Abstract

In cancer, cellular senescence is a complex process that leads to inhibition of proliferation of cells that may develop a neoplastic phenotype. A plethora of signaling pathways, when dysregulated, have been shown to elicit a senescence response. Two well-known tumor suppressor pathways, controlled by the p53 and retinoblastoma proteins, have been implicated in maintaining the cellular senescence phenotype. Kindlin-2, a member of an actin cytoskeleton organizing and integrin activator proteins, has been shown to play a key role in the regulation of several hallmarks of several cancers, including breast cancer (BC). The molecular mechanisms whereby Kindlin-2 regulates cellular senescence in BC tumors remains largely unknown. Here we show that Kindlin-2 regulates cellular senescence in part through its interaction with p53, whereby it regulates the expression of the p53-responsive genes; i.e., SerpinB2 and p21, during the induction of senescence. Our data show that knockout of Kindlin-2 via CRISPR/Cas9 in several BC cell lines significantly increases expression levels of both SerpinB2 and p21 resulting in the activation of hallmarks of cellular senescence. Mechanistically, interaction between Kindlin-2 and p53 at the promotor level is critical for the regulated expression of SerpinB2 and p21. These findings identify a previously unknown Kindlin-2/p53/SerpinB2 signaling axis that regulates cellular senescence and intervention in this axis may serve as a new therapeutic window for BCs treatment.

Highlights

  • Physiological cellular senescence is defined as the irreversible arrest of cell proliferation[1]

  • To further explore the role of Kindlin-2 in breast cancer (BC) pathology, we performed an RNA-seq analysis to identify genes affected by loss of Kindlin-2

  • Nuclear function of Kindlin-2 was previously reported to form a transcriptional complex with β-catenin to regulate Wnt signaling in BC29

Read more

Summary

Introduction

Physiological cellular senescence is defined as the irreversible arrest of cell proliferation[1]. A plethora of signaling pathways, when dysregulated, have been identified to elicit a senescence response[3] These include DNA damage, dysfunctional telomeres, perturbations to chromatin organization, activation of certain oncogenes, and inhibition of certain tumor suppressor genes[2,3,4]. In both physiological and Kindlin-2 (FERMT2) is one of three kindlin family members of FERM domain-containing proteins. It is an adapter protein with many different binding partners including the cytoplasmic tail of integrin beta subunits, an interaction that permits kindlins to function as key regulators in integrin activation[6,7,8]. In BC, Kindlin-2 is involved in the regulation of the tumor microenvironment by influencing the recruitment of macrophages to the Official journal of the Cell Death Differentiation Association

Methods
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