scholarly journals 007 GILT-mediated antigen processing in thymic epithelial cells diminishes T cell-mediated protection from melanoma through promoting thymic deletion and regulatory T cells

2016 ◽  
Vol 136 (5) ◽  
pp. S2
Author(s):  
M.P. Rausch ◽  
T.C. Metzger ◽  
M. Waterfield ◽  
J. Cortez ◽  
M.S. Anderson ◽  
...  
2002 ◽  
Vol 195 (10) ◽  
pp. 1349-1358 ◽  
Author(s):  
Karen Honey ◽  
Terry Nakagawa ◽  
Christoph Peters ◽  
Alexander Rudensky

CD4+ T cells are positively selected in the thymus on peptides presented in the context of major histocompatibility complex class II molecules expressed on cortical thymic epithelial cells. Molecules regulating this peptide presentation play a role in determining the outcome of positive selection. Cathepsin L mediates invariant chain processing in cortical thymic epithelial cells, and animals of the I-Ab haplotype deficient in this enzyme exhibit impaired CD4+ T cell selection. To determine whether the selection defect is due solely to the block in invariant chain cleavage we analyzed cathepsin L–deficient mice expressing the I-Aq haplotype which has little dependence upon invariant chain processing for peptide presentation. Our data indicate the cathepsin L defect in CD4+ T cell selection is haplotype independent, and thus imply it is independent of invariant chain degradation. This was confirmed by analysis of I-Ab mice deficient in both cathepsin L and invariant chain. We show that the defect in positive selection in the cathepsin L−/− thymus is specific for CD4+ T cells that can be selected in a wild-type and provide evidence that the repertoire of T cells selected differs from that in wild-type mice, suggesting cortical thymic epithelial cells in cathepsin L knockout mice express an altered peptide repertoire. Thus, we propose a novel role for cathepsin L in regulating positive selection by generating the major histocompatibility complex class II bound peptide ligands presented by cortical thymic epithelial cells.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 835-835
Author(s):  
Phillip M. Garfin ◽  
Patrick Viatour ◽  
Dullei Min ◽  
Jerrod Bryson ◽  
Kenneth I. Weinberg ◽  
...  

Abstract Abstract 835 The establishment of the thymic microenvironment early in life is crucial for the production functional T cells. Conversely, thymic involution results in a decreased T cell output. Thymic involution has important health implications especially following bone marrow transplant. Our objective is to determine molecular and cellular mechanisms that will allow for regeneration of involuted thymic tissue, restore production of naïve T cells, and improve immune function while improving our understanding of immunobiology. In this pursuit, we have focused on the Retinoblastoma family of tumor suppressor proteins. The main function of the RB pathway is to restrict passage through the G1/S transition of the cell cycle. RB and its two family members, p107 and p130, mediate the action of a broad range of cellular signals to control the proliferation, survival, and differentiation status of a large number of mammalian cell types. We found that inactivation of the RB pathway in the thymus by early deletion of RB family genes prevents thymic involution, promotes expansion of functional thymic epithelial cells (TECs), and increases thymic T cell output. Moreover, we have identified a direct regulatory relationship between RB and the Foxn1 transcription factor Via E2F transcription factors, where RB/E2F complexes directly repress the Foxn1 promoter, thereby promoting involution. Thus, the RB family is a critical mediator of extra- and intra-cellular signals to regulate thymic epithelial cells and thymus function, and decreasing RB pathway function may promote regeneration of the involuted thymus and restoration of naïve T cell production in patients. Disclosures: No relevant conflicts of interest to declare.


1990 ◽  
Vol 171 (4) ◽  
pp. 1101-1121 ◽  
Author(s):  
E K Gao ◽  
D Lo ◽  
J Sprent

T cell tolerance induction was examined in long-term H-2-heterozygous parent----F1 chimeras prepared with supralethal irradiation (1,300 rad). Although these chimeras appeared to be devoid of host-type APC, the donor T cells developing in the chimeras showed marked tolerance to host-type H-2 determinants. Tolerance to the host appeared to be virtually complete in four assay systems: (a) primary mixed lymphocyte reactions (MLR) of purified lymph node (LN) CD8+ cells (+/- IL-2); (b) primary MLR of CD4+ (CD8-) thymocytes; (c) skin graft rejection; and (d) induction of lethal graft-vs.-host disease by CD4+ cells. Similar tolerance was observed in chimeras given double irradiation. The only assay in which the chimera T cells failed to show near-total tolerance to the host was the primary MLR of post-thymic CD4+ cells. In this assay, LN CD4+ cells regularly gave a significant antihost MLR. The magnitude of this response was two- to fourfold less than the response of normal parental strain CD4+ cells and, in I-E(-)----I-E+ chimeras, was paralleled by approximately 70% deletion of V beta 11+ cells. Since marked tolerance was evident at the level of mature thymocytes, tolerance induction in the chimeras presumably occurred in the thymus itself. The failure to detect host APC in the thymus implies that tolerance reflected contact with thymic epithelial cells (and/or other non-BM-derived cells in the thymus). To account for the residual host reactivity of LN CD4+ cells and the incomplete deletion of V beta 11+ cells, it is suggested that T cell contact with thymic epithelial cells induced clonal deletion of most of the host-reactive T cells but spared a proportion of these cells (possibly low affinity cells). Since these latter cells appeared to be functionally inert in the thymus (in contrast to LN), we suggest that the thymic epithelial cells induced a temporary form of anergy in the remaining host-reactive thymocytes. This anergic state disappeared when the T cells left the thymus and reached LN.


Blood ◽  
2006 ◽  
Vol 108 (4) ◽  
pp. 1298-1305 ◽  
Author(s):  
Mikael Maksimow ◽  
Mari Miiluniemi ◽  
Fumiko Marttila-Ichihara ◽  
Sirpa Jalkanen ◽  
Arno Hänninen

Abstract Lymphoma cells are malignant cells of the T- or B-cell lineage that often express many surface markers inappropriately, yet are not recognized as abnormal by the immune system. We modeled this situation by inoculating ovalbumin-expressing E.G7-OVA lymphoma cells into mice that expressed ovalbumin as a self antigen in pancreatic islets, and investigated the efficacy of dendritic cell (DC) vaccination in these mice. Although vaccination with DC-expressing ovalbumin induced strong cytotoxic T-cell immunity, which led to clearance of E.G7-OVA lymphoma cells in naive C57BL/6 mice, DC vaccination was ineffective in mice expressing ovalbumin as a self antigen. Antigen modification to increase its processing via the endosomal processing pathway dramatically increased CD4 T-cell activation but paradoxically, impaired the protective effect of DC vaccination even in naive mice. Depletion of CD25+ T cells (regulatory T cells [Tregs]) prior to vaccination restored the efficacy of DC vaccination and allowed eradication of lymphoma also in mice expressing ovalbumin as a self antigen. We conclude that lymphoma cells may be eradicated using DC vaccination if activation of CD25+ Tregs is simultaneously inhibited, and that intentionally enhanced endosomal antigen processing in DC vaccines may shift the balance from CD4 T-cell help toward stimulation of Tregs.


2021 ◽  
Vol 118 (37) ◽  
pp. e2100542118
Author(s):  
Balázs Koncz ◽  
Gergő M. Balogh ◽  
Benjamin T. Papp ◽  
Leó Asztalos ◽  
Lajos Kemény ◽  
...  

Adaptive immune recognition is mediated by the binding of peptide–human leukocyte antigen complexes by T cells. Positive selection of T cells in the thymus is a fundamental step in the generation of a responding T cell repertoire: only those T cells survive that recognize human peptides presented on the surface of cortical thymic epithelial cells. We propose that while this step is essential for optimal immune function, the process results in a defective T cell repertoire because it is mediated by self-peptides. To test our hypothesis, we focused on amino acid motifs of peptides in contact with T cell receptors. We found that motifs rarely or not found in the human proteome are unlikely to be recognized by the immune system just like the ones that are not expressed in cortical thymic epithelial cells or not presented on their surface. Peptides carrying such motifs were especially dissimilar to human proteins. Importantly, we present our main findings on two independent T cell activation datasets and directly demonstrate the absence of naïve T cells in the repertoire of healthy individuals. We also show that T cell cross-reactivity is unable to compensate for the absence of positively selected T cells. Additionally, we show that the proposed mechanism could influence the risk for different infectious diseases. In sum, our results suggest a side effect of T cell positive selection, which could explain the nonresponsiveness to many nonself peptides and could improve the understanding of adaptive immune recognition.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3347-3347
Author(s):  
Karlie R. Sharma ◽  
Sarah E Bryant ◽  
Sherry Koontz ◽  
Harry L. Malech ◽  
Elizabeth M. Kang

Abstract The thymus plays a crucial role in preventing autoimmune reactions in a variety of species. Through the expression of the autoimmune regulator (Aire), a protein found in medullary thymic epithelial cells (mTECs), the thymus is able to express tissue-restricted peripheral self-antigens (TRAs). Through their interaction with these TRAs, developing thymocytes that target host antigens can be tagged for clonal deletion. Graft versus host disease (GvHD), an autoimmune reaction occurring in approximately 40% of allogeneic stem cell transplantation patients, is an immune mediated reaction in which donor T cells recognize the host as antigenically foreign, causing donor T cells to expand and attack host tissues. GvHD is often associated with significant morbidity and mortality, and because the thymus is a target tissue of T-cell alloimmunity and can be severely damaged in many autoimmune diseases, we focused on significant morphological and cellular changes within the thymus as possible contributors to the continuing proliferation and survival of alloimmune T-cells in GvHD. Previous data collected from a mouse model of GvHD showed a reduction in expression of Aire and a reduced diversity of TRAs in the thymus. We also found that the thymus is significantly smaller in size when compared to wild-type mice, which correlates to overall thymic involution as both the cortical and medullary TEC levels were reduced. This reduction of Aire expression also correlated with a reduction in thymically-derived FoxP3+NrpI+regulatory T-cells, which were significantly reduced in both spleen and blood of GvHD mice when compared to bone marrow transplant controls (blood p < 0.0005, spleen p < 0.0001). SEMA4A, a transmembrane protein known to interact with Nrp1 to promote regulatory T-cell function and survival, was also significantly changed in GvHD compared to healthy controls (p = 0.014). In addition, cytokine data collected from a GvHD mouse model also showed increases in several chemokines associated with thymic atrophy and aging, indicating that thymii found in GvHD mice are subject to significant damage due to autoimmune reactions. Interestingly, there is evidence of thymic damage in mouse models of colitis as well. Ulcerative colitis is a form of inflammatory bowel disease caused by T-cell infiltration into the colon, resembling some of the pathophysiology of G.I. GvHD. Thymii from DSS colitis mice showed a similarly significant reduction of Aire and FoxP3 expression, suggesting a new, more prominent role for thymic damage in colitis severity (p < 0.005). This damage was observed to occur in a generalized way, as FACS analysis of thymic epithelial cell types showed that both medullary and cortical epithelial cells in diseased thymus were reduced, not just mTECs. The reduction in expression of Aire protein is due directly to RNA reduction, with a mean 8 fold reduction of Aire RNA expression in colitis mice compared to healthy mice (p = 0.0154). While SEMA4A was seen to be increased in GvHD, there was a significant reduction in expression in colitis thymii (p=0.0138). Reductions in SEMA4A have previously been shown to cause impaired immune cell differentiation and function, followed by impaired homeostasis and function of FoxP3+T-cells, indicating that this reduction in SEMA4A may be the cause of reduced FoxP3 expression in the colitis thymus. It was also observed that NrpI expressing FoxP3 cells in the spleen and blood were unchanged in colitis mice, indicating that reduction in FoxP3 expressing cells in colitis are not entirely due to thymic damage. This is supported by cytokine data, in which no inflammatory or aging related cytokines were changed in colitis thymii. The significant thymic involution and reduced expression of the Aire protein in the thymus is a likely contributor to overall increases of autoimmune T-cells in both colitis and GvHD. Though colitis and GvHD clearly use different pathways to affect the thymus, thymic damage plays a significant role in both diseases. The reduced expression of the FoxP3 protein also indicates a role for loss of Aire in lowered immune tolerance, contributing to the overall autoimmune phenotype in both diseases. Disclosures No relevant conflicts of interest to declare.


2006 ◽  
Vol 19 (5) ◽  
pp. 404-414 ◽  
Author(s):  
Yimin Sun ◽  
Bao-Sheng Ge ◽  
Michiyuki Kasai ◽  
Clara Diffendaffer ◽  
Nancy Parks ◽  
...  

2020 ◽  
Vol 10 ◽  
Author(s):  
Hong-Xia Wang ◽  
Wenrong Pan ◽  
Lei Zheng ◽  
Xiao-Ping Zhong ◽  
Liang Tan ◽  
...  

The thymus is the primary lymphoid organ responsible for the generation and maturation of T cells. Thymic epithelial cells (TECs) account for the majority of thymic stromal components. They are further divided into cortical and medullary TECs based on their localization within the thymus and are involved in positive and negative selection, respectively. Establishment of self-tolerance in the thymus depends on promiscuous gene expression (pGE) of tissue-restricted antigens (TRAs) by TECs. Such pGE is co-controlled by the autoimmune regulator (Aire) and forebrain embryonic zinc fingerlike protein 2 (Fezf2). Over the past two decades, research has found that TECs contribute greatly to thymopoiesis and T cell development. In turn, signals from T cells regulate the differentiation and maturation of TECs. Several signaling pathways essential for the development and maturation of TECs have been discovered. New technology and animal models have provided important observations on TEC differentiation, development, and thymopoiesis. In this review, we will discuss recent advances in classification, development, and maintenance of TECs and mechanisms that control TEC functions during thymic involution and central tolerance.


Sign in / Sign up

Export Citation Format

Share Document