Your browser doesn't support javascript.
loading
Relocation of CLIC1 promotes tumor cell invasion and colonization of fibrin.
Gurski, Lisa A; Knowles, Lynn M; Basse, Per H; Maranchie, Jodi K; Watkins, Simon C; Pilch, Jan.
Afiliação
  • Gurski LA; Department of Urology, University of Pittsburgh School of Medicine, Shadyside Medical Center, Pittsburgh, Pennsylvania.
  • Knowles LM; Department of Urology, University of Pittsburgh School of Medicine, Shadyside Medical Center, Pittsburgh, Pennsylvania.
  • Basse PH; University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
  • Maranchie JK; Department of Urology, University of Pittsburgh School of Medicine, Shadyside Medical Center, Pittsburgh, Pennsylvania. University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
  • Watkins SC; University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania. Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
  • Pilch J; Department of Urology, University of Pittsburgh School of Medicine, Shadyside Medical Center, Pittsburgh, Pennsylvania. University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania. Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University Medical Center, Homburg, Saa
Mol Cancer Res ; 13(2): 273-80, 2015 Feb.
Article em En | MEDLINE | ID: mdl-25205595
ABSTRACT
UNLABELLED Chloride intracellular channel 1 (CLIC1) has been shown to be upregulated in various malignancies but its exact function remains unclear. Here, it is revealed that CLIC1 is critical for the stability of invadopodia in endothelial and tumor cells embedded in a 3-dimensional (3D) matrix of fibrin. Invadopodia stability was associated with the capacity of CLIC1 to induce stress fiber and fibronectin matrix formation following its ß3 integrin (ITGB3)-mediated recruitment into invadopodia. This pathway, in turn, was relevant for fibrin colonization as well as slug (SNAI2) expression and correlated with a significant role of CLIC1 in metastasis in vivo. Mechanistically, a reduction of myosin light chain kinase (MYLK) in CLIC1-depleted as well as ß3 integrin-depleted cells suggests an important role of CLIC1 for integrin-mediated actomyosin dynamics in cells embedded in fibrin. Overall, these results indicate that CLIC1 is an important contributor to tumor invasion, metastasis, and angiogenesis. IMPLICATIONS This study uncovers an important new function of CLIC1 in the regulation of cell-extracellular matrix interactions and ability of tumor cells to metastasize to distant organs.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Canais de Cloreto / Matriz Extracelular / Metástase Neoplásica / Neovascularização Patológica Limite: Adult / Aged / Female / Humans / Male / Middle aged Idioma: En Ano de publicação: 2015 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Canais de Cloreto / Matriz Extracelular / Metástase Neoplásica / Neovascularização Patológica Limite: Adult / Aged / Female / Humans / Male / Middle aged Idioma: En Ano de publicação: 2015 Tipo de documento: Article