Your browser doesn't support javascript.
loading
Loss of oncogenic miR-155 in tumor cells promotes tumor growth by enhancing C/EBP-ß-mediated MDSC infiltration.
Kim, Sinae; Song, Jin Hoi; Kim, Seokho; Qu, Peng; Martin, Betty K; Sehareen, Waheed S; Haines, Diana C; Lin, Pengnian C; Sharan, Shyam K; Chang, Suhwan.
Afiliación
  • Kim S; Department of Biomedical Sciences, Department of Physiology, University of Ulsan School of Medicine, Seoul, South Korea.
  • Song JH; Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Taejeon, South Korea.
  • Kim S; Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Taejeon, South Korea.
  • Qu P; Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA.
  • Martin BK; Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA.
  • Sehareen WS; Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA.
  • Haines DC; Pathology Histotechnology Laboratory, Leidos Inc., Frederick National Laboratory for Cancer, Frederick, MD, USA.
  • Lin PC; Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA.
  • Sharan SK; Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA.
  • Chang S; Department of Biomedical Sciences, Department of Physiology, University of Ulsan School of Medicine, Seoul, South Korea.
Oncotarget ; 7(10): 11094-112, 2016 Mar 08.
Article en En | MEDLINE | ID: mdl-26848978
ABSTRACT
The oncogenic role of microRNA-155 (miR-155) in leukemia is well established but its role in other cancers, especially breast cancer, is gradually emerging. In this study we examined the effect of mir-155 loss in a well-characterized spontaneous breast cancer mouse model where Brca1 and Trp53 are deleted by K14-Cre. miR-155 is known to be up-regulated in BRCA1-deficient tumors. Surprisingly, complete loss of miR-155 (miR-155ko/ko) did not alter the tumor free survival of the mutant mice. However, we found increased infiltration of myeloid derived suppressor cells (MDSCs) in miR-155 deficient tumors. In addition, cytokine/chemokine array analysis revealed altered level of cytokines that are implicated in the recruitment of MDSCs. Mechanistically, we identified C/EBP-ß, a known miR-155 target, to regulate the expression of these cytokines in the miR-155-deficient cells. Furthermore, using an allograft model, we showed that inhibition of miR-155 in cancer cells suppressed in vivo growth, which was restored by the loss of miR-155 in the microenvironment. Taken together, we have uncovered a novel tumor suppressive function of miR-155 in the tumor microenvironment, which is also dependent on miR-155 expression in the tumor cells. Because of the oncogenic as well as tumor suppressive roles of miR-155, our findings warrant caution against a systemic inhibition of miR-155 for anticancer therapy.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: MicroARNs / Microambiente Tumoral / Células Supresoras de Origen Mieloide / Neoplasias Mamarias Experimentales Tipo de estudio: Prognostic_studies Límite: Animals Idioma: En Revista: Oncotarget Año: 2016 Tipo del documento: Article País de afiliación: Corea del Sur

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: MicroARNs / Microambiente Tumoral / Células Supresoras de Origen Mieloide / Neoplasias Mamarias Experimentales Tipo de estudio: Prognostic_studies Límite: Animals Idioma: En Revista: Oncotarget Año: 2016 Tipo del documento: Article País de afiliación: Corea del Sur