Abstract
Atherosclerotic cardiovascular disease (ASCVD) is a chronic inflammatory disease of the arterial walls and is characterized by the accumulation of lipoproteins that are insufficiently cleared by phagocytes. Following the initiation of atherosclerosis, the pathological progression is accelerated by engagement of the adaptive immune system. Atherosclerosis triggers the breakdown of tolerance to self-components. This loss of tolerance is reflected in defective expression of immune checkpoint molecules, dysfunctional antigen presentation, and aberrations in T cell populations — most notably in regulatory T (Treg) cells — and in the production of autoantibodies. The breakdown of tolerance to self-proteins that is observed in ASCVD may be linked to the conversion of Treg cells to ‘exTreg’ cells because many Treg cells in ASCVD express T cell receptors that are specific for self-epitopes. Alternatively, or in addition, breakdown of tolerance may trigger the activation of naive T cells, resulting in the clonal expansion of T cell populations with pro-inflammatory and cytotoxic effector phenotypes. In this Perspective, we review the evidence that atherosclerosis is associated with a breakdown of tolerance to self-antigens, discuss possible immunological mechanisms and identify knowledge gaps to map out future research. Rational approaches aimed at re-establishing immune tolerance may become game changers in treating ASCVD and in preventing its downstream sequelae, which include heart attacks and strokes.
Introduction
Atherosclerosis is an inflammatory disease affecting the arterial walls of large and medium-sized arteries and is characterized by the accumulation of low-density oxidized lipoproteins and immune cell-rich plaques1. Rupture or erosion of these plaques causes catastrophic thrombosis, which triggers immediate ischaemia in the heart, brain or extremities, subsequently culminating in tissue infarction1. According to the latest report of the American Heart Association, the global age-standardized mortality rate of cardiovascular disease (CVD) is 240 per 100,000 (20 million cases per year) whereas morbidity rate is 7,354 per 100,000 (608 million cases per year)2. The incidence of atherosclerotic cardiovascular disease (ASCVD) can be reduced by regulating low-density lipoprotein (LDL) cholesterol through statins and PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors3. Despite the success of statins and PCSK9 inhibitors, there is a significant residual inflammatory risk even when LDL cholesterol is treated to target4; the risk of major adverse cardiovascular events (MACEs) remains at 47% to 53% (refs. 4,5). Anti-inflammatory therapies such as the IL-1β inhibitor canakinumab have shown promise in reducing MACEs6. However, such therapies impair host defence mechanisms, resulting in other complications including lethal infections6. Other anti-inflammatory drugs, such as colchicine, have also shown promise in lowering MACEs in patients with stable coronary disease7,8.
Although it has been known for more than 20 years that patients with atherosclerosis have circulating autoantibodies against apolipoprotein B (ApoB)-containing lipoproteins, including LDL9,10, it was only recently that this observation was linked with a potential systemic loss of tolerance to self-components in atherosclerosis11,12. In mouse models of atherosclerosis, self-reactive CD4+ T cells4 enable cytotoxic CD8+ T cell-mediated killing of host cells13. Other self-reactive CD4+ T cells enter germinal centres, which promotes B cell affinity maturation, isotype switching and the development of plasma cells secreting high-affinity antibodies9. In some studies, high levels of IgG antibodies to LDL predict poor outcomes of ASCVD10, suggesting that the autoimmune response in atherosclerosis is maladaptive. However, the correlation of IgG and IgM levels to oxidized LDL and to ApoB peptides varies among studies. In most studies, no significant correlation was shown between the presence of IgG antibodies specific for oxidized low-density lipoprotein (oxLDL) and the occurrence of ASCVD. One study14 has actually reported that oxLDL-specific IgG antibody titres negatively correlated with Gensini scores (used to evaluate the severity of coronary artery disease) in patients undergoing coronary angiography. However, in another study15, oxLDL-specific IgG titres positively correlated with coronary artery stenosis (that is, narrowing of the blood vessels) in patients with myocardial infarction. In all studies reporting IgM against oxLDL, the levels of these antibodies were negatively correlated with disease severity in patients16–19, which supports findings made in mouse models of disease20. Therefore, there is data suggesting that atherosclerosis is associated with a loss of tolerance, but how this contributes to ASCVD outcomes is still unclear. In this Perspective, we review the emerging evidence that atherosclerosis is associated with a break of tolerance to self-antigens, discuss possible immunological mechanisms and identify knowledge gaps to map out future research.
Antigen presentation in atherosclerosis
Self-antigens recognized by T cells
In atherosclerosis lesions, LDL particles entrapped within the sub-endothelial space undergo oxidation. This process leads to the formation of malondialdehyde and 4-hydroxynonenal. This oxidative transformation correlates with significant structural alterations in LDL, involving fragmentation of ApoB and the creation of diverse aldehyde and phospholipid adducts bound to ApoB-derived peptides. Modified LDL can be taken up by scavenger receptors such as CD36, but CD4+ T cell responses to native ApoB sequences have also been demonstrated21,22. It is improbable that ApoB is the only relevant autoantigen; for example, heat shock proteins have also been proposed to be autoantigens in atherosclerosis23. In atherosclerosis, ApoB-specific CD4+ T cells show clonal expansion24. In a single-cell RNA sequencing (scRNA-seq) study with T cell receptor (TCR) sequencing, the ApoB-specific CD4+ T cells were identified by using tetramers to an ApoB epitope. Uniform Manifold Approximation and Projection for Dimension Reduction (UMAP) showed that the transcriptomes of most ApoB-specific cells were more similar to those of memory T cells than regulatory T cells24. Other atherosclerosis-associated autoantigens include oxLDL, malondialdehyde-modified LDL, β2-glycoprotein I and heat shock protein 60 (HSP60)21,23,25. However, the epitopes of these autoantigens that are targeted by T cells remain to be identified. It is also probable that more atherosclerosis-associated autoantigens remain to be discovered.
Antigen presentation
Atherosclerosis in humans and model organisms is always accompanied by autoantibody production. These atherosclerosis-associated autoantibodies are commonly measured by reactivity to oxLDL9, which contains many epitopes. Isotype-switched IgG antibodies specific for oxLDL require the assistance of T cells, specifically CD4+ follicular helper T (TFH) cells, which recognize antigenic epitopes displayed by MHC class II molecules. Circumstantial evidence suggests that antigen presentation occurs in the lymph nodes draining arteries affected by atherosclerosis (Fig. 1). In mice, the axillar and cervical lymph nodes draining the aortic and carotid arches become very large in response to atherosclerosis, suggesting fulminant antigen presentation1. Later, antigen-experienced T cells can be re-activated in a recall response in the atherosclerotic plaque proper26. The arterial adventitia is endowed with lymphatics, which have been shown to be involved in reverse cholesterol transport and provide a plausible pathway for the migration of dendritic cells (DCs) from atherosclerotic plaques to draining lymph nodes27. However, DCs carrying atherosclerosis-associated antigens to draining lymph nodes have not been shown experimentally. It should be noted that it is technically very challenging to trace these DCs. First, the vascular lymphatics are only incompletely described27. From what we know, the afferent lymphatics are very small and, therefore, hard to cannulate. Second, the kinetics of DC recruitment from the artery wall are not known.
Fig. 1 |. Antigen presentation in atherosclerosis.
The adventitia of large arteries has afferent lymphatics that drain into local lymph nodes, but in the setting of atherosclerosis, how immune cells can migrate from the plaque to the adventitia is unknown. Antigen-presenting cells (APCs) may carry antigen from the artery to the draining lymph node, wherein they enter germinal centres (indicated by dashed lines) that are populated by B cells (blue) and some T cells (red). High endothelial venules (HEVs) are the gateways enabling entry of naive T cells from the blood into lymph nodes. In atherosclerosis, plaque forms a neointima (yellow) and low-density lipoprotein (LDL) accumulates and becomes oxidized. In the draining lymph node, some T cells undergo clonal expansion in response to arterial antigens including ApoB. Some T cells leave via efferent lymphatics and traverse the thoracic duct (dark blue-shaded area) to enter the blood circulation. Clonally expanded T cells home back to the plaque (arrows), some via CC-chemokine receptor 5 (CCR5)-mediated trafficking. Over time, arterial tertiary lymphatic organs form in the adventitia, wherein APCs encounter naive and antigen-experienced B cells and T cells. T cell clonal expansion is more extensive in arterial tertiary lymphoid organs (ATLOs) than in lymph nodes, suggesting that ATLOs may provide a ‘shortcut’ that accelerates autoimmunity in atherosclerosis. Some germinal centre B cells become plasma cells and secrete antibodies to atherosclerosis-associated antigens including oxidized LDL (not shown).
Failure of self-tolerance in atherosclerosis
Normally, autoimmune T cell responses are kept in check owing to central and peripheral tolerance mechanisms (see Boxes 1 and 2). Failure of peripheral T cell tolerance in atherosclerosis was first formally shown in studies by Wang et al.11 and Depuydt et al.12, both published in 2023. The main findings of these studies were that several factors were associated with this failure of peripheral T cell tolerance: (1) defective immune checkpoint expression, (2) clonal expansion of CD4+ T cell, CD8+ T cell and regulatory T (Treg) cell populations, (3) T cell exhaustion, (4) the probable conversion of Treg cells to T helper 17 (TH17) cells, and (5) dysfunctional antigen presentation11,12. Both of these recent studies have used scRNA-seq with TCR-seq, in which TCRα and TCRβ sequences are matched with transcriptomes in the same cells. In a published mouse study11, the highest degree of plaque-specific clonal T cell expansion was seen in effector CD8+ T cells, and these clonally expanded T cells expressed genes such as Cd69, Fos and Fosb, indicative of recent TCR engagement11. Similar observations were made in a very recent study of human CD8+ T cells in subjects with coronary artery disease28. In the mouse study11, T cell transcriptomes with paired TCRα and TCRβ sequences were assessed by scRNA-seq in aorta-draining renal lymph nodes (RLNs) of healthy or atherosclerotic mice, arterial tertiary lymphoid organs (ATLOs) and atherosclerotic plaques11. scRNA-Seq with TCR-Seq also revealed transcriptomes of ApoB-tetramer-specific CD4+ T cells in humans24. There were progressive changes from wild-type RLNs to Apoe−/− RLNs, ATLOs and atherosclerotic plaques, reflected in a notable reduction in the proportions of Treg cells and naive CD4+ T cells, coupled with a significant increase in the proportions of effector memory (TEM) and central memory (TCM) CD8+ T cells. The majority of TCRα and TCRβ CDR3 sequences observed in CD8+ T cells within atherosclerotic plaques shared patterns with those seen in CD8+ T cell populations expanded in the aorta-draining RLNs of Apoe−/− mice, but not in the RLNs of wild-type mice. Single-cell TCRα and TCRβ CDR3 sequences showed clonal expansion, which progressively increased from wild-type RLNs to Apoe−/− RLNs to ATLOs and finally to plaques.
Box 1. T cell development and central tolerance.
T cell development in the thymus
T cells develop from bone marrow-derived progenitors that home to the thymus and undergo a complex and irreversible differentiation process67. Following T cell lineage commitment and population expansion, T cell receptor (TCR) gene rearrangement occurs, leading to the development of either γδ or αβ T cell progenitors. At this stage, all T cell precursors are double-negative (DN) for the expression of CD4 and CD8 (ref. 67). The somatic recombination of TCR V, D and J genes forms a highly diverse array of unique TCRs with stochastic specificities. The fate of αβ T cell progenitors is determined by the affinity of their TCR for self-peptides presented by MHC class I or MHC class II molecules67. Most αβ TCR+ DN thymocytes fail positive selection and die by apoptosis. Other αβ TCR+ DN thymocytes pass positive selection67, proliferate and generate a large population of CD4+CD8+ double-positive (DP) T cells67. During this maturation, many T cells are eliminated by negative selection. Negative selection eliminates cells whose TCR has high affinity for self-peptide–MHC complexes. Cells with lower-affinity TCRs pass negative selection, which is the mechanism for central tolerance67,68.
Central tolerance
One of the most important mechanisms of central tolerance in the thymus is the negative selection, also called clonal deletion, of immature T cells that bear high-affinity TCRs against self-antigens69. However, negative selection is incomplete70–75. A second layer of central tolerance, known as clonal diversion, ensures that self-reactive T cells that survive past negative selection are imprinted with a regulatory phenotype64,65. These thymic-derived regulatory T (tTreg) cells suppress T cell activation and effector function in the periphery76. tTreg cells are thought to express TCRs with intermediate affinity for self-epitopes. Both negative selection and clonal diversion depend upon the recognition of self-peptide–MHC complexes displayed on thymic antigen-presenting cells, particularly the medullary thymic epithelial cells that specialize in the ectopic expression of many antigens in the thymus77,78. Insufficient expression or presentation of self-antigens in the thymus and engagement of TCRs with low affinity and avidity allow the developing T cells to escape central tolerance.
Box 2. Mechanisms of peripheral tolerance.
Mature autoreactive T cells that escape thymic negative selection79 are restrained by peripheral tolerance mechanisms37,67, namely, ignorance80, anergy81, expression of inhibitory molecules82 and active suppression by regulatory T (Treg) cells83.
Ignorance
When self-antigens are exclusively expressed in immune-privileged sites, such as the brain or the eye, they are effectively hidden from circulating immune cells by anatomical barriers72,80,84. When the levels of expression or presentation of these self-antigens in peripheral tissue and lymphoid organs are too low to trigger T cell activation, autoreactive T cells against these self-antigens are maintained in a quiescent state. Hidden and low-expressed self-antigens are ignored by the immune system72,80,84. However, antigen-inexperienced naive T cells against such self-antigens can be activated when sufficient quantities of cognate antigen are presented to them by activated antigen-presenting cells (APCs). This can happen under conditions of tissue damage and chronic inflammation85–87, which is relevant to atherosclerosis.
Anergy
T cell anergy occurs when naive T cells encounter self-antigens in the absence of proper co-stimulation88. They enter a state of hypo-responsiveness characterized by low IL-2 production and poor proliferation89. This anergic state results from improper regulation of T cell receptor (TCR) proximal signalling events (for example, excessive calcium–NFAT signalling and defective mTOR and RAS–MAPK activation) and involves active gene silencing, for example, at the IL-2 locus, as well as transcriptional upregulation of anergy-specific genes (for example, GRAIL and Cbl-b)81. T cell anergy can be reversed by cytokines such as IL-2 and IL-15 that induce proliferation and cell survival86,90.
Inhibitory molecules
Activation-dependent upregulation of co-inhibitory checkpoint molecules such as CTLA4 and PD1 help to control excessive autoreactive T cell responses91. CTLA4 predominantly controls naive T cell activation in lymphoid organs92,93, whereas PD1 primarily exerts its regulatory effect during recall responses in peripheral tissues94. CTLA4 interacts with CD80 and CD86 on APCs and blocks co-stimulation from CD28 (ref. 95). CTLA4 inhibits T cell activation by triggering T cell anergy96. PD1 ligands PDL1 and PDL2 are constitutively expressed on APCs and are induced in non-immune cells in inflamed tissues97. PD1 limits ongoing effector T cell activation and proliferation by inducing a dysfunctional state known as T cell exhaustion98. Blocking PD1 or CTLA4 can overcome this, a mechanism used to enhance cancer immunotherapy. In the context of atherosclerosis, such as in autoimmune diseases, PD1 and CTLA4 are probably beneficial and dampen disease progression99,100.
Regulatory T cells
The principal mechanism of peripheral tolerance is based on suppression of effector T cell responses by Treg cells. Treg cells express the transcription factor FOXP3 and the high-affinity IL-2 receptor CD25 but do not express the IL-7 receptor CD127 (refs. 101,102). They exert their suppressive effects through various mechanisms, including the secretion of immunosuppressive cytokines such as IL-10 and transforming growth factor-β (TGFβ), direct cell–cell contact inhibition, and modulation of APCs103,104. These mechanisms collectively work to maintain immune tolerance and prevent autoimmunity. Treg cells exhibit considerable heterogeneity and have tissue-specific, characteristic transcriptomes105,106. Most Treg cells originate in the thymus (tTreg cells) during T cell development107. Peripherally induced regulatory T cells (pTreg cells) develop from conventional CD4+ T cells in the periphery in response to cytokine signals, specifically TGFβ and retinoic acid103,108. Both tTreg cells and pTreg cells display a broad range of antigen specificities including many self-antigens.
Treg cells are not naive. Their cell surface phenotype and transcriptome suggest that they repeatedly engage their TCRs. At steady state, Treg cells proliferate in vivo29,109. Treg cells increase in number during inflammation, a mechanism that avoids excessive inflammation associated with disease. However, Tregcells are not stable. In the presence of repeated TCR engagement and inflammatory stimuli, Treg cells become ‘exTreg’ cells40. Such exTreg cells can express either T-bet and acquire a T helper 1 (TH1)-like transcriptome110,111, express RORγt and acquire a TH17-like transcriptome40, express GATA3 and acquire a TH2-like transcriptome112,113, express BCL-6 and acquire a follicular helper T (TFH) cell-like phenotype32, or express CD16 and CD56 on the cell surface and become cytotoxic exTreg cells29.
Several defects in the expression of immune checkpoint molecules and other immune receptors were previously reported11, but none have been validated so far. There was indirect evidence for Treg cells in ATLOs acquiring a TH17-like phenotype, a finding that needs to be confirmed by Treg cell lineage tracking11. Breakdown of tolerance was also seen in scRNA-Seq data from human coronary and carotid artery plaques11, which was associated with a strong cytotoxic gene signature including CST1, GZMB, GZMK, GZMM, NKG7 and PRF1 (ref. 12).
In a recent human carotid plaque study12, clonal expansion of effector CD4+ and CD8+ T cells was reported in the plaque, with evidence for recent TCR engagement supported by expression of CD69, FOS and FOSB (ref. 12). Of particular interest was the expansion of TEMRA (terminally differentiated effector memory cells re-expressing CD45RA) clusters, which are a type of T cells that express CD45RA and are thought to be terminally differentiated. These cells expressed GZMB, PRF1 and NKG7 and lacked CD27 and CD28 (ref. 12). Another TEMRA cluster expressed KLRD1, KLRG1 and FCGR3A (ref. 12), the gene encoding CD16. We recently identified this signature as indicative of terminally differentiated exTreg cells29. The percentage of large (1 to 10%) and medium (0.1 to 1%) clones was higher in plaques than in peripheral blood mononuclear cells (PBMCs). One CD8+ T cell clone that was very highly expanded in PBMCs (>10%) had a TCRα sequence known to be specific for cytomegalovirus and showed no evidence of recent TCR engagement12. On the basis on these findings, the authors proposed atherosclerosis as an ‘autoimmune-like disease’12. In 2018, we suggested that atherosclerosis is “a chronic inflammatory disease with an autoimmune component”4. The idea that atherosclerosis is an autoimmune disease has previously been discussed23. Previous evidence was limited to the presence of autoantibodies to (modified) LDL9. A recent study has provided evidence of clonal T cell expansion in plaque and ATLOs11.
Mechanisms for tolerance breakdown in atherosclerosis
The mechanisms involved in breaking tolerance to self-antigens in the setting of atherosclerosis are unknown. However, potential candidate mechanisms include Treg cell instability and dysfunction or the activation of self-reactive naive T cells.
Treg cell instability in atherosclerosis
Treg cell instability has been observed in the context of atherosclerosis, whereby Treg cells convert to pro-inflammatory exTreg cell phenotypes21,24. Such exTreg cells have been described in many mouse studies of atherosclerosis30,31. Three early studies have provided evidence that Treg cells acquire a TH1-like gene expression signature30,31 that includes expression of the chemokine receptor CCR5, a receptor that was shown to promote CD4+ T cell homing to atherosclerotic arteries in vivo31. In another mouse atherosclerosis study, Treg cells were shown to switch to a TFH cell-like phenotype32. In a study of patients with atherosclerosis, ApoB-specific T cells were shown to express FOXP3 and RORγt or FOXP3 and T-bet, assessed at the protein level by flow cytometry33.
In a recent study, we mapped exTreg cell transcriptomes from the Apoe−/− mouse model of atherosclerosis — obtained from sorting CD4+ T cells from Treg lineage tracker mice onto human scRNA-seq data — and showed that human exTreg cells express CD16, CD56 and an array of cytotoxic genes29. These exTreg cells cannot regulate effector T cell proliferation. Instead, they kill target cells with remarkable efficiency, comparable to killing mediated by CD8+ cytotoxic T lymphocytes (CTLs)29. Thus, it appears that exTreg cells in atherosclerosis exist in multiple versions. Some still express FOXP3 and acquire additional TH1-like, TH17-like or TFH-like gene signatures34. The near-complete loss of FOXP3 in exTreg cells reported previously29 suggests that these cells may be the most terminally differentiated exTreg cells. The breakdown of tolerance to self may be related to the conversion of Treg cells to exTreg cells because many Treg cells express TCRs specific for self-epitopes (Fig. 2).
Fig. 2 |. Conversion of regulatory T cells in atherosclerosis.
FOXP3+CD25+CD127− regulatory T (Treg) cells (blue) can convert into at least four types of ‘exTreg’ cells in atherosclerosis. In studies of the Apoe−/− mouse model, T helper 1 (TH1)-likeTreg cells (yellow) have been described with a FOXP3lowCD25− T-bet+ CCR5+ phenotype30,31. Another study has shown that Treg cells become increasingly TH17-like (orange) in response to a western diet21. A similar T cell phenotype (FOXP3+RORγt+ TH17-like) was shown by flow cytometry in patients with atherosclerosis33. In Apoe−/− mice, some exTreg cells can acquire a TFH-like phenotype (pink) and express CXCR5, PD1, BCL-6 and ICOS32. In humans and mice with atherosclerosis, terminally differentiated cytotoxic exTreg cells (red) express very low levels of FOXP3 and no CD25, but they acquire expression of CCL3, CCL4 and CCL5, TBX21, and a cytotoxic signature including NKG7 (ref. 29). The mechanisms that drive the conversion of Treg cells to exTreg cells are still unknown.
The mechanisms leading to Treg cell instability in the context of atherosclerosis remain to be defined, but previous studies have shown that chronic antigen exposure, pro-inflammatory signalling and metabolic reprogramming can destabilize Treg cells (see Box 3).
Box 3. What drives regulatory T cell instability?
Several lines of evidence suggest that chronic antigen exposure, pro-inflammatory signalling and metabolic reprogramming at inflamed tissue sites work in concert to cause regulatory (Treg)cell instability47,114,115.
Dysregulation of the FOXP3 locus
A stable Treg cell programme is maintained both by Foxp3 expression and regulation of its target genes, as well as by preserving a hypomethylated or demethylated epigenetic landscape at specific chromatin sites116. Treg cell-specific deletion of TET2 and TET3 proteins that normally help to maintain demethylated chromatin states in the Foxp3 upstream enhancer region triggers loss of Foxp3 expression and Treg cell instability117,118. Treg cells activated in the absence of the chromatin-modifying enzyme EZH2 were unable to resolve inflammation at tissue sites, thereby leading to multi-organ autoimmunity119.
Prolonged or unregulated TCR signalling
Human and mouse Treg cells proliferate in vivo29. Treg cell maintenance in the periphery requires a basal level of continuous activation through TCR-mediated, IL-2R-mediated and CD28-mediated co-stimulation that induces a Treg cell-specific suppressive gene programme and a permissive epigenetic landscape120. However, prolonged engagement of TCR–CD28 signalling complexes121–123 can cause hyperactivation of the PI3K–AKT–mTOR pathways122. This can trigger chromatin modifications at the Foxp3 locus110,122. Treg cells that are deficient in NRP1 (ref. 124) or PTEN125 are more susceptible to pathogenic conversion. NRP1 deficiency makes Treg cells lose their suppressive capacity, although FOXP3 expression remains intact60. These cells express TH1 cell-associated markers such as IFNγ, T-bet, CXCR3 and Eomes60. PTEN deletion in Treg cells also drives instability and loss of function via CD25 and FOXP3 downregulation and increased production of IFNγ, resulting in systemic lupus-like autoimmunity126. Downregulation of the transcription factor Eos reprogrammes Treg cells into FOXP3+ helper-like T cells, called Eos-labile Treg cells127.
Toll-like receptor signalling
Tol-like receptor (TLR) signalling can promote autoimmune conditions85 and TLR engagement has been shown to block the suppressive action of CD4+CD25+ Treg cells128. TCR engagement and TLR signalling can mediate Treg cell instability by activating common downstream pathways. One example is autoantigen-mediated TCR stimulation combined with TLR activation by LPS, which can destabilize Treg cells by strongly activating the MEK–ERK pathway129. Increased glucose uptake and glycolytic activity impairs the FOXP3-dependent suppressive programme of Treg cells130.
Pro-inflammatory cytokine signalling
Pro-inflammatory cytokines may also contribute to destabilizing Treg cells. In the inflamed synovial joints of patients with rheumatoid arthritis, tumour necrosis factor (TNF) upregulated the expression and activity of protein phosphatase 1 (PP1), which specifically dephosphorylated the Ser418 residue on FOXP3 in Treg cells131. This compromised the transcriptional activity of FOXP3 and converted the Treg cell-suppressive programme into an effector programme, characterized by IL-17 and IFNγ expression131. FOXP3 phosphorylation and Treg cell suppressive function were restored by treating patients with anti-TNF therapy (infliximab)131. IL-12 was shown to promote TH1-like Treg cells44,45 (Fig. 2). Mechanistically, this was mediated by enhanced phosphorylation of AKT and its downstream targets FOXO1 and FOXO3, which triggered IFNγ production and loss of suppressive ability132. IL-12, together with a Treg cell-intrinsic hyperactive Notch signalling pathway, led to STAT4 phosphorylation and IFNγ production133. IL-1β can also reprogramme Treg cells into IL-17-producing effector cells134,135, and treatment of mouse and human Treg cells with IFNγ resulted in a loss of their suppressor programme60. Treg cells from inflamed tissues, such as the tumour microenvironment, were shown to be more susceptible to loss of suppressive functions owing to their increased expression of IFNGR1 (ref. 60), which is directly relevant to Treg cell instability induced by cytokine signalling.
TGFβ, retinoic acid and IDO protect Treg cell stability
The destabilizing effects of prolonged TCR activation and pro-inflammatory cytokines can be counterbalanced by Treg cell-promoting factors such as TGFβ121 and all-trans acid136. Indoleamine-pyrrole 2,3-dioxygenase (IDO), produced by a tolerogenic subset of activated plasmacytoid DCs, can also block Treg cell plasticity127. In IDO-deficient mice, ligation of TLR9 by CpG, particularly on plasmacytoid DCs, triggered IL-6 production that caused reprogramming of Treg cells into TH17-like exTreg cells137. Immunization of mice in the presence of IDO blockade converted Treg cells into IL-17-producing and IL-22-producing non-suppressive effector T cells138. Most of them also co-expressed IL-2, CD40L and TNF127,138,139.
Regulatory T cell dysfunction
Although Treg cells are known to be stable under homoeostatic conditions35, several recent reports have shown Treg cell dysfunction36 and instability in many autoimmune diseases37,38. Fate-mapping experiments in mouse models have confirmed that Treg cells downregulate Foxp3 during severe lymphopenia39, in experimentally induced autoimmune encephalomyelitis40, in collagen-induced autoimmune arthritis41, in type 1 diabetes42 and in atherosclerosis29. These exTreg cells produce effector cytokines such as IL-2 (ref. 39), IFNγ (refs. 40,42) and IL-17 (ref. 41), and when adoptively transferred to naive mice, they trigger tissue damage and promote disease39–42. IL-17A+ FOXP3+ human Treg cells are present in the skin lesions of patients with psoriasis43 and in the synovium of patients with rheumatoid arthritis41. Significantly higher frequencies of IFNγ+ FOXP3+ Treg cells were also reported in patients with relapsing remitting multiple sclerosis44 or with type 1 diabetes45 than in healthy controls. Chronic tissue inflammation and antigen-driven T cell activation, which are hallmarks of these diseases46, rewire Treg cells and drive the conversion of their suppressive programme to a pathogenic one47,48. In summary, Treg cells appear to become dysfunctional in almost all autoimmune disease models that have been studied.
In mouse models of atherosclerosis, fate-mapping studies demonstrated the accumulation of T-bet+ TH1-like30,31 and BCL-6+ TFH-like32 exTreg cells34, particularly in the inflamed aortic wall and in organ-draining lymph nodes. Transcriptomic analysis of Treg and exTreg cells from the spleen and lymph nodes of 20-week-old atherosclerotic mice revealed an upregulation of cytotoxic molecules, such as granzymes and perforin, in the exTreg cells, and similar results were observed in exTreg cells from patients with atherosclerosis29. In all these studies29–32, exTreg cells were reported to have compromised suppressive capacity. Instead, they acquired effector programmes such as cytotoxicity29 and secretion of inflammatory cytokines such as TNF and IFNγ24,29–31. Consistent with previous observations that downregulation of CD25 has an important role in Treg cell instability49, exTreg cells in atherosclerosis displayed low levels of CD25 expression29–32. In most cases, this was accompanied by a concomitant loss in Foxp3 expression29,31,32. Both human29 and mouse30,31 exTreg cells expressed pro-inflammatory chemokine receptors such as CXCR3 and CCR5. Adoptive transfer of the reprogrammed Treg cells promoted lesion formation and systemic IFNγ production in atherosclerotic mice24,31, suggesting that pathogenic conversion of Treg cells to exTreg cells in atherosclerosis may have a causal association with disease progression.
Antigen-mediated activation of T cells has been suggested to be a driver of Treg cell instability in mouse models of multiple sclerosis40 and autoimmune arthritis41. TCR activation seems to be necessary but not sufficient for Treg cells to covert to exTreg cells. An inflammatory microenvironment also has a pivotal role as >90% of adoptively transferred Treg cells remained stable in the absence of an inflammatory setting in a mouse model of graft-versus-host disease50. Therefore, it is probable that both TCR-mediated antigen-specific T cell activation and a local inflammatory milieu promote autoreactivity in atherosclerosis51.
RNA sequencing of mouse CD4+ T cells specific for the atherosclerosis-associated autoantigen ApoB revealed lower expression of Treg cell-associated genes — such as Foxp3, Il2ra, Ctla4 and Il10 — in ApoB-specific CD4+ T cells than in non-ApoB-specific bulk CD4+ T cells21. Instead, ApoB-specific T cells were enriched in genes encoding transcription factors, cytokines and chemokine receptors typically associated with TH17, TH1 and TFH cells including Rora, Tbx21, Bcl6, Il17a, Il21, Ifng, Cxcr3 and Cxcr5 (ref. 21). Overall, the transcriptomes of mouse ApoB-tetramer+ cells21 resembled those of the CCR5+FOXP3+T-bet+ reprogrammed Treg cells that were previously described31. Thus, atherosclerosis-associated antigen-specific Treg cells may be more plastic than other Treg cells. Within the ApoB-tetramer+ subset, T cells that expressed FOXP3, but not other transcription factors associated with effector T cell responses, were no longer detectable by flow cytometry in old (more than 20 weeks old) atherosclerotic Apoe-deficient mice21. Human ApoB-reactive CD4+ T cells from patients with cardiovascular disease also exhibited heightened co-expression of the TH1 and TH17 lineage-defining transcription factors T-bet and RORγt in FOXP3+ CD4+ T cells33. Their transcriptomes displayed signatures of effector and memory T cells, rather than of Treg cells24. Indeed, single-cell sequencing of atherosclerotic tissue samples have shown that clonally expanded populations of pro-inflammatory effector and memory T cell subsets accumulate in mouse11,52 and human12,53,54 lesions. In humans12,53 and mice11, T cells found in plaques expressed the genes encoding the pro-inflammatory chemokine receptors CCR5, CXCR6 and CX3CR1, which have an important role in T cell infiltration into inflamed aortas31,55. All three chemokine receptors were shown to be expressed at the protein level29. All three receptors are known to be causally involved in atherosclerosis, based on knockout studies31,55,56. CCR5 is involved in CD4+ T cell homing to atherosclerotic aorta31 and CXCR6 was also shown to be involved in T cell homing55. CCR5, CXCR6 and CX3CR1 are also expressed on human and mouse exTreg cells29–31.
MHC class II-dependent interaction between Treg cells and activated APCs can trigger Treg cell reprogramming, requiring TCR engagement and an inflammatory environment57. Pro-inflammatory APCs, such as dendritic cells, that accumulate in progressing atherosclerotic plaque can drive Treg cell activation and tune their phenotypes by expressing a diverse array of co-stimulatory molecules, cytokines and chemokines58. CCL17 secreted by a subset of dendritic cell has a vital part in progression of atherosclerosis by recruiting inflammatory T cells to lesion sites and hindering Treg cell maintenance and expansion in lymphatic tissue59.
RNA-seq analysis of mouse ApoB+ CD4+ T cells from young (8 weeks) versus old (20 weeks) Apoe−/− mice showed activation of inflammatory signalling pathways in autoreactive T cells from old atherosclerotic mice21. Specific enrichment of CCR5 (refs. 30,31) and IL-6Rα32 on the surface of exTreg cells in mice suggests that these cells have acquired increased responsiveness to pro-inflammatory ligands such as CCL5 and IL-6. Mouse11 and human53 plaque CD4+ T cells were also enriched in the receptor for the IFNγ receptor IFNGR1, whose expression has been shown to drive loss of suppressive function of Treg cells in a tumour microenvironment60.
A recent study has reported that inflammatory DCs are the predominant DC type found in in atherosclerotic plaque tissue in mouse models of the disease, whereas tolerogenic DCs dominated in lymph nodes11. Human carotid artery plaque macrophages from patients with symptomatic disease also exhibit higher expression of CCL5 than those from the asymptomatic group53. These and other studies52,61–63 have shown that DC and macrophage subsets in atherosclerotic plaques express inflammatory molecules such as TLR2, IL1B, CCL3, CCL4 and TNF, all of which could contribute to the loss of T cell tolerance within the plaque microenvironment49. As such, these are candidate pathways for mediating the loss of tolerance in atherosclerosis and warrant further investigation. It is also possible that breakdown of tolerance may be achieved by aberrant activation of naive T cells, resulting in clonal expansion and a possibly aggressive, pro-inflammatory effector phenotype (Fig. 3).
Fig. 3 |. Activation and expansion of naive T cells.
Dendritic cells (DCs) derived from plaques may enter draining lymph nodes through afferent lymphatics and present self-antigens on MHC class I (MHC-I) or MHC-II molecules to T cells in the context of co-stimulation. This can initiate the activation and clonal expansion of naive CD8+ or CD4+ T cells. These cells may acquire effector phenotypes, leave the lymph node through efferent lymphatics, and enter the circulation, homing back to atherosclerotic arteries26,31. Whether such newly activated T cells acquire their effector programme at the site of initial antigen encounter (that is, in the draining lymph node) or in the peripheral (plaque) tissue is not known66. HEV, high endothelial venule; TCR, T cell receptor.
However, there is currently no evidence for this mechanism being operative in atherosclerosis. As such, whether the autoimmune component of atherosclerosis is based on exTreg cells or on self-reactive effector T cells, or even both, remains to be clarified.
Future directions
Currently, it is not known how the different subpopulations of exTreg cells (namely TH1-like, TH17-like, TFH-like and cytotoxic exTreg cells) are related to each other. The trigger responsible for converting Treg cells that are specific for atherosclerosis-associated autoantigens into exTreg cells is also unknown. It is not even clear whether there is one triggering pathway or whether multiple pathways interact. On the APC side, it is possible that the migration or function of tolerogenic DCs may be defective in atherosclerosis64, and there may be atherosclerosis-associated factors that result in fewer to lerogenic APCs and more pro-inflammatory APCs. Discovering the factors that promote the shift of Treg cells to exTreg cells and tolerogenic APCs to inflammatory APCs will be of paramount importance for designing possible immune-based therapeutic or preventative strategies for atherosclerosis. Capitalizing on these insights may enable the development of tolerogenic vaccines to restore tolerance to self, an approach that has shown promise in mouse models of atherosclerosis65.
Footnotes
Competing interests
K.L. is a co-founder of Atherovax, a biotech company developing a tolerogenic vaccine for atherosclerosis. The other authors declare no competing interests.
Additional information
Peer review information Nature Reviews Immunology thanks C. Weber, J. Nilsson and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
References
- 1.Wolf D & Ley K Immunity and inflammation in atherosclerosis. Circ. Res. 124, 315–327 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tsao CW et al. Heart Disease and Stroke Statistics — 2023 update: a report from the American Heart Association. Circulation 147, e93–e621 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Michos ED, McEvoy JW & Blumenthal RS Lipid management for the prevention of atherosclerotic cardiovascular disease. N. Engl. J. Med. 381, 1557–1567 (2019). [DOI] [PubMed] [Google Scholar]
- 4.Kobiyama K & Ley K Atherosclerosis. Circ. Res. 123, 1118–1120 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ridker PM et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N. Engl. J. Med. 359, 2195–2207 (2008). [DOI] [PubMed] [Google Scholar]
- 6.Ridker PM et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–1131 (2017). [DOI] [PubMed] [Google Scholar]
- 7.Tardif JC et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N. Engl. J. Med. 381, 2497–2505 (2019). [DOI] [PubMed] [Google Scholar]
- 8.Nidorf SM, Eikelboom JW, Budgeon CA & Thompson PL Low-dose colchicine for secondary prevention of cardiovascular disease. J. Am. Coll. Cardiol. 61, 404–410 (2013). [DOI] [PubMed] [Google Scholar]
- 9.Tsimikas S, Palinski W & Witztum JL Circulating autoantibodies to oxidized LDL correlate with arterial accumulation and depletion of oxidized LDL in LDL receptor-deficient mice. Arterioscler. Thromb. Vasc. Biol. 21, 95–100 (2001). [DOI] [PubMed] [Google Scholar]
- 10.Tsimikas S et al. Relationship of IgG and IgM autoantibodies to oxidized low density lipoprotein with coronary artery disease and cardiovascular events. J. Lipid Res. 48, 425–433 (2007). [DOI] [PubMed] [Google Scholar]
- 11.Wang Z et al. Pairing of single-cell RNA analysis and T cell antigen receptor profiling indicates breakdown of T cell tolerance checkpoints in atherosclerosis. Nat. Cardiovasc. Res. 2, 290–306 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Depuydt MAC et al. Single-cell T cell receptor sequencing of paired human atherosclerotic plaques and blood reveals autoimmune-like features of expanded effector T cells. Nat. Cardiovasc. Res. 2, 112–125 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Saigusa R, Winkels H & Ley K T cell subsets and functions in atherosclerosis. Nat. Rev. Cardiol. 17, 387–401 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Che J et al. Serum autoantibodies against human oxidized low-density lipoproteins are inversely associated with severity of coronary stenotic lesions calculated by Gensini score. Cardiol. J. 18, 364–370 (2011). [PubMed] [Google Scholar]
- 15.Gruzdeva O et al. Multivessel coronary artery disease, free fatty acids, oxidized LDL and its antibody in myocardial infarction. Lipids Health Dis. 13, 111 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chen Q et al. Association of anti-oxidized LDL and candidate genes with severity of coronary stenosis in the Women’s Ischemia Syndrome Evaluation study. J. Lipid Res. 52, 801–807 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Garrido-Sánchez L, Chinchurreta P, García-Fuentes E, Mora M & Tinahones FJ A higher level of IgM anti-oxidized LDL antibodies is associated with a lower severity of coronary atherosclerosis in patients on statins. Int. J. Cardiol. 145, 263–264 (2010). [DOI] [PubMed] [Google Scholar]
- 18.Soto Y et al. Autoantibodies to oxidized low density lipoprotein in relation with coronary artery disease. Hum. Antib. 18, 109–117 (2009). [DOI] [PubMed] [Google Scholar]
- 19.van den Berg VJ et al. IgM anti-malondialdehyde low density lipoprotein antibody levels indicate coronary heart disease and necrotic core characteristics in the Nordic Diltiazem (NORDIL) study and the Integrated Imaging and Biomarker Study 3 (IBIS-3). EBioMedicine 36, 63–72 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Que X et al. Oxidized phospholipids are proinflammatory and proatherogenic in hypercholesterolaemic mice. Nature 558, 301–306 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wolf D et al. Pathogenic autoimmunity in atherosclerosis evolves from initially protective apolipoprotein B100-reactive CD4+ T-regulatory cells. Circulation 142, 1279–1293 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Roy P et al. Immunodominant MHC-II (major histocompatibility complex II) restricted epitopes in human apolipoprotein B. Circ. Res. 131, 258–276 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wick G, Knoflach M & Xu Q Autoimmune and inflammatory mechanisms in atherosclerosis. Annu. Rev. Immunol. 22, 361–403 (2004). [DOI] [PubMed] [Google Scholar]
- 24.Saigusa R et al. Single cell transcriptomics and TCR reconstruction reveal CD4 T cell response to MHC-II-restricted APOB epitope in human cardiovascular disease. Nat. Cardiovasc. Res. 1, 462–475 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bentzon JF, Otsuka F, Virmani R & Falk E Mechanisms of plaque formation and rupture. Circ. Res. 114, 1852–1866 (2014). [DOI] [PubMed] [Google Scholar]
- 26.Koltsova EK et al. Dynamic T cell-APC interactions sustain chronic inflammation in atherosclerosis. J. Clin. Invest. 122, 3114–3126 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Martel C et al. Lymphatic vasculature mediates macrophage reverse cholesterol transport in mice. J. Clin. Invest. 123, 1571–1579 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Iqneibi S et al. Single cell transcriptomics reveals recent CD8T cell receptor signaling in patients with coronary artery disease. Front. Immunol. 14, 1239148 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Freuchet A et al. Identification of human exTreg cells as CD16+CD56+ cytotoxic CD4+ T cells. Nat. Immunol. 24, 1748–1761 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Butcher MJ et al. Atherosclerosis-driven Treg plasticity results in formation of a dysfunctional subset of plastic IFNγ+ Th1/Tregs. Circ. Res. 119, 1190–1203 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Li J et al. CCR5+T-bet+FoxP3+ effector CD4 T cells drive atherosclerosis. Circ. Res. 118, 1540–1552 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gaddis DE et al. Apolipoprotein AI prevents regulatory to follicular helper T cell switching during atherosclerosis. Nat. Commun. 9, 1095 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kimura T et al. Regulatory CD4+ T cells recognize major histocompatibility complex class II molecule-restricted peptide epitopes of apolipoprotein B. Circulation 138, 1130–1143 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ali AJ, Makings J & Ley K Regulatory T cell stability and plasticity in atherosclerosis. Cells 9, 2665 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rubtsov YP et al. Stability of the regulatory T cell lineage in vivo. Science 329, 1667–1671 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Korn T et al. Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation. Nat. Med. 13, 423–431 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Theofilopoulos AN, Kono DH & Baccala R The multiple pathways to autoimmunity. Nat. Immunol. 18, 716–724 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Buckner JH Mechanisms of impaired regulation by CD4+CD25+FOXP3+ regulatory T cells in human autoimmune diseases. Nat. Rev. Immunol. 10, 849–859 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Duarte JH, Zelenay S, Bergman M-L, Martins AC & Demengeot J Natural Treg cells spontaneously differentiate into pathogenic helper cells in lymphopenic conditions. Eur. J. Immunol. 39, 948–955 (2009). [DOI] [PubMed] [Google Scholar]
- 40.Bailey-Bucktrout SL et al. Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response. Immunity 39, 949–962 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Komatsu N et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat. Med. 20, 62–68 (2014). [DOI] [PubMed] [Google Scholar]
- 42.Zhou X et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat. Immunol. 10, 1000–1007 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bovenschen HJ et al. Foxp3+ regulatory T cells of psoriasis patients easily differentiate into IL-17A-producing cells and are found in lesional skin. J. Invest. Dermatol. 131, 1853–1860 (2011). [DOI] [PubMed] [Google Scholar]
- 44.Dominguez-Villar M, Baecher-Allan CM & Hafler DA Identification of T helper type 1-like, Foxp3+ regulatory T cells in human autoimmune disease. Nat. Med. 17, 673–675 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.McClymont SA et al. Plasticity of human regulatory T cells in healthy subjects and patients with type 1 diabetes. J. Immunol. 186, 3918–3926 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rosenblum MD, Remedios KA & Abbas AK Mechanisms of human autoimmunity. J. Clin. Invest. 125, 2228–2233 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Barbi J, Pardoll D & Pan F Treg functional stability and its responsiveness to the microenvironment. Immunol. Rev. 259, 115–139 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Guo J & Zhou X Regulatory T cells turn pathogenic. Cell Mol. Immunol. 12, 525–532 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chinen T et al. An essential role for the IL-2 receptor in Treg cell function. Nat. Immunol. 17, 1322–1333 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Laurence A et al. STAT3 transcription factor promotes instability of nTreg cells and limits generation of iTreg cells during acute murine graft-versus-host disease. Immunity 37, 209–222 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Edsfeldt A & Nilsson J Understanding autoimmunity in atherosclerosis paves the way for novel therapies. Nat. Cardiovasc. Res. 2, 227–229 (2023). [DOI] [PubMed] [Google Scholar]
- 52.Winkels H et al. Atlas of the immune cell repertoire in mouse atherosclerosis defined by single-cell RNA-sequencing and mass cytometry. Circ. Res. 122, 1675–1688 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Fernandez DM et al. Single-cell immune landscape of human atherosclerotic plaques. Nat. Med. 25, 1576–1588 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chowdhury RR et al. Human coronary plaque T cells are clonal and cross-react to virus and self. Circ. Res. 130, 1510–1530 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Galkina E et al. CXCR6 promotes atherosclerosis by supporting T-cell homing, interferon-γ production, and macrophage accumulation in the aortic wall. Circulation 116, 1801–1811 (2007). [DOI] [PubMed] [Google Scholar]
- 56.Combadière C et al. Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double knockout mice. Circulation 107, 1009–1016 (2003). [DOI] [PubMed] [Google Scholar]
- 57.Munn DH, Sharma MD & Johnson TS Treg destabilization and reprogramming: implications for cancer immunotherapy. Cancer Res. 78, 5191–5199 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Roy P, Orecchioni M & Ley K How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat. Rev. Immunol. 22, 251–265 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Weber C et al. CCL17-expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice. J. Clin. Invest. 121, 2898–2910 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Overacre-Delgoffe AE et al. Interferon-γ drives Treg fragility to promote anti-tumor immunity. Cell 169, 1130–1141.e11 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Depuydt MAC et al. Microanatomy of the human atherosclerotic plaque by single-cell transcriptomics. Circ. Res. 127, 1437–1455 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cole JE et al. Immune cell census in murine atherosclerosis: cytometry by time of flight illuminates vascular myeloid cell diversity. Cardiovasc. Res. 114, 1360–1371 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kim K et al. Transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models. Circ. Res. 123, 1127–1142 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Baratin M et al. Homeostatic NF-κB signaling in steady-state migratory dendritic cells regulates immune homeostasis and tolerance. Immunity 42, 627–639 (2015). [DOI] [PubMed] [Google Scholar]
- 65.Roy P, Ali AJ, Kobiyama K, Ghosheh Y & Ley K Opportunities for an atherosclerosis vaccine: from mice to humans. Vaccine 38, 4495–4506 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ley K The second touch hypothesis: T cell activation, homing and polarization. F1000Research 3, 37 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Xing Y & Hogquist KA T-cell tolerance: central and peripheral. Cold Spring Harb. Perspect. Biol. 4, a006957 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bluestone JA Mechanisms of tolerance. Immunol. Rev. 241, 5–19 (2011). [DOI] [PubMed] [Google Scholar]
- 69.Wirnsberger G, Hinterberger M & Klein L Regulatory T-cell differentiation versus clonal deletion of autoreactive thymocytes. Immunol. Cell Biol. 89, 45–53 (2011). [DOI] [PubMed] [Google Scholar]
- 70.Bouneaud C, Kourilsky P & Bousso P Impact of negative selection on the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones escapes clonal deletion. Immunity 13, 829–840 (2000). [DOI] [PubMed] [Google Scholar]
- 71.Yu W et al. Clonal deletion prunes but does not eliminate self-specific αβ CD8+ T lymphocytes. Immunity 42, 929–941 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Malhotra D et al. Tolerance is established in polyclonal CD4+ T cells by distinct mechanisms, according to self-peptide expression patterns. Nat. Immunol. 17, 187–195 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Legoux FP et al. CD4+ T cell tolerance to tissue-restricted self antigens is mediated by antigen-specific regulatory T cells rather than deletion. Immunity 43, 896–908 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Tai X et al. How autoreactive thymocytes differentiate into regulatory versus effector CD4+ T cells after avoiding clonal deletion. Nat. Immunol. 24, 637–651 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Owen DL et al. Thymic regulatory T cells arise via two distinct developmental programs. Nat. Immunol. 20, 195–205 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Shevach EM & Thornton AM tTregs, pTregs, and iTregs: similarities and differences. Immunol. Rev. 259, 88–102 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Metzger TC & Anderson MS Control of central and peripheral tolerance by Aire. Immunol. Rev. 241, 89–103 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Klein L, Kyewski B, Allen PM & Hogquist KA Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat. Rev. Immunol. 14, 377–391 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Cebula A et al. Dormant pathogenic CD4+ T cells are prevalent in the peripheral repertoire of healthy mice. Nat. Commun. 10, 4882 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Parish IA & Heath WR Too dangerous to ignore: self-tolerance and the control of ignorant autoreactive T cells. Immunol. Cell Biol. 86, 146–152 (2008). [DOI] [PubMed] [Google Scholar]
- 81.Zheng Y, Zha Y & Gajewski TF Molecular regulation of T-cell anergy. EMBO Rep. 9, 50–55 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Nurieva RI, Liu X & Dong C Molecular mechanisms of T-cell tolerance. Immunol. Rev. 241, 133–144 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Shevach EM Mechanisms of Foxp3+ T regulatory cell-mediated suppression. Immunity 30, 636–645 (2009). [DOI] [PubMed] [Google Scholar]
- 84.Kurts C et al. CD8 T cell ignorance or tolerance to islet antigens depends on antigen dose. Proc. Natl Acad. Sci. USA 96, 12703–12707 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lang KS et al. Toll-like receptor engagement converts T-cell autoreactivity into overt autoimmune disease. Nat. Med. 11, 138–145 (2005). [DOI] [PubMed] [Google Scholar]
- 86.Ramanathan S et al. Exposure to IL-15 and IL-21 enables autoreactive CD8 T cells to respond to weak antigens and cause disease in a mouse model of autoimmune diabetes. J. Immunol. 186, 5131–5141 (2011). [DOI] [PubMed] [Google Scholar]
- 87.Millar DG et al. Hsp70 promotes antigen-presenting cell function and converts T-cell tolerance to autoimmunity in vivo. Nat. Med. 9, 1469–1476 (2003). [DOI] [PubMed] [Google Scholar]
- 88.Schwartz RH T cell anergy. Annu. Rev. Immunol. 21, 305–334 (2003). [DOI] [PubMed] [Google Scholar]
- 89.Jenkins MK & Schwartz RH Antigen presentation by chemically modified splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo. J. Exp. Med. 165, 302–319 (1987). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Oelert T et al. Irradiation and IL-15 promote loss of CD8 T-cell tolerance in response to lymphopenia. Blood 115, 2196–2202 (2010). [DOI] [PubMed] [Google Scholar]
- 91.Bour-Jordan H et al. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/B7 family. Immunol. Rev. 241, 180–205 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.He X & Xu C Immune checkpoint signaling and cancer immunotherapy. Cell Res. 30, 660–669 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Walunas TL et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity 1, 405–413 (1994). [DOI] [PubMed] [Google Scholar]
- 94.Francisco LM, Sage PT & Sharpe AH The PD-1 pathway in tolerance and autoimmunity. Immunol. Rev. 236, 219–242 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Linsley PS et al. CTLA-4 is a second receptor for the B cell activation antigen B7. J. Exp. Med. 174, 561–569 (1991). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Krummel MF & Allison JP CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J. Exp. Med. 183, 2533–2540 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Keir ME, Butte MJ, Freeman GJ & Sharpe AH PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schietinger A & Greenberg PD Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol. 35, 51–60 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Poels K et al. Immune checkpoint inhibitor treatment and atherosclerotic cardiovascular disease: an emerging clinical problem. J. Immunother. Cancer 9, e002916 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Postow MA, Sidlow R & Hellmann MD Immune-related adverse events associated with immune checkpoint blockade. N. Engl. J. Med. 378, 158–168 (2018). [DOI] [PubMed] [Google Scholar]
- 101.Georgiev P, Charbonnier L-M & Chatila TA Regulatory T cells: the many faces of Foxp3. J. Clin. Immunol. 39, 623–640 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Liu W et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J. Exp. Med. 203, 1701–1711 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Josefowicz SZ, Lu L-F & Rudensky AY Regulatory T cells: mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531–564 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Sakaguchi S, Yamaguchi T, Nomura T & Ono M Regulatory T cells and immune tolerance. Cell 133, 775–787 (2008). [DOI] [PubMed] [Google Scholar]
- 105.Zemmour D et al. Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat. Immunol. 19, 291–301 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Benoist C & Mathis D Treg cells, life history, and diversity. Cold Spring Harb. Perspect. Biol. 4, a007021 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hori S, Nomura T & Sakaguchi S Control of regulatory T cell development by the transcription factor Foxp3. Science 299, 1057–1061 (2003). [DOI] [PubMed] [Google Scholar]
- 108.Coombes JL et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J. Exp. Med. 204, 1757–1764 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Walker LSK CD4+ CD25+ Treg: divide and rule? Immunology 111, 129–137 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhang J et al. Identification of the E3 deubiquitinase ubiquitin-specific peptidase 21 (USP21) as a positive regulator of the transcription factor GATA3. J. Biol. Chem. 288, 9373–9382 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li Y et al. USP21 prevents the generation of T-helper-1-like Treg cells. Nat. Commun. 7, 13559 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Delacher M et al. Rbpj expression in regulatory T cells is critical for restraining TH2 responses. Nat. Commun. 10, 1621 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Delacher M et al. Genome-wide DNA-methylation landscape defines specialization of regulatory T cells in tissues. Nat. Immunol. 18, 1160–1172 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Saxena V, Lakhan R, Iyyathurai J & Bromberg JS Mechanisms of exTreg induction. Eur. J. Immunol. 51, 1956–1967 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Shi H & Chi H Metabolic control of Treg cell stability, plasticity, and tissue-specific heterogeneity. Front. Immunol. 10, 2716 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ohkura N et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 37, 785–799 (2012). [DOI] [PubMed] [Google Scholar]
- 117.Nakatsukasa H et al. Loss of TET proteins in regulatory T cells promotes abnormal proliferation, Foxp3 destabilization and IL-17 expression. Int. Immunol. 31, 335–347 (2019). [DOI] [PubMed] [Google Scholar]
- 118.Yue X, Lio C-WJ, Samaniego-Castruita D, Li X & Rao A Loss of TET2 and TET3 in regulatory T cells unleashes effector function. Nat. Commun. 10, 2011 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.DuPage M et al. The chromatin-modifying enzyme Ezh2 is critical for the maintenance of regulatory T cell identity after activation. Immunity 42, 227–238 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Li MO & Rudensky AY T cell receptor signalling in the control of regulatory T cell differentiation and function. Nat. Rev. Immunol. 16, 220–233 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Gabryšová L et al. Integrated T-cell receptor and costimulatory signals determine TGF-β-dependent differentiation and maintenance of Foxp3+ regulatory T cells. Eur. J. Immunol. 41, 1242–1248 (2011). [DOI] [PubMed] [Google Scholar]
- 122.Sauer S et al. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc. Natl Acad. Sci. USA 105, 7797–7802 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hoffmann P et al. Loss of FOXP3 expression in natural human CD4+CD25+ regulatory T cells upon repetitive in vitro stimulation. Eur. J. Immunol. 39, 1088–1097 (2009). [DOI] [PubMed] [Google Scholar]
- 124.Hua J et al. Pathological conversion of regulatory T cells is associated with loss of allotolerance. Sci. Rep. 8, 7059 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Huynh A et al. Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability. Nat. Immunol. 16, 188–196 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Shrestha S et al. Treg cells require the phosphatase PTEN to restrain TH1 and TFH cell responses. Nat. Immunol. 16, 178–187 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Sharma MD et al. An inherently bifunctional subset of Foxp3+ T helper cells is controlled by the transcription factor eos. Immunity 38, 998–1012 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Pasare C & Medzhitov R Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science 299, 1033–1036 (2003). [DOI] [PubMed] [Google Scholar]
- 129.Guo J et al. Constitutive activation of MEK1 promotes Treg cell instability in vivo. J. Biol. Chem. 289, 35139–35148 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Gerriets VA et al. Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat. Immunol. 17, 1459–1466 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Nie H et al. Phosphorylation of FOXP3 controls regulatory T cell function and is inhibited by TNF-α in rheumatoid arthritis. Nat. Med. 19, 322–328 (2013). [DOI] [PubMed] [Google Scholar]
- 132.Kitz A et al. AKT isoforms modulate Th1-like Treg generation and function in human autoimmune disease. EMBO Rep. 17, 1169–1183 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Charbonnier L-M, Wang S, Georgiev P, Sefik E & Chatila TA Control of peripheral tolerance by regulatory T cell-intrinsic Notch signaling. Nat. Immunol. 16, 1162–1173 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Feldhoff LM et al. IL-1β induced HIF-1α inhibits the differentiation of human FOXP3+ T cells. Sci. Rep. 7, 465 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Chen L, Wu J, Pier E, Zhao Y & Shen Z mTORC2-PKBα/Akt1 serine 473 phosphorylation axis is essential for regulation of FOXP3 stability by chemokine CCL3 in psoriasis. J. Invest. Dermatol. 133, 418–428 (2013). [DOI] [PubMed] [Google Scholar]
- 136.Lu L et al. Critical role of all-trans retinoic acid in stabilizing human natural regulatory T cells under inflammatory conditions. Proc. Natl Acad. Sci. USA 111, E3432–E3440 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Baban B et al. IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J. Immunol. 183, 2475–2483 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Sharma MD et al. Indoleamine 2,3-dioxygenase controls conversion of Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes. Blood 113, 6102–6111 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sharma MD et al. Reprogrammed Foxp3+ regulatory T cells provide essential help to support cross-presentation and CD8+ T cell priming in naive mice. Immunity 33, 942–954 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]