Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
J Natl Cancer Inst ; 106(4): dju043, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24627270

RESUMO

BACKGROUND: The c-Myc oncoprotein is activated in the majority of colorectal cancers (CRCs), whereas the TGF-ß pathway is frequently affected by loss-of-function mutations, for example in SMAD2/3/4 genes. The canonical model places Myc downstream of inhibitory TGF-ß signaling. However, we previously demonstrated that Myc also inhibits TGF-ß signaling through the miR-17~92 microRNA cluster, raising the question about functional relationships between these two pathways. METHODS: We engineered a series of genetically complex murine and human CRC cell lines in which Myc and TGF-ß activities could be manipulated simultaneously. This was achieved through retroviral expression of the Myc-estrogen receptor fusion protein and through Smad4 short hairpin RNA knockdown. Cell lines thus modified were injected subcutaneously in immunocompromised mice, and the resultant tumors (n = 5-10 per treatment group) were analyzed for overall growth and neovascularization. Additionally, the distribution of MYC and TGF-ß pathway mutations was analyzed in previously profiled human CRC samples. RESULTS: In kras-mutated/trp53-deleted murine colonocytes, either Myc activation or TGF-ß inactivation increased tumor sizes and microvascular densities approximately 1.5- to 2.5-fold, chiefly through downregulation of thrombospondin-1 and related type I repeat-containing proteins. Combining Myc activation with TGF-ß inactivation did not further accelerate tumorigenesis. This redundancy and the negative effect of TGF-ß signaling on angiogenesis were also demonstrated using xenografts of human CRC cell lines. Furthermore, the analysis of the Cancer Genome Atlas data revealed that in CRC without microsatellite instability, overexpression of Myc and inactivation of Smads (including acquired mutations in SMAD2) are mutually exclusive, with odds ratio less than 0.1. CONCLUSIONS: In human CRC, gain-of-function alterations in Myc and loss-of-function alterations in TGF-ß exhibit a masking epistatic interaction and are functionally redundant.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias Colorretais/metabolismo , Epistasia Genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Xenoenxertos , Humanos , Imuno-Histoquímica , Camundongos , Mutação , Proteínas Proto-Oncogênicas c-myc/genética , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética
2.
Cancer Res ; 71(24): 7490-501, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22028325

RESUMO

Thrombospondin-1 (TSP-1) is an endogenous inhibitor of angiogenesis encoded by the THBS1 gene, whose promoter is activated by p53. In advanced colorectal cancers (CRC), its expression is sustained or even slightly increased despite frequent loss of p53. Here, we determined that in HCT116 CRC cells, p53 activates the THBS1 primary transcript, but fails to boost THBS1 mRNA or protein levels, implying posttranscriptional regulation by microRNAs (miRNA). In a global miRNA gain-of-function screen done in the Dicer-deficient HCT116 variant, several miRNAs negatively regulated THBS1 mRNA and protein levels, one of them being miR-194. Notably, in agreement with published data, p53 upregulated miR-194 expression in THBS1 retrovirus-transduced HCT116 cells, leading to decreased TSP-1 levels. This negative effect was mediated by a single miR-194 complementary site in the THBS1 3'-untranslated region, and its elimination resulted in TSP-1 reactivation, impaired angiogenesis in Matrigel plugs, and reduced growth of HCT116 xenografts. Conversely, transient overexpression of miR-194 in HCT116/THBS1 cells boosted Matrigel angiogenesis, and its stable overexpression in Ras-induced murine colon carcinomas increased microvascular densities and vessel sizes. Although the overall contribution of miR-194 to neoplastic growth is context dependent, p53-induced activation of this GI tract-specific miRNA during ischemia could promote angiogenesis and facilitate tissue repair.


Assuntos
Neoplasias do Colo/genética , MicroRNAs/genética , Neovascularização Patológica/genética , Trombospondina 1/genética , Proteína Supressora de Tumor p53/genética , Regiões 3' não Traduzidas/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Camundongos , MicroRNAs/metabolismo , Mutação , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombospondina 1/metabolismo , Transcrição Gênica , Transdução Genética , Transplante Heterólogo , Proteína Supressora de Tumor p53/metabolismo
3.
Cancer Res ; 70(20): 8233-46, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20940405

RESUMO

c-Myc stimulates angiogenesis in tumors through mechanisms that remain incompletely understood. Recent work indicates that c-Myc upregulates the miR-17∼92 microRNA cluster and downregulates the angiogenesis inhibitor thrombospondin-1, along with other members of the thrombospondin type 1 repeat superfamily. Here, we show that downregulation of the thrombospondin type 1 repeat protein clusterin in cells overexpressing c-Myc and miR-17∼92 promotes angiogenesis and tumor growth. However, clusterin downregulation by miR-17∼92 is indirect. It occurs as a result of reduced transforming growth factor-ß (TGFß) signaling caused by targeting of several regulatory components in this signaling pathway. Specifically, miR-17-5p and miR-20 reduce the expression of the type II TGFß receptor and miR-18 limits the expression of Smad4. Supporting these results, in human cancer cell lines, levels of the miR-17∼92 primary transcript MIR17HG negatively correlate with those of many TGFß-induced genes that are not direct targets of miR-17∼92 (e.g., clusterin and angiopoietin-like 4). Furthermore, enforced expression of miR-17∼92 in MIR17HG(low) cell lines (e.g., glioblastoma) results in impaired gene activation by TGFß. Together, our results define a pathway in which c-Myc activation of miR-17∼92 attenuates the TGFß signaling pathway to shut down clusterin expression, thereby stimulating angiogenesis and tumor cell growth.


Assuntos
Inibidores da Angiogênese/biossíntese , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-myc/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Inibidores da Angiogênese/antagonistas & inibidores , Animais , Sequência de Bases , Clusterina/genética , Neoplasias do Colo/genética , Regulação para Baixo , Genes Reporter , Humanos , Luciferases/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/fisiologia , Dados de Sequência Molecular , Mutação , Receptores de Fatores de Crescimento Transformadores beta/genética , Ribonuclease III/genética , Fator de Crescimento Transformador beta/genética , Regiões não Traduzidas
4.
J Virol ; 80(18): 9000-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16940512

RESUMO

Mouse mammary tumor virus (MMTV) induces breast cancer with almost 100% efficiency in susceptible strains through insertional activation of protooncogenes, such as members of the wnt and fibroblast growth factor (fgf) families. We previously showed that expression of the MMTV envelope protein (Env) in normal immortalized mammary epithelial cells grown in three-dimensional cultures caused their morphological transformation, and that this phenotype depended on an immunoreceptor tyrosine-based activation motif (ITAM) present in Env and signaling through the Syk tyrosine kinase (E. Katz, M. H. Lareef, J. C. Rassa, S. M. Grande, L. B. King, J. Russo, S. R. Ross, and J. G. Monroe, J. Exp. Med. 201:431-439, 2005). Here, we examined the role of the Env protein in virus-induced mammary tumorigenesis in vivo. Similar to the effect seen in vitro, Env expression in the mammary glands of transgenic mice bearing either full-length wild-type provirus or only Env transgenes showed increased lobuloalveolar budding. Introduction of the ITAM mutation into the env of an infectious, replication-competent MMTV or into MMTV/murine leukemia virus pseudotypes had no effect on incorporation of Env into virus particles or on in vitro infectivity. Moreover, replication-competent MMTV bearing the ITAM mutation in Env infected lymphoid and mammary tissue at the same level as wild-type MMTV and was transmitted through milk. However, mammary tumor induction was greatly attenuated, and the pattern of oncogene activation was altered. Taken together, these studies indicate that the MMTV Env protein participates in mammary epithelial cell transformation in vivo and that this requires a functional ITAM in the envelope protein.


Assuntos
Neoplasias Mamárias Animais/virologia , Vírus do Tumor Mamário do Camundongo/metabolismo , Tirosina/química , Proteínas do Envelope Viral/química , Motivos de Aminoácidos , Animais , Feminino , Humanos , Linfócitos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA