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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Methods ; 12(3): 211-4, 3 p following 214, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25581801

RESUMO

We present SEEK (search-based exploration of expression compendia; http://seek.princeton.edu/), a query-based search engine for very large transcriptomic data collections, including thousands of human data sets from many different microarray and high-throughput sequencing platforms. SEEK uses a query-level cross-validation-based algorithm to automatically prioritize data sets relevant to the query and a robust search approach to identify genes, pathways and processes co-regulated with the query. SEEK provides multigene query searching with iterative metadata-based search refinement and extensive visualization-based analysis options.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Ferramenta de Busca , Transcriptoma , Algoritmos , Bases de Dados Genéticas , Ontologia Genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , RNA
2.
Physiol Genomics ; 46(6): 191-4, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24474443

RESUMO

The Systems Biology of Cell State Regulation Section is dedicated to considering how we can define a cellular state and how cells transition between states. One important decision that a cell makes is whether to cycle, that is, replicate DNA and generate daughter cells, or to exit the cell cycle in a reversible manner. The members of the Systems Biology of Cell State Regulation Editorial Board have an interest in the role of epigenetics and the commitment to a dividing or nondividing state. The ability of cells to transition between proliferating and nonproliferating states is essential for the proper formation of tissues. The ability to enter the cell cycle when needed is necessary for complex multicellular processes, such as healing injuries or mounting an immune response. Cells that fail to quiesce properly can contribute to the formation of tumors. In this perspective piece, we focus on research exploring the relationship between epigenetics and the cell cycle.


Assuntos
Ciclo Celular/genética , Cromatina/genética , Epigênese Genética , Transdução de Sinais/genética , Proliferação de Células , Cromatina/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Modelos Genéticos , Interferência de RNA
3.
Proc Natl Acad Sci U S A ; 108(34): 14240-5, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21831840

RESUMO

Recent observations suggest that p53 mutations are responsible not only for growth of primary tumors but also for their dissemination. However, mechanisms involved in p53-mediated control of cell motility and invasion remain poorly understood. By using the primary ovarian surface epithelium cell culture, we show that conditional inactivation of p53 or expression of its mutant forms results in overexpression of MET receptor tyrosine kinase, a crucial regulator of invasive growth. At the same time, cells acquire increased MET-dependent motility and invasion. Wild-type p53 negatively regulates MET expression by two mechanisms: (i) transactivation of MET-targeting miR-34, and (ii) inhibition of SP1 binding to MET promoter. Both mechanisms are not functional in p53 absence, but mutant p53 proteins retain partial MET promoter suppression. Accordingly, MET overexpression, cell motility, and invasion are particularly high in p53-null cells. These results identify MET as a critical effector of p53 and suggest that inhibition of MET may be an effective antimetastatic approach to treat cancers with p53 mutations. These results also show that the extent of advanced cancer traits, such as invasion, may be determined by alterations in individual components of p53/MET regulatory network.


Assuntos
Movimento Celular , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas c-met/genética , Receptores de Fatores de Crescimento/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Epitélio/metabolismo , Epitélio/patologia , Feminino , Inativação Gênica , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Mutantes/metabolismo , Invasividade Neoplásica , Ovário/metabolismo , Ovário/patologia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Fator de Transcrição Sp1/metabolismo
4.
Cell Rep ; 31(12): 107782, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32579914

RESUMO

Tumor cells are characterized by unlimited proliferation and perturbed differentiation. Using single-cell RNA sequencing, we demonstrate that tumor cells in medulloblastoma (MB) retain their capacity to differentiate in a similar way as their normal originating cells, cerebellar granule neuron precursors. Once they differentiate, MB cells permanently lose their proliferative capacity and tumorigenic potential. Differentiated MB cells highly express NeuroD1, a helix-loop-helix transcription factor, and forced expression of NeuroD1 promotes the differentiation of MB cells. The expression of NeuroD1 in bulk MB cells is repressed by trimethylation of histone 3 lysine-27 (H3K27me3). Inhibition of the histone lysine methyltransferase EZH2 prevents H3K27 trimethylation, resulting in increased NeuroD1 expression and enhanced differentiation in MB cells, which consequently reduces tumor growth. These studies reveal the mechanisms underlying MB cell differentiation and provide rationales to treat MB (potentially other malignancies) by stimulating tumor cell differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Neoplasias Cerebelares/patologia , Meduloblastoma/patologia , Proteínas do Tecido Nervoso/metabolismo , Animais , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteínas Hedgehog/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Receptor Patched-1/metabolismo , Transdução de Sinais , Análise de Célula Única
5.
Cancer Res ; 67(18): 8433-8, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17823410

RESUMO

MicroRNAs (miRNA) are a recently discovered class of noncoding RNAs that negatively regulate gene expression. Recent evidence indicates that miRNAs may play an important role in cancer. However, the mechanism of their deregulation in neoplastic transformation has only begun to be understood. To elucidate the role of tumor suppressor p53 in regulation of miRNAs, we have analyzed changes in miRNA microarray expression profile immediately after conditional inactivation of p53 in primary mouse ovarian surface epithelium cells. Among the most significantly affected miRNAs were miR-34b and miR-34c, which were down-regulated 12-fold according to quantitative reverse transcription-PCR analysis. Computational promoter analysis of the mir-34b/mir-34c locus identified the presence of evolutionarily conserved p53 binding sites approximately 3 kb upstream of the miRNA coding sequence. Consistent with evolutionary conservation, mir-34b/mir-34c were also down-regulated in p53-null human ovarian carcinoma cells. Furthermore, as expected from p53 binding to the mir-34b/c promoter, doxorubicin treatment of wild-type, but not p53-deficient, cells resulted in an increase of mir-34b/mir-34c expression. Importantly, miR-34b and miR-34c cooperate in suppressing proliferation and soft-agar colony formation of neoplastic epithelial ovarian cells, in agreement with the partially overlapping spectrum of their predicted targets. Taken together, these results show the existence of a novel mechanism by which p53 suppresses such critical components of neoplastic growth as cell proliferation and adhesion-independent colony formation.


Assuntos
MicroRNAs/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Animais , Sequência de Bases , Adesão Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Genes p53 , Humanos , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Ovário/citologia , Ovário/metabolismo , Ovário/fisiologia , Regiões Promotoras Genéticas , Transcrição Gênica , Transfecção , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Mol Biol Cell ; 29(15): 1825-1838, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29874120

RESUMO

Cell migration is a highly conserved process involving cytoskeletal reorganization and restructuring of the surrounding extracellular matrix. Although there are many studies describing mechanisms underlying cell motility, little has been reported about the contribution of alternative isoform use toward cell migration. Here, we investigated whether alternative isoform use can affect cell migration focusing on reversion-inducing-cysteine-rich protein with Kazal motifs (RECK), an established inhibitor of cell migration. We found that a shorter isoform of RECK is more highly expressed in proliferating fibroblasts, in TGF-ß-treated fibroblasts, and in tumors compared with differentiated tissue. Knockdown of this short RECK isoform reduces fibroblast migration through Matrigel. Thus, this short isoform of RECK generated by a combination of alternative splicing and alternative polyadenylation plays an opposing role to the canonical RECK isoform, as knockdown of canonical RECK results in faster cell migration through Matrigel. We show that the short RECK protein competes with matrix metalloprotease 9 (MMP9) for binding to the Kazal motifs of canonical RECK, thus liberating MMP9 from an inactivating interaction with canonical RECK. Our studies provide a new paradigm and a detailed mechanism for how alternative isoform use can regulate cell migration by producing two proteins with opposing effects from the same genetic locus.


Assuntos
Movimento Celular , Proteínas Ligadas por GPI/metabolismo , Motivos de Aminoácidos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Proteínas Ligadas por GPI/química , Técnicas de Silenciamento de Genes , Humanos , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Isoformas de Proteínas/metabolismo , Via Secretória/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia
7.
Genome Biol ; 19(1): 176, 2018 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-30360761

RESUMO

BACKGROUND: In response to a wound, fibroblasts are activated to migrate toward the wound, to proliferate and to contribute to the wound healing process. We hypothesize that changes in pre-mRNA processing occurring as fibroblasts enter the proliferative cell cycle are also important for promoting their migration. RESULTS: RNA sequencing of fibroblasts induced into quiescence by contact inhibition reveals downregulation of genes involved in mRNA processing, including splicing and cleavage and polyadenylation factors. These genes also show differential exon use, especially increased intron retention in quiescent fibroblasts compared to proliferating fibroblasts. Mapping the 3' ends of transcripts reveals that longer transcripts from distal polyadenylation sites are more prevalent in quiescent fibroblasts and are associated with increased expression and transcript stabilization based on genome-wide transcript decay analysis. Analysis of dermal excisional wounds in mice reveals that proliferating cells adjacent to wounds express higher levels of cleavage and polyadenylation factors than quiescent fibroblasts in unwounded skin. Quiescent fibroblasts contain reduced levels of the cleavage and polyadenylation factor CstF-64. CstF-64 knockdown recapitulates changes in isoform selection and gene expression associated with quiescence, and results in slower migration. CONCLUSIONS: Our findings support cleavage and polyadenylation factors as a link between cellular proliferation state and migration.


Assuntos
Ciclo Celular , Movimento Celular , Fibroblastos/fisiologia , Poli A/metabolismo , Poliadenilação , Pele/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Células Cultivadas , Fibroblastos/citologia , Humanos , Poli A/genética , Splicing de RNA , Pele/citologia , Fatores de Poliadenilação e Clivagem de mRNA/genética
8.
Hum Pathol ; 68: 79-86, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28882697

RESUMO

Some patients with eosinophilic esophagitis (EoE) do not respond to therapy. The clinicopathologic characteristics and gene expression profile at time of presentation could help predict response to therapy. Refractory EoE was defined as persistence of symptoms and biopsies with histologic features of EoE after 6 months of therapy with proton pump inhibitors and topical corticosteroids. Initial biopsies from refractory EoE patients (n=21), responder to therapy (n=8), patients who relapsed (n=6), and reflux controls (n=24) were studied. RNA was isolated from a subset of cases, and gene expression analysis of 285 genes involved in inflammation was performed using NanoString technology. There was no difference in the presenting symptoms among groups. The number of eosinophils/high-power field among nonresponders was higher (66±15) than responders (39±8; P<.0001) and similar to patients who relapsed (62±11). Six genes were expressed by at least 4-fold compared with reflux at a false discovery rate < 0.05, including overexpression of ALOX15, CCL26, FCER2, RTNLB, and RNASE2, and underexpression of DSG1. EoE patients refractory to therapy or who relapsed showed a trend toward higher ALOX15 expression compared with patients with good response to therapy (364.4- and 425-fold change, P=.097 and P=.07). RTNLB was significantly overexpressed in patients who were refractory to therapy versus those who responded favorably (10-fold versus 3-fold; P<.01). In conclusion, the number of eosinophils/high-power field in the initial biopsy inversely correlates with therapy response. Overexpression of RTNLB in refractory-to-therapy patients and overexpression of ALOX15 and CCL26 suggest that they are critical in the EoE pathogenesis.


Assuntos
Corticosteroides/uso terapêutico , Resistência a Medicamentos/genética , Esofagite Eosinofílica/genética , Esofagite Eosinofílica/patologia , Eosinófilos/patologia , Esôfago/patologia , Inibidores da Bomba de Prótons/uso terapêutico , Transcriptoma , Adolescente , Araquidonato 15-Lipoxigenase/genética , Biópsia , Quimiocina CCL26/genética , Criança , Pré-Escolar , Esofagite Eosinofílica/tratamento farmacológico , Eosinófilos/efeitos dos fármacos , Esôfago/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Marcadores Genéticos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Regulação para Cima
9.
Nat Med ; 23(4): 450-460, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28288111

RESUMO

Although blood-brain barrier (BBB) compromise is central to the etiology of diverse central nervous system (CNS) disorders, endothelial receptor proteins that control BBB function are poorly defined. The endothelial G-protein-coupled receptor (GPCR) Gpr124 has been reported to be required for normal forebrain angiogenesis and BBB function in mouse embryos, but the role of this receptor in adult animals is unknown. Here Gpr124 conditional knockout (CKO) in the endothelia of adult mice did not affect homeostatic BBB integrity, but resulted in BBB disruption and microvascular hemorrhage in mouse models of both ischemic stroke and glioblastoma, accompanied by reduced cerebrovascular canonical Wnt-ß-catenin signaling. Constitutive activation of Wnt-ß-catenin signaling fully corrected the BBB disruption and hemorrhage defects of Gpr124-CKO mice, with rescue of the endothelial gene tight junction, pericyte coverage and extracellular-matrix deficits. We thus identify Gpr124 as an endothelial GPCR specifically required for endothelial Wnt signaling and BBB integrity under pathological conditions in adult mice. This finding implicates Gpr124 as a potential therapeutic target for human CNS disorders characterized by BBB disruption.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Glioblastoma/genética , Infarto da Artéria Cerebral Média/genética , Hemorragias Intracranianas/genética , Receptores Acoplados a Proteínas G/genética , Junções Íntimas/metabolismo , Animais , Barreira Hematoencefálica/ultraestrutura , Modelos Animais de Doenças , Células Endoteliais/ultraestrutura , Matriz Extracelular/metabolismo , Citometria de Fluxo , Imunofluorescência , Glioblastoma/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Hemorragias Intracranianas/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Microvasos , Pericitos/ultraestrutura , Reação em Cadeia da Polimerase em Tempo Real , Junções Íntimas/ultraestrutura , Via de Sinalização Wnt
10.
Cell Stem Cell ; 21(1): 78-90.e6, 2017 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-28686870

RESUMO

Several cell populations have been reported to possess intestinal stem cell (ISC) activity during homeostasis and injury-induced regeneration. Here, we explored inter-relationships between putative mouse ISC populations by comparative RNA-sequencing (RNA-seq). The transcriptomes of multiple cycling ISC populations closely resembled Lgr5+ ISCs, the most well-defined ISC pool, but Bmi1-GFP+ cells were distinct and enriched for enteroendocrine (EE) markers, including Prox1. Prox1-GFP+ cells exhibited sustained clonogenic growth in vitro, and lineage-tracing of Prox1+ cells revealed long-lived clones during homeostasis and after radiation-induced injury in vivo. Single-cell mRNA-seq revealed two subsets of Prox1-GFP+ cells, one of which resembled mature EE cells while the other displayed low-level EE gene expression but co-expressed tuft cell markers, Lgr5 and Ascl2, reminiscent of label-retaining secretory progenitors. Our data suggest that the EE lineage, including mature EE cells, comprises a reservoir of homeostatic and injury-inducible ISCs, extending our understanding of cellular plasticity and stemness.


Assuntos
Antígenos de Diferenciação/metabolismo , Células Enteroendócrinas/metabolismo , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Jejuno/lesões , Jejuno/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos de Diferenciação/genética , Células Enteroendócrinas/patologia , Regulação da Expressão Gênica , Mucosa Intestinal/patologia , Jejuno/patologia , Camundongos , Camundongos Transgênicos , Células-Tronco/patologia
11.
Cell Rep ; 6(6): 1000-1007, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24630988

RESUMO

The miR-34 family was originally found to be a direct target of p53 and is a group of putative tumor suppressors. Surprisingly, mice lacking all mir-34 genes show no increase in cancer formation by 18 months of age, hence placing the physiological relevance of previous studies in doubt. Here, we report that mice with prostate epithelium-specific inactivation of mir-34 and p53 show expansion of the prostate stem cell compartment and develop early invasive adenocarcinomas and high-grade prostatic intraepithelial neoplasia, whereas no such lesions are observed after inactivation of either the mir-34 or p53 genes alone by 15 months of age. Consistently, combined deficiency of p53 and miR-34 leads to acceleration of MET-dependent growth, self-renewal, and motility of prostate stem/progenitor cells. Our study provides direct genetic evidence that mir-34 genes are bona fide tumor suppressors and identifies joint control of MET expression by p53 and miR-34 as a key component of prostate stem cell compartment regulation, aberrations in which may lead to cancer.


Assuntos
MicroRNAs/genética , Células-Tronco Neoplásicas/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteína Supressora de Tumor p53/genética , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Processos de Crescimento Celular/fisiologia , Masculino , Camundongos , MicroRNAs/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Proteína Supressora de Tumor p53/metabolismo
12.
Nat Med ; 20(7): 769-77, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24859528

RESUMO

The application of primary organoid cultures containing epithelial and mesenchymal elements to cancer modeling holds promise for combining the accurate multilineage differentiation and physiology of in vivo systems with the facile in vitro manipulation of transformed cell lines. Here we used a single air-liquid interface culture method without modification to engineer oncogenic mutations into primary epithelial and mesenchymal organoids from mouse colon, stomach and pancreas. Pancreatic and gastric organoids exhibited dysplasia as a result of expression of Kras carrying the G12D mutation (Kras(G12D)), p53 loss or both and readily generated adenocarcinoma after in vivo transplantation. In contrast, primary colon organoids required combinatorial Apc, p53, Kras(G12D) and Smad4 mutations for progressive transformation to invasive adenocarcinoma-like histology in vitro and tumorigenicity in vivo, recapitulating multi-hit models of colorectal cancer (CRC), as compared to the more promiscuous transformation of small intestinal organoids. Colon organoid culture functionally validated the microRNA miR-483 as a dominant driver oncogene at the IGF2 (insulin-like growth factor-2) 11p15.5 CRC amplicon, inducing dysplasia in vitro and tumorigenicity in vivo. These studies demonstrate the general utility of a highly tractable primary organoid system for cancer modeling and driver oncogene validation in diverse gastrointestinal tissues.


Assuntos
Transformação Celular Neoplásica/genética , Trato Gastrointestinal/patologia , Oncogenes , Animais , Neoplasias Gastrointestinais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos
14.
Clin Cancer Res ; 16(4): 1119-28, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20145172

RESUMO

PURPOSE: The miR-34 family is directly transactivated by tumor suppressor p53, which is frequently mutated in human epithelial ovarian cancer (EOC). We hypothesized that miR-34 expression would be decreased in EOC and that reconstituted miR-34 expression might reduce cell proliferation and invasion of EOC cells. EXPERIMENTAL DESIGNS: miR-34 expression was determined by quantitative reverse transcription-PCR and in situ hybridization in a panel of 83 human EOC samples. Functional characterization of miR-34 was accomplished by reconstitution of miR-34 expression in EOC cells with synthetic pre-miR molecules followed by determining changes in proliferation, apoptosis, and invasion. RESULTS: miR-34a expression is decreased in 100%, and miR-34b*/c in 72%, of EOC with p53 mutation, whereas miR-34a is also downregulated in 93% of tumors with wild-type p53. Furthermore, expression of miR-34b*/c is significantly reduced in stage IV tumors compared with stage III (P = 0.0171 and P = 0.0029, respectively). Additionally, we observed promoter methylation and copy number variations at mir-34. In situ hybridization showed that miR-34a expression is inversely correlated with MET immunohistochemical staining, consistent with translational inhibition by miR-34a. Finally, miR-34 reconstitution experiments in p53 mutant EOC cells resulted in reduced proliferation, motility, and invasion, the latter of which was dependent on MET expression. CONCLUSIONS: Our work suggests that miR-34 family plays an important role in EOC pathogenesis and reduced expression of miR-34b*/c may be particularly important for progression to the most advanced stages. Part of miR-34 effects on motility and invasion may be explained by regulation of MET, which is frequently overexpressed in EOC.


Assuntos
MicroRNAs/genética , Neoplasias Ovarianas/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proliferação de Células , Metilação de DNA , Regulação para Baixo , Feminino , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica
15.
Histol Histopathol ; 23(9): 1161-9, 2008 09.
Artigo em Inglês | MEDLINE | ID: mdl-18581287

RESUMO

Ovarian cancer remains a leading cause of morbidity and mortality, with little change in survival rates over the past 30 years. Research in the molecular biology underlying the disease demonstrates frequent mutation in the p53/Rb/p16 tumor suppressor pathways and activation of c-myc, K-ras and Akt oncogenic signaling. Recently, miRNAs have been demonstrated to play an important role in controlling proliferation, apoptosis and many other processes altered in the cancer state. In this review we discuss a number of recent publications that implicate a role for microRNAs in ovarian cancer and assess how this new field may improve our fundamental understanding of the disease and provide improved diagnostic and therapeutic approaches.


Assuntos
Adenocarcinoma/genética , MicroRNAs/fisiologia , Neoplasias Ovarianas/genética , RNA Neoplásico , Adenocarcinoma/patologia , Animais , Feminino , Humanos , Neoplasias Ovarianas/patologia
16.
Cancer Res ; 66(16): 7889-98, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16912162

RESUMO

Pathways mediated by p53 and Rb are frequently altered in aggressive human cancers, including prostate carcinoma. To test directly the roles of p53 and Rb in prostate carcinogenesis, we have conditionally inactivated these genes in the prostate epithelium of the mouse. Inactivation of either p53 or Rb leads to prostatic intraepithelial neoplasia developing from the luminal epithelium by 600 days of age. In contrast, inactivation of both genes results in rapidly developing (median survival, 226 days) carcinomas showing both luminal epithelial and neuroendocrine differentiation. The resulting neoplasms are highly metastatic, resistant to androgen depletion from the early stage of development, and marked with multiple gene expression signatures commonly found in human prostate carcinomas. Interestingly, gains at 4qC3 and 4qD2.2 and loss at 14qA2-qD2 have been consistently found by comparative genomic hybridization. These loci contain such human cancer-related genes as Nfib, L-myc, and Nkx3.1, respectively. Our studies show a critical role for p53 and Rb deficiency in prostate carcinogenesis and identify likely secondary genetic alterations. The new genetically defined model should be particularly valuable for providing new molecular insights into the pathogenesis of human prostate cancer.


Assuntos
Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteína do Retinoblastoma/deficiência , Proteína Supressora de Tumor p53/deficiência , Animais , Modelos Animais de Doenças , Células Epiteliais/patologia , Genes do Retinoblastoma , Genes p53 , Genótipo , Masculino , Camundongos , Camundongos Knockout , Metástase Neoplásica , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA