Abstract

Thymocytes and thymic epithelial cells (TECs) cross-talk is essential to support T cell development and preserve thymic architecture and maturation of TECs and Foxp3+ natural regulatory T cells. Accordingly, disruption of thymic lymphostromal cross-talk may have major implications on the thymic mechanisms that govern T cell tolerance. Several genetic defects have been described in humans that affect early stages of T cell development [leading to severe combined immune deficiency (SCID)] or late stages in thymocyte maturation (resulting in combined immunodeficiency). Hypomorphic mutations in SCID-causing genes may allow for generation of a limited pool of T lymphocytes with a restricted repertoire. These conditions are often associated with infiltration of peripheral tissues by activated T cells and immune dysregulation, as best exemplified by Omenn syndrome (OS). In this review, we will discuss our recent findings on abnormalities of thymic microenvironment in OS with a special focus of defective maturation of TECs, altered distribution of thymic dendritic cells and impairment of deletional and non-deletional mechanisms of central tolerance. Here, taking advantage of mouse models of OS and atypical SCID, we will discuss how modifications in stromal compartment impact and shape lymphocyte differentiation, and vice versa how inefficient T cell signaling results in defective stromal maturation. These findings are instrumental to understand the extent to which novel therapeutic strategies should act on thymic stroma to achieve full immune reconstitution.

Highlights

  • Thymocytes and thymic epithelial cells (TECs) cross-talk is essential to support T cell development and preserve thymic architecture and maturation of TECs and Foxp3+ natural regulatory T cells

  • Deletion of self-reactive thymocytes in the thymic medulla is based on the recognition of self-antigens that are presented by medullary TECs and thymic dendritic cells (DCs)

  • A key role is played by the autoimmune regulator (AIRE), a transcription factor expressed by a subset of mature medullary TECs (mTECs) that drives the expression of tissue-restricted antigens (TRAs), mediating negative selection of autoreactive thymocytes [1, 2]

Read more

Summary

INTRODUCTION

Thymocytes and thymic epithelial cells (TECs) cross-talk is essential to support T cell development and preserve thymic architecture and maturation of TECs and Foxp3+ natural regulatory T (nTreg) cells. MM mice showed skin redness when shaved, no T cells infiltration was observed in the tissues and no obvious signs were reported, making this mutant strain a model of leaky SCID, in which T and B cells are present in low number and T cells are predominantly activated, but no obvious signs of autoimmunity are present [26] In another mouse model, homozygosity for the Rag S721C mutation was associated with impaired T cell development, presence of oligoclonal, activated T cells, profound B cell lymphopenia, and yet significant serum levels of immunoglobulin [15, 17, 18]. The study of animal models carrying hypomorphic Rag mutations has demonstrated that defective T cell lymphopoiesis affects maturation and function of thymic stroma, and impinges on both deletional and non-deletional mechanisms of immune tolerance, thereby providing important insights on the pathophysiology of OS

ANIMAL STUDIES TO TARGET THYMIC STROMA IN Rag DEFICIENCIES
Findings
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