Abstract

The tumor suppressor p53 is induced by genotoxic stress in both normal and transformed cells and serves to transcriptionally coordinate cell cycle checkpoint control and programmed cell death responses. Macrophage migration inhibitory factor (MIF) is an autocrine and paracrine acting cytokine/growth factor that promotes lung adenocarcinoma cell motility, anchorage-independence and neo-angiogenic potential. Several recent studies indicate that the only known homolog of MIF, D-dopachrome tautomerase (D-DT - also referred to as MIF-2), has functionally redundant activities with MIF and cooperatively promotes MIF-dependent pro-tumorigenic phenotypes. We now report that MIF and D-DT synergistically inhibit steady state p53 phosphorylation, stabilization and transcriptional activity in human lung adenocarcinoma cell lines. The combined loss of MIF and D-DT by siRNA leads to dramatically reduced cell cycle progression, anchorage independence, focus formation and increased programmed cell death when compared to individual loss of MIF or D-DT. Importantly, p53 mutant and p53 null lung adenocarcinoma cell lines were only nominally rescued from the cell growth effects of MIF/D-DT combined deficiency suggesting only a minor role for p53 in these transformed cell growth phenotypes. Finally, increased p53 activation was found to be independent of aberrantly activated AMP-activated protein kinase (AMPK) that occurs in response to MIF/D-DT-deficiency but is dependent on reactive oxygen species (ROS) that mediate aberrant AMPK activation in these cells. Combined, these findings suggest that both p53 wildtype and mutant human lung adenocarcinoma tumors rely on MIF family members for maximal cell growth and survival.

Highlights

  • The tumor suppressor p53 is induced by genotoxic stress in both normal and transformed cells and serves to transcriptionally coordinate cell cycle checkpoint control and programmed cell death responses

  • More recent studies demonstrate that migration inhibitory factor (MIF) and D-DT additively antagonize the tumor suppressive activities of AMP-activated protein kinase (AMPK) in lung adenocarcinoma cells resulting in maximal mTOR pathway activation [15]

  • Using previously characterized and validated siRNA oligos targeting MIF and D-DT [6,15], we knocked down both MIF family members - individually and jointly - and evaluated relative p53 responses in human lung adenocarcinoma cell lines

Read more

Summary

Introduction

P53 is the most commonly mutated tumor suppressor in human cancers, with a mutation rate higher than 50% [1]. The p53 pathway is mutated at a high frequency in non-small cell lung carcinoma (NSCLC) lesions (50–70%), suggesting an important contribution to tumorigenic initiation and progression [4] Those NSCLC lesions that harbor wildtype alleles of p53 are thought to have developed alternative mechanisms that serve to suppress p53 activity. More recent studies demonstrate that MIF and D-DT additively antagonize the tumor suppressive activities of AMP-activated protein kinase (AMPK) in lung adenocarcinoma cells resulting in maximal mTOR pathway activation [15]. Our data indicate that MIF and D-DT cooperate to maintain low steady state p53 expression and activity in human NSCLC cell lines, and that this inhibition partially accounts for MIF/D-DT-dependent promotion of pro-growth phenotypes. The functional overlap of MIF and D-DT in NSCLC pro-tumorigenic pathways provides strong rationale for the simultaneous therapeutic targeting of MIF and D-DT in lung adenocarcinoma malignant disease

Experimental Procedures
Results
Discussion
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