Abstract

Ca2+ influx through Orai1 channels is crucial for several T cell functions, but a role in regulating basal cellular motility has not been described. Here, we show that inhibition of Orai1 channel activity increases average cell velocities by reducing the frequency of pauses in human T cells migrating through confined spaces, even in the absence of extrinsic cell contacts or antigen recognition. Utilizing a novel ratiometric genetically encoded cytosolic Ca2+ indicator, Salsa6f, which permits real-time monitoring of cytosolic Ca2+ along with cell motility, we show that spontaneous pauses during T cell motility in vitro and in vivo coincide with episodes of cytosolic Ca2+ signaling. Furthermore, lymph node T cells exhibited two types of spontaneous Ca2+ transients: short-duration 'sparkles' and longer duration global signals. Our results demonstrate that spontaneous and self-peptide MHC-dependent activation of Orai1 ensures random walk behavior in T cells to optimize immune surveillance.

Highlights

  • To initiate the adaptive immune response, T cells must make direct contact with antigen-presenting cells (APCs) in the lymph node, enabling T cell receptors (TCRs) to engage peptide-bound MHC molecules presented on the APC surface

  • These experiments show that Enhanced green fluorescent protein (eGFP)-tagged Orai1-E106A expression can serve as a robust tool to assess cellular roles of Orai1 channel activity and that transfected cells without detectable eGFP fluorescence can be used as an internal control

  • Human T cells expressing the dominant-negative Orai1-E106A construct migrated with higher average velocities than controls, both in reconstituted mouse lymph nodes in vivo and in confined microchannels in vitro

Read more

Summary

Introduction

To initiate the adaptive immune response, T cells must make direct contact with antigen-presenting cells (APCs) in the lymph node, enabling T cell receptors (TCRs) to engage peptide-bound MHC molecules presented on the APC surface. Optimizing T cell motility to balance search sensitivity, specificity, and speed is crucial for efficient antigen search and proper immune function (Cahalan and Parker, 2005; Krummel et al, 2016). Both cell-intrinsic and environmental factors have been proposed to regulate T cell motility within lymph nodes and peripheral tissues (Miller et al, 2002; Bousso and Robey, 2003; Mempel et al, 2004; Mrass et al, 2010). Whereas the basic signaling mechanisms for cell-intrinsic induction of random motility have been previously explored in fibroblasts and neuroblastoma cells (Petrie et al, 2009), it remains unclear if such mechanisms apply in T cells

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