Abstract

Chemokines and their receptors play diverse roles in regulating cancer growth and progression. The receptor CXCR3 can have two splice variants with opposite functions. CXCR3-A promotes cell growth, whereas CXCR3-B mediates growth-inhibitory signals. However, the negative signals through CXCR3-B in cancer cells are not well characterized. In this study, we found that CXCR3-B-mediated signaling in MCF-7 and T47D breast cancer cells induced apoptotic cell death. Signals through CXCR3-B decreased the levels of the antiapoptotic proteins Bcl-2 and Bcl-xL and increased the expression of apoptotic cleaved poly(ADP-ribose) polymerase. Along with up-regulation in apoptosis, CXCR3-B signals were associated with a decrease in cellular autophagy with reduced levels of the autophagic markers Beclin-1 and LC3B. Notably, CXCR3-B down-regulated the expression of the cytoprotective and antiapoptotic molecule heme oxygenase-1 (HO-1) at the transcriptional level. There was an increased nuclear localization of Bach-1 and nuclear export of Nrf2, which are important negative and positive transcription factors, respectively, for HO-1 expression. We also observed that CXCR3-B promoted the activation of p38 MAPK and the inhibition of ERK-1/2. CXCR3-B could not induce cancer cell apoptosis at the optimal level when we either inhibited p38 activity or knocked down Bach-1. Further, CXCR3-B-induced apoptosis was down-regulated when we overexpressed HO-1. Together, our data suggest that CXCR3-B mediates a growth-inhibitory signal in breast cancer cells through the modulations of nuclear translocation of Bach-1 and Nrf2 and down-regulation of HO-1. We suggest that the induction of CXCR3-B-mediated signaling can serve as a novel therapeutic approach where the goal is to promote tumor cell apoptosis.

Highlights

  • The chemokine receptor CXCR3-B initiates inhibitory signals

  • Induction of CXCR3-B-medited Signaling Promotes Apoptosis and Inhibits Autophagy of Breast Cancer Cells—We observed that the expression of CXCR3-B is much lower in breast cancer cells compared with normal breast epithelial cells (Fig. 1A, top left panel)

  • Our findings suggest that the CXCR3-B-mediated signal can induce nuclear translocation of Bach-1 and nuclear export of nuclear factor E2-related factor 2 (Nrf2), which can be associated with the down-regulation of heme oxygenase-1 (HO-1) transcription

Read more

Summary

Background

Results: CXCR3-B-mediated signal induced apoptosis and inhibited autophagy of breast cancer cells It is associated with nuclear translocation of Bach-1, nuclear export of Nrf, and down-regulation of HO-1. Significance: Induction of CXCR3-B-mediated signaling can serve as a novel therapeutic approach for the treatment of breast cancer Chemokines and their receptors play diverse roles in regulating cancer growth and progression. Our data suggest that CXCR3-B mediates a growth-inhibitory signal in breast cancer cells through the modulations of nuclear translocation of Bach-1 and Nrf and down-regulation of HO-1. We show that CXCR3-B mediates a growthinhibitory signal in human breast cancer cells through the down-regulation of antiapoptotic HO-1 It is associated with decreased phosphorylation of ERK-1/2 and increased phosphorylation of p38 MAPK. CXCR3-B-induced signals promote increased nuclear translocation of Bach-1 and nuclear export of Nrf

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