Your browser doesn't support javascript.
loading
Non-cell-autonomous suppression of tumor growth by RECK in immunocompetent mice.
Matsuzaki, Tomoko; Inoue, Joe; Minato, Nagahiro; Noda, Makoto.
Afiliação
  • Matsuzaki T; Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
  • Inoue J; Institute of Laboratory Animals, Kyoto University Graduate School of Medicine, Kyoto, Japan.
  • Minato N; Departments of Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
  • Noda M; Departments of Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
J Cell Physiol ; : e31396, 2024 Aug 05.
Article em En | MEDLINE | ID: mdl-39104026
ABSTRACT
RECK is a candidate tumor suppressor gene isolated as a gene that induces flat reversion in a cell line transformed by the KRAS oncogene. Since RECK knockout mice die in utero, they are not suitable for studying the effects of RECK on tumor formation. In this study, we found an increased incidence of spontaneous pulmonary adenomas in mice with reduced RECK expression (RECK-Hypo mice). To evaluate the effects of RECK expressed by either tumor cells or host cells on tumor growth, we established a tumorigenic cell line (MKER) from the kidney of a C57BL/6 mouse and performed syngeneic transplantation experiments. Our results indicate that when RECK expression is low in host cells, transplanted MKER cells grow faster and kill the animal more rapidly. Since RECK is required for the formation of proper fibrillin fibers that serve as a tissue reservoir for precursors of TGFß-family cytokines, we assessed the levels of TGFß1 in the peripheral blood. We found a significant increase in TGFß1 in RECK-Hypo mice compared to wild-type mice. We also found that the proportion of FOXP3-positive regulatory T (Treg) cells among splenocytes was higher in RECK-Hypo mice compared to the control mice. Furthermore, the number of FOXP3-positive cells in spontaneous hematopoietic neoplasms in the lungs as well as tumors that formed after MKER transplantation was significantly higher in RECK-Hypo mice compared to the control mice. These findings indicate that RECK-mediated tumor suppression involves a non-cell-autonomous mechanism and that possible roles of TGFß1 and Treg cells in such a mechanism warrant further study.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: J Cell Physiol Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Japão

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: J Cell Physiol Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Japão