Next Article in Journal
PTPN1 Deficiency Modulates BMPR2 Signaling and Induces Endothelial Dysfunction in Pulmonary Arterial Hypertension
Next Article in Special Issue
Large-Scale Polymorphism Analysis of Dog Leukocyte Antigen Class I and Class II Genes (DLA-88, DLA-12/88L and DLA-DRB1) and Comparison of the Haplotype Diversity between Breeds in Japan
Previous Article in Journal
Triphasic 3D In Vitro Model of Bone-Tendon-Muscle Interfaces to Study Their Regeneration
Previous Article in Special Issue
The Parallel Presentation of Two Functional CTL Epitopes Derived from the O and Asia 1 Serotypes of Foot-and-Mouth Disease Virus and Swine SLA-2*HB01: Implications for Universal Vaccine Development
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MHC Class II Presentation in Autoimmunity

by
Irina A. Ishina
1,*,
Maria Y. Zakharova
1,*,
Inna N. Kurbatskaia
1,
Azad E. Mamedov
1,
Alexey A. Belogurov, Jr.
1,2 and
Alexander G. Gabibov
1,3,4
1
Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
2
Department of Biological Chemistry, Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
3
Department of Life Sciences, Higher School of Economics, 101000 Moscow, Russia
4
Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
*
Authors to whom correspondence should be addressed.
Cells 2023, 12(2), 314; https://doi.org/10.3390/cells12020314
Submission received: 6 December 2022 / Revised: 10 January 2023 / Accepted: 12 January 2023 / Published: 14 January 2023
(This article belongs to the Special Issue Major Histocompatibility Complex (MHC) in Health and Disease 2022)

Abstract

:
Antigen presentation by major histocompatibility complex class II (MHC-II) molecules is crucial for eliciting an efficient immune response by CD4+ T cells and maintaining self-antigen tolerance. Some MHC-II alleles are known to be positively or negatively associated with the risk of the development of different autoimmune diseases (ADs), including those characterized by the emergence of autoreactive T cells. Apparently, the MHC-II presentation of self-antigens contributes to the autoimmune T cell response, initiated through a breakdown of central tolerance to self-antigens in the thymus. The appearance of autoreactive T cell might be the result of (i) the unusual interaction between T cell receptors (TCRs) and self-antigens presented on MHC-II; (ii) the posttranslational modifications (PTMs) of self-antigens; (iii) direct loading of the self-antigen to classical MHC-II without additional nonclassical MHC assistance; (iv) the proinflammatory environment effect on MHC-II expression and antigen presentation; and (v) molecular mimicry between foreign and self-antigens. The peculiarities of the processes involved in the MHC-II-mediated presentation may have crucial importance in the elucidation of the mechanisms of triggering and developing ADs as well as for clarification on the protective effect of MHC-II alleles that are negatively associated with ADs.

1. Introduction

The efficacy of the immune response, as well as the severity of the disease, is strongly associated with the genetics of individual patients. One of the most important genetic determinants specifying the predisposition to different diseases is the MHC locus. Recently, the undoubted existence of genetic gateways was demonstrated for viral infections such as SARS-CoV-2. In particular, distinct MHC-I and -II alleles increase the probability of the severe clinical course of the infection [1,2,3,4]. The existing cohorts of so-called “disease controllers” and “progressors” patients in HIV with confirmed genetic resistance to infection were evidently shown in numerous studies [5,6,7]. Summarizing, genome-encoded MHC alleles are decision-makers in terms of the dynamics and amplitude of the immune response, including autoimmune abnormalities.
A highly polymorphic MHC genomic region is linked with a broad variety of autoimmune pathologies, particularly characterized by the presence of autoreactive T cell clones [8]. One of the triggers of this autoreactive T cell response might be the presentation of self-antigens by MHC-II molecules in the periphery. One may say that autoimmune diseases may be initiated directly by MHC molecules due to their structural features independent of antigen presentation [9,10,11], but such speculations are beyond the scope of our review.
Processing of proteins captured outside the cell is the conventional method for generating antigenic peptides presented by MHC-II molecules on the cell surface. Structural polymorphism of the binding groove pockets determines a broad range of peptide ligands that can be presented by MHC-II molecules to CD4+ T cells. The repertoire of T cells is formed via positive and negative selection occurring in the cortex and the thymic medulla, respectively (Figure 1). Depending on T cell receptor (TCR) affinity or avidity to the peptide–MHC-II complex (pMHC), T cells undergo elimination or differentiation into the Treg or naïve CD4+ T cell lineage [12,13,14]. However, there are some additional factors influencing the T cell differentiation pathway during central tolerance establishment and potentially facilitating the appearance of autoreactive T cells: the topology of self-antigen presentation [15], the topology of recognition of TCR within the pMHC complex [16], the frequency of pMHC–TCR interaction [17], and costimulatory interactions [18]. Moreover, the modifications of antigens and the cytokine milieu might affect the presentation of antigenic fragments in the periphery and thus enhance the development of autoreactive T cells.
Extensive genotypic medical statistics indicate that certain MHC-II alleles are associated, either positively or negatively, with definite autoimmune diseases (ADs). The HLA-DRB1*01:01 (DR1) and 04:01 (DR4) alleles are positively associated with rheumatoid arthritis (RA) [19]. HLA-DRB1*15:01 (DR15) is the risk allele for multiple sclerosis (MS) [20] and Goodpasture syndrome [21], while HLA-DRB1*01:01 is protective against these two diseases [22,23,24]. The HLA-DQA1*05:01/DQB1*02:01 (DQ2.5) and HLA-DQA1*03:01/DQB1*03:02 (DQ8.1) alleles are positively associated with type 1 diabetes (T1D), while the HLA-DQA1*01:02/DQB1*06:02 (DQ6.2) allele is negatively associated with this disease [25,26,27]. Although a significant body of MHC-II association data has been accumulated, the correlation between such associations and autoreactive T cell response development has not yet been completely studied. Here, we attempt to establish several scenarios causing the emergence or lack of autoreactive T cells due to the pMHC engagement.

2. MHC-II Presentation of Low-Affinity Self-Antigens

MHC-II molecules are synthesized in the endoplasmic reticulum (ER); the peptide-binding groove is occupied by the invariant chain Ii (CD74 fragment) to prevent degradation and premature binding of self-antigens by the complex. As they migrate to the late endosomal compartments with an acidic environment, proteolytic enzymes shorten the invariant chain to a shorter class II-associated invariant chain peptide (CLIP) [28]. Protein antigens are proteolytically processed in endosomes as well (Figure 2). The peptide-binding groove of an MHC class II molecule has nine pockets that can accommodate certain amino acid residues of the peptide, typically stabilized by noncovalent bonds [29]. The P1, P4, P6, and P9 anchor residues interact with the groove of MHC-II, thus forming a binding register, while the remaining residues of the peptide are oriented in the opposite direction for TCR binding. Presumably, if there is a shift in the binding register between antigenic peptides and MHC molecules, the pMHC complex can interact with an entirely different TCR.
The nonclassical MHC molecules HLA-DM (DM) and HLA-DO (DO) in humans and H2-DM and H2-DO in mice play an important role in exchanging CLIP for endosomal antigens [30]. By interacting with MHC-II, DM has the “editing” function: it ensures the binding of peptides with higher affinity for the MHC molecule compared with CLIP, as well as increases the loading/dissociation rate of the antigen, but it causes no effect on the equilibrium affinity [31,32]. Apparently, DM facilitates the presentation of antigen epitopes with the optimal binding register on MHC-II [33,34]. DO also performs an editing action, interacting with DM and regulating its catalytic function by preventing its binding to MHC-II [35]. Comparing MHC-II immunopeptidomes from two cell lines, DO knockout or not, it was shown that only the DR1+, DM+, and DO+ lymphoblastoid cell lines presented specific antigens on MHC-II [36]. Thus, DO contributes to the diversification of the MHC-II-presented antigenic repertoire. In experiments with DM-knockout mice, CLIP was not efficiently exchanged with other peptides, thus limiting the MHC-II antigen diversity and causing incomplete negative selection of CD4+ T cells in the thymus [37]. The repertoire of antigens presented by MHC-II can vary significantly depending on the DM/DO ratio in the cell [38]. Presumably, DM “editing” decreases the number of antigens, characterized by a low affinity for MHC-II, on the antigen-presenting cell (APC) surface, as was shown for DQ1 and DQ6 [39], as well as for DR3 alleles [31]. Analysis of the immunopeptidome of thymic APCs revealed that most antigens detected on MHC-II have a high affinity for MHC-II molecules [40,41]. Most likely, the effect of nonclassical MHC molecules is one of the important factors ensuring the highly competitive conditions for MHC-II ligand binding, which eventually results in the presentation of higher-affinity antigens.
It should be emphasized that the presentation of preferentially high-affinity antigens in the thymus may have its own shortcomings. It has been demonstrated that the MHC-II-presented antigen repertoire from tissues affected by autoimmune processes mostly consists of low-affinity peptides [42,43]. The abundance of self-antigens increases in peripheral tissues (e.g., myelin basic protein (MBP) in the central nervous system (CNS) [44,45,46] and insulin in the pancreas [47]). These protein self-antigens can be processed in the extracellular environment outside APCs. Next, the resulting antigenic fragments, characterized by a low affinity for MHC-II molecules, can be loaded directly on the APC surface or can enter early endosomes with low DM levels, thus escaping DM editing, and as a result, can be presented on the cell surface (Figure 3A,B). Certain MHC-II risk alleles bind CLIP with diminished affinity, which additionally promotes the binding of low-affinity antigenic peptides [48,49]. Rapid dissociation of CLIP enables the generation of empty complexes without DM involvement, which may potentially bind low-affinity antigenic peptides. Due to the spontaneous release of CLIP, low-affinity antigenic peptides are able to bind MHC-II in early endosomes without DM assistance or directly on the cell surface. Therefore, pMHC complexes on the APC surface at the periphery can activate TCRs that have not undergone negative selection due to the low abundance of this pMHC in the thymus. For MS patients, there was detected an immune response to full-length MS autoantigen proteolipid protein (PLP), naturally processed by APCs, with 2 immunodominant epitopes generation. Alternatively, the T cell response to several PLP synthetic fragments with high affinity to MS-associated MHC-II alleles was also observed after additional T cell stimulation by these peptides [50]. Since these fragments are normally hidden in a protein fold and are inaccessible for the endosomal proteases, the T cell activation seems to be the result of extracellular PLP processing and loading on MHC-II. Thus, the classical intracellular processing of antigen involves its capture by the APC with proteolytic processing and generation of fragments with optimal binding registers with the participation of HLA-DM in late endosomes, followed by subsequent exposure of pMHCs on the APC surface. These pMHCs take part in the negative selection process in the thymus, induce T cell clonal deletion and, after all, no autoreactive T cell response is observed at the periphery. Since pMHCs with “suboptimal” fragments were present in the thymus at low levels, autoreactive T cells might engage them at the periphery due to the failure of negative selection in the thymus.
Furthermore, a single antigen fragment can have several MHC-II binding registers. As T1D develops in non-obese diabetic (NOD) mice (the mouse model of T1D), autoreactive T cells recognize insulin (Ins. B:9–23) fragments presented on the product of diabetogenic allele I-Ag7 [51,52]. The Ins. B:9–23 peptide has several binding registers in the context of I-Ag7. The antigen with binding register 12–20 is characterized by low affinity and an increased dissociation rate in the presence of DM, while fragment 13–21 forms a more stable complex with I-Ag7, which is almost insusceptible to DM editing. Immunization with antigen fragments 12–20 elicits an autoreactive T cell response, while immunization with antigen fragments 13–21 causes no response [53]. Presumably, the autoreactive TCR, specific to the antigen fragment 13–21, undergoes negative selection in the thymus, while the TCR, interacting with the antigen fragment 12–20, escapes the central tolerance mechanisms due to the potentially low abundance of the pMHC complex on the medullary APCs. Additionally, multiple autoreactive insulin-specific T cells were reported to recognize Ins. B:9–23 bound to I-Ag7 in a low-affinity register 3 (Ins. B:14–22). The Ins. B:9–23 bound to I-Ag7 activates diabetogenic CD4+ T cells and binds insulin-specific pancreatic T cells from NOD mice [54,55]. Thus, peptides may bind MHC-II molecules in low-affinity register in early endosomes or on the cell surface in the absence of DM due to the abundance of certain antigens in the autoimmunity-affected peripheral sites. Derived pMHC complexes engage autoreactive T cells escaping negative selection owing to the underrepresentation of unstable pMHCs in the thymus. Human DQ8 shares structural similarities with mouse I-Ag7. The Ins. B:11–23 binds DQ8 with a low affinity and engages autoreactive peripheral blood CD4+ T cells of subjects with T1D [56].
Concluding, nonclassical MHC molecules help to present the most high-affinity antigens with the optimal binding register by MHC-II on thymic APCs. Nevertheless, when there is an excess of a certain self-antigen in peripheral tissues, its presentation on MHC-II molecules can occur without DM editing, allowing the presentation of low-affinity self-antigen fragments on the surface of APCs. The pMHC complexes loaded by low-affinity antigens are unstable and have a short lifespan; however, if their abundance on the surface of APCs is sufficiently high, they can potentially activate T cells, because the strength of the autoreactive T cell response is not directly correlated with antigen affinity to MHC-II [57].

3. The Peculiarities of the Interaction between TCRs and Self-pMHC Complexes: Links with Autoimmunity

Generally, CD4+ T cell TCRs recognize foreign antigens within MHC-II molecules [58,59,60] with CD4 receptors playing the key role in the induction of the T cell signaling cascade [61,62,63]. The analysis of trimolecular complexes between TCRs, MHC-II, and exogenous antigens has revealed some structural similarity in the TCR binding topology [64]. The TCR is arranged diagonally with respect to the antigen residing in the peptide-binding groove limited by α-helices. The variable regions of α and β TCR chains interact with the β and α chains of the MHC-II molecule, respectively [65]. The most structurally diverse CDR3 fragments of α and β TCR chains are located above the P5 residue of the bound peptide, while the CDR1 and CDR2 fragments interact with the α-helices of MHC-II molecules. The binding of TCR HA1.7 to the fragment of hemagglutinin HA protein within DR1 is an example of canonical interaction [66].
It was shown that, by contrast with TCRs binding bacterial or viral antigens on MHC-II, some autoreactive TCRs have an alternative binding topology. Thus, the TCR from an MS patient (Ob.1A12) specific for the fragment of myelin basic protein (MBP), which is presented on the HLA-DR2b risk allele molecule, interacts mainly with the N-terminus of the MBP fragment [16]. Furthermore, this TCR does not reside in the canonical diagonal position: its orientation angle is 110°, as opposed to 70° for HA1.7. The main interaction with the pMHC complex occurs due to the binding of the CDR3 region of TCR with the P2 residue of the MBP peptide fragment. Other TCRs, Ob.2F3 and Ob.3D1, obtained from the same donor with MS, also have an alternative binding topology, as examined using computational simulation [67]. Another TCR, 3A6, binds the MBP fragment in the complex with different DR15 allomorph DR2a, which is positively associated with the risk of developing MS. The 3A6 also has a suboptimal topology and low affinity for pMHC, similar to other autoreactive TCRs [68]. Its CDRs are shifted toward the N-terminus of the antigen, and CDR3 is also located above P2 of the MBP fragment. Nevertheless, similar to HA1.7, 3A6 binds diagonally with respect to pMHC. Therefore, Ob.1A12 and 3A6 have low affinity for pMHC, and their binding topology is alternate to anti-viral and anti-bacterial TCRs, which may potentially facilitate the escape from negative selection mechanisms in the thymus. Low-affinity TCRs were also reported in the pathogenesis of T1D. The repertoire of the autoreactive T cells, infiltrating pancreatic islets, exhibited low self-reactivity and promoted the development of T1D [69]. Presumably, the interaction between these T1D TCRs and self-antigen-carrying pMHCs is also characterized by alternate topology. The MBP fragment presented by DQ1 is recognized by the Hy.1B11 TCR with relatively high affinity [70]. These TCRs have canonical localization but are significantly shifted with respect to the DQ1α chain so that their interaction with the antigen is substantially limited. Only the CDR3 region of the Hy.1B11α chain contacts the MBP fragment. Presumably, the unusual topology of autoreactive TCRs binding to pMHC impedes activation by the CD4 molecule and, therefore, clonal deletion of T cells at the negative selection stage. This may explain the existence of T cells with similar TCRs (such as Hy.1B11) in the periphery [71]. The unusual topology of the trimolecular complex often implies the participation of a low-affinity TCR. However, autoimmune TCRs with a high affinity for pMHCs have also been reported. It is known that DQ molecules have a lower expression level than DR molecules. Therefore, appropriate autoreactive TCRs might require higher affinities for DQ pMHC complexes to overcome clonal deletion in the thymus [70]. Another possible explanation of high-affinity autoreactive TCR emergence at the periphery is that the autoantigenic fragment may be a weak binder in the pMHC complex. As previously discussed, pMHC carrying low-affinity peptides allows T cells to escape negative selection. Nevertheless, they can further bind autoreactive TCRs at the periphery. Interestingly, the weak interaction between the MBP fragment and the product of the risk allele DR4 is stabilized by the MS2-3C8 TCR involved in canonical high-affinity interaction with pMHC [72]. The structures of the trimolecular complexes of three autoreactive TCRs in model mice with experimental autoimmune encephalomyelitis (EAE), which bind the MBP fragment in complex with I-Au, have revealed canonical binding. However, the interaction between the MBP antigen and the I-Au molecule is characterized by low affinity due to partial occupation of the peptide-binding cleft [73,74,75]. The affinities of self-peptides for MHC-II molecules of risk alleles and autoreactive TCRs for pMHC complexes of risk alleles are summarized in Table 1.

4. The Effect of MHC-II-Presented Self-Antigen Modifications on Autoreactive TCR Recognition

Antigens processed for further presentation by MHC-II molecules can undergo various posttranslational modifications (PTMs), including glycosylation, iodination, citrullination, etc. (Figure 4). PTMs can either occur spontaneously or be induced by various enzymes. Autoreactive T cell responses to modified self-antigens have been observed for many ADs. Supposedly, the modified antigens (neoantigens) are either totally not present in the thymus or their quantity is extremely low. These modifications presumably take place in peripheral tissues and are absent when the antigen is presented by mTEC cells in the thymus, which has been demonstrated for glycosylated type II collagen for the development of RA [76]. Although modified antigens are available in the thymus due to the presence of migratory APCs, their quantity is probably insufficient for negative T cell selection in the thymus. Thus, thyroglobulin, which is the autoantigen in patients with Hashimoto’s thyroiditis, is present in the thymus in its nonmodified form, whereas the level of its iodinated form, which seems to initiate the autoreactive T cell response, is extremely low in the thymus and is insufficient for central tolerance to be established [77].
Citrullination of antigens is described for a number of ADs, especially for RA; the positive charge of arginine is lost due to the conversion to citrulline, which ultimately alters the epitope affinity to MHC-II and TCR [78]. The products of risk MHC-II alleles for RA carry a consensus amino acid sequence in the antigen-binding groove, forming a positively charged P4 pocket–shared epitope (SE), which binds the polar amino acid residues of peptides at the P4 position (e.g., citrulline) and induces the activation of autoreactive T cells [79,80]. In addition to hosting a polar-neutral citrulline residue, SE can itself serve as the main contact zone for autoreactive TCRs and represent citrullinated fragments of the fibrinogen autoantigen that directly interact with autoreactive TCRs via the P2-citrulline residue [81]. Autoreactive T cells specific for citrullinated tenascin-C antigenic peptides, presented by DR4 molecules, were revealed in RA patients [82]. Additionally, glycosylation was reported to be responsible for the pathogenesis of RA. Analysis of the crystal structure of the trimolecular complex of an autoreactive TCR and type II collagen (Col2) neoantigen presented on DR4 has shown that lysine residues subjected to galactosylation are the key sites for TCR recognition [83]. Furthermore, the autoreactive T cell response is dependent on the galactosylation of lysine in the Col2259–273 fragment at position 264. Modified autoantigenic peptides also participate in T1D development. Several citrullinated and transglutaminated GAD65 epitopes are recognized by autoreactive CD4+ T cells in T1D preferentially to their unmodified form [84]. Autoreactive T cells specific for citrullinated glucokinase epitopes are linked to T1D pathogenesis [85]. Another example of the modified antigens impacting the pathogenesis of T1D is the generation of a disulfide bond. T cells recognize insulin fragments presented on DR4 molecules in T1D upon the assembly of a disulfide bridge between neighboring cysteine residues [86]. The citrullinated fragments of MBP and myelin oligodendrocyte glycoprotein (MOG) cause EAE due to the activation of autoreactive T cells [87,88,89]. In EAE, acetylation of the MBP fragment elicits an encephalitogenic T cell response [73,74,75].
Hybrid insulin peptides (HIPs) are identified as neoantigens involved in the pathogenesis of T1D [90]. HIPs are formed via a peptide bond formation between insulin fragments and other secretory granule peptides. Mass spectrometry analysis of pancreatic islets revealed the presence of HIPs in humans and mice [91]. The risk MHC-II molecules are reported to present various HIPs. The HIPs formed by proinsulin C-peptide and chromogranin A or islet amyloid polypeptide 2 (IAPP2) were present on I-Ag7 molecules in the NOD mouse pancreas and recognized by pathogenic T cells [92]. Autoreactive T cells are able to recognize HIPs in the context of DQ2 and/or DQ8 and produce proinflammatory cytokines during the development of T1D [93,94]. The fusion of proinsulin C peptide and neuropeptide Y presented by DQ8+ and DR4+ B cells elicits an autoreactive T cell response in T1D [95]. DR4 presents different HIPs and is recognized by effector memory T cells [96]. The structures of trimolecular complexes of hybrid antigens formed by C-peptide and IAPP2, which are presented on DQ8, have revealed that the TCRα chain interacts exclusively with the C-peptide, while the TCR β chain interacts with IAPP2 only [97].
Multiple antigens with PTMs existing at the periphery cannot be inefficiently presented on thymic APCs, which results in the abrogation of autoreactive T cell clonal deletion. Concluding, PTM-modified antigens can be responsible for the induction of autoreactive T cells and for developing autoimmunity.

5. The Effect of the Proinflammatory Environment on MHC-II Antigen Presentation and Autoreactive T Cell Engagement

The generation and expansion of pathological T cells may be attributed to the increase in MHC-II expression levels due to the proinflammatory environment induced by viral or bacterial infections. The impacts of viral and bacterial infections on antigen presentation are the proposed triggers of pathological CD4+ T cell expansion in ADs such as MS and T1D [98,99,100,101,102]. Earlier, a proinflammatory environment was implicated in TCR-independent bystander activation in different ADs [103,104]. Proinflammatory cytokine release and bacterial antigen presence trigger the elevated synthesis of MHC-II molecules to maximize the presentation of foreign antigens for an efficient CD4+ T cell response. High IFN-γ levels increase the number of MHC-II molecules on professional and nonprofessional APCs, thereby altering the composition of the MHC peptidome in the inflammatory area [105]. Additionally, APCs, activated in pathogenic conditions, express an increased number of costimulatory molecules [106,107]. Therefore, rare self-antigen pMHC, which cannot induce an autoreactive T cell response under normal conditions, might engage autoreactive TCRs in a proinflammatory environment (Figure 5). Concluding, cytokines induced by the inflammatory environment may potentially activate antigen-specific autoreactive T cells.
A correlation between the inflammatory milieu and elevated MHC-II levels has been observed for several ADs. It was shown that MHC-II transcripts were upregulated by proinflammatory cytokine expression in β-cells of T1D patients [108,109]. IFN-γ induces the expression of I-Ag7 molecules on beta cells of NOD mice, leading to the emergence of CD4+ autoreactive T cells and the promotion of T1D [110]. In addition, IFN-γ promotes the expression of MHC-II on islet endothelial cells in NOD mice [111]. The transcription factors responsible for the expression of MHC-II are elevated in MS lesions of human brain tissue [112]. Aberrant MHC-II expression was also found on oligodendrocytes in EAE mice and MS patients [113]. The expression of MHC-II in mouse joints led to the development of severe erosive inflammatory polyarthritis [114]. Additionally, the inflammatory environment promotes the expression of MHC-II molecules on hepatocytes during autoimmune hepatitis and pMHC transfer via trogocytosis to interacting CD4+ T cells, which further amplifies the autoimmune response [115].
The onset of autoimmunity often coincides with exposure to viral and bacterial infections. The T1D progression is probably linked with Coxsackievirus B, rotavirus, and other viruses [116,117,118]. MS development correlates with several infections, such as Epstein–Barr virus (EBV) and herpesvirus-6 [119,120,121]. The proinflammatory cytokines, potentially induced by infections, may reshape the repertoire of antigens presented on MHC-II in inflammation-affected sites. As previously discussed, some autoreactive T cells are characterized by altered binding topology to the cognate pMHCs, which results in inefficient clonal deletion of autoreactive T cells in the thymus. Therefore, autoreactive T cells mildly engaging with pMHCs under homeostatic conditions might develop an autoimmune response under inflammatory conditions due to the general expansion of pMHCs in the inflamed area.

6. Molecular Mimicry between Self- and Foreign MHC-II Antigens May Lead to T Cell-Mediated Autoimmunity

Molecular mimicry refers to a structural similarity between self and exogenous antigens and potentially leads to autoimmunity [122,123,124]. The structural resemblance of antigens might result in the occasional misactivation of T cells (Figure 6). Autoreactive T cells from MS patients specific for MBP can cross-react with EBV nuclear antigen 1 (EBVNA1) [125,126]. Apparently, the virus-specific TCR was able to bind structurally similar self-antigen fragments presented on MHC-II and initiate an autoreactive T cell response. The probability of MS development is significantly increased when EBV infection is combined with the MHC-II risk allele HLA-DRB1*15:01. It was shown that CD4+ T cells specific for DR15-presented EBV antigens could cross-react with MBP [127].
Narcolepsy is a rare autoimmune chronic neurological disorder positively associated with the HLA-DQB1*06:02 allele and characterized by targeting hypocretin neurons [128]. Cross-reactive T cells for hypocretin autoantigen (HCRTNH2) and flu HA (H1N1 2009 strain) antigens were shown to influence narcolepsy development [129]. T cell cross-reactivity between viral epitope neuraminidase (NA175–189) and self-epitope protein-O-mannosyltransferase 1 (POMT1675–689) is also involved in narcolepsy pathogenesis [130].
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is an autoimmune disease targeting myeloperoxidase (MPO). Autoreactive T cells specific for the MPO409–428 cross-react with the 6PGD391–410 epitopes from Staphylococcus aureus [131]. Exogenous/self MHC-II antigen mimicry was also noted between streptococcal M protein and human cardiac myosin in the pathogenesis of rheumatic heart disease [132]. A bacterial L-asparaginase antigen fragment (L-ASNase67-81), mimicking a type II collagen epitope, may be presented on DR4 and elicits a CD4+ T cell response in blood samples of RA patients [133].
The gut microbiome potentially influences the development of various autoimmune pathologies [134,135]. It was shown that carrying certain risk or protective MHC-II alleles correlates with the composition of the gut microbiome in patients with AD [136,137,138]. Supposedly, distinct foreign antigenic peptides structurally similar to self-epitopes might cross-react with pathological T cells. Peptides from Lactobacillus reuteri mimic MOG antigenic fragments and elicit autoreactive T cell responses in an EAE model [139]. The DR53 (HLA-DRB4*01:03) molecule, presenting epitopes from Roseburia intestinalis, may bind beta-2 glycoprotein I (b2GPI) antigenic peptide and activate autoreactive T cells in antiphospholipid syndrome [140]. The antigenic peptide hprt4-18 from gut bacteria Parabacteroides distasonis, which is structurally similar to the fragment insulin B:9–23, activates human and NOD mouse insulin-specific T cells ex vivo [141,142].
Summarizing, bacterial and viral infections or gut microbiota composition seem to be the possible triggers of AD development through the mechanism of molecular mimicry, involving the recognition of structurally similar exogenous and endogenous antigenic fragments presented by MHC-II by CD4+ T cells.

7. The Role of Protective MHC-II Alleles in the Development of Autoimmune Diseases

Certain MHC alleles provide so-called “protection” toward the development of immune-related diseases due to peculiarities of antigen presentation, which affect the engagement of T cells [143,144]. “Protective” allele means that the risk of AD initiation in individuals carrying this allele is statistically lower than that for healthy donors, representing the average population [145,146,147,148]. Interestingly, the protein products of MHC-II protective alleles often differ from the products of risk alleles only by several amino acid residues localized in the TCR contact sites or near the key positions of the peptide-binding groove [149]. Supposedly, the protective effect can be ensured due to the deletion of autoreactive T cells in the thymus and/or induction of Treg cells (Figure 7A).
The expression of the I-E protective allele in NOD mice normally having the H-2g7 haplotype seems to prevent the development of T1D [150]. The protective effect of the I-E allele is reasoned by the deletion of autoreactive T cells [151]. Autoreactive TCR (4.1 TCR), expressed in transgenic NOD mice, undergoes negative selection in mice with the H-2g7/b, H-2g7/k, H-2g7/q, and H-2g7/nb1 haplotypes due to interaction with the products of protective MHC-II alleles [152]. Nevertheless, some studies cast doubt on the theory of deletion of autoreactive T cell clones as the only explanation for the protectivity of distinct MHC-II alleles in the development of ADs [153,154]. ADs are often characterized by a broadened repertoire of autoreactive T cells; therefore, protection against the development of autoimmune pathology cannot be solely attributed to the negative selection of a limited number of clones.
Tregs play a crucial role in maintaining peripheral tolerance and preventing ADs and tissue damage by controlling T cell expansion [155,156]. Supposedly, Tregs attenuate the immune response by engaging pMHCs encoded by protective alleles. However, the exact molecular mechanisms, involved in the interaction between Treg TCRs and the protective allele pMHC are still enigmatic. DR15 risk and DR1 protective molecules in Goodpasture syndrome present the α3 chain of type IV collagen (α3135–145) antigen with different binding registers [21]. Autoreactive T cells bound this self-antigen pMHC in DR15-positive patients with Goodpasture syndrome, and immunization with this antigen in DR15 humanized transgenic mice led to the development of the model disease. On the other hand, DR1 presenting α3135–145 interacts with Tregs in healthy donors. The DR1 humanized transgenic mice were similarly characterized by Treg differentiation and resistance to disease development. Presumably, changes in the register of antigen binding to the MHC-II molecules alter the antigen’s amino acid binding pattern, engaging TCR, which results in the predetermination of the phenotype of bound T cells.
Tregs may also play a role in the protection toward T1D development [157]. The protective (DQ6) T1D MHC-II molecules prevent the binding of islet-specific antigens to predisposing (DQ8) MHC-II molecules by engaging epitopes in different binding registers [158]. The proposed “epitope stealing” mechanism mediated by DQ6 molecules prevents the development of an autoreactive T cell response in T1D, elicited by autoantigen presentation on DQ8 molecules. It is possible that the antigenic peptides, presented by DQ6, promote the selection of Tregs in the thymus, which subsequently attenuates T1D development at peripheral sites. Healthy donors with the DR15/DQ6 haplotype had a higher level of T1D self-antigen-specific Tregs compared to those carrying neutral or risk MHC-II alleles [27]. Protective MHC-II alleles are also involved in the shaping of the gut microbial community and differentiation of Tregs. NOD mice, expressing the protective MHC-II Eα gene, do not develop T1D due to the change in gut microbiome composition and increased Treg proportions in the cecum [137].
Some studies revealed that autoreactive T cells can bind a number of antidiabetogenic MHC-II molecules, but differentiation into Tregs occurs due to engaging pMHCs of protective alleles exclusively [159,160]. The interaction with several pMHC complexes can be explained by TCR promiscuity and the unusual binding topology of some autoreactive TCRs, which was discussed previously. Interestingly, the trimolecular complex formed between Tregs and the proinsulin complex, presented by DR4 molecules, is characterized by 180° reversed polarity docking of TCR compared to canonical binding [161]. Thus, the TCRα and TCRβ chains interact with the α chain and β chain of the MHC-II molecule, respectively, while opposite interactions take place upon canonical binding. There is a limited number of reported crystal structures of trimolecular complexes of Tregs TCRs, making it difficult to accurately determine whether this binding topology is typical for most Tregs. However, canonical binding was revealed by analyzing the TCR structure of neonatal Tregs binding Padi4 peptide in the complex with I-Ab [162]. The crystal structures of trimolecular complexes of TCR and self-antigens presented by MHC-II protective alleles, which could possibly elucidate the molecular mechanism of protectivity, have not yet been reported.
The protective effect provided by MHC-II can also be related to the low rate of self-antigen loading to MHC-II molecules (Figure 7B). Antigen binding to MHC-II is a time-limited and competitive process controlled by nonclassical HLA molecules, as discussed above. It has been demonstrated that the self-antigen fragment MBP151-164 may be presented by the DR1 MHC-II allele, which is protective for patients with MS but is characterized by a slow loading rate in comparison with the exogenous peptide of influenza virus hemagglutinin protein HA306–318 [22]. This kinetic discrimination is structurally reasoned by decelerated docking of MBP residues in the P4 pocket. Taking into consideration the additional DM editing and the presence of other exogenous antigens competing with MBP peptide for DR1 in late endosomes, this fragment possibly cannot reach the surface of APCs. Therefore, the self-antigen MBP peptide is not presented or underrepresented by the protective MHC-II allele and does not elicit the possible autoreactive T cell response. However, the hypothesis of kinetic discrimination of self-antigens binding to molecules of protective MHC alleles needs additional clarification in terms of extended studies of MHC-II alleles relevant to the variety of ADs.

8. Conclusions

In this review, we systematized the data about MHC-II self-antigen presentation as an important pathway involved in the induction of the autoreactive CD4+ T cells. MHC-II allele polymorphisms are responsible for the diversity of the T cell repertoire. Normally, autoantigen-specific T cells are eliminated during negative selection in the thymus. However, pathologic autoreactive T cell clones can be expanded upon the development of ADs. Autoreactive T cells seem to appear due to the breakdown in the mechanisms of central tolerance in the thymus and may be activated as a result of interaction with pMHC carrying self-antigen on APCs. Normally, self-antigens, characterized by high affinity to MHC-II, are efficiently presented on APCs in the thymus and cause strong binding with the TCR of CD4+ T cells leading to the elimination of autoreactive T cells by negative selection. Self-antigens with low affinity for MHC-II molecules are limitedly presented in the thymus because of preliminary antigen repertoire editing by nonclassical MHC-II molecules. During inflammation, the abundance of self-antigens could be increased in the periphery (for example, in the inflammation locus), and low-affinity self-antigens can probably be loaded on MHC-II molecules without HLA-DM catalysis. In addition, many autoreactive TCRs have an unusual topology of binding to self-antigens presented on risk allele molecules, which can also potentially explain why autoreactive T cells escape negative selection in the thymus. The PTMs of autoantigens in peripheral tissue also may lead to recognition by T cells which have not undergone negative selection in the thymus. Finally, viral or bacterial infections might play a role in the development of ADs via the generation of a special proinflammatory environment or molecular mimicry between self- and pathogen antigens.
Here we have also discussed the presentation of self-antigens on protective MHC-II allele products and its significance for the subsequent induction of CD4+ T cell response. Summarizing, its protective role may be realized by the following scenarios: (i) deletion of autoreactive T cell clones, specific to protective pMHC in the thymus, during negative selection; (ii) induction of Treg development; and (iii) low rate loading of self-antigen on the protective MHC-II molecules and its kinetic discrimination in the concurrence with infection exogenous antigens.
Future determination of the etiology of ADs and steps toward targeted therapeutic approaches require the identification of a broader repertoire of self-antigens capable of binding presentation to the products of risk and protective MHC-II alleles along with the identification of pathogenic TCRs connected with the recognition of auto-pMHC. On the other hand, the basis of the thymic negative selection is directly linked with the MHC-II-mediated presentation of self-antigens; therefore, balancing between central and peripheral tolerance versus autoreactivity may be caused by thermodynamic and kinetic peculiarities of the autoantigen loading on the MHC-II molecules. Elucidation of the molecular mechanisms underlying the assembly of the autoreactive trimolecular pMHC-II-TCR complexes and its further signaling may result in the elaboration of novel therapeutic modalities in terms of the induction of the immunological tolerance [46,163,164,165], topological blockade of the self-antigens [166,167,168], and specific depletion of the autoreactive T cell clones [169,170].

Author Contributions

Conceptualization, I.A.I.; writing—original draft preparation, I.A.I., M.Y.Z., and I.N.K.; writing—review and editing, I.A.I., M.Y.Z., I.N.K., A.E.M., A.A.B.J., and A.G.G.; supervision, A.A.B.J. and A.G.G.; project administration, A.A.B.J. and A.G.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Russian Science Foundation (grant No. 17-74-30019). A.A.B.J. would like to acknowledge personal fellowship MD-5902.2021.1.4 and project 075-15-2021-1033.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Dmitriy M. Chudakov for valuable advice during manuscript preparation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Debnath, M.; Banerjee, M.; Berk, M. Genetic Gateways to COVID-19 Infection: Implications for Risk, Severity, and Outcomes. FASEB J. 2020, 34, 8787–8795. [Google Scholar] [CrossRef] [PubMed]
  2. Colona, V.L.; Biancolella, M.; Novelli, A.; Novelli, G. Will GWAS Eventually Allow the Identification of Genomic Biomarkers for COVID-19 Severity and Mortality? J. Clin. Investig. 2021, 131, e155011. [Google Scholar] [CrossRef] [PubMed]
  3. Andreakos, E.; Abel, L.; Vinh, D.C.; Kaja, E.; Drolet, B.A.; Zhang, Q.; O’Farrelly, C.; Novelli, G.; Rodríguez-Gallego, C.; Haerynck, F.; et al. A Global Effort to Dissect the Human Genetic Basis of Resistance to SARS-CoV-2 Infection. Nat. Immunol. 2022, 23, 159–164. [Google Scholar] [CrossRef] [PubMed]
  4. Nersisyan, S.; Zhiyanov, A.; Zakharova, M.; Ishina, I.; Kurbatskaia, I.; Mamedov, A.; Galatenko, A.; Shkurnikov, M.; Gabibov, A.; Tonevitsky, A. Alterations in SARS-CoV-2 Omicron and Delta Peptides Presentation by HLA Molecules. Peer J. 2022, 10, e13354. [Google Scholar] [CrossRef] [PubMed]
  5. The International HIV Controllers Study. The Major Genetic Determinants of HIV-1 Control Affect HLA Class I Peptide Presentation. Science (1979) 2010, 330, 1551–1557. [Google Scholar] [CrossRef] [Green Version]
  6. Košmrlj, A.; Read, E.L.; Qi, Y.; Allen, T.M.; Altfeld, M.; Deeks, S.G.; Pereyra, F.; Carrington, M.; Walker, B.D.; Chakraborty, A.K. Effects of Thymic Selection of the T-Cell Repertoire on HLA Class I-Associated Control of HIV Infection. Nature 2010, 465, 350–354. [Google Scholar] [CrossRef] [Green Version]
  7. Monel, B.; McKeon, A.; Lamothe-Molina, P.; Jani, P.; Boucau, J.; Pacheco, Y.; Jones, R.B.; le Gall, S.; Walker, B.D. HIV Controllers Exhibit Effective CD8+ T Cell Recognition of HIV-1-Infected Non-Activated CD4+ T Cells. Cell. Rep. 2019, 27, 142–153. [Google Scholar] [CrossRef] [Green Version]
  8. Ishigaki, K.; Lagattuta, K.A.; Luo, Y.; James, E.A.; Buckner, J.H.; Raychaudhuri, S. HLA Autoimmune Risk Alleles Restrict the Hypervariable Region of T Cell Receptors. Nat. Genet. 2022, 54, 393–402. [Google Scholar] [CrossRef]
  9. Scavuzzi, B.M.; van Drongelen, V.; Holoshitz, J. HLA-G and the MHC Cusp Theory. Front. Immunol. 2022, 13, 814967. [Google Scholar] [CrossRef]
  10. van Drongelen, V.; Scavuzzi, B.M.; Nogueira, S.V.; Miller, F.W.; Sawalha, A.H.; Holoshitz, J. HLA-DRB1 Allelic Epitopes That Associate with Autoimmune Disease Risk or Protection Activate Reciprocal Macrophage Polarization. Sci. Rep. 2021, 11, 2599. [Google Scholar] [CrossRef]
  11. Jin, H.; Kishida, K.; Arase, N.; Matsuoka, S.; Nakai, W.; Kohyama, M.; Suenaga, T.; Yamamoto, K.; Sasazuki, T.; Arase, H. Abrogation of Self-Tolerance by Misfolded Self-Antigens Complexed with MHC Class II Molecules. Sci. Adv. 2022, 8, abj9867. [Google Scholar] [CrossRef] [PubMed]
  12. Lee, H.-M.; Bautista, J.L.; Scott-Browne, J.; Mohan, J.F.; Hsieh, C.-S. A Broad Range of Self-Reactivity Drives Thymic Regulatory T Cell Selection to Limit Responses to Self. Immunity 2012, 37, 475–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cozzo Picca, C.; Oh, S.; Panarey, L.; Aitken, M.; Basehoar, A.; Caton, A.J. Thymocyte Deletion Can Bias Treg Formation toward Low-Abundance Self-Peptide. Eur. J. Immunol. 2009, 39, 3301–3306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lagattuta, K.A.; Kang, J.B.; Nathan, A.; Pauken, K.E.; Jonsson, A.H.; Rao, D.A.; Sharpe, A.H.; Ishigaki, K.; Raychaudhuri, S. Repertoire Analyses Reveal T Cell Antigen Receptor Sequence Features That Influence T Cell Fate. Nat. Immunol. 2022, 23, 446–457. [Google Scholar] [CrossRef] [PubMed]
  15. Malhotra, D.; Linehan, J.L.; Dileepan, T.; Lee, Y.J.; Purtha, W.E.; Lu, J.V.; Nelson, R.W.; Fife, B.T.; Orr, H.T.; Anderson, M.S.; et al. Tolerance Is Established in Polyclonal CD4+ T Cells by Distinct Mechanisms, According to Self-Peptide Expression Patterns. Nat. Immunol. 2016, 17, 187–195. [Google Scholar] [CrossRef]
  16. Hahn, M.; Nicholson, M.J.; Pyrdol, J.; Wucherpfennig, K.W. Unconventional Topology of Self Peptide–Major Histocompatibility Complex Binding by a Human Autoimmune T Cell Receptor. Nat. Immunol. 2005, 6, 490–496. [Google Scholar] [CrossRef] [Green Version]
  17. Mariathasan, S.; Zakarian, A.; Bouchard, D.; Michie, A.M.; Zúñiga-Pflücker, J.C.; Ohashi, P.S. Duration and Strength of Extracellular Signal-Regulated Kinase Signals Are Altered During Positive Versus Negative Thymocyte Selection. J. Immunol. 2001, 167, 4966–4973. [Google Scholar] [CrossRef] [Green Version]
  18. Itano, A.; Salmon, P.; Kioussis, D.; Tolaini, M.; Corbella, P.; Robey, E. The Cytoplasmic Domain of CD4 Promotes the Development of CD4 Lineage T Cells. J. Exp. Med. 1996, 183, 731–741. [Google Scholar] [CrossRef]
  19. Fugger, L.; Svejgaard, A. Association of MHC and Rheumatoid Arthritis: HLA-DR4 and Rheumatoid Arthritis—Studies in Mice and Men. Arthritis. Res. 2000, 2, 208. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, J.; Jelcic, I.; Mühlenbruch, L.; Haunerdinger, V.; Toussaint, N.C.; Zhao, Y.; Cruciani, C.; Faigle, W.; Naghavian, R.; Foege, M.; et al. HLA-DR15 Molecules Jointly Shape an Autoreactive T Cell Repertoire in Multiple Sclerosis. Cell 2020, 183, 1264–1281.e20. [Google Scholar] [CrossRef]
  21. Ooi, J.D.; Petersen, J.; Tan, Y.H.; Huynh, M.; Willett, Z.J.; Ramarathinam, S.H.; Eggenhuizen, P.J.; Loh, K.L.; Watson, K.A.; Gan, P.Y.; et al. Dominant Protection from HLA-Linked Autoimmunity by Antigen-Specific Regulatory T Cells. Nature 2017, 545, 243–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Mamedov, A.; Vorobyeva, N.; Filimonova, I.; Zakharova, M.; Kiselev, I.; Bashinskaya, V.; Baulina, N.; Boyko, A.; Favorov, A.; Kulakova, O.; et al. Protective Allele for Multiple Sclerosis HLA-DRB1*01:01 Provides Kinetic Discrimination of Myelin and Exogenous Antigenic Peptides. Front Immunol. 2020, 10, 3088. [Google Scholar] [CrossRef] [PubMed]
  23. Mamedov, A.E.; Zakharova, M.Y.; Favorova, O.O.; Kulakova, O.G.; Boyko, A.N.; Knorre, V.D.; Vorobieva, N.A.; Khurs, E.N.; Kiselev, I.S.; Baulina, N.M.; et al. Loading Rate of Exogenous and Autoantigenic Determinants on Major Histocompatibility Complex Class II Mediates Resistance to Multiple Sclerosis. Dokl. Biochem. Biophys. 2019, 485, 115–118. [Google Scholar] [CrossRef] [PubMed]
  24. Mamedov, A.E.; Filimonova, I.N.; Smirnov, I.V.; Belogurov, A.A. Peculiarities of the Presentation of the Encephalitogenic MBP Peptide by HLA-DR Complexes Providing Protection and Predisposition to Multiple Sclerosis. Acta Nat. 2021, 13, 127. [Google Scholar] [CrossRef] [PubMed]
  25. Fowler, M.J. Diabetes: Magnitude and Mechanisms. Clin. Diabetes 2010, 28, 42–46. [Google Scholar] [CrossRef] [Green Version]
  26. Pociot, F.; Lernmark, Å. Genetic Risk Factors for Type 1 Diabetes. Lancet 2016, 387, 2331–2339. [Google Scholar] [CrossRef]
  27. Xiaomin, W.; Junbao, Y.; Eddie, J.; I-Ting, C.; Helena, R.; Kwork, W.W. Increased Islet Antigen–Specific Regulatory and Effector CD4+ T Cells in Healthy Individuals with the Type 1 Diabetes–Protective Haplotype. Sci. Immunol. 2020, 5, eaax8767. [Google Scholar] [CrossRef]
  28. Blum, J.S.; Wearsch, P.A.; Cresswell, P. Pathways of Antigen Processing. Annu Rev. Immunol. 2013, 31, 443–473. [Google Scholar] [CrossRef] [Green Version]
  29. Ishina, I.A.; Filimonova, I.N.; Zakharova, M.Y.; Ovchinnikova, L.A.; Mamedov, A.E.; Lomakin, Y.A.; Belogurov, A.A. Exhaustive Search of the Receptor Ligands by the CyCLOPS (Cytometry Cell-Labeling Operable Phage Screening) Technique. Int. J. Mol. Sci. 2020, 21, 6258. [Google Scholar] [CrossRef]
  30. Álvaro-Benito, M.; Freund, C. Revisiting Nonclassical HLA II Functions in Antigen Presentation: Peptide Editing and Its Modulation. HLA 2020, 96, 415–429. [Google Scholar] [CrossRef]
  31. Álvaro-Benito, M.; Morrison, E.; Abualrous, E.T.; Kuropka, B.; Freund, C. Quantification of HLA-DM-Dependent Major Histocompatibility Complex of Class II Immunopeptidomes by the Peptide Landscape Antigenic Epitope Alignment Utility. Front. Immunol. 2018, 9, 872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Vogt, A.B.; Kropshofer, H.; Moldenhauer, G.; Hämmerling, G.J. Kinetic Analysis of Peptide Loading onto HLA-DR Molecules Mediated by HLA-DM. Proc. Natl. Acad. Sci. USA 1996, 93, 9724–9729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Álvaro-Benito, M.; Morrison, E.; Ebner, F.; Abualrous, E.T.; Urbicht, M.; Wieczorek, M.; Freund, C. Distinct Editing Functions of Natural HLA-DM Allotypes Impact Antigen Presentation and CD4+ T Cell Activation. Cell. Mol. Immunol. 2020, 17, 133–142. [Google Scholar] [CrossRef] [PubMed]
  34. Reyes-Vargas, E.; Barker, A.P.; Zhou, Z.; He, X.; Jensen, P.E. HLA-DM Catalytically Enhances Peptide Dissociation by Sensing Peptide–MHC Class II Interactions throughout the Peptide-Binding Cleft. J. Biol. Chem. 2020, 295, 2959–2973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Poluektov, Y.; Kim, A.; Sadegh-Nasseri, S. HLA-DO and Its Role in MHC Class II Antigen Presentation. Front. Immunol. 2013, 4, 260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Nanaware, P.P.; Jurewicz, M.M.; Leszyk, J.D.; Shaffer, S.A.; Stern, L.J. HLA-DO Modulates the Diversity of the MHC-II Self-Peptidome. Mol. Cell. Proteom. 2019, 18, 490–503. [Google Scholar] [CrossRef] [Green Version]
  37. Surh, C.D.; Lee, D.-S.; Fung-Leung, W.; Karlsson, L.; Sprent, J. Thymic Selection by a Single MHC/Peptide Ligand Produces a Semidiverse Repertoire of CD4+ T Cells. Immunity 1997, 7, 209–219. [Google Scholar] [CrossRef] [Green Version]
  38. Olsson, N.; Jiang, W.; Adler, L.N.; Mellins, E.D.; Elias, J.E. Tuning DO: DM Ratios Modulates MHC Class II Immunopeptidomes. Mol. Cell. Proteom. 2022, 21, 100204. [Google Scholar] [CrossRef]
  39. Zhou, Z.; Reyes-Vargas, E.; Escobar, H.; Chang, K.Y.; Barker, A.P.; Rockwood, A.L.; Delgado, J.C.; He, X.; Jensen, P.E. Peptidomic Analysis of Type 1 Diabetes Associated HLA-DQ Molecules and the Impact of HLA-DM on Peptide Repertoire Editing. Eur. J. Immunol. 2017, 47, 314–326. [Google Scholar] [CrossRef]
  40. Collado, J.A.; Alvarez, I.; Ciudad, M.T.; Espinosa, G.; Canals, F.; Pujol-Borrell, R.; Carrascal, M.; Abian, J.; Jaraquemada, D. Composition of the HLA-DR-Associated Human Thymus Peptidome. Eur. J. Immunol. 2013, 43, 2273–2282. [Google Scholar] [CrossRef]
  41. Nielsen, M.; Justesen, S.; Lund, O.; Lundegaard, C.; Buus, S. NetMHCIIpan-2.0—Improved Pan-Specific HLA-DR Predictions Using a Novel Concurrent Alignment and Weight Optimization Training Procedure. Immunome Res. 2010, 6, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Muixí, L.; Carrascal, M.; Alvarez, I.; Daura, X.; Martí, M.; Armengol, M.P.; Pinilla, C.; Abian, J.; Pujol-Borrell, R.; Jaraquemada, D. Thyroglobulin Peptides Associate In Vivo to HLA-DR in Autoimmune Thyroid Glands. J. Immunol. 2008, 181, 795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Fissolo, N.; Haag, S.; de Graaf, K.L.; Drews, O.; Stevanovic, S.; Rammensee, H.G.; Weissert, R. Naturally Presented Peptides on Major Histocompatibility Complex I and II Molecules Eluted from Central Nervous System of Multiple Sclerosis Patients. Mol. Cell. Proteom. 2009, 8, 2090–2101. [Google Scholar] [CrossRef] [Green Version]
  44. Martinsen, V.; Kursula, P. Multiple Sclerosis and Myelin Basic Protein: Insights into Protein Disorder and Disease. Amino. Acids 2022, 54, 99–109. [Google Scholar] [CrossRef] [PubMed]
  45. Belogurov, A.A.; Kurkova, I.N.; Friboulet, A.; Thomas, D.; Misikov, V.K.; Zakharova, M.Y.; Suchkov, S.V.; Kotov, S.V.; Alehin, A.I.; Avalle, B.; et al. Recognition and Degradation of Myelin Basic Protein Peptides by Serum Autoantibodies: Novel Biomarker for Multiple Sclerosis. J. Immunol. 2008, 180, 1258–1267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Belogurov, A.; Zakharov, K.; Lomakin, Y.; Surkov, K.; Avtushenko, S.; Kruglyakov, P.; Smirnov, I.; Makshakov, G.; Lockshin, C.; Gregoriadis, G.; et al. CD206-Targeted Liposomal Myelin Basic Protein Peptides in Patients with Multiple Sclerosis Resistant to First-Line Disease-Modifying Therapies: A First-in-Human, Proof-of-Concept Dose-Escalation Study. Neurotherapeutics 2016, 13, 895–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Nokoff, N.J.; Rewers, M.; Cree Green, M. The Interplay of Autoimmunity and Insulin Resistance in Type 1 Diabetes. Discov. Med. 2012, 13, 115–122. [Google Scholar]
  48. Ito, Y.; Ashenberg, O.; Pyrdol, J.; Luoma, A.M.; Rozenblatt-Rosen, O.; Hofree, M.; Christian, E.; Ferrari de Andrade, L.; Tay, R.E.; Teyton, L.; et al. Rapid CLIP Dissociation from MHC II Promotes an Unusual Antigen Presentation Pathway in Autoimmunity. J. Exp. Med. 2018, 215, 2617–2635. [Google Scholar] [CrossRef]
  49. Busch, R.; Kollnberger, S.; Mellins, E.D. HLA Associations in Inflammatory Arthritis: Emerging Mechanisms and Clinical Implications. Nat. Rev. Rheumatol. 2019, 15, 364–381. [Google Scholar] [CrossRef] [Green Version]
  50. Markovic-Plese, S.; Fukaura, H.; Zhang, J.; al-Sabbagh, A.; Southwood, S.; Sette, A.; Kuchroo, V.K.; Hafler, D.A. T Cell Recognition of Immunodominant and Cryptic Proteolipid Protein Epitopes in Humans. J. Immunol. 1995, 155, 982. [Google Scholar] [CrossRef]
  51. Nakayama, M.; Beilke, J.N.; Jasinski, J.M.; Kobayashi, M.; Miao, D.; Li, M.; Coulombe, M.G.; Liu, E.; Elliott, J.F.; Gill, R.G.; et al. Priming and Effector Dependence on Insulin B:9-23 Peptide in NOD Islet Autoimmunity. J. Clin. Investig. 2007, 117, 1835–1843. [Google Scholar] [CrossRef] [PubMed]
  52. Levisetti, M.G.; Suri, A.; Petzold, S.J.; Unanue, E.R. The Insulin-Specific T Cells of Nonobese Diabetic Mice Recognize a Weak MHC-Binding Segment in More Than One Form. J. Immunol. 2007, 178, 6051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Mohan, J.F.; Petzold, S.J.; Unanue, E.R. Register Shifting of an Insulin Peptide-MHC Complex Allows Diabetogenic T Cells to Escape Thymic Deletion. J. Exp. Med. 2011, 208, 2375–2383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Stadinski, B.D.; Zhang, L.; Crawford, F.; Marrack, P.; Eisenbarth, G.S.; Kappler, J.W. Diabetogenic T Cells Recognize Insulin Bound to IAg7 in an Unexpected, Weakly Binding Register. Proc. Natl. Acad. Sci. USA 2010, 107, 10978–10983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Frances, C.; Brian, S.; Niyun, J.; Aaron, M.; Maki, N.; Philip, P.; Philippa, M.; George, E.; Kappler, J.W. Specificity and Detection of Insulin-Reactive CD4+ T Cells in Type 1 Diabetes in the Nonobese Diabetic (NOD) Mouse. Proc. Natl. Acad. Sci. USA 2011, 108, 16729–16734. [Google Scholar] [CrossRef] [Green Version]
  56. Junbao, Y.; I-Ting, C.; Tomasz, S.; Nadia, T.-C.; Carla, J.G.; James, E.A.; Kappler, J.W.; Davidson, H.W.; Kwork, W.W. Autoreactive T Cells Specific for Insulin B:11-23 Recognize a Low-Affinity Peptide Register in Human Subjects with Autoimmune Diabetes. Proc. Natl. Acad. Sci. USA 2014, 111, 14840–14845. [Google Scholar] [CrossRef] [Green Version]
  57. Wan, X.; Vomund, A.N.; Peterson, O.J.; Chervonsky, A.V.; Lichti, C.F.; Unanue, E.R. The MHC-II Peptidome of Pancreatic Islets Identifies Key Features of Autoimmune Peptides. Nat. Immunol. 2020, 21, 455–463. [Google Scholar] [CrossRef]
  58. Loyal, L.; Braun, J.; Henze, L.; Kruse, B.; Dingeldey, M.; Reimer, U.; Kern, F.; Schwarz, T.; Mangold, M.; Unger, C.; et al. Cross-Reactive CD4+ T Cells Enhance SARS-CoV-2 Immune Responses upon Infection and Vaccination. Science (1979) 2022, 374, eabh1823. [Google Scholar] [CrossRef]
  59. Gao, Y.; Cai, C.; Grifoni, A.; Müller, T.R.; Niessl, J.; Olofsson, A.; Humbert, M.; Hansson, L.; Österborg, A.; Bergman, P.; et al. Ancestral SARS-CoV-2-Specific T Cells Cross-Recognize the Omicron Variant. Nat. Med. 2022, 28, 472–476. [Google Scholar] [CrossRef]
  60. Molodtsov, I.A.; Kegeles, E.; Mitin, A.N.; Mityaeva, O.; Musatova, O.E.; Panova, A.E.; Pashenkov, M.V.; Peshkova, I.O.; Alsalloum, A.; Asaad, W.; et al. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)–Specific T Cells and Antibodies in Coronavirus Disease 2019 (COVID-19) Protection: A Prospective Study. Clin. Infect. Dis. 2022, 75, e1–e9. [Google Scholar] [CrossRef]
  61. Yiyuan, Y.; Xiang, W.X.; Mariuzza, R.A.A. Crystal Structure of a Complete Ternary Complex of T-Cell Receptor, Peptide–MHC, and CD4. Proc. Natl. Acad. Sci. USA 2012, 109, 5405–5410. [Google Scholar] [CrossRef] [PubMed]
  62. Zareie, P.; Farenc, C.; la Gruta, N.L. MHC Restriction: Where Are We Now? Viral. Immunol. 2020, 33, 179–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Mørch, A.M.; Bálint, Š.; Santos, A.M.; Davis, S.J.; Dustin, M.L. Coreceptors and TCR Signaling—The Strong and the Weak of It. Front Cell. Dev. Biol. 2020, 8, 597627. [Google Scholar] [CrossRef] [PubMed]
  64. Rudolph, M.G.; Stanfield, R.L.; Wilson, I.A. How TCRs bind MHCs, peptides, and coreceptors. Annu. Rev. Immunol. 2006, 24, 419–466. [Google Scholar] [CrossRef] [PubMed]
  65. Rossjohn, J.; Gras, S.; Miles, J.J.; Turner, S.J.; Godfrey, D.I.; McCluskey, J. T Cell Antigen Receptor Recognition of Antigen-Presenting Molecules. Annu. Rev. Immunol. 2015, 33, 169–200. [Google Scholar] [CrossRef]
  66. Hennecke, J.; Carfi, A.; Wiley, D.C. Structure of a Covalently Stabilized Complex of a Human Aβ T-Cell Receptor, Influenza HA Peptide and MHC Class II Molecule, HLA-DR1. EMBO J. 2000, 19, 5611–5624. [Google Scholar] [CrossRef] [Green Version]
  67. Kato, Z.; Stern, J.N.H.; Nakamura, H.K.; Kuwata, K.; Kondo, N.; Strominger, J.L. Positioning of Autoimmune TCR-Ob.2F3 and TCR-Ob.3D1 on the MBP85–99/HLA-DR2 Complex. Proc. Natl. Acad. Sci. USA 2008, 105, 15523–15528. [Google Scholar] [CrossRef] [Green Version]
  68. Li, Y.; Huang, Y.; Lue, J.; Quandt, J.A.; Martin, R.; Mariuzza, R.A. Structure of a Human Autoimmune TCR Bound to a Myelin Basic Protein Self-Peptide and a Multiple Sclerosis-Associated MHC Class II Molecule. EMBO J. 2005, 24, 2968–2979. [Google Scholar] [CrossRef] [Green Version]
  69. Kong, Y.; Jing, Y.; Allard, D.; Scavuzzo, M.A.; Sprouse, M.L.; Borowiak, M.; Bettini, M.L.; Bettini, M. A Dormant T-Cell Population with Autoimmune Potential Exhibits Low Self-Reactivity and Infiltrates Islets in Type 1 Diabetes. Eur. J. Immunol. 2022, 52, 1158–1170. [Google Scholar] [CrossRef]
  70. Sethi, D.K.; Schubert, D.A.; Anders, A.-K.; Heroux, A.; Bonsor, D.A.; Thomas, C.P.; Sundberg, E.J.; Pyrdol, J.; Wucherpfennig, K.W. A Highly Tilted Binding Mode by a Self-Reactive T Cell Receptor Results in Altered Engagement of Peptide and MHC. J. Exp. Med. 2011, 208, 91–102. [Google Scholar] [CrossRef] [Green Version]
  71. Adams, J.J.; Narayanan, S.; Liu, B.; Birnbaum, M.E.; Kruse, A.C.; Bowerman, N.A.; Chen, W.; Levin, A.M.; Connolly, J.M.; Zhu, C.; et al. T Cell Receptor Signaling Is Limited by Docking Geometry to Peptide-Major Histocompatibility Complex. Immunity 2011, 35, 681–693. [Google Scholar] [CrossRef] [PubMed]
  72. Yin, Y.; Li, Y.; Kerzic, M.C.; Martin, R.; Mariuzza, R.A. Structure of a TCR with High Affinity for Self-Antigen Reveals Basis for Escape from Negative Selection. EMBO J. 2011, 30, 1137–1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Maynard, J.; Petersson, K.; Wilson, D.H.; Adams, E.J.; Blondelle, S.E.; Boulanger, M.J.; Wilson, D.B.; Garcia, K.C. Structure of an Autoimmune T Cell Receptor Complexed with Class II Peptide-MHC: Insights into MHC Bias and Antigen Specificity. Immunity 2005, 22, 81–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Feng, D.; Bond, C.J.; Ely, L.K.; Maynard, J.; Garcia, K.C. Structural Evidence for a Germline-Encoded T Cell Receptor–Major Histocompatibility Complex Interaction “Codon”. Nat. Immunol. 2007, 8, 975–983. [Google Scholar] [CrossRef]
  75. Zamvil, S.S.; Mitchell, D.J.; Moore, A.C.; Kitamura, K.; Steinman, L.; Rothbard, J.B. T-Cell Epitope of the Autoantigen Myelin Basic Protein That Induces Encephalomyelitis. Nature 1986, 324, 258–260. [Google Scholar] [CrossRef]
  76. Raposo, B.; Merky, P.; Lundqvist, C.; Yamada, H.; Urbonaviciute, V.; Niaudet, C.; Viljanen, J.; Kihlberg, J.; Kyewski, B.; Ekwall, O.; et al. T Cells Specific for Post-Translational Modifications Escape Intrathymic Tolerance Induction. Nat. Commun. 2018, 9, 353. [Google Scholar] [CrossRef] [Green Version]
  77. Carayanniotis, G. Recognition of Thyroglobulin by T Cells: The Role of Iodine. Thyroid 2007, 17, 963–973. [Google Scholar] [CrossRef]
  78. Kwon, E.-J.; Ju, J.H. Impact of Posttranslational Modification in Pathogenesis of Rheumatoid Arthritis: Focusing on Citrullination, Carbamylation, and Acetylation. Int. J. Mol. Sci. 2021, 22, 10576. [Google Scholar] [CrossRef]
  79. Hill, J.A.; Southwood, S.; Sette, A.; Jevnikar, A.M.; Bell, D.A.; Cairns, E. Cutting Edge: The Conversion of Arginine to Citrulline Allows for a High-Affinity Peptide Interaction with the Rheumatoid Arthritis-Associated HLA-DRB1*0401 MHC Class II Molecule. J. Immunol. 2003, 171, 538. [Google Scholar] [CrossRef] [Green Version]
  80. Scherer, H.U.; Häupl, T.; Burmester, G.R. The Etiology of Rheumatoid Arthritis. J. Autoimmun. 2020, 110, 102400. [Google Scholar] [CrossRef]
  81. Lim, J.J.; Jones, C.M.; Loh, T.J.; Ting, Y.T.; Zareie, P.; Loh, K.L.; Rossjohn, J.; Felix, N.; Supi, A.; Baker, D.G. The Shared Susceptibility Epitope of HLA-DR4 Binds Citrullinated Self-Antigens and the TCR. Sci. Immunol. 2021, 6, eabe0896. [Google Scholar] [CrossRef] [PubMed]
  82. Song, J.; Schwenzer, A.; Wong, A.; Turcinov, S.; Rims, C.; Martinez, L.R.; Arribas-Layton, D.; Gerstner, C.; Muir, V.S.; Midwood, K.S. Shared Recognition of Citrullinated Tenascin-C Peptides by T and B Cells in Rheumatoid Arthritis. JCI Insight 2021, 6, e145217. [Google Scholar] [CrossRef] [PubMed]
  83. Ge, C.; Weisse, S.; Xu, B.; Dobritzsch, D.; Viljanen, J.; Kihlberg, J.; Do, N.-N.; Schneider, N.; Lanig, H.; Holmdahl, R.; et al. Key Interactions in the Trimolecular Complex Consisting of the Rheumatoid Arthritis—Associated DRB1 * 04:01 Molecule, the Major Glycosylated Collagen II Peptide and the T-Cell Receptor. Ann. Rheum. Dis. 2022, 81, 480. [Google Scholar] [CrossRef]
  84. McGinty, J.W.; Chow, I.-T.; Greenbaum, C.; Odegard, J.; Kwok, W.W.; James, E.A. Recognition of Posttranslationally Modified GAD65 Epitopes in Subjects With Type 1 Diabetes. Diabetes 2014, 63, 3033–3040. [Google Scholar] [CrossRef] [Green Version]
  85. Yang, M.-L.; Horstman, S.; Gee, R.; Guyer, P.; Lam, T.T.; Kanyo, J.; Perdigoto, A.L.; Speake, C.; Greenbaum, C.J.; Callebaut, A.; et al. Citrullination of Glucokinase Is Linked to Autoimmune Diabetes. Nat. Commun. 2022, 13, 1870. [Google Scholar] [CrossRef] [PubMed]
  86. Mannering, S.I.; Harrison, L.C.; Williamson, N.A.; Morris, J.S.; Thearle, D.J.; Jensen, K.P.; Kay, T.W.H.; Rossjohn, J.; Falk, B.A.; Nepom, G.T.; et al. The Insulin A-Chain Epitope Recognized by Human T Cells Is Posttranslationally Modified. J. Exp. Med. 2005, 202, 1191–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Cao, L.; Sun, D.; Whitaker, J.N. Citrullinated Myelin Basic Protein Induces Experimental Autoimmune Encephalomyelitis in Lewis Rats through a Diverse T Cell Repertoire. J. Neuroimmunol. 1998, 88, 21–29. [Google Scholar] [CrossRef]
  88. Carrillo-Vico, A.; Leech, M.D.; Anderton, S.M. Contribution of Myelin Autoantigen Citrullination to T Cell Autoaggression in the Central Nervous System. J. Immunol. 2010, 184, 2839. [Google Scholar] [CrossRef] [Green Version]
  89. Valdivia, A.O.; Agarwal, P.K.; Bhattacharya, S.K. Myelin Basic Protein Phospholipid Complexation Likely Competes with Deimination in Experimental Autoimmune Encephalomyelitis Mouse Model. ACS Omega 2020, 5, 15454–15467. [Google Scholar] [CrossRef]
  90. Mitchell, A.M.; Alkanani, A.A.; McDaniel, K.A.; Pyle, L.; Waugh, K.; Steck, A.K.; Nakayama, M.; Yu, L.; Gottlieb, P.A.; Rewers, M.J.; et al. T-Cell Responses to Hybrid Insulin Peptides Prior to Type 1 Diabetes Development. Proc. Natl. Acad. Sci. USA 2021, 118, e2019129118. [Google Scholar] [CrossRef]
  91. Aaron Wiles, T.; Powell, R.; Michel, C.R.; Scott Beard, K.; Hohenstein, A.; Bradley, B.; Reisdorph, N.; Haskins, K.; Delong, T. Identification of Hybrid Insulin Peptides (HIPs) in Mouse and Human Islets by Mass Spectrometry. J. Proteome Res. 2019, 18, 814–825. [Google Scholar] [CrossRef] [PubMed]
  92. Baker, R.L.; Jamison, B.L.; Wiles, T.A.; Lindsay, R.S.; Barbour, G.; Bradley, B.; Delong, T.; Friedman, R.S.; Nakayama, M.; Haskins, K. CD4 T Cells Reactive to Hybrid Insulin Peptides Are Indicators of Disease Activity in the NOD Mouse. Diabetes 2018, 67, 1836–1846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Baker, R.L.; Rihanek, M.; Hohenstein, A.C.; Nakayama, M.; Michels, A.; Gottlieb, P.A.; Haskins, K.; Delong, T. Hybrid Insulin Peptides Are Autoantigens in Type 1 Diabetes. Diabetes 2019, 68, 1830–1840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Wiles, T.A.; Hohenstein, A.; Landry, L.G.; Dang, M.; Powell, R.; Guyer, P.; James, E.A.; Nakayama, M.; Haskins, K.; Delong, T.; et al. Characterization of Human CD4 T Cells Specific for a C-Peptide/C-Peptide Hybrid Insulin Peptide. Front. Immunol. 2021, 12, 668680. [Google Scholar] [CrossRef] [PubMed]
  95. Delong, T.; Wiles, T.A.; Baker, R.L.; Bradley, B.; Barbour, G.; Reisdorph, R.; Haskins, K.; Elso, C.M.; Kumer, N.; Kent, S.C.; et al. Pathogenic CD4 T Cells in Type 1 Diabetes Recognize Epitopes Formed by Peptide Fusion. Science (1979) 2016, 351, 711–714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Arribas-Layton, D.; Guyer, P.; Delong, T.; Dang, M.; Chow, I.-T.; Speake, C.; Greenbaum, C.J.; Kwok, W.W.; Baker, R.L.; Haskins, K.; et al. Hybrid Insulin Peptides Are Recognized by Human T Cells in the Context of DRB1 * 04:01. Diabetes 2020, 69, 1492–1502. [Google Scholar] [CrossRef]
  97. Tran, M.T.; Faridi, P.; Lim, J.J.; Ting, Y.T.; Onwukwe, G.; Bhattacharjee, P.; Jones, C.M.; Tresoldi, E.; Cameron, F.J.; la Gruta, N.L.; et al. T Cell Receptor Recognition of Hybrid Insulin Peptides Bound to HLA-DQ8. Nat. Commun. 2021, 12, 5110. [Google Scholar] [CrossRef]
  98. Christen, U.; von Herrath, M.G. Infections and Autoimmunity—Good or Bad? J. Immunol. 2005, 174, 7481. [Google Scholar] [CrossRef] [Green Version]
  99. Markovic-Plese, S.; Hemmer, B.; Zhao, Y.; Simon, R.; Pinilla, C.; Martin, R. High Level of Cross-Reactivity in Influenza Virus Hemagglutinin-Specific CD4+ T-Cell Response: Implications for the Initiation of Autoimmune Response in Multiple Sclerosis. J. Neuroimmunol. 2005, 169, 31–38. [Google Scholar] [CrossRef]
  100. Hiemstra, H.S.; Schloot, N.C.; van Veelen, P.A.; Willemen, S.J.M.; Franken, K.L.M.C.; van Rood, J.J.; de Vries, R.R.P.; Chaudhuri, A.; Behan, P.O.; Drijfhout, J.W.; et al. Cytomegalovirus in Autoimmunity: T Cell Crossreactivity to Viral Antigen and Autoantigen Glutamic Acid Decarboxylase. Proc. Natl. Acad. Sci. USA 2001, 98, 3988–3991. [Google Scholar] [CrossRef] [Green Version]
  101. Bellucci, G.; Rinaldi, V.; Buscarinu, M.C.; Reniè, R.; Bigi, R.; Pellicciari, G.; Morena, E.; Romano, C.; Marrone, A.; Mechelli, R.; et al. Multiple Sclerosis and SARS-CoV-2: Has the Interplay Started? Front. Immunol. 2021, 12, 755333. [Google Scholar] [CrossRef] [PubMed]
  102. Satheesh, N.J.; Salloum-Asfar, S.; Abdulla, S.A. The Potential Role of COVID-19 in the Pathogenesis of Multiple Sclerosis—A Preliminary Report. Viruses 2021, 13, 2091. [Google Scholar] [CrossRef] [PubMed]
  103. Brennan, F.M.; Smith, N.M.G.; Owen, S.; Li, C.; Amjadi, P.; Green, P.; Andersson, A.; Palfreeman, A.C.; Hillyer, P.; Foey, A.; et al. Resting CD4+ effector Memory T Cells Are Precursors of Bystander-Activated Effectors: A Surrogate Model of Rheumatoid Arthritis Synovial T-Cell Function. Arthritis Res. 2008, 10, R36. [Google Scholar] [CrossRef] [Green Version]
  104. Martino, G.; Grohovaz, F.; Brambilla, E.; Codazzi, F.; Consiglio, A.; Clementi, E.; Filippi, M.; Comi, G.; Grimaldi, L.M.E. Proinflammatory Cytokines Regulate Antigen-Independent T-Cell Activation by Two Separate Calcium-Signaling Pathways in Multiple Sclerosis Patients. Ann. Neurol. 1998, 43, 340–349. [Google Scholar] [CrossRef]
  105. Steimle, V.; Siegrist, C.-A.; Mottet, A.; Lisowska-Grospierre, B.; Mach, B. Regulation of MHC Class II Expression by Interferon-γ Mediated by the Transactivator Gene CIITA. Science (1979) 1994, 265, 106–109. [Google Scholar] [CrossRef] [PubMed]
  106. Iglesias, B.M.; Cerase, J.; Ceracchini, C.; Levi, G.; Aloisi, F. Analysis of B7-1 and B7-2 Costimulatory Ligands in Cultured Mouse Microglia: Upregulation by Interferon-γ and Lipopolysaccharide and Downregulation by Interleukin-10, Prostaglandin E2 and Cyclic AMP-Elevating Agents. J. Neuroimmunol. 1997, 72, 83–93. [Google Scholar] [CrossRef] [PubMed]
  107. Nikcevich, K.M.; Gordon, K.B.; Tan, L.; Hurst, S.D.; Kroepfl, J.F.; Gardinier, M.; Barrett, T.A.; Miller, S.D. IFN-Gamma-Activated Primary Murine Astrocytes Express B7 Costimulatory Molecules and Prime Naive Antigen-Specific T Cells. J. Immunol. 1997, 158, 614. [Google Scholar] [CrossRef]
  108. Russell, M.A.; Redick, S.D.; Blodgett, D.M.; Richardson, S.J.; Leete, P.; Krogvold, L.; Dahl-Jørgensen, K.; Bottino, R.; Brissova, M.; Spaeth, J.M.; et al. HLA Class II Antigen Processing and Presentation Pathway Components Demonstrated by Transcriptome and Protein Analyses of Islet β-Cells From Donors With Type 1 Diabetes. Diabetes 2019, 68, 988–1001. [Google Scholar] [CrossRef] [Green Version]
  109. Quesada-Masachs, E.; Zilberman, S.; Rajendran, S.; Chu, T.; McArdle, S.; Kiosses, W.B.; Lee, J.-H.M.; Yesildag, B.; Benkahla, M.A.; Pawlowska, A.; et al. Upregulation of HLA Class II in Pancreatic Beta Cells from Organ Donors with Type 1 Diabetes. Diabetologia 2022, 65, 387–401. [Google Scholar] [CrossRef]
  110. Zhao, Y.; Scott, N.A.; Quah, H.S.; Krishnamurthy, B.; Bond, F.; Loudovaris, T.; Mannering, S.I.; Kay, T.W.H.; Thomas, H.E. Mouse Pancreatic Beta Cells Express MHC Class II and Stimulate CD4+ T Cells to Proliferate. Eur. J. Immunol. 2015, 45, 2494–2503. [Google Scholar] [CrossRef]
  111. Scott, N.A.; Zhao, Y.; Krishnamurthy, B.; Mannering, S.I.; Kay, T.W.H.; Thomas, H.E. IFNγ-Induced MHC Class II Expression on Islet Endothelial Cells Is an Early Marker of Insulitis but Is Not Required for Diabetogenic CD4+ T Cell Migration. Front. Immunol. 2018, 9, 2800. [Google Scholar] [CrossRef] [PubMed]
  112. Gobin, S.J.P.; Montagne, L.; van Zutphen, M.; van der Valk, P.; van den Elsen, P.J.; de Groot, C.J.A. Upregulation of Transcription Factors Controlling MHC Expression in Multiple Sclerosis Lesions. Glia 2001, 36, 68–77. [Google Scholar] [CrossRef] [PubMed]
  113. Falcão, A.M.; van Bruggen, D.; Marques, S.; Meijer, M.; Jäkel, S.; Agirre, E.; Samudyata; Floriddia, E.M.; Vanichkina, D.P.; ffrench-Constant, C.; et al. Disease-Specific Oligodendrocyte Lineage Cells Arise in Multiple Sclerosis. Nat. Med. 2018, 24, 1837–1844. [Google Scholar] [CrossRef] [PubMed]
  114. Kanazawa, S.; Ota, S.; Sekine, C.; Tada, T.; Otsuka, T.; Okamoto, T.; Sønderstrup, G.; Peterlin, B.M. Aberrant MHC Class II Expression in Mouse Joints Leads to Arthritis with Extraarticular Manifestations Similar to Rheumatoid Arthritis. Proc. Natl. Acad. Sci. USA 2006, 103, 14465–14470. [Google Scholar] [CrossRef] [Green Version]
  115. Fasano, R.; Malerba, E.; Prete, M.; Solimando, A.G.; Buonavoglia, A.; Silvestris, N.; Leone, P.; Racanelli, V. Impact of Antigen Presentation Mechanisms on Immune Response in Autoimmune Hepatitis. Front. Immunol. 2022, 12, 814155. [Google Scholar] [CrossRef]
  116. Serreze, D.V.; Wasserfall, C.; Ottendorfer, E.W.; Stalvey, M.; Pierce, M.A.; Gauntt, C.; O’Donnell, B.; Flanagan, J.B.; Campbell-Thompson, M.; Ellis, T.M.; et al. Diabetes Acceleration or Prevention by a Coxsackievirus B4 Infection: Critical Requirements for Both Interleukin-4 and Gamma Interferon. J. Virol. 2005, 79, 1045–1052. [Google Scholar] [CrossRef] [Green Version]
  117. Nekoua, M.P.; Alidjinou, E.K.; Hober, D. Persistent Coxsackievirus B Infection and Pathogenesis of Type 1 Diabetes Mellitus. Nat. Rev. Endocrinol. 2022, 18, 503–516. [Google Scholar] [CrossRef]
  118. Isaacs, S.R.; Foskett, D.B.; Maxwell, A.J.; Ward, E.J.; Faulkner, C.L.; Luo, J.Y.X.; Rawlinson, W.D.; Craig, M.E.; Kim, K.W. Viruses and Type 1 Diabetes: From Enteroviruses to the Virome. Microorganisms 2021, 9, 1519. [Google Scholar] [CrossRef]
  119. Gabibov, A.G.; Belogurov, A.A., Jr.; Lomakin, Y.A.; Zakharova, M.Y.; Avakyan, M.E.; Dubrovskaya, V.V.; Smirnov, I.V.; Ivanov, A.S.; Molnar, A.A.; Gurtsevitch, V.E.; et al. Combinatorial Antibody Library from Multiple Sclerosis Patients Reveals Antibodies That Cross-React with Myelin Basic Protein and EBV Antigen. FASEB J. 2011, 25, 4211–4221. [Google Scholar] [CrossRef]
  120. Abdul-Ghaffar, A.Y.; Samaha, D.Y.; Wahba, N.S.; Hussein, A.E.A. Relation between Epstein-Barr Virus Infection and Multiple Sclerosis. QJM: Int. J. Med. 2021, 114, hcab091. [Google Scholar] [CrossRef]
  121. Tao, C.; Simpson-Yap, S.; Taylor, B.; Blizzard, L.; Lucas, R.; Ponsonby, A.-L.; Broadley, S.; van der Mei, I. Markers of Epstein-Barr Virus and Human Herpesvirus-6 Infection and Multiple Sclerosis Clinical Progression. Mult. Scler. Relat. Disord. 2022, 59, 103561. [Google Scholar] [CrossRef] [PubMed]
  122. Lanz, T.V.; Brewer, R.C.; Ho, P.P.; Moon, J.-S.; Jude, K.M.; Fernandez, D.; Fernandes, R.A.; Gomez, A.M.; Nadj, G.-S.; Bartley, C.M.; et al. Clonally Expanded B Cells in Multiple Sclerosis Bind EBV EBNA1 and GlialCAM. Nature 2022, 603, 321–327. [Google Scholar] [CrossRef] [PubMed]
  123. Smatti, M.K.; Cyprian, F.S.; Nasrallah, G.K.; al Thani, A.A.; Almishal, R.O.; Yassine, H.M. Viruses and Autoimmunity: A Review on the Potential Interaction and Molecular Mechanisms. Viruses 2019, 11, 762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Tengvall, K.; Huang, J.; Hellström, C.; Kammer, P.; Biström, M.; Ayoglu, B.; Bomfim, I.L.; Stridh, P.; Butt, J.; Brenner, N.; et al. Molecular Mimicry between Anoctamin 2 and Epstein-Barr Virus Nuclear Antigen 1 Associates with Multiple Sclerosis Risk. Proc. Natl. Acad. Sci. USA 2019, 116, 16955–16960. [Google Scholar] [CrossRef] [Green Version]
  125. Wucherpfennig, K.W.; Strominger, J.L. Molecular Mimicry in T Cell-Mediated Autoimmunity: Viral Peptides Activate Human T Cell Clones Specific for Myelin Basic Protein. Cell 1995, 80, 695–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Lünemann, J.D.; Jelčić, I.; Roberts, S.; Lutterotti, A.; Tackenberg, B.; Martin, R.; Munz, C. EBNA1-Specific T Cells from Patients with Multiple Sclerosis Cross React with Myelin Antigens and Co-Produce IFN-γ and IL-2. J. Exp. Med. 2008, 205, 1763–1773. [Google Scholar] [CrossRef] [Green Version]
  127. Zdimerova, H.; Murer, A.; Engelmann, C.; Raykova, A.; Deng, Y.; Gujer, C.; Rühl, J.; McHugh, D.; Caduff, N.; Naghavian, R.; et al. Attenuated Immune Control of Epstein–Barr Virus in Humanized Mice Is Associated with the Multiple Sclerosis Risk Factor HLA-DR15. Eur. J. Immunol. 2021, 51, 64–75. [Google Scholar] [CrossRef]
  128. Latorre, D.; Kallweit, U.; Armentani, E.; Foglierini, M.; Mele, F.; Cassotta, A.; Jovic, S.; Jarrossay, D.; Mathis, J.; Zellini, F.; et al. T Cells in Patients with Narcolepsy Target Self-Antigens of Hypocretin Neurons. Nature 2018, 562, 63–68. [Google Scholar] [CrossRef]
  129. Luo, G.; Ambati, A.; Lin, L.; Bonvalet, M.; Partinen, M.; Ji, X.; Maecker, H.T.; Mignot, E.J.-M. Autoimmunity to Hypocretin and Molecular Mimicry to Flu in Type 1 Narcolepsy. Proc. Natl. Acad. Sci. USA 2018, 115, E12323–E12332. [Google Scholar] [CrossRef] [Green Version]
  130. Vuorela, A.; Freitag, T.L.; Leskinen, K.; Pessa, H.; Härkönen, T.; Stracenski, I.; Kirjavainen, T.; Olsen, P.; Saarenpää-Heikkilä, O.; Ilonen, J.; et al. Enhanced Influenza A H1N1 T Cell Epitope Recognition and Cross-Reactivity to Protein-O-Mannosyltransferase 1 in Pandemrix-Associated Narcolepsy Type 1. Nat. Commun. 2021, 12, 2283. [Google Scholar] [CrossRef]
  131. Ooi, J.D.; Jiang, J.-H.; Eggenhuizen, P.J.; Chua, L.L.; van Timmeren, M.; Loh, K.L.; O’Sullivan, K.M.; Gan, P.Y.; Zhong, Y.; Tsyganov, K.; et al. A Plasmid-Encoded Peptide from Staphylococcus Aureus Induces Anti-Myeloperoxidase Nephritogenic Autoimmunity. Nat. Commun. 2019, 10, 3392. [Google Scholar] [CrossRef]
  132. Ellis, N.M.J.; Li, Y.; Hildebrand, W.; Fischetti, V.A.; Cunningham, M.W. T Cell Mimicry and Epitope Specificity of Cross-Reactive T Cell Clones from Rheumatic Heart Disease. J. Immunol. 2005, 175, 5448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Moten, D.; Teneva, I.; Apostolova, D.; Batsalova, T.; Dzhambazov, B. Molecular Mimicry of the Rheumatoid Arthritis-Related Immunodominant T-Cell Epitope within Type II Collagen (CII260-270) by the Bacterial L-Asparaginase. Int. J. Mol. Sci. 2022, 23, 9149. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, Y.; Lin, J.; Xiao, L.; Zhang, X.; Zhao, L.; Wang, M.; Li, L. Gut Microbiota in Systemic Lupus Erythematosus: A Fuse and a Solution. J. Autoimmun. 2022, 132, 102867. [Google Scholar] [CrossRef] [PubMed]
  135. Garabatos, N.; Santamaria, P. Gut Microbial Antigenic Mimicry in Autoimmunity. Front. Immunol. 2022, 13, 873607. [Google Scholar] [CrossRef] [PubMed]
  136. Russell, J.T.; Roesch, L.F.W.; Ördberg, M.; Ilonen, J.; Atkinson, M.A.; Schatz, D.A.; Triplett, E.W.; Ludvigsson, J. Genetic Risk for Autoimmunity Is Associated with Distinct Changes in the Human Gut Microbiome. Nat. Commun. 2019, 10, 3621. [Google Scholar] [CrossRef] [Green Version]
  137. Silverman, M.; Kua, L.; Tanca, A.; Pala, M.; Palomba, A.; Tanes, C.; Bittinger, K.; Uzzau, S.; Benoist, C.; Mathis, D. Protective Major Histocompatibility Complex Allele Prevents Type 1 Diabetes by Shaping the Intestinal Microbiota Early in Ontogeny. Proc. Natl. Acad. Sci. USA 2017, 114, 9671–9676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Mullaney, J.A.; Stephens, J.E.; Costello, M.-E.; Fong, C.; Geeling, B.E.; Gavin, P.G.; Wright, C.M.; Spector, T.D.; Brown, M.A.; Hamilton-Williams, E.E. Type 1 Diabetes Susceptibility Alleles Are Associated with Distinct Alterations in the Gut Microbiota. Microbiome 2018, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  139. Miyauchi, E.; Kim, S.-W.; Suda, W.; Kawasumi, M.; Onawa, S.; Taguchi-Atarashi, N.; Morita, H.; Taylor, T.D.; Hattori, M.; Ohno, H. Gut Microorganisms Act Together to Exacerbate Inflammation in Spinal Cords. Nature 2020, 585, 102–106. [Google Scholar] [CrossRef]
  140. Ruff, W.E.; Dehner, C.; Kim, W.J.; Pagovich, O.; Aguiar, C.L.; Yu, A.T.; Roth, A.S.; Vieira, S.M.; Kriegel, C.; Adeniyi, O.; et al. Pathogenic Autoreactive T and B Cells Cross-React with Mimotopes Expressed by a Common Human Gut Commensal to Trigger Autoimmunity. Cell Host Microbe 2019, 26, 100–113.e8. [Google Scholar] [CrossRef]
  141. Girdhar, K.; Huang, Q.; Chow, I.-T.; Brady, C.; Raisingani, A.; Tran, T.U.; Kwok, W.; Atkinson, M.A.; Kahn, C.R.; Altindis, E. 938-P: A Human Gut Commensal, Parabacteroides Distasonis, Enhances Type 1 Diabetes Autoimmunity via Molecular Mimicry. Diabetes 2021, 70, 938. [Google Scholar] [CrossRef]
  142. Girdhar, K.; Huang, Q.; Chow, I.-T.; Vatanen, T.; Brady, C.; Raisingani, A.; Autissier, P.; Atkinson, M.A.; Kwok, W.W.; Kahn, C.R.; et al. A Gut Microbial Peptide and Molecular Mimicry in the Pathogenesis of Type 1 Diabetes. Proc. Natl. Acad. Sci. USA 2022, 119, e2120028119. [Google Scholar] [CrossRef] [PubMed]
  143. Koning, D.; Quakkelaar, E.D.; Schellens, I.M.M.; Spierings, E.; van Baarle, D. Protective HLA Alleles Recruit Biased and Largely Similar Antigen-Specific T Cell Repertoires across Different Outcomes in HIV Infection. J. Immunol. 2022, 208, 3–15. [Google Scholar] [CrossRef] [PubMed]
  144. Abualrous, E.T.; Sticht, J.; Freund, C. Major Histocompatibility Complex (MHC) Class I and Class II Proteins: Impact of Polymorphism on Antigen Presentation. Curr. Opin. Immunol. 2021, 70, 95–104. [Google Scholar] [CrossRef] [PubMed]
  145. Poomarimuthu, M.; Ramasamy, T.; Govindan, R.; Andiappan, R.; Nagarajan, G.; Kadiam, S.; Mariakuttikan, J. Association of HLA-DRB1 Alleles with Rheumatic Fever and Rheumatic Heart Disease: A Meta-Analysis. Immunol. Investig. 2022, 51, 221–232. [Google Scholar] [CrossRef]
  146. Panhuber, A.; Lamorte, G.; Bruno, V.; Cetin, H.; Bauer, W.; Höftberger, R.; Erber, A.C.; Frommlet, F.; Koneczny, I. A Systematic Review and Meta-Analysis of HLA Class II Associations in Patients with IgG4 Autoimmunity. Sci. Rep. 2022, 12, 9229. [Google Scholar] [CrossRef]
  147. Zhao, L.P.; Papadopoulos, G.K.; Moustakas, A.K.; Bondinas, G.P.; Carlsson, A.; Larsson, H.E.; Ludvigsson, J.; Marcus, C.; Persson, M.; Samuelsson, U.; et al. Nine Residues in HLA-DQ Molecules Determine with Susceptibility and Resistance to Type 1 Diabetes among Young Children in Sweden. Sci. Rep. 2021, 11, 8821. [Google Scholar] [CrossRef]
  148. Zhao, L.P.; Papadopoulos, G.K.; Kwok, W.W.; Moustakas, A.K.; Bondinas, G.P.; Carlsson, A.; Elding Larsson, H.; Ludvigsson, J.; Marcus, C.; Samuelsson, U.; et al. Next-Generation HLA Sequence Analysis Uncovers Seven HLA-DQ Amino Acid Residues and Six Motifs Resistant to Childhood Type 1 Diabetes. Diabetes 2020, 69, 2523–2535. [Google Scholar] [CrossRef]
  149. Jones, E.Y.; Fugger, L.; Strominger, J.L.; Siebold, C. MHC Class II Proteins and Disease: A Structural Perspective. Nat. Rev. Immunol. 2006, 6, 271–282. [Google Scholar] [CrossRef]
  150. Nishimoto, H.; Kikutani, H.; Yamamura, K.; Kishimoto, T. Prevention of Autoimmune Insulitis by Expression of I–E Molecules in NOD Mice. Nature 1987, 328, 432–434. [Google Scholar] [CrossRef]
  151. Reich, E.-P.; Sherwin, R.S.; Kanagawa, O.; Janeway, C.A. An Explanation for the Protective Effect of the MHC Class II I–E Molecule in Murine Diabetes. Nature 1989, 341, 326–328. [Google Scholar] [CrossRef] [PubMed]
  152. Schmidt, D.; Verdaguer, J.; Averill, N.; Santamaria, P. A Mechanism for the Major Histocompatibility Complex–Linked Resistance to Autoimmunity. J. Exp. Med. 1997, 186, 1059–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Jan, B.; Brigitte, S.; Osami, K.; Christophe, B.; Diane, M. MHC-Linked Protection from Diabetes Dissociated from Clonal Deletion of T Cells. Science (1979) 1990, 249, 293–295. [Google Scholar] [CrossRef]
  154. Katz, J.D.; Wang, B.; Haskins, K.; Benoist, C.; Mathis, D. Following a Diabetogenic T Cell from Genesis through Pathogenesis. Cell 1993, 74, 1089–1100. [Google Scholar] [CrossRef] [PubMed]
  155. Wing, J.B.; Tanaka, A.; Sakaguchi, S. Human FOXP3+ Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer. Immunity 2019, 50, 302–316. [Google Scholar] [CrossRef] [Green Version]
  156. Sakaguchi, S.; Mikami, N.; Wing, J.B.; Tanaka, A.; Ichiyama, K.; Ohkura, N. Regulatory T Cells and Human Disease. Annu Rev. Immunol. 2020, 38, 541–566. [Google Scholar] [CrossRef] [Green Version]
  157. Mitchell, A.M.; Michels, A.W. Self-Antigens Targeted by Regulatory T Cells in Type 1 Diabetes. Int. J. Mol. Sci. 2022, 23, 3155. [Google Scholar] [CrossRef]
  158. van Lummel, M.; Buis, D.T.P.; Ringeling, C.; de Ru, A.H.; Pool, J.; Papadopoulos, G.K.; van Veelen, P.A.; Reijonen, H.; Drijfhout, J.W.; Roep, B.O. Epitope Stealing as a Mechanism of Dominant Protection by HLA-DQ6 in Type 1 Diabetes. Diabetes 2019, 68, 787–795. [Google Scholar] [CrossRef] [Green Version]
  159. Tsai, S.; Serra, P.; Clemente-Casares, X.; Slattery, R.M.; Santamaria, P. Dendritic Cell–Dependent In Vivo Generation of Autoregulatory T Cells by Antidiabetogenic MHC Class II. J. Immunol. 2013, 191, 70. [Google Scholar] [CrossRef] [Green Version]
  160. Sue, T.; Pau, S.; Xavier, C.-C.; Jun, Y.; Shari, T.; Slattery, R.M.; Elliott, J.F.; Pere, S. Antidiabetogenic MHC Class II Promotes the Differentiation of MHC-Promiscuous Autoreactive T Cells into FOXP3+ Regulatory T Cells. Proc. Natl. Acad. Sci. USA 2013, 110, 3471–3476. [Google Scholar] [CrossRef] [Green Version]
  161. Beringer, D.X.; Kleijwegt, F.S.; Wiede, F.; van der Slik, A.R.; Loh, K.L.; Petersen, J.; Dudek, N.L.; Duinkerken, G.; Laban, S.; Joosten, A.; et al. T Cell Receptor Reversed Polarity Recognition of a Self-Antigen Major Histocompatibility Complex. Nat. Immunol. 2015, 16, 1153–1161. [Google Scholar] [CrossRef] [PubMed]
  162. Stadinski, B.D.; Blevins, S.J.; Spidale, N.A.; Duke, B.R.; Huseby, P.G.; Stern, L.J.; Huseby, E.S. A Temporal Thymic Selection Switch and Ligand Binding Kinetics Constrain Neonatal Foxp3+ Treg Cell Development. Nat. Immunol. 2019, 20, 1046–1058. [Google Scholar] [CrossRef] [PubMed]
  163. Ivanova, V.V.; Khaiboullina, S.F.; Gomzikova, M.O.; Martynova, E.V.; Ferreira, A.M.; Garanina, E.E.; Sakhapov, D.I.; Lomakin, Y.A.; Khaibullin, T.I.; Granatov, E.V.; et al. Divergent Immunomodulation Capacity of Individual Myelin Peptides—Components of Liposomal Therapeutic against Multiple Sclerosis. Front. Immunol. 2017, 8, 1335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Belogurov, A.A., Jr.; Stepanov, A.V.; Smirnov, I.V.; Melamed, D.; Bacon, A.; Mamedov, A.E.; Boitsov, V.M.; Sashchenko, L.P.; Ponomarenko, N.A.; Sharanova, S.N.; et al. Liposome-Encapsulated Peptides Protect against Experimental Allergic Encephalitis. FASEB J. 2013, 27, 222–231. [Google Scholar] [CrossRef] [Green Version]
  165. Selck, C.; Dominguez-Villar, M. Antigen-Specific Regulatory T Cell Therapy in Autoimmune Diseases and Transplantation. Front. Immunol. 2021, 12, 661875. [Google Scholar] [CrossRef]
  166. Gupta, S.; Li, D.; Ostrov, D.A.; Nguyen, C.Q. Blocking IAg7 Class II Major Histocompatibility Complex by Drug-like Small Molecules Alleviated Sjögren’s Syndrome in NOD Mice. Life Sci. 2022, 288, 120182. [Google Scholar] [CrossRef]
  167. Ostrov, D.A.; Gottlieb, P.A.; Michels, A.W. Rationally Designed Small Molecules to Prevent Type 1 Diabetes. Curr. Opin. Endocrinol. Diabetes Obes. 2019, 26, 90. [Google Scholar] [CrossRef]
  168. Ostrov, D.A.; Alkanani, A.; McDaniel, K.A.; Case, S.; Baschal, E.E.; Pyle, L.; Ellis, S.; Pöllinger, B.; Seidl, K.J.; Shah, V.N.; et al. Methyldopa Blocks MHC Class II Binding to Disease-Specific Antigens in Autoimmune Diabetes. J. Clin. Investig. 2018, 128, 1888–1902. [Google Scholar] [CrossRef] [Green Version]
  169. Yi, J.; Miller, A.T.; Archambault, A.S.; Jones, A.J.; Bradstreet, T.R.; Bandla, S.; Hsu, Y.-S.; Edelson, B.T.; Zhou, Y.W.; Fremont, D.H.; et al. Antigen-Specific Depletion of CD4+ T Cells by CAR T Cells Reveals Distinct Roles of Higher- and Lower-Affinity TCRs during Autoimmunity. Sci. Immunol. 2022, 7, eabo0777. [Google Scholar] [CrossRef]
  170. Kobayashi, S.; Thelin, M.A.; Parrish, H.L.; Deshpande, N.R.; Lee, M.S.; Karimzadeh, A.; Niewczas, M.A.; Serwold, T.; Kuhns, M.S. A Biomimetic Five-Module Chimeric Antigen Receptor (5MCAR) Designed to Target and Eliminate Antigen-Specific T Cells. Proc. Natl. Acad. Sci. USA 2020, 117, 28950–28959. [Google Scholar] [CrossRef]
Figure 1. Stages of T cell selection during the establishment of central tolerance. T cell precursor double-negative (DN) T cells enter the cortex of the thymus through a blood vessel to undergo positive selection. Due to the interaction between antigen-presenting cells (APCs) present on cortical thymic epithelial cells (cTECs) and migratory dendritic cells (DCs), double-positive (DP) T cells are differentiated into CD4+ or CD8+ single-positive (SP) T cells and migrate to the medulla of the thymus to undergo negative selection. As they interact with APCs (medulla thymic epithelial cells (mTECs), migratory or resident DCs, and B cells), CD4+ T cells become CD4+ naïve T cells or T regulatory cells (Tregs) and leave the thymus. Negative selection is schematically shown only for SP CD4+ T cells.
Figure 1. Stages of T cell selection during the establishment of central tolerance. T cell precursor double-negative (DN) T cells enter the cortex of the thymus through a blood vessel to undergo positive selection. Due to the interaction between antigen-presenting cells (APCs) present on cortical thymic epithelial cells (cTECs) and migratory dendritic cells (DCs), double-positive (DP) T cells are differentiated into CD4+ or CD8+ single-positive (SP) T cells and migrate to the medulla of the thymus to undergo negative selection. As they interact with APCs (medulla thymic epithelial cells (mTECs), migratory or resident DCs, and B cells), CD4+ T cells become CD4+ naïve T cells or T regulatory cells (Tregs) and leave the thymus. Negative selection is schematically shown only for SP CD4+ T cells.
Cells 12 00314 g001
Figure 2. MHC-II maturation and antigenic peptide loading. MHC-II molecules are synthesized in the endoplasmic reticulum (ER) and loaded with an invariant chain (Ii). The MHC-II complex with Ii is transported through the Golgi to the late endosome. Endosomal proteases process antigens to short peptides and Ii to shorter class II-associated invariant chain peptide (CLIP). The binding of a nonclassical HLA-DM (DM) molecule to MHC-II promotes CLIP exchange to the antigenic peptide with an optimal binding register. The formed pMHC complex is transported to the surface of the APC for CD4+ T cell recognition.
Figure 2. MHC-II maturation and antigenic peptide loading. MHC-II molecules are synthesized in the endoplasmic reticulum (ER) and loaded with an invariant chain (Ii). The MHC-II complex with Ii is transported through the Golgi to the late endosome. Endosomal proteases process antigens to short peptides and Ii to shorter class II-associated invariant chain peptide (CLIP). The binding of a nonclassical HLA-DM (DM) molecule to MHC-II promotes CLIP exchange to the antigenic peptide with an optimal binding register. The formed pMHC complex is transported to the surface of the APC for CD4+ T cell recognition.
Cells 12 00314 g002
Figure 3. The role of a nonclassical DM molecule in the editing of the self-antigen repertoire presented by MHC-II. (A) In a thymic APC, the self-antigen is processed through a classical pathway under exposure to cellular proteolytic enzymes. Next, CLIP is exchanged to the high-affinity fragment in late endosomes under DM control. The stable pMHC complex is then transferred to the APC surface, where the T cells specific to this pMHC undergo clonal deletion. (B) If an excessive amount of certain self-antigens, processed outside APCs, is present in peripheral tissues, then these self-antigen fragments can be loaded on MHC-II molecules in early endosomes or on the surface of APCs without the recruitment of DM. The absence of DM editing allows the emergence of either stable or unstable pMHC complexes. The pMHCs, containing high-affinity fragments, do not elicit a T cell response due to the deletion of autoreactive T cells in the thymus. Unstable complexes with low-affinity fragments can bind autoreactive T cells since such T cells are not susceptible to clonal deletion.
Figure 3. The role of a nonclassical DM molecule in the editing of the self-antigen repertoire presented by MHC-II. (A) In a thymic APC, the self-antigen is processed through a classical pathway under exposure to cellular proteolytic enzymes. Next, CLIP is exchanged to the high-affinity fragment in late endosomes under DM control. The stable pMHC complex is then transferred to the APC surface, where the T cells specific to this pMHC undergo clonal deletion. (B) If an excessive amount of certain self-antigens, processed outside APCs, is present in peripheral tissues, then these self-antigen fragments can be loaded on MHC-II molecules in early endosomes or on the surface of APCs without the recruitment of DM. The absence of DM editing allows the emergence of either stable or unstable pMHC complexes. The pMHCs, containing high-affinity fragments, do not elicit a T cell response due to the deletion of autoreactive T cells in the thymus. Unstable complexes with low-affinity fragments can bind autoreactive T cells since such T cells are not susceptible to clonal deletion.
Cells 12 00314 g003
Figure 4. Effect of posttranslational modifications of self-antigens on trimolecular complex assembly. Certain self-antigen fragments do not bind to MHC-II molecules, or their binding affinity is low. Similar self-antigen with posttranslational modifications (iodination, acetylation, glycosylation, citrullination, generation of hybrid peptides, etc.) exhibiting increased affinity for MHC-II molecules, can appear in peripheral tissues. The resulting pMHC complexes can bind autoreactive T cells that have not undergone negative selection in the thymus.
Figure 4. Effect of posttranslational modifications of self-antigens on trimolecular complex assembly. Certain self-antigen fragments do not bind to MHC-II molecules, or their binding affinity is low. Similar self-antigen with posttranslational modifications (iodination, acetylation, glycosylation, citrullination, generation of hybrid peptides, etc.) exhibiting increased affinity for MHC-II molecules, can appear in peripheral tissues. The resulting pMHC complexes can bind autoreactive T cells that have not undergone negative selection in the thymus.
Cells 12 00314 g004
Figure 5. The development of autoreactive CD4+ T cells in a proinflammatory environment. The proinflammatory environment caused by viral or bacterial infection entails IFN-γ production, which results in increased MHC-II and costimulatory molecule expression compared to normal conditions. Therefore, rare self-antigens are present in a greater number, which can lead to the potential activation of autoreactive T cells.
Figure 5. The development of autoreactive CD4+ T cells in a proinflammatory environment. The proinflammatory environment caused by viral or bacterial infection entails IFN-γ production, which results in increased MHC-II and costimulatory molecule expression compared to normal conditions. Therefore, rare self-antigens are present in a greater number, which can lead to the potential activation of autoreactive T cells.
Cells 12 00314 g005
Figure 6. The expansion of autoreactive CD4+ T cells due to the molecular mimicry between foreign and self-antigens. The presentation of viral or bacterial antigenic peptides on MHC-II molecules, structurally similar to self-antigens, may result in the expansion of CD4+ T cells, specific to exogenous pMHC. Expanded CD4+ T cells, specific for foreign peptides, can bind the self-antigen pMHC, which results in the development of autoimmunity.
Figure 6. The expansion of autoreactive CD4+ T cells due to the molecular mimicry between foreign and self-antigens. The presentation of viral or bacterial antigenic peptides on MHC-II molecules, structurally similar to self-antigens, may result in the expansion of CD4+ T cells, specific to exogenous pMHC. Expanded CD4+ T cells, specific for foreign peptides, can bind the self-antigen pMHC, which results in the development of autoimmunity.
Cells 12 00314 g006
Figure 7. The role of protective MHC-II molecules in the suppression of autoreactive T cell development. (A) Recognition of the self-antigen presented on the product of the protective allele by CD4+ T cell results in negative selection or Treg development in the thymus. (B) The slow loading rate of self-antigen on the MHC-II molecule of the protective allele results in the failure of pMHC complex assembly on the surface of thymic or peripheral APCs.
Figure 7. The role of protective MHC-II molecules in the suppression of autoreactive T cell development. (A) Recognition of the self-antigen presented on the product of the protective allele by CD4+ T cell results in negative selection or Treg development in the thymus. (B) The slow loading rate of self-antigen on the MHC-II molecule of the protective allele results in the failure of pMHC complex assembly on the surface of thymic or peripheral APCs.
Cells 12 00314 g007
Table 1. Autoantigens and autoimmune TCRs in autoimmune diseases.
Table 1. Autoantigens and autoimmune TCRs in autoimmune diseases.
ADSpeciesMHC-II AlleleAntigenic PeptideProposed Molecular Mechanisms of AD SusceptibilityReference
Low-affinity autoantigenic peptides
T1DMouseI-Ag7Ins. B:12–20, Ins. B:13–21Weak binding of CLIP by MHC-II molecule promotes loading of low-affinity peptides.[48]
T1DMouseI-Ag7Ins. B:12–20, Ins. B:14–22MHC-II binding of antigenic peptide in a low-affinity register leads to recognition by autoreactive TCRs.[52,53,54,55]
T1DHumanHLA-DQA1*03:01, HLA-DQB1*03:02Ins. B:11–23MHC-II binding of antigenic peptide in a low-affinity register leads to recognition by autoreactive TCRs.[56]
Low-affinity autoreactive TCRs
MSHumanHLA-DRB1*15:01MBP:85–99Autoreactive TCRs bind pMHC complexes with low affinity and alternate topology.[16,67]
MSHumanHLA-DRB5*01:01MBP:84–102[68]
High or intermediate affinity autoreactive TCRs
MSHumanHLA-DQA1*01:02, HLA-DQB1*05:02MBP:85–99The unusual topology of trimolecular complex impedes T cell activation by CD4 receptor.[70]
MSHumanHLA-DRB1*04:01MBP:111–129Autoreactive TCR stabilizes weak interaction between antigenic peptide and MHC-II. [72]
EAEMouseI-AuMBP:1–11 (acetylated)Autoreactive TCRs bind pMHC complexes with acetylated autoantigens in unusual binding register, where the part of peptide-binding groove is empty.[73,74,75]
Abbreviations: AD, autoimmune disease; T1D, type 1 diabetes; Ins, insulin; CLIP, class II-associated invariant chain peptide; MS, multiple sclerosis; MBP, myelin basic protein; TCR, T cell receptor; EAE, experimental autoimmune encephalomyelitis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ishina, I.A.; Zakharova, M.Y.; Kurbatskaia, I.N.; Mamedov, A.E.; Belogurov, A.A., Jr.; Gabibov, A.G. MHC Class II Presentation in Autoimmunity. Cells 2023, 12, 314. https://doi.org/10.3390/cells12020314

AMA Style

Ishina IA, Zakharova MY, Kurbatskaia IN, Mamedov AE, Belogurov AA Jr., Gabibov AG. MHC Class II Presentation in Autoimmunity. Cells. 2023; 12(2):314. https://doi.org/10.3390/cells12020314

Chicago/Turabian Style

Ishina, Irina A., Maria Y. Zakharova, Inna N. Kurbatskaia, Azad E. Mamedov, Alexey A. Belogurov, Jr., and Alexander G. Gabibov. 2023. "MHC Class II Presentation in Autoimmunity" Cells 12, no. 2: 314. https://doi.org/10.3390/cells12020314

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop