Abstract

Persistent T cell antigen receptor (TCR) signaling by CD8 T cells is a feature of cancer and chronic infections and results in the sustained expression of, and signaling by, inhibitory receptors, which ultimately impair cytotoxic activity via poorly characterized mechanisms. We have previously determined that the LPA5 GPCR expressed by CD8 T cells, upon engaging the lysophosphatidic acid (LPA) bioactive serum lipid, functions as an inhibitory receptor able to negatively regulate TCR signaling. Notably, the levels of LPA and autotaxin (ATX), the phospholipase D enzyme that produces LPA, are often increased in chronic inflammatory disorders such as chronic infections, autoimmune diseases, obesity, and cancer. In this report, we demonstrate that LPA engagement selectively by LPA5 on human and mouse CD8 T cells leads to the inhibition of several early TCR signaling events including intracellular calcium mobilization and ERK activation. We further show that, as a consequence of LPA5 suppression of TCR signaling, the exocytosis of perforin-containing granules is significantly impaired and reflected by repressed in vitro and in vivo CD8 T cell cytolytic activity. Thus, these data not only document LPA5 as a novel inhibitory receptor but also determine the molecular and biochemical mechanisms by which a naturally occurring serum lipid that is elevated under settings of chronic inflammation signals to suppress CD8 T cell killing activity in both human and murine cells. As diverse tumors have repeatedly been shown to aberrantly produce LPA that acts in an autocrine manner to promote tumorigenesis, our findings further implicate LPA in activating a novel inhibitory receptor whose signaling may be therapeutically silenced to promote CD8 T cell immunity.

Highlights

  • CD8 T cell function often deteriorates in the presence of prolonged antigen-specific stimulation that accompanies chronic infections and cancer [1]

  • We have previously reported that naïve murine CD8 T cells express the LPA2, LPA5, and LPA6 G-protein coupled receptors (GPCRs) and that LPA5 was able to negatively regulate mouse CD8 T cell T cell antigen receptor (TCR) signaling [39]

  • Whether human CD8 T cell TCR signaling is subject to LPA5 regulation is not established so we initially determined if human CD8 T cells expressed the same orthologous lysophosphatidic acid (LPA) receptors using qPCR analysis of CD8 T cells isolated from peripheral blood

Read more

Summary

Introduction

CD8 T cell function often deteriorates in the presence of prolonged antigen-specific stimulation that accompanies chronic infections and cancer [1]. The in vivo therapeutic interference of these inhibitory receptors, referred to as immune checkpoint blockade, is able to restore CD8 T cell function and has achieved success in the treatment of certain cancers [15, 16]. Despite these achievements, current checkpoint blockade therapy has been successful for only a minority of patients and a subset of cancers indicating that different cancers use multiple and/or diverse mechanisms to suppress CD8 T cell cytotoxicity and evade anti-tumor immunity. It is evident that a better mechanistic understanding of the signaling pathways and inhibitory mechanisms used by inhibitory receptors will facilitate the targeting of multiple inhibitory signaling pathways and would be expected to lead to enhanced combination checkpoint blockade therapies [18]

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.