Your browser doesn't support javascript.
loading
ROBO1, a tumor suppressor and critical molecular barrier for localized tumor cells to acquire invasive phenotype: study in African-American and Caucasian prostate cancer models.
Parray, Aijaz; Siddique, Hifzur R; Kuriger, Jacquelyn K; Mishra, Shrawan K; Rhim, Johng S; Nelson, Heather H; Aburatani, Hiroyuki; Konety, Badrinath R; Koochekpour, Shahriar; Saleem, Mohammad.
Afiliação
  • Parray A; Section of Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, MN.
Int J Cancer ; 135(11): 2493-506, 2014 Dec 01.
Article em En | MEDLINE | ID: mdl-24752651
ABSTRACT
High-risk populations exhibit early transformation of localized prostate cancer (CaP) disease to metastasis which results in the mortality of such patients. The paucity of knowledge about the molecular mechanism involved in acquiring of metastatic behavior by primary tumor cells and non-availability of reliable phenotype-discriminating biomarkers are stumbling blocks in the management of CaP disease. Here, we determine the role and translational relevance of ROBO1 (an organogenesis-associated gene) in human CaP. Employing CaP-progression models and prostatic tissues of Caucasian and African-American patients, we show that ROBO1 expression is localized to cell-membrane and significantly lost in primary and metastatic tumors. While Caucasians exhibited similar ROBO1 levels in primary and metastatic phenotype, a significant difference was observed between tumor phenotypes in African-Americans. Epigenetic assays identified promoter methylation of ROBO1 specific to African-American metastatic CaP cells. Using African-American CaP models for further studies, we show that ROBO1 negatively regulates motility and invasiveness of primary CaP cells, and its loss causes these cells to acquire invasive trait. To understand the underlying mechanism, we employed ROBO1-expressing/ROBO1-C2C3-mutant constructs, immunoprecipitation, confocal-microscopy and luciferase-reporter techniques. We show that ROBO1 through its interaction with DOCK1 (at SH3-SH2-domain) controls the Rac-activation. However, loss of ROBO1 results in Rac1-activation which in turn causes E-Cadherin/ß-catenin cytoskeleton destabilization and induction of cell migration. We suggest that ROBO1 is a predictive biomarker that has potential to discriminate among CaP types, and could be exploited as a molecular target to inhibit the progression of disease as well as treat metastasis in high-risk populations such as African-Americans.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 6_ODS3_enfermedades_notrasmisibles Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Negro ou Afro-Americano / Receptores Imunológicos / Genes Supressores de Tumor / População Branca / Proteínas do Tecido Nervoso Tipo de estudo: Etiology_studies / Incidence_studies / Observational_studies / Prognostic_studies / Risk_factors_studies Idioma: En Revista: Int J Cancer Ano de publicação: 2014 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 6_ODS3_enfermedades_notrasmisibles Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Negro ou Afro-Americano / Receptores Imunológicos / Genes Supressores de Tumor / População Branca / Proteínas do Tecido Nervoso Tipo de estudo: Etiology_studies / Incidence_studies / Observational_studies / Prognostic_studies / Risk_factors_studies Idioma: En Revista: Int J Cancer Ano de publicação: 2014 Tipo de documento: Article