Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Nat Commun ; 14(1): 5118, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37612286

RESUMEN

To date, single-nucleotide polymorphisms (SNPs) have been the most intensively investigated class of polymorphisms in genome wide associations studies (GWAS), however, other classes such as insertion-deletion or multiple nucleotide length polymorphism (MNLPs) may also confer disease risk. Multiple reports have shown that the 5p15.33 prostate cancer risk region is a particularly strong expression quantitative trait locus (eQTL) for Iroquois Homeobox 4 (IRX4) transcripts. Here, we demonstrate using epigenome and genome editing that a biallelic (21 and 47 base pairs (bp)) MNLP is the causal variant regulating IRX4 transcript levels. In LNCaP prostate cancer cells (homozygous for the 21 bp short allele), a single copy knock-in of the 47 bp long allele potently alters the chromatin state, enabling de novo functional binding of the androgen receptor (AR) associated with increased chromatin accessibility, Histone 3 lysine 27 acetylation (H3K27ac), and ~3-fold upregulation of IRX4 expression. We further show that an MNLP is amongst the strongest candidate susceptibility variants at two additional prostate cancer risk loci. We estimated that at least 5% of prostate cancer risk loci could be explained by functional non-SNP causal variants, which may have broader implications for other cancers GWAS. More generally, our results underscore the importance of investigating other classes of inherited variation as causal mediators of human traits.


Asunto(s)
Neoplasias , Polimorfismo de Nucleótido Simple , Humanos , Masculino , Cromatina/genética , Acetilación , Alelos , Nucleótidos
3.
Nat Commun ; 14(1): 346, 2023 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-36681680

RESUMEN

While the mutational and transcriptional landscapes of renal cell carcinoma (RCC) are well-known, the epigenome is poorly understood. We characterize the epigenome of clear cell (ccRCC), papillary (pRCC), and chromophobe RCC (chRCC) by using ChIP-seq, ATAC-Seq, RNA-seq, and SNP arrays. We integrate 153 individual data sets from 42 patients and nominate 50 histology-specific master transcription factors (MTF) to define RCC histologic subtypes, including EPAS1 and ETS-1 in ccRCC, HNF1B in pRCC, and FOXI1 in chRCC. We confirm histology-specific MTFs via immunohistochemistry including a ccRCC-specific TF, BHLHE41. FOXI1 overexpression with knock-down of EPAS1 in the 786-O ccRCC cell line induces transcriptional upregulation of chRCC-specific genes, TFCP2L1, ATP6V0D2, KIT, and INSRR, implicating FOXI1 as a MTF for chRCC. Integrating RCC GWAS risk SNPs with H3K27ac ChIP-seq and ATAC-seq data reveals that risk-variants are significantly enriched in allelically-imbalanced peaks. This epigenomic atlas in primary human samples provides a resource for future investigation.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Epigenómica , Factores de Transcripción/genética , Oncogenes , Factores de Transcripción Forkhead/genética
4.
J Correct Health Care ; 28(6): 396-404, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36383107

RESUMEN

This study sought to evaluate the association of a solitary confinement ban with self-harm among adolescents in New York City's jail system. Data were extracted from medical records on 5,038 adolescent incarcerations from October 1, 2013, through July 12, 2016, and compared incarcerations before the ban (February 20, 2015) with those after the ban. Of the 2,503 adolescent incarcerations pre-ban, there were 171 self-harm gestures among 106 incarcerations (4.2% of incarcerations). Post-ban, there were 2,100 adolescent incarcerations and 105 self-harm gestures among 71 incarcerations (3.4% of incarcerations; p < .01). The post-ban group experienced significantly lower self-harm risk compared with the pre-ban (Crude incident rate ratio, 1.35 vs. 1.81; p < .05). In adjusted analysis, the hazard of self-harm associated with solitary confinement exposure was 1.51 times that of incarcerations with no solitary exposure (p < .05) . This signifies negative health outcomes of adolescent solitary confinement and the need for policy change consideration.


Asunto(s)
Cárceles Locales , Conducta Autodestructiva , Adolescente , Humanos , Ciudad de Nueva York/epidemiología , Conducta Autodestructiva/epidemiología
5.
Cancer Res ; 82(16): 2848-2859, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35731919

RESUMEN

African-American (AA) men are more likely to be diagnosed with and die from prostate cancer than European American (EA) men. Despite the central role of the androgen receptor (AR) transcription factor in prostate cancer, little is known about the contribution of epigenetics to observed racial disparities. We performed AR chromatin immunoprecipitation sequencing on primary prostate tumors from AA and EA men, finding that sites with greater AR binding intensity in AA relative to EA prostate cancer are enriched for lipid metabolism and immune response genes. Integration with transcriptomic and metabolomic data demonstrated coinciding upregulation of lipid metabolism gene expression and increased lipid levels in AA prostate cancer. In a metastatic prostate cancer cohort, upregulated lipid metabolism associated with poor prognosis. These findings offer the first insights into ancestry-specific differences in the prostate cancer AR cistrome. The data suggest a model whereby increased androgen signaling may contribute to higher levels of lipid metabolism, immune response, and cytokine signaling in AA prostate tumors. Given the association of upregulated lipogenesis with prostate cancer progression, our study provides a plausible biological explanation for the higher incidence and aggressiveness of prostate cancer observed in AA men. SIGNIFICANCE: With immunotherapies and inhibitors of metabolic enzymes in clinical development, the altered lipid metabolism and immune response in African-American men provides potential therapeutic opportunities to attenuate racial disparities in prostate cancer.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Negro o Afroamericano/genética , Humanos , Inmunidad , Metabolismo de los Lípidos/genética , Masculino , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Regulación hacia Arriba
6.
JCI Insight ; 7(10)2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35603787

RESUMEN

The androgen receptor (AR) is a master transcription factor that regulates prostate cancer (PC) development and progression. Inhibition of AR signaling by androgen deprivation is the first-line therapy with initial efficacy for advanced and recurrent PC. Paradoxically, supraphysiological levels of testosterone (SPT) also inhibit PC progression. However, as with any therapy, not all patients show a therapeutic benefit, and responses differ widely in magnitude and duration. In this study, we evaluated whether differences in the AR cistrome before treatment can distinguish between SPT-responding (R) and -nonresponding (NR) tumors. We provide the first preclinical evidence to our knowledge that SPT-R tumors exhibit a distinct AR cistrome when compared with SPT-NR tumors, indicating a differential biological role of the AR. We applied an integrated analysis of ChIP-Seq and RNA-Seq to the pretreatment tumors and identified an SPT-R signature that distinguishes R and NR tumors. Because transcriptomes of SPT-treated clinical specimens are not available, we interrogated available castration-resistant PC (CRPC) transcriptomes and showed that the SPT-R signature is associated with improved survival and has the potential to identify patients who would respond to SPT. These findings provide an opportunity to identify the subset of patients with CRPC who would benefit from SPT therapy.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Receptores Androgénicos , Antagonistas de Andrógenos , Humanos , Masculino , Recurrencia Local de Neoplasia , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/genética , Testosterona
7.
Nat Commun ; 13(1): 2559, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35562350

RESUMEN

c-MYC (MYC) is a major driver of prostate cancer tumorigenesis and progression. Although MYC is overexpressed in both early and metastatic disease and associated with poor survival, its impact on prostate transcriptional reprogramming remains elusive. We demonstrate that MYC overexpression significantly diminishes the androgen receptor (AR) transcriptional program (the set of genes directly targeted by the AR protein) in luminal prostate cells without altering AR expression. Analyses of clinical specimens reveal that concurrent low AR and high MYC transcriptional programs accelerate prostate cancer progression toward a metastatic, castration-resistant disease. Data integration of single-cell transcriptomics together with ChIP-seq uncover an increase in RNA polymerase II (Pol II) promoter-proximal pausing at AR-dependent genes following MYC overexpression without an accompanying deactivation of AR-bound enhancers. Altogether, our findings suggest that MYC overexpression antagonizes the canonical AR transcriptional program and contributes to prostate tumor initiation and progression by disrupting transcriptional pause release at AR-regulated genes.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Genes myc , Humanos , Masculino , Próstata/patología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-myc , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo
8.
Am J Hum Genet ; 108(11): 2071-2085, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34699744

RESUMEN

Genome-wide association studies (GWASs) of prostate cancer have identified >250 significant risk loci, but the causal variants and mechanisms for these loci remain largely unknown. Here, we sought to identify and characterize risk-harboring regulatory elements by integrating epigenomes from primary prostate tumor and normal tissues of 27 individuals across the H3K27ac, H3K4me3, and H3K4me2 histone marks and FOXA1 and HOXB13 transcription factors. We identified 7,371 peaks with significant allele specificity (allele-specific quantitative trait locus [asQTL] peaks). Showcasing their relevance to prostate cancer risk, H3K27ac T-asQTL peaks were the single annotation most enriched for prostate cancer GWAS heritability (40×), significantly higher than corresponding non-asQTL H3K27ac peaks (14×) or coding regions (14×). Surprisingly, fine-mapped GWAS risk variants were most significantly enriched for asQTL peaks observed in tumors, including asQTL peaks that were differentially imbalanced with respect to tumor-normal states. These data pinpointed putative causal regulatory elements at 20 GWAS loci, of which 11 were detected only in the tumor samples. More broadly, tumor-specific asQTLs were enriched for expression QTLs in benign tissues as well as accessible regions found in stem cells, supporting a hypothesis where some germline variants become reactivated during or after transformation and can be captured by epigenomic profiling of the tumor. Our study demonstrates the power of allele specificity in chromatin signals to uncover GWAS mechanisms, highlights the relevance of tumor-specific regulation in the context of cancer risk, and prioritizes multiple loci for experimental follow-up.


Asunto(s)
Alelos , Epigénesis Genética , Predisposición Genética a la Enfermedad , Próstata/metabolismo , Neoplasias de la Próstata/genética , Elementos de Facilitación Genéticos , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Sitios de Carácter Cuantitativo
9.
Nat Commun ; 12(1): 1979, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785741

RESUMEN

Lineage plasticity, the ability of a cell to alter its identity, is an increasingly common mechanism of adaptive resistance to targeted therapy in cancer. An archetypal example is the development of neuroendocrine prostate cancer (NEPC) after treatment of prostate adenocarcinoma (PRAD) with inhibitors of androgen signaling. NEPC is an aggressive variant of prostate cancer that aberrantly expresses genes characteristic of neuroendocrine (NE) tissues and no longer depends on androgens. Here, we investigate the epigenomic basis of this resistance mechanism by profiling histone modifications in NEPC and PRAD patient-derived xenografts (PDXs) using chromatin immunoprecipitation and sequencing (ChIP-seq). We identify a vast network of cis-regulatory elements (N~15,000) that are recurrently activated in NEPC. The FOXA1 transcription factor (TF), which pioneers androgen receptor (AR) chromatin binding in the prostate epithelium, is reprogrammed to NE-specific regulatory elements in NEPC. Despite loss of dependence upon AR, NEPC maintains FOXA1 expression and requires FOXA1 for proliferation and expression of NE lineage-defining genes. Ectopic expression of the NE lineage TFs ASCL1 and NKX2-1 in PRAD cells reprograms FOXA1 to bind to NE regulatory elements and induces enhancer activity as evidenced by histone modifications at these sites. Our data establish the importance of FOXA1 in NEPC and provide a principled approach to identifying cancer dependencies through epigenomic profiling.


Asunto(s)
Adenocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Tumores Neuroendocrinos/genética , Neoplasias de la Próstata/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/terapia , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Epigenómica/métodos , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Masculino , Mutación , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/terapia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Interferencia de ARN , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo
10.
Nat Genet ; 52(8): 790-799, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32690948

RESUMEN

Epigenetic processes govern prostate cancer (PCa) biology, as evidenced by the dependency of PCa cells on the androgen receptor (AR), a prostate master transcription factor. We generated 268 epigenomic datasets spanning two state transitions-from normal prostate epithelium to localized PCa to metastases-in specimens derived from human tissue. We discovered that reprogrammed AR sites in metastatic PCa are not created de novo; rather, they are prepopulated by the transcription factors FOXA1 and HOXB13 in normal prostate epithelium. Reprogrammed regulatory elements commissioned in metastatic disease hijack latent developmental programs, accessing sites that are implicated in prostate organogenesis. Analysis of reactivated regulatory elements enabled the identification and functional validation of previously unknown metastasis-specific enhancers at HOXB13, FOXA1 and NKX3-1. Finally, we observed that prostate lineage-specific regulatory elements were strongly associated with PCa risk heritability and somatic mutation density. Examining prostate biology through an epigenomic lens is fundamental for understanding the mechanisms underlying tumor progression.


Asunto(s)
Neoplasias de la Próstata/genética , Línea Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Epigenómica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , Masculino , Próstata/patología , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética
11.
Psychiatr Serv ; 71(6): 547-554, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32041509

RESUMEN

OBJECTIVE: New York City's (NYC) Correctional Health Services has introduced specialized treatment units for patients with serious mental illness in the NYC jail system. With multidisciplinary mental health staffing and a coordinated approach with NYC's Department of Correction, these units expand therapeutic options for patients vulnerable to clinical instability and physical harm, including those at high risk of psychiatric medication nonadherence and those returning to jail from psychiatric hospitalization. This study evaluated the extent to which these units improve clinical outcomes for this population. METHODS: This retrospective, observational cohort study included adult males with serious mental illness with a length of jail stay of ≥14 days between January 1, 2016, and March 31, 2018. Patients on treatment units were matched with patients of similar characteristics (control group) by using propensity score matching (N=302 pairs). Rates of self-injury, injury due to violence, and psychiatric medication adherence were analyzed. RESULTS: Most patients on treatment units had diagnoses of schizophrenia spectrum and other psychotic disorders (81%), and most had a violent felony as their most severe charge (68%). Compared with patients in a control group, those on the treatment units had lower rates per 100 person-days of injury due to violence at 30 and 60 days (0.04, 95% confidence interval [CI]=0.02-0.07, and 0.03, 95% CI=0.02-0.06, respectively) and higher mean medication adherence (77% versus 55%, p<0.001). They also experienced lower rates of self-injury, although the difference was not statistically significant. CONCLUSIONS: Initial outcomes indicate substantial benefits to patients, demonstrating the value of a rehabilitative approach to psychiatric care in jail.


Asunto(s)
Cárceles Locales/organización & administración , Cumplimiento de la Medicación/estadística & datos numéricos , Servicios de Salud Mental/organización & administración , Trastornos Psicóticos/terapia , Conducta Autodestructiva/prevención & control , Adolescente , Adulto , Unidades Hospitalarias , Humanos , Masculino , Persona de Mediana Edad , Ciudad de Nueva York , Estudios Retrospectivos , Adulto Joven
12.
Am J Hum Genet ; 106(2): 170-187, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32004450

RESUMEN

Although quantitative trait locus (QTL) associations have been identified for many molecular traits such as gene expression, it remains challenging to distinguish the causal nucleotide from nearby variants. In addition to traditional QTLs by association, allele-specific (AS) QTLs are a powerful measure of cis-regulation that are concordant with traditional QTLs but typically less susceptible to technical/environmental noise. However, existing methods for estimating causal variant probabilities (i.e., fine mapping) cannot produce valid estimates from asQTL signals due to complexities in linkage disequilibrium (LD). We introduce PLASMA (Population Allele-Specific Mapping), a fine-mapping method that integrates QTL and asQTL information to improve accuracy. In simulations, PLASMA accurately prioritizes causal variants over a wide range of genetic architectures. Applied to RNA-seq data from 524 kidney tumor samples, PLASMA achieves a greater power at 50 samples than conventional QTL-based fine mapping at 500 samples, with more than 17% of loci fine mapped to within five causal variants, compared to 2% by QTL-based fine mapping, and a 6.9-fold overall reduction in median credible set size compared to QTL-based fine mapping when applied to H3K27AC ChIP-seq from just 28 prostate tumor/normal samples. Variants in the PLASMA credible sets for RNA-seq and ChIP-seq were enriched for open chromatin and chromatin looping, respectively, at a comparable or greater degree than credible variants from existing methods while containing far fewer markers. Our results demonstrate how integrating AS activity can substantially improve the detection of causal variants from existing molecular data.


Asunto(s)
Algoritmos , Desequilibrio Alélico , Biomarcadores de Tumor/genética , Mapeo Cromosómico/métodos , Neoplasias Renales/genética , Neoplasias de la Próstata/genética , Sitios de Carácter Cuantitativo , Simulación por Computador , Interpretación Estadística de Datos , Humanos , Neoplasias Renales/patología , Desequilibrio de Ligamiento , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/patología
13.
Nat Med ; 25(10): 1615-1626, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31591588

RESUMEN

Oncogenesis is driven by germline, environmental and stochastic factors. It is unknown how these interact to produce the molecular phenotypes of tumors. We therefore quantified the influence of germline polymorphisms on the somatic epigenome of 589 localized prostate tumors. Predisposition risk loci influence a tumor's epigenome, uncovering a mechanism for cancer susceptibility. We identified and validated 1,178 loci associated with altered methylation in tumoral but not nonmalignant tissue. These tumor methylation quantitative trait loci influence chromatin structure, as well as RNA and protein abundance. One prominent tumor methylation quantitative trait locus is associated with AKT1 expression and is predictive of relapse after definitive local therapy in both discovery and validation cohorts. These data reveal intricate crosstalk between the germ line and the epigenome of primary tumors, which may help identify germline biomarkers of aggressive disease to aid patient triage and optimize the use of more invasive or expensive diagnostic assays.


Asunto(s)
Metilación de ADN/genética , Epigenoma/genética , Mutación de Línea Germinal/genética , Neoplasias de la Próstata/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Genoma Humano/genética , Humanos , Masculino , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/genética , Sitios de Carácter Cuantitativo/genética
14.
Clin Cancer Res ; 25(14): 4480-4492, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30996073

RESUMEN

PURPOSE: Defects in genes in the DNA repair pathways significantly contribute to prostate cancer progression. We hypothesize that overexpression of DNA repair genes may also drive poorer outcomes in prostate cancer. The ribonucleotide reductase small subunit M2 (RRM2) is essential for DNA synthesis and DNA repair by producing dNTPs. It is frequently overexpressed in cancers, but very little is known about its function in prostate cancer. EXPERIMENTAL DESIGN: The oncogenic activity of RRM2 in prostate cancer cells was assessed by inhibiting or overexpressing RRM2. The molecular mechanisms of RRM2 function were determined. The clinical significance of RRM2 overexpression was evaluated in 11 prostate cancer clinical cohorts. The efficacy of an RRM2 inhibitor (COH29) was assessed in vitro and in vivo. Finally, the mechanism underlying the transcriptional activation of RRM2 in prostate cancer tissue and cells was determined. RESULTS: Knockdown of RRM2 inhibited its oncogenic function, whereas overexpression of RRM2 promoted epithelial mesenchymal transition in prostate cancer cells. The prognostic value of RRM2 RNA levels in prostate cancer was confirmed in 11 clinical cohorts. Integrating the transcriptomic and phosphoproteomic changes induced by RRM2 unraveled multiple oncogenic pathways downstream of RRM2. Targeting RRM2 with COH29 showed excellent efficacy. Thirteen putative RRM2-targeting transcription factors were bioinformatically identified, and FOXM1 was validated to transcriptionally activate RRM2 in prostate cancer. CONCLUSIONS: We propose that increased expression of RRM2 is a mechanism driving poor patient outcomes in prostate cancer and that its inhibition may be of significant therapeutic value.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Reparación del ADN , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , Ribonucleósido Difosfato Reductasa/metabolismo , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Células Cultivadas , Estudios de Cohortes , Proteína Forkhead Box M1/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Ribonucleósido Difosfato Reductasa/genética , Tasa de Supervivencia , Activación Transcripcional , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Correct Health Care ; 24(4): 365-370, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30092737

RESUMEN

Individuals with serious mental illness detained in jail may require frequent psychiatric hospitalization due to the destabilizing nature of the jail environment. This study examined the impact of a pilot treatment program involving continuity of patient care across jail and hospital settings aimed at reducing hospitalizations and negative health outcomes for a population of high-risk, incarcerated individuals with mental illness. This study examined rate ratios of psychiatric hospitalizations, injuries, and suicide watches, comparing 15 patients in the treatment program to themselves pretreatment and to a control group of 15 frequently rehospitalized patients. Patients in treatment experienced significant decreases in overall hospitalizations ( p < .001), 15-day rehospitalizations ( p < .002), and suicide watches in jail ( p < .02), compared to themselves pretreatment. A boundary-spanning treatment program lowered hospitalization rates and need for suicide watch for a small, yet clinically complicated and challenging group of patients.


Asunto(s)
Continuidad de la Atención al Paciente/organización & administración , Administración Hospitalaria , Trastornos Mentales/epidemiología , Trastornos Mentales/terapia , Prisiones/organización & administración , Adulto , Humanos , Masculino , Readmisión del Paciente/estadística & datos numéricos , Proyectos Piloto , Conducta Autodestructiva/epidemiología , Suicidio/estadística & datos numéricos
16.
Cell ; 174(2): 422-432.e13, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29909987

RESUMEN

Increased androgen receptor (AR) activity drives therapeutic resistance in advanced prostate cancer. The most common resistance mechanism is amplification of this locus presumably targeting the AR gene. Here, we identify and characterize a somatically acquired AR enhancer located 650 kb centromeric to the AR. Systematic perturbation of this enhancer using genome editing decreased proliferation by suppressing AR levels. Insertion of an additional copy of this region sufficed to increase proliferation under low androgen conditions and to decrease sensitivity to enzalutamide. Epigenetic data generated in localized prostate tumors and benign specimens support the notion that this region is a developmental enhancer. Collectively, these observations underscore the importance of epigenomic profiling in primary specimens and the value of deploying genome editing to functionally characterize noncoding elements. More broadly, this work identifies a therapeutic vulnerability for targeting the AR and emphasizes the importance of regulatory elements as highly recurrent oncogenic drivers.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo , Acetilación , Adulto , Anciano , Antineoplásicos/farmacología , Benzamidas , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Metilación de ADN , Edición Génica , Histonas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA