulfóxido Reductasas/metabolismo , Estrés Oxidativo , Animales , Drosophila melanogaster/enzimología , Humanos , Metionina Sulfóxido Reductasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA