Your browser doesn't support javascript.
loading
Quantitatively controlling expression of miR-17~92 determines colon tumor progression in a mouse tumor model.
Jiang, Hong; Wang, Ping; Wang, Qilong; Wang, Baomei; Mu, Jingyao; Zhuang, Xiaoying; Zhang, Lifeng; Yan, Jun; Miller, Donald; Zhang, Huang-Ge.
Afiliação
  • Jiang H; Louisville Veterans Administration Medical Center, Louisville, Kentucky; James Brown Cancer Center, University of Louisville, Louisville, Kentucky; Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky; Department of General Surgery, First Affiliated Hospital of N
  • Wang P; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.
  • Wang Q; James Brown Cancer Center, University of Louisville, Louisville, Kentucky.
  • Wang B; James Brown Cancer Center, University of Louisville, Louisville, Kentucky.
  • Mu J; James Brown Cancer Center, University of Louisville, Louisville, Kentucky.
  • Zhuang X; James Brown Cancer Center, University of Louisville, Louisville, Kentucky.
  • Zhang L; James Brown Cancer Center, University of Louisville, Louisville, Kentucky.
  • Yan J; James Brown Cancer Center, University of Louisville, Louisville, Kentucky.
  • Miller D; James Brown Cancer Center, University of Louisville, Louisville, Kentucky.
  • Zhang HG; Louisville Veterans Administration Medical Center, Louisville, Kentucky; James Brown Cancer Center, University of Louisville, Louisville, Kentucky; Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky. Electronic address: h0zhan17@louisville.edu.
Am J Pathol ; 184(5): 1355-68, 2014 May.
Article em En | MEDLINE | ID: mdl-24681249
ABSTRACT
The miRNA cluster miR-17~92 targets mRNAs involved in distinct pathways that either promote or inhibit tumor progression. However, the cellular and molecular mechanisms underlying miR-17~92 cluster-mediated protumorigenic or anti-tumorigenic effects have not been studied. Herein, we determined that inhibition of colon cancer progression is dictated by quantitatively controlling expression of the miR-17~92 cluster. miR-19 in the context of the miR-17~92 cluster at medium levels promoted tumor metastasis through induction of Wnt/ß-catenin-mediated epithelial-mesenchymal transition by targeting to the tumor-suppressor gene, PTEN. However, higher levels of the miR-17~92 cluster switched from PTEN to oncogenes, including Ctnnb1 (ß-catenin) via miR-18a, which resulted in inhibition of tumor growth and metastasis. However, overexpression of Ctnnb1in tumor cells with high-level miR-17~92 did not lead to an increase in the levels of ß-catenin protein, suggesting that other factors regulated by higher levels of miR-17~92 might also contribute to inhibition of tumor growth and metastasis. Those unidentified factors may negatively regulate the production of ß-catenin protein. Collectively, the data presented in this study revealed that higher levels of miR-17~92 were a critical negative regulator for activation of the Wnt/ß-catenin pathway and could have a potential therapeutic application.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Regulação Neoplásica da Expressão Gênica / Neoplasias do Colo / Progressão da Doença / MicroRNAs Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2014 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Regulação Neoplásica da Expressão Gênica / Neoplasias do Colo / Progressão da Doença / MicroRNAs Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2014 Tipo de documento: Article