The prostate metastasis suppressor gene NDRG1 differentially regulates cell motility and invasion

Anup Sharma, Janet Mendonca, James Ying, Hea Soo Kim, James E. Verdone, Jelani C. Zarif, Michael Carducci, Hans Hammers, Kenneth J. Pienta, Sushant Kachhap

Research output: Contribution to journalArticlepeer-review

31 Scopus citations

Abstract

Experimental and clinical evidence suggests that N-myc downregulated gene 1 (NDRG1) functions as a suppressor of prostate cancer metastasis. Elucidating pathways that drive survival and invasiveness of NDRG1-deficient prostate cancer cells can help in designing therapeutics to target metastatic prostate cancer cells. However, the molecular mechanisms that lead NDRG1-deficient prostate cancer cells to increased invasiveness remain largely unknown. In this study, we demonstrate that NDRG1-deficient prostate tumors have decreased integrin expression and reduced cell adhesion and motility. Our data indicate that loss of NDRG1 differentially affects Rho GTPases. Specifically, there is a downregulation of active RhoA and Rac1 GTPases with a concomitant upregulation of active Cdc42 in NDRG1-deficient cells. Live cell imaging using a fluorescent sensor that binds to polymerized actin revealed that NDRG1-deficient cells have restricted actin dynamics, thereby affecting cell migration. These cellular and molecular characteristics are in sharp contrast to what is expected after loss of a metastasis suppressor. We further demonstrate that NDRG1-deficient cells have increased resistance to anoikis and increased invasiveness which is independent of its elevated Cdc42 activity. Furthermore, NDRG1 regulates expression and glycosylation of EMMPRIN, a master regulator of matrix metalloproteases. NDRG1 deficiency leads to an increase in EMMPRIN expression with a concomitant increase in matrix metalloproteases and thus invadopodial activity. Using a three-dimensional invasion assay and an in vivo metastasis assay for human prostate xenografts, we demonstrate that NDRG1-deficient prostate cancer cells exhibit a collective invasion phenotype and are highly invasive. Thus, our findings provide novel insights suggesting that loss of NDRG1 leads to a decrease in actinmediated cellular motility but an increase in cellular invasion, resulting in increased tumor dissemination which positively impacts metastatic outcome.

Original languageEnglish (US)
Pages (from-to)655-669
Number of pages15
JournalMolecular oncology
Volume11
Issue number6
DOIs
StatePublished - Jun 2017

Keywords

  • Actin cytoskeleton
  • Matrix metalloproteases
  • Metastasis
  • Prostate cancer

ASJC Scopus subject areas

  • Genetics
  • Molecular Medicine
  • Oncology
  • Cancer Research

Fingerprint

Dive into the research topics of 'The prostate metastasis suppressor gene NDRG1 differentially regulates cell motility and invasion'. Together they form a unique fingerprint.

Cite this