Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell ; 161(2): 319-32, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25843629

RESUMO

Research over the past decade has suggested important roles for pseudogenes in physiology and disease. In vitro experiments demonstrated that pseudogenes contribute to cell transformation through several mechanisms. However, in vivo evidence for a causal role of pseudogenes in cancer development is lacking. Here, we report that mice engineered to overexpress either the full-length murine B-Raf pseudogene Braf-rs1 or its pseudo "CDS" or "3' UTR" develop an aggressive malignancy resembling human diffuse large B cell lymphoma. We show that Braf-rs1 and its human ortholog, BRAFP1, elicit their oncogenic activity, at least in part, as competitive endogenous RNAs (ceRNAs) that elevate BRAF expression and MAPK activation in vitro and in vivo. Notably, we find that transcriptional or genomic aberrations of BRAFP1 occur frequently in multiple human cancers, including B cell lymphomas. Our engineered mouse models demonstrate the oncogenic potential of pseudogenes and indicate that ceRNA-mediated microRNA sequestration may contribute to the development of cancer.


Assuntos
Linfoma Difuso de Grandes Células B/genética , Proteínas Proto-Oncogênicas B-raf/genética , Pseudogenes , RNA/metabolismo , Animais , Sequência de Bases , Humanos , Linfoma Difuso de Grandes Células B/metabolismo , Camundongos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas B-raf/metabolismo
2.
Hepatology ; 51(4): 1383-90, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20044804

RESUMO

UNLABELLED: The mitogen-inducible gene-6 (mig-6) is a multi-adaptor protein implicated in the regulation of the HER family of receptor tyrosine kinases. We have reported recently that mig-6 is a negative regulator of epidermal growth factor receptor (EGFR)-dependent skin morphogenesis and tumor formation in vivo. In the liver, ablation of mig-6 leads to an increase in EGFR protein levels, suggesting that mig-6 is a negative regulator of EGFR function. In line with this observation, primary hepatocytes isolated from mig-6 knockout and wild-type control mice display sustained mitogenic signaling in response to EGF. In order to explore the role of mig-6 in the liver in vivo, we analyzed liver regeneration in mig-6 knockout and wild-type control mice. Interestingly, mig-6 knockout mice display enhanced hepatocyte proliferation in the initial phases after partial hepatectomy. This phenotype correlates with activation of endogenous EGFR signaling, predominantly through the protein kinase B pathway. In addition, mig-6 is an endogenous inhibitor of EGFR signaling and EGF-induced tumor cell migration in human liver cancer cell lines. Moreover, mig-6 is down-regulated in human hepatocellular carcinoma and this correlates with increased EGFR expression. CONCLUSION: Our data implicate mig-6 as a regulator of EGFR activity in hepatocytes and as a suppressor of EGFR signaling in human liver cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Carcinoma Hepatocelular/patologia , Receptores ErbB/fisiologia , Hepatócitos/fisiologia , Neoplasias Hepáticas/patologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Supressoras de Tumor
3.
Nat Med ; 24(2): 165-175, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29309058

RESUMO

Multiple immune-cell types can infiltrate tumors and promote progression and metastasis through different mechanisms, including immunosuppression. How distinct genetic alterations in tumors affect the composition of the immune landscape is currently unclear. Here, we characterized the immune-cell composition of prostate cancers driven by the loss of the critical tumor suppressor gene Pten, either alone or in combination with the loss of Trp53, Zbtb7a or Pml. We observed a striking quantitative and qualitative heterogeneity that was directly dependent on the specific genetic events in the tumor and ranged from 'cold', noninflamed tumors to massively infiltrated landscapes-results with important therapeutic implications. Further, we showed these qualitative differences in transcriptomic analysis of human prostate cancer samples. These data suggest that patient stratification on the basis of integrated genotypic-immunophenotypic analyses may be necessary for successful clinical trials and tailored precision immunological therapies.


Assuntos
Proteínas de Ligação a DNA/genética , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/imunologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Animais , Linfócitos B/imunologia , Proteínas de Ligação a DNA/imunologia , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Heterogeneidade Genética , Genótipo , Humanos , Imunoterapia , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Knockout , Células Mieloides/imunologia , PTEN Fosfo-Hidrolase/imunologia , Proteína da Leucemia Promielocítica/genética , Proteína da Leucemia Promielocítica/imunologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Linfócitos T/imunologia , Fatores de Transcrição/imunologia , Transcriptoma/genética , Transcriptoma/imunologia , Proteína Supressora de Tumor p53/imunologia
4.
Mol Cancer Ther ; 16(7): 1279-1289, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28500233

RESUMO

The use of PARP inhibitors in combination with radiotherapy is a promising strategy to locally enhance DNA damage in tumors. Here we show that radiation-resistant cells and tumors derived from a Pten/Trp53-deficient mouse model of advanced prostate cancer are rendered radiation sensitive following treatment with NanoOlaparib, a lipid-based injectable nanoformulation of olaparib. This enhancement in radiosensitivity is accompanied by radiation dose-dependent changes in γ-H2AX expression and is specific to NanoOlaparib alone. In animals, twice-weekly intravenous administration of NanoOlaparib results in significant tumor growth inhibition, whereas previous studies of oral olaparib as monotherapy have shown no therapeutic efficacy. When NanoOlaparib is administered prior to radiation, a single dose of radiation is sufficient to triple the median mouse survival time compared to radiation only controls. Half of mice treated with NanoOlaparib + radiation achieved a complete response over the 13-week study duration. Using ferumoxytol as a surrogate nanoparticle, MRI studies revealed that NanoOlaparib enhances the intratumoral accumulation of systemically administered nanoparticles. NanoOlaparib-treated tumors showed up to 19-fold higher nanoparticle accumulation compared to untreated and radiation-only controls, suggesting that the in vivo efficacy of NanoOlaparib may be potentiated by its ability to enhance its own accumulation. Together, these data suggest that NanoOlaparib may be a promising new strategy for enhancing the radiosensitivity of radiation-resistant tumors lacking BRCA mutations, such as those with PTEN and TP53 deletions. Mol Cancer Ther; 16(7); 1279-89. ©2017 AACR.


Assuntos
PTEN Fosfo-Hidrolase/genética , Ftalazinas/administração & dosagem , Piperazinas/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Proteína Supressora de Tumor p53/genética , Animais , Proteína BRCA1/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Nanoestruturas/administração & dosagem , Nanoestruturas/química , PTEN Fosfo-Hidrolase/deficiência , Ftalazinas/química , Piperazinas/química , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Proteína Supressora de Tumor p53/deficiência , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Discov ; 5(5): 550-63, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25653093

RESUMO

UNLABELLED: The ETS family of transcription factors has been repeatedly implicated in tumorigenesis. In prostate cancer, ETS family members, such as ERG, ETV1, ETV4, and ETV5, are frequently overexpressed due to chromosomal translocations, but the molecular mechanisms by which they promote prostate tumorigenesis remain largely undefined. Here, we show that ETS family members, such as ERG and ETV1, directly repress the expression of the checkpoint kinase 1 (CHK1), a key DNA damage response cell-cycle regulator essential for the maintenance of genome integrity. Critically, we find that ERG expression correlates with CHK1 downregulation in human patients and demonstrate that Chk1 heterozygosity promotes the progression of high-grade prostatic intraepithelial neoplasia into prostatic invasive carcinoma in Pten(+) (/-) mice. Importantly, CHK1 downregulation sensitizes prostate tumor cells to etoposide but not to docetaxel treatment. Thus, we identify CHK1 as a key functional target of the ETS proto-oncogenic family with important therapeutic implications. SIGNIFICANCE: Genetic translocation and aberrant expression of ETS family members is a common event in different types of human tumors. Here, we show that through the transcriptional repression of CHK1, ETS factors may favor DNA damage accumulation and consequent genetic instability in proliferating cells. Importantly, our findings provide a rationale for testing DNA replication inhibitor agents in ETS-positive TP53-proficient tumors.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Dano ao DNA , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Sequência Conservada , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/genética , Etoposídeo/farmacologia , Regulação Neoplásica da Expressão Gênica , Genótipo , Humanos , Masculino , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ligação Proteica , Proteínas Quinases/genética , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Regulador Transcricional ERG , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Cancer Discov ; 4(8): 896-904, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24866151

RESUMO

UNLABELLED: Prostate cancer is the most prevalent cancer in males, and treatment options are limited for advanced forms of the disease. Loss of the PTEN and TP53 tumor suppressor genes is commonly observed in prostate cancer, whereas their compound loss is often observed in advanced prostate cancer. Here, we show that PARP inhibition triggers a p53-dependent cellular senescence in a PTEN-deficient setting in the prostate. Surprisingly, we also find that PARP-induced cellular senescence is morphed into an apoptotic response upon compound loss of PTEN and p53. We further show that superactivation of the prosurvival PI3K-AKT signaling pathway limits the efficacy of a PARP single-agent treatment, and that PARP and PI3K inhibitors effectively synergize to suppress tumorigenesis in human prostate cancer cell lines and in a Pten/Trp53-deficient mouse model of advanced prostate cancer. Our findings, therefore, identify a combinatorial treatment with PARP and PI3K inhibitors as an effective option for PTEN-deficient prostate cancer. SIGNIFICANCE: The paucity of therapeutic options in advanced prostate cancer displays an urgent need for the preclinical assessment of novel therapeutic strategies. We identified differential therapeutic vulnerabilities that emerge upon the loss of both PTEN and p53, and observed that combined inhibition of PARP and PI3K provides increased efficacy in hormone-insensitive advanced prostate cancer.


Assuntos
Elafina/genética , PTEN Fosfo-Hidrolase/genética , Poli(ADP-Ribose) Polimerases/genética , Neoplasias da Próstata/tratamento farmacológico , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Elafina/antagonistas & inibidores , Humanos , Masculino , Camundongos , Terapia de Alvo Molecular , Inibidores de Poli(ADP-Ribose) Polimerases , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
7.
Cell Rep ; 4(6): 1276-87, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24055062

RESUMO

Increasing evidence points to an important role for the ribosome in the regulation of biological processes and as a target for deregulation in disease. Here, we describe a SILAC (stable isotope labeling by amino acids in cell culture)-based mass spectrometry approach to probing mammalian riboproteomes. Using a panel of cell lines, as well as genetic and pharmacological perturbations, we obtained a comparative characterization of the cellular riboproteome. This analysis identified a set of riboproteome components, consisting of a diverse array of proteins with a strong enrichment for RNA-binding proteins. Importantly, this global analysis uncovers a high incidence of genetic alterations to riboproteome components in cancer, with a distinct bias toward genetic amplification. We further validated association with polyribosomes for several riboproteome components and demonstrate that enrichment at the riboproteome can depend on cell type, genetics, or cellular stimulus. Our results have important implications for the understanding of how ribosomes function and provide a platform for uncovering regulators of translation.


Assuntos
Neoplasias da Próstata/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Ribossomos/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Amplificação de Genes , Humanos , Masculino , Mamíferos , Espectrometria de Massas , Neoplasias da Próstata/genética , Proteoma/genética , Ribossomos/genética , Transcriptoma
8.
Cancer Res ; 70(2): 802-12, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20068154

RESUMO

In the growth factor receptor gene FGFR4 the presence of the common single nucleotide polymorphism Arg388 has been associated with progression of various types of cancer including breast cancer. However, a causative relationship is not readily assigned due to genetic heterogeneity in different patient cohorts. To address this issue, we compared the effects of this allele on malignant progression in the WAP-TGFalpha transgenic mouse model of breast cancer. A knock-in strain was generated to introduce an analogous Arg385 allele into the murine FGFR4 gene. Mouse embryonic fibroblasts derived from this strain displayed accelerated cell transformation, with transformed cells exhibiting greater motility and invasive behavior. In the in vivo context of TGFalpha-induced mammary carcinogenesis, tumor development and progression was significantly advanced in tumor mass, size, and onset of pulmonary metastases. Our findings definitively identify the FGFR4 Arg388 allele as a functional prognostic marker for breast cancer progression.


Assuntos
Neoplasias Mamárias Experimentais/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Células 3T3 , Alelos , Animais , Adesão Celular/genética , Movimento Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Genoma , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Leite/genética , Polimorfismo de Nucleotídeo Único , Fator de Crescimento Transformador alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA