Molecular Medicine Israel

PSGL-1 attenuates early TCR signaling to suppress CD8+ T cell progenitor differentiation and elicit terminal CD8+ T cell exhaustion

Highlights

  • PSGL-1 restrains TCR signaling and glycolytic potential of CD8+ T cells
  • PSGL-1 deficiency limits CD8+ T cell exhaustion and supports precursor populations
  • Co-ligation of PSGL-1 and the TCR promotes T cell exhaustion in CD8+ T cells
  • PSGL-1 therapeutic blockade promotes T cell responses and melanoma tumor control

Summary

PSGL-1 (P-selectin glycoprotein-1) is a T cell-intrinsic checkpoint regulator of exhaustion with an unknown mechanism of action. Here, we show that PSGL-1 acts upstream of PD-1 and requires co-ligation with the T cell receptor (TCR) to attenuate activation of mouse and human CD8+ T cells and drive terminal T cell exhaustion. PSGL-1 directly restrains TCR signaling via Zap70 and maintains expression of the Zap70 inhibitor Sts-1. PSGL-1 deficiency empowers CD8+ T cells to respond to low-affinity TCR ligands and inhibit growth of PD-1-blockade-resistant melanoma by enabling tumor-infiltrating T cells to sustain an elevated metabolic gene signature supportive of increased glycolysis and glucose uptake to promote effector function. This outcome is coupled to an increased abundance of CD8+ T cell stem cell-like progenitors that maintain effector functions. Additionally, pharmacologic blockade of PSGL-1 curtails T cell exhaustion, indicating that PSGL-1 represents an immunotherapeutic target for PD-1-blockade-resistant tumors.

Introduction

CD8+ effector T cells (TEFF) are vital in protecting against and clearing virally infected cells and tumor cells. However, in settings of chronic viral infection and cancer, multiple factors including sustained antigen exposure promote T cell exhaustion. Accompanied by progressive decline in effector functions and proliferative capacity, this state is linked to increased co-expression of multiple inhibitory receptors including PD-1, CTLA-4, LAG3, and TIM-3.1 The discovery of immune checkpoint blockade (ICB) monoclonal antibody therapy to promote antitumor immunity by inhibiting CTLA-4 and/or PD-1 on T cells is one of most significant cancer therapeutic breakthroughs of the last decade, with efficacy shown across many cancers.2 Despite dramatic success in some patients, a significant population fails to respond to ICB. It is therefore essential to identify novel mechanisms regulating the differentiation of exhausted T cells (TEX) that could expand ICB’s applicability to more patients. In particular, it will be critical to develop ICB strategies that support the development and maintenance of stem cell-like (TSC) or progenitor TEX (TPEX) that retain the capacity for proliferation and TEFF function.3

PSGL-1 (P-selectin glycoprotein ligand-1) is expressed on most hematopoietic cells, including T cells.4,5 We identified that genetic deletion of PSGL-1 prevented development of TEX and supported viral clearance in the chronic lymphocytic choriomeningitis virus (LCMV) clone 13 (Cl13) model6 due to increased TEFF function that was accompanied by decreased inhibitory receptor expression. PSGL-1 deficiency also supported growth control of a PD-1-blockade-resistant melanoma tumor.6 Furthermore, PSGL-1 deficiency with acute LCMV Armstrong infection promoted greater TEFF and memory progenitor T cell formation,7 underscoring the fundamental role PSGL-1 plays as a regulator of T cell responses. In this study, we investigated cellular and molecular mechanisms by which PSGL-1 signaling intrinsically regulates TEX differentiation including the impact of PSGL-1 on T cell receptor (TCR) signaling, glycolysis, and the promotion of TSC and TPEX. We demonstrate herein that PSGL-1 ligation concomitant with TCR ligation drives TEX differentiation in human and mouse CD8+ T cells. Critically, we show that blockade of PSGL-1 recapitulates PSGL-1 deficiency in antitumor and antiviral responses, and its distinct mechanism of T cell inhibition underscores the translational potential of targeting PSGL-1 by ICB.

Results

PSGL-1 restrains TCR signaling magnitude and duration

Since PSGL-1 deficiency enhanced TEFF responses, we addressed whether PSGL-1 modulates CD8+ T cell responses to TCR engagement. TCR stimulation of naive wild-type (WT) and PSGL-1−/− OT-I CD8+ T cells showed that PSGL-1 deficiency enabled higher expression of the T cell activation markers CD25, CD69, CD44, and PD-1 (Figure 1A)8,9,10,11 and greater functionality (Figure S1A). To directly evaluate TCR signaling, we assessed phosphorylation of Zap70, Erk1/2, and Akt by western blot (Figure 1B) and found greater activation of each of these signaling molecules in PSGL-1−/− T cells (Figure S1B), with the greatest effect on pAkt levels in PSGL-1−/− compared with WT T cells (Figure 1C), demonstrating that PSGL-1 expression inherently limits T cell activation from the time of initial TCR engagement…

Sign up for our Newsletter