Your browser doesn't support javascript.
loading
Tumor Cell-Intrinsic c-Myb Upregulation Stimulates Antitumor Immunity in a Murine Colorectal Cancer Model.
van Gogh, Merel; Glaus Garzon, Jesus F; Sahin, Dilara; Knopfova, Lucia; Benes, Petr; Boyman, Onur; Jurisica, Igor; Borsig, Lubor.
Afiliación
  • van Gogh M; Institute of Physiology, University of Zurich, Zurich, Switzerland.
  • Glaus Garzon JF; Institute of Physiology, University of Zurich, Zurich, Switzerland.
  • Sahin D; Department of Immunology, University Hospital Zurich, Zurich, Switzerland.
  • Knopfova L; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
  • Benes P; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
  • Boyman O; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
  • Jurisica I; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
  • Borsig L; Department of Immunology, University Hospital Zurich, Zurich, Switzerland.
Cancer Immunol Res ; 11(10): 1432-1444, 2023 Oct 04.
Article en En | MEDLINE | ID: mdl-37478172
ABSTRACT
The transcription factor c-Myb is overexpressed in many different types of solid tumors, including colorectal cancer. However, its exact role in tumorigenesis is unclear. In this study, we show that tumor-intrinsic c-Myb expression in mouse models of colon cancer and melanoma suppresses tumor growth. Although no differences in proliferation, apoptosis, and angiogenesis of tumors were evident in tumors with distinct levels of c-Myb expression, we observed changes in intratumoral immune cell infiltrates. MC38 tumors with upregulated c-Myb expression showed increased numbers of CD103+ dendritic cells and eosinophils, but decreased tumor-associated macrophages (TAM). Concomitantly, an increase in the number of activated cytotoxic CD8+ T cells upon c-Myb upregulation was observed, which correlated with a pro-inflammatory tumor microenvironment and increased numbers of M1 polarized TAMs. Mechanistically, c-Myb upregulation in immunogenic MC38 colon cancer cells resulted in enhanced expression of immunomodulatory genes, including those encoding ß2-microglobulin and IFNß, and decreased expression of the gene encoding the chemokine receptor CCR2. The increased numbers of activated cytotoxic CD8+ T cells contributed to tumor growth attenuation. In poorly immunogenic CT26, LLC, and B16-BL6 tumor cells, c-Myb upregulation did not affect the immunomodulatory gene expression. Despite this, c-Myb upregulation led to reduced B16-BL6 tumor growth but it did not affect tumor growth of CT26 and LLC tumors. Altogether, we postulate that c-Myb functions as a tumor suppressor in a tumor cell-type specific manner and modulates antitumor immunity.

Texto completo: 1 Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Cancer Immunol Res Año: 2023 Tipo del documento: Article País de afiliación: Suiza

Texto completo: 1 Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Cancer Immunol Res Año: 2023 Tipo del documento: Article País de afiliación: Suiza