Abstract

Persistent antigen stimulation in chronic infections has been associated with antigen-specific T cell dysfunction and upregulation of inhibitory receptors, including programmed cell death protein 1 (PD-1). Pulmonary tuberculosis (TB) disease is characterized by high levels of Mycobacterium tuberculosis (Mtb), yet the relationship between bacterial load, PD-1 expression, and Mtb-specific T cell function in human TB has not been well-defined. Using peripheral blood samples from adults with LTBI and with pulmonary TB disease, we tested the hypothesis that PD-1 expression is associated with bacterial load and functional capacity of Mtb-specific T cell responses. We found that PD-1 was expressed at significantly higher levels on Th1 cytokine-producing Mtb-specific CD4 T cells from patients with smear-positive TB, compared with smear-negative TB and LTBI, which decreased after completion of anti-TB treatment. By contrast, expression of PD-1 on Mtb-specific CD8 T cells was significantly lower than on Mtb-specific CD4 T cells and did not differ by Mtb infection and disease status. In vitro stimulation of PBMC with Mtb antigens demonstrated that PD-1 is induced on proliferating Mtb-specific CD4 T cells and that Th1 cytokine production capacity is preferentially maintained within PD-1+ proliferating CD4 T cells, compared with proliferating Mtb-specific CD4 T cells that lack PD-1 expression. Together, these data indicate that expression of PD-1 on Mtb-specific CD4 T cells is indicative of mycobacterial antigen exposure and identifies a population of effector cells with Th1 cytokine production capacity. These studies provide novel insights into the role of the PD-1 pathway in regulating CD4 and CD8 T cell responses in Mtb infection and provide rationale for future studies to evaluate PD-1 expression on antigen-specific CD4 T cells as a potential biomarker for bacterial load and treatment response in human TB.

Highlights

  • Infection with Mycobacterium tuberculosis (Mtb) is responsible for over 10 million cases of tuberculosis (TB) and approximately 1.7 million deaths each year [1]

  • On day 6 of the assay, cells were re-stimulated with a CFP-10/ESAT-6 peptide pool for 6 h, and analyzed by flow cytometry for expression of PD-1 and intracellular IFN-γ and TNF-α. (A) Flow cytometry data indicating IFN-γ production capacity by proliferating

  • Plots are shown gated on viable CD3+CD4+ lymphocytes; data are shown from an individual with latent Mtb infection (LTBI) and a patient with smear+ TB. (B) Flow cytometry data indicating co-expression of PD-1 and IFN-γ by OGlo CD4 T cells following stimulation with a CFP-10/ESAT-6 peptide pool

Read more

Summary

INTRODUCTION

Infection with Mycobacterium tuberculosis (Mtb) is responsible for over 10 million cases of tuberculosis (TB) and approximately 1.7 million deaths each year [1]. Mtb infection of PD-1−/− mice leads to increased frequencies of Mtb-specific CD4 T cells; PD-1−/− mice display enhanced susceptibility to TB disease, characterized by increased bacterial loads, increased inflammatory and necrotic responses in the lungs, and reduced survival [34,35,36]. These studies in PD-1-deficient mice demonstrate a necessary role for PD-1 in limiting excessive IFN-γ production by CD4 T cells, which has been associated with exacerbated disease in murine models of TB [37]. To test the hypothesis that PD-1 expression is associated with bacterial load and Mtb-specific T cell functional capacity, we conducted a comprehensive analysis of PD-1 expression on Mtb-specific CD4 and CD8 T cells in clinically well-characterized cohorts of South African adults across a spectrum of Mtb infection and disease

MATERIALS AND METHODS
Ethics Statement
Participants
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