Your browser doesn't support javascript.
loading
Proteomic and Phosphoproteomic Reprogramming in Epithelial Ovarian Cancer Metastasis.
Frederick, Mallory I; Hovey, Owen F J; Kakadia, Jenica H; Shepherd, Trevor G; Li, Shawn S C; Heinemann, Ilka U.
Afiliação
  • Frederick MI; Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
  • Hovey OFJ; Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
  • Kakadia JH; Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
  • Shepherd TG; Department of Obstetrics & Gynaecology, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada.
  • Li SSC; Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada. Electronic address: sli@uwo.ca.
  • Heinemann IU; Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada. Electronic address: ilka.heinemann@uwo.ca.
Mol Cell Proteomics ; 22(11): 100660, 2023 Nov.
Article em En | MEDLINE | ID: mdl-37820923
ABSTRACT
Epithelial ovarian cancer (EOC) is a high-risk cancer presenting with heterogeneous tumors. The high incidence of EOC metastasis from primary tumors to nearby tissues and organs is a major driver of EOC lethality. We used cellular models of spheroid formation and readherence to investigate cellular signaling dynamics in each step toward EOC metastasis. In our system, adherent cells model primary tumors, spheroid formation represents the initiation of metastatic spread, and readherent spheroid cells represent secondary tumors. Proteomic and phosphoproteomic analyses show that spheroid cells are hypoxic and show markers for cell cycle arrest. Aurora kinase B abundance and downstream substrate phosphorylation are significantly reduced in spheroids and readherent cells, explaining their cell cycle arrest phenotype. The proteome of readherent cells is most similar to spheroids, yet greater changes in the phosphoproteome show that spheroid cells stimulate Rho-associated kinase 1 (ROCK1)-mediated signaling, which controls cytoskeletal organization. In spheroids, we found significant phosphorylation of ROCK1 substrates that were reduced in both adherent and readherent cells. Application of the ROCK1-specific inhibitor Y-27632 to spheroids increased the rate of readherence and altered spheroid density. The data suggest ROCK1 inhibition increases EOC metastatic potential. We identified novel pathways controlled by Aurora kinase B and ROCK1 as major drivers of metastatic behavior in EOC cells. Our data show that phosphoproteomic reprogramming precedes proteomic changes that characterize spheroid readherence in EOC metastasis.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Ovarianas Limite: Female / Humans Idioma: En Revista: Mol Cell Proteomics Assunto da revista: BIOLOGIA MOLECULAR / BIOQUIMICA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Canadá

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Ovarianas Limite: Female / Humans Idioma: En Revista: Mol Cell Proteomics Assunto da revista: BIOLOGIA MOLECULAR / BIOQUIMICA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Canadá