Your browser doesn't support javascript.
loading
Rapalogs can promote cancer cell stemness in vitro in a Galectin-1 and H-ras-dependent manner.
Posada, Itziar M D; Lectez, Benoit; Sharma, Mukund; Oetken-Lindholm, Christina; Yetukuri, Laxman; Zhou, Yong; Aittokallio, Tero; Abankwa, Daniel.
Afiliação
  • Posada IMD; Turku Center for Biotechnology, Åbo Akademi University, Tykistökatu 6B, Turku, Finland.
  • Lectez B; Turku Center for Biotechnology, Åbo Akademi University, Tykistökatu 6B, Turku, Finland.
  • Sharma M; Turku Center for Biotechnology, Åbo Akademi University, Tykistökatu 6B, Turku, Finland.
  • Oetken-Lindholm C; Turku Center for Biotechnology, Åbo Akademi University, Tykistökatu 6B, Turku, Finland.
  • Yetukuri L; Turku Center for Biotechnology, Åbo Akademi University, Tykistökatu 6B, Turku, Finland.
  • Zhou Y; Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland.
  • Aittokallio T; Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States of America.
  • Abankwa D; Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland.
Oncotarget ; 8(27): 44550-44566, 2017 Jul 04.
Article em En | MEDLINE | ID: mdl-28562352
ABSTRACT
Currently several combination treatments of mTor- and Ras-pathway inhibitors are being tested in cancer therapy. While multiple feedback loops render these central signaling pathways robust, they complicate drug targeting.Here, we describe a novel H-ras specific feedback, which leads to an inadvertent rapalog induced activation of tumorigenicity in Ras transformed cells. We find that rapalogs specifically increase nanoscale clustering (nanoclustering) of oncogenic H-ras but not K-ras on the plasma membrane. This increases H-ras signaling output, promotes mammosphere numbers in a H-ras-dependent manner and tumor growth in ovo. Surprisingly, also other FKBP12 binders, but not mTor-inhibitors, robustly decrease FKBP12 levels after prolonged (>2 days) exposure. This leads to an upregulation of the nanocluster scaffold galectin-1 (Gal-1), which is responsible for the rapamycin-induced increase in H-ras nanoclustering and signaling output. We provide evidence that Gal-1 promotes stemness features in tumorigenic cells. Therefore, it may be necessary to block inadvertent induction of stemness traits in H-ras transformed cells by specific Gal-1 inhibitors that abrogate its effect on H-ras nanocluster. On a more general level, our findings may add an important mechanistic explanation to the pleiotropic physiological effects that are observed with rapalogs.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Proteínas ras / Galectina 1 / Autorrenovação Celular / Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Proteínas ras / Galectina 1 / Autorrenovação Celular / Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article