Abstract

Previous studies have reported the up-regulation of the two-pore domain K+ channel K2P5.1 in the CD4+ T cells of patients with multiple sclerosis (MS) and rheumatoid arthritis (RA), as well as in a mouse model of inflammatory bowel disease (IBD). However, the mechanisms underlying this up-regulation remain unclear. Inflammation-associated hypoxia is involved in the pathogenesis of autoimmune diseases, such as IBD, MS, and RA, and T cells are exposed to a hypoxic environment during their recruitment from inflamed tissues to secondary lymphoid tissues. We herein investigated whether inflammation-associated hypoxia is attributable to the increased expression and activity of K2P5.1 in the splenic CD4+ T cells of chemically-induced IBD model mice. Significant increases in hypoxia-inducible factor (HIF)-1α transcripts and proteins were found in the splenic CD4+ T cells of the IBD model. In the activated splenic CD4+ T cells, hypoxia (1.5% O2) increased K2P5.1 expression and activity, whereas a treatment with the HIF inhibitor FM19G11 but not the selective HIF-2 inhibitor exerted the opposite effect. Hypoxia-exposed K2P5.1 up-regulation was also detected in stimulated thymocytes and the mouse T-cell line. The class III histone deacetylase sirtuin-1 (SIRT1) is a downstream molecule of HIF-1α signaling. We examined the effects of the SIRT1 inhibitor NCO-01 on K2P5.1 transcription in activated CD4+ T cells, and we found no significant effects on the K2P5.1 transcription. No acute compensatory responses of K2P3.1–K2P5.1 up-regulation were found in the CD4+ T cells of the IBD model and the hypoxia-exposed T cells. Collectively, these results suggest a mechanism for K2P5.1 up-regulation via HIF-1 in the CD4+ T cells of the IBD model.

Highlights

  • Alkaline pH-activated K+ channels (K2P5.1, 16.1, 17.1) belonging to the two-pore domain K+ (K2P) channel superfamily contribute to setting the resting potential and control of Ca2+ signaling, and they have been implicated in inflammation and cancer development [1,2,3]

  • The present study demonstrated that 1) a hypoxic characteristic in inflamed colonic tissues generates hypoxia-inducible factor (HIF)-1α activation in the splenic CD4+ T cells of inflammatory bowel disease (IBD) model mice, and 2) the K2P5.1 channel is an HIF-1α target gene in splenic CD4+ T cells

  • Similar results were obtained in other T-cell lineage that were exposed to hypoxia

Read more

Summary

Introduction

Alkaline pH-activated K+ channels (K2P5.1, 16.1, 17.1) belonging to the two-pore domain K+ (K2P) channel superfamily contribute to setting the resting potential and control of Ca2+ signaling, and they have been implicated in inflammation and cancer development [1,2,3]. K2P5.1 expression and activity is up-regulated in the CD4+ T cells of patients with autoimmune diseases such as rheumatoid arthritis (RA) [7], and multiple sclerosis (MS) [8] and those from inflammatory bowel disease (IBD) model mice [9]; the mechanisms that underly inflammatory response-mediated K2P5.1 up-regulation in CD4+ T cells remain unclear. We recently reported the histone deacetylase (HDAC)-mediated up-regulation of KCa3.1 in the CD4+ T cells of IBD model mice [13,14]. No significant changes were observed in the expression levels of K2P5.1 transcripts following a treatment with selective class I HDAC inhibitors, suggesting that the mechanisms responsible for KCa3.1 and K2P5.1 transcription in T cells under inflammatory conditions differ

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