Abstract

KAI1/CD82 protein is a member of the tetraspanin superfamily and has been rediscovered as a cancer metastasis suppressor. The mechanism of KAI1/CD82-mediated suppression of cancer metastasis remains to be established. In this study, we found that migration of the metastatic prostate cancer cell line Du145 was substantially inhibited when KAI1/CD82 was expressed. The expression of focal adhesion kinase (FAK) and Lyn, a Src family tyrosine kinase and substrate of FAK, was up-regulated at both RNA and protein levels upon KAI1/CD82 expression. The activation of FAK and Lyn, however, remained unchanged in Du145-KAI1/CD82 cells. As a downstream target of FAK-Lyn signaling, the p130CAS (Crk-associated substrate) protein was decreased upon the expression of KAI1/CD82. Consequently, less p130CAS-CrkII complex, which functions as a "molecular switch" in cell motility, was formed in Du145-KAI1/CD82 cells. To confirm that the p130CAS-CrkII complex is indeed important for the motility inhibition by KAI1/CD82, overexpression of p130CAS in Du145-KAI1/CD82 cells increased the formation of p130CAS-CrkII complex and largely reversed the KAI1/CD82-mediated inhibition of cell motility. Taken together, our studies indicate the following: 1) signaling of FAK-Lyn-p130CAS-CrkII pathway is altered in KAI1/CD82-expressing cells, and 2) p130CAS-CrkII coupling is required for KAI1/CD82-mediated suppression of cell motility.

Highlights

  • Understanding the molecular determinants that govern cancer invasiveness and metastasis is fundamentally important for successfully diagnosing and treating metastatic cancers

  • Other transmembrane 4 superfamily (TM4SF) proteins such as CD81 and CD151 were equivalently expressed on the cell surfaces of both transfectants, and their levels were not altered by the KAI1/CD82 expression in Du145 cells

  • The diminished haptotactic cell migration in Du145-KAI1/CD82 cells was observed on both FN- and LN5-coated substrata (Fig. 2A), indicating that Du145-KAI1/CD82 has general suppressive effects on cell migration mediated by both FN- and LM-binding integrins such as ␣5␤1 and ␣3␤1, respectively

Read more

Summary

Introduction

Understanding the molecular determinants that govern cancer invasiveness and metastasis is fundamentally important for successfully diagnosing and treating metastatic cancers. Tially inhibits cell motility and/or invasiveness of these cancer cells in vitro and dramatically suppresses metastasis in animal models [1, 10, 14, 16] These observations indicate that the KAI1/CD82 expression level may determine the invasive and metastatic potential of various malignant tumors. Clustering KAI1/CD82 with its monoclonal antibody (mAb) induces cytoskeletal rearrangement and elongated cellular extension [26, 27] This signaling event is dependent on the activities of protein kinase A, protein kinase C (PKC), and Rho small GTPases but independent of Src kinase activity [26, 27]. The signaling pathway that involves Shc, MEK, and MAP kinase ERKs determines random cell migration through regulating actin-myosin assembly and cell contraction [28, 29]. The phosphatidylinositol 3-kinase (PI3-K)-Akt/PKB pathway plays an important role in the events of cell movement, such as cellular polarization during chemotaxis, by modulating FAK and Rho signaling pathways (34 –36)

Objectives
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call