Your browser doesn't support javascript.
loading
Tumor Cell Autonomous RON Receptor Expression Promotes Prostate Cancer Growth Under Conditions of Androgen Deprivation.
Brown, Nicholas E; Paluch, Andrew M; Nashu, Madison A; Komurov, Kakajan; Waltz, Susan E.
Afiliação
  • Brown NE; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Paluch AM; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Nashu MA; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Komurov K; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA.
  • Waltz SE; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45267, USA. Electronic address: susan.waltz@uc.edu.
Neoplasia ; 20(9): 917-929, 2018 09.
Article em En | MEDLINE | ID: mdl-30121008
ABSTRACT
Current treatment strategies provide minimal results for patients with castration-resistant prostate cancer (CRPC). Attempts to target the androgen receptor have shown promise, but resistance ultimately develops, often due to androgen receptor reactivation. Understanding mechanisms of resistance, including androgen receptor reactivation, is crucial for development of more efficacious CRPC therapies. Here, we report that the RON receptor tyrosine kinase is highly expressed in the majority of human hormone-refractory prostate cancers. Further, we show that exogenous expression of RON in human and murine prostate cancer cells circumvents sensitivity to androgen deprivation and promotes prostate cancer cell growth in both in vivo and in vitro settings. Conversely, RON loss induces sensitivity of CRPC cells to androgen deprivation. Mechanistically, we demonstrate that RON overexpression leads to activation of multiple oncogenic transcription factors (namely, ß-catenin and NF-κB), which are sufficient to drive androgen receptor nuclear localization and activation of AR responsive genes under conditions of androgen deprivation and support castration-resistant growth. In total, this study demonstrates the functional significance of RON during prostate cancer progression and provides a strong rationale for targeting RON signaling in prostate cancer as a means to limit resistance to androgen deprivation therapy.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Regulação Neoplásica da Expressão Gênica / Receptores Proteína Tirosina Quinases / Androgênios Limite: Animals / Humans / Male Idioma: En Revista: Neoplasia Assunto da revista: NEOPLASIAS Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Regulação Neoplásica da Expressão Gênica / Receptores Proteína Tirosina Quinases / Androgênios Limite: Animals / Humans / Male Idioma: En Revista: Neoplasia Assunto da revista: NEOPLASIAS Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Estados Unidos