Abstract

CD8+ T cells are critical for controlling HIV infection. During the chronic phase of lentiviral infection, CD8+ T cells lose their proliferative capacity and exhibit impaired antiviral function. This loss of CD8+ T cell function is due, in part, to CD4+CD25+ T regulatory (Treg) cell-mediated suppression. Our research group has demonstrated that lentivirus-activated CD4+CD25+ Treg cells induce the repressive transcription factor forkhead box P3 (Foxp3) in autologous CD8+ T cells following co-culture. We have recently reported that Treg-induced Foxp3 binds the interleukin-2 (IL-2), interferon-γ (IFN- γ), and tumor necrosis factor-α (TNF-α) promoters in virus-specific CD8+ T cells. These data suggest an important role of Foxp3-mediated CD8+ T cell dysfunction in lentiviral infection. To elucidate the mechanism of this suppression, we previously reported that decreased methylation facilitates Foxp3 binding in mitogen-activated CD8+ T cells from feline immunodeficiency virus (FIV)-infected cats. We demonstrated the reduced binding of Foxp3 to the IL-2 promoter by increasing methylation of CD8+ T cells. In the studies presented here, we ask if another form of epigenetic modulation might alleviate Foxp3-mediated suppression in CD8+ T cells. We hypothesized that decreasing histone acetylation in virus-specific CD8+ T cells would decrease Treg-induced Foxp3 binding to the IL-2 promoter. Indeed, using anacardic acid (AA), a known histone acetyl transferase (HAT) inhibitor, we demonstrate a reduction in Foxp3 binding to the IL-2 promoter in virus-specific CD8+ T cells co-cultured with autologous Treg cells. These data identify a novel mechanism of Foxp3-mediated CD8+ T cell dysfunction during lentiviral infection.

Highlights

  • IntroductionImmunodeficiency Virus (HIV) and Feline Immunodeficiency Virus (FIV) infection are marked by robust expansion of CD8+ T cells during the acute phase which is followed by a steady state lower level of virus-specific CD8+ T cells during the chronic phase of infection

  • Acquired Immunodeficiency Syndrome (AIDS) lentiviral infections such as HumanImmunodeficiency Virus (HIV) and Feline Immunodeficiency Virus (FIV) infection are marked by robust expansion of CD8+ T cells during the acute phase which is followed by a steady state lower level of virus-specific CD8+ T cells during the chronic phase of infection

  • We demonstrated that treatment of virus-specific CD8+ T cells of feline immunodeficiency virus (FIV)-infected cats with anacardic acid (AA) for 24 h followed by co-culture with autologous T regulatory (Treg) cells resulted in a reduction in forkhead box P3 (Foxp3) binding at the IL-2 promoter

Read more

Summary

Introduction

Immunodeficiency Virus (HIV) and Feline Immunodeficiency Virus (FIV) infection are marked by robust expansion of CD8+ T cells during the acute phase which is followed by a steady state lower level of virus-specific CD8+ T cells during the chronic phase of infection. CD8+ T cells in the chronic phase of infection are characterized by impaired proliferative capacity, reduced cytolytic function and dysfunctional effector function [1,2,3]. Naive CD8+ T cells undergo rapid expansion and differentiation based on the strength and duration of IL-2 signals. Antigen-specific CD8+ T cells respond to autocrine and paracrine IL-2 signals via the high affinity trimer IL-2-R to expand and differentiate to mount a potent immune response [7].

Objectives
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