Abstract

Regulatory T cells (Tregs) are the major determinant of peripheral immune tolerance. Many Treg subsets have been described, however thymus-derived and peripherally induced Tregs remain the most important subpopulations. In multiple sclerosis, a prototypical autoimmune disorder of the central nervous system, Treg dysfunction is a pathogenic hallmark. In contrast, induction of Treg proliferation and enhancement of their function are central immune evasion mechanisms of infectious pathogens. In accordance, Treg expansion is compartmentalized to tissues with high viral replication and prolonged in chronic infections. In friend retrovirus infection, Treg expansion is mainly based on excessive interleukin-2 production by infected effector T cells. Moreover, pathogens seem also to enhance Treg functions as shown in human immunodeficiency virus infection, where Tregs express higher levels of effector molecules such as cytotoxic T-lymphocyte-associated protein 4, CD39 and cAMP and show increased suppressive capacity. Thus, insights into the molecular mechanisms by which intracellular pathogens alter Treg functions might aid to find new therapeutic approaches to target central nervous system autoimmunity. In this review, we summarize the current knowledge of the role of pathogens for Treg function in the context of autoimmune neuroinflammation. We discuss the mechanistic implications for future therapies and provide an outlook for new research directions.

Highlights

  • In the context of infections, Tregs mediate beneficial and detrimental effects on short- and long-term disease outcomes

  • We investigate the impact of pathogens on immune cell distributions, profiles, and functionality - Treg functions - in the setting of neuroinflammation

  • Dysregulated expression of human endogenous retroviruses (HERVs) may contribute to central nervous system (CNS) damage such as observed in a severe combined immunodeficiency mouse model [302]. This dysregulated HERV-W activity is likely to involve binding of its envelope protein (ENV) to TLR4 and its co-receptor CD14 which triggers the release of pro-inflammatory cytokines such as TNFa, IL-1b or IL-6 fostering the autoimmune response [303,304,305]

Read more

Summary

INTRODUCTION

In the context of infections, Tregs mediate beneficial and detrimental effects on short- and long-term disease outcomes. Tregs are generally found to express CD4 and either or both the highaffinity receptor for interleukin (IL)-2 CD25+ as well as the forkhead box protein P3 (Foxp3) [3] Their expression of intracellular and surface markers, such as CD25, glucocorticoid-induced tumor necrosis factor receptor (GITR) and the inhibitory cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) define their phenotype and function [4,5,6]. Defining the mechanisms by which intracellular pathogens alter Treg function might pave the way toward new therapeutic approaches in the settings of infections, and in autoimmune neuroinflammation. We discuss how the complex changes in Tregs lead to altered function and that the underlying mechanisms could contribute to better understand the pathophysiology of neuroinflammatory diseases and their treatments. We discuss established and potential therapeutic strategies in MS resulting from this new molecular understanding

Janus-Faced Nature
One Janus Face
Other Janus Face
Role of Tregs in Multiple Sclerosis
Results
Tregs in the Advent of the Era of Immunometabolomics
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