Following syngeneic or autologous stem cell transplantation, a syndrome closely resembling allogeneic graft versus host disease (GvHD) occurs in 5% to 20% of recipients,1,2 although there are no genetic differences between the cell therapy product and the host tissues. In a cohort of patients receiving transplants from an identical twin to treat aplastic anemia, GvHD-like features only developed among those recipients treated with pretransplant conditioning.3 Moreover, in allogeneic stem cell transplantation, host resident memory T cells persist in peripheral tissues even after myeloablative conditioning and contribute to the pathogenesis of GvHD.4 These findings imply that the inflammatory response consequent upon pretransplant conditioning can trigger T-cell autoreactivity. Among the factors that have been suggested to contribute to this phenomenon are destruction with inadequate reconstitution of T regulatory cells (Treg) in the peritransplant period, and disruption of the gut microbiome with reduced diversity and impaired production of short-chain fatty acids (reviewed in Rafei and Jenq5). Here, we propose an additional mechanism to account for these findings.
Differences in minor histocompatibility antigens (MiHA) between the donor and recipient of hematopoietic stem cell transplants are well-recognized precipitants of GvHD in the setting of HLA-matched sibling and unrelated donation. MiHA arise through genetically encoded polymorphism in the source proteins of the endogenous peptide antigens that are presented by donor and recipient HLA. The capacity of MiHA to precipitate GvHD reflects the sensitivity of the donor immune cell precursors toward a small proportion of peptide antigens. By analogy, we hypothesize that pretransplant conditioning can trigger donor T-cell reactivity against the host through the generation of both conventional and unconventional peptide epitopes that are not present in the thymus during T-cell development. Autoreactive T cells able to recognize this “induced/altered self” are not removed by negative selection6 and persist within the mature T-cell repertoires of both the donor and recipient. Observations made in the context of autologous or syngeneic transplantation are also relevant to matched and haploidentical stem cell transplantation and solid organ transplantation in which matched and allogeneic HLA are often coexpressed by the donor organ. In these settings, autoreactive T cells could synergize with T cells responding to MiHA and with those directly or indirectly recognizing allogeneic major histocompatibility complex (MHC) to augment damage to target tissues. Both memory T cells and previously naive T cells could contribute to this phenomenon. In addition to self-restricted self-peptides and indirectly presented peptides processed from allogeneic donor MHC, self-peptides directly presented by donor MHC or MiHA could also be subject to induction/modification but, although they may influence the alloresponse, they would not be targets for autoreactivity.
Transplantable organs are subjected to many sources of cellular stress (Figure 1). Donor brain death is accompanied by a catecholamine storm, erratic perfusion of peripheral tissues, and a surge in proinflammatory cytokines and chemokines.7 Circulatory death exposes donor organs to an extended period of warm ischemia,8 whereas some degree of ischemia-reperfusion injury affects all solid organ transplants. Recipient preparation for hematopoietic stem cell transplantation typically involves both irradiation and administration of cytoreductive chemotherapy. The early stages of alloreactive T-cell responses, whether directed toward the graft or the host, are marked by the release of cytokines and other soluble mediators that act upon the target tissues. These factors may alter tissue proteomes and immunopeptidomes in various ways. Among these are the generation of proteasome-spliced, or “hybrid” peptides,9 and immunoproteasome-dependent peptides, modification of the amino acid side chains of peptides or their source proteins,10-13 and the utilization of noncanonical start codons and alternative open reading frames (ORFs)14,15 for protein translation, in parallel with the standard templates. The resulting “dark proteome” yields novel peptide epitopes termed “cryptic” because standard analysis pipelines do not reveal them.16 Beyond directly altering the interacting surfaces of the T-cell receptor-peptide-MHC (pMHC), posttranslational modification (PTM) of proteins can perturb classical antigen processing pathways, exposing epitopes that are normally degraded before MHC loading. An increased density of previously rare epitopes may be sufficient to stimulate weakly reactive T cells past the threshold of activation.17,18
FIGURE 1.: Factors promoting the generation of induced/altered self-peptides in transplantation. Ischemia-reperfusion injury, pretransplant conditioning with cytoreductive agents, and irradiation and exposure to cytokines all contribute to oxidative stress. Cytokines such as IFN-γ increase the expression of immunoproteasome-dependent peptides, plus spliced or hybrid peptides generated in the proteasome, whereas PTMs of amino acid residues result from specific enzyme activity or exposure to a chemically reactive milieu or can be spontaneous. Oxidative stress increases the usage of noncanonical start codons (CUG, UUG), and proteins translated from these alternative ORFs give rise to cryptic peptides. Both native and altered self-peptides presented by allogeneic MHC are recognized by alloreactive T cells, whereas presentation of altered-self by donor–recipient matched MHC may give rise to de novo autoreactivity. IFN-γ, interferon gamma; MHC, major histocompatibility complex; ORF, open reading frame; PTM, posttranslational modification.
Exposure of cells to interferon gamma (IFN-γ) increases the diversity of the peptide antigens presented by MHC molecules (ie, the immunopeptidome), substantially augmenting presentation of both conventional linear and proteasome-spliced peptides.9 IFN-γ upregulates expression of numerous potential source proteins and induces transcription of the specialized immunoproteasome subunits β1i, β2i, and β5i.19 Immunoproteasomes differ in proteolytic activity from constitutive proteasomes, giving rise to more hydrophobic peptides,19 some of which are exclusively produced by the immunoproteasome. Although medullary thymic epithelial cells express both constitutive proteasomes and immunoproteasomes,20 participation of immunoproteasome-dependent self-peptides in negative selection may be limited. Immunoproteasome-dependent antigens have been implicated in the development of autoimmunity,21 whereas selective inhibition of the b5i subunit blocks development of diabetes in a model in which an immunoproteasome-dependent epitope is expressed in pancreatic islets.22 Consistent with immunoproteasome activity, conventional self-peptides that do not cause negative selection in the thymus have been detected in cells treated with IFN and in rejecting grafts, where they were presented by syngeneic MHC.23 Proteasome-mediated peptide splicing involves the joining of 2 noncontiguous peptide segments, either from the same protein (cis-splicing) or from different proteins (trans-splicing), resulting in novel, nongenomically templated peptide antigens.24
PTM of amino acids can occur spontaneously, through exposure to reactive oxygen species, or via the action of enzymes such as acetyl or methyltransferases, kinases, ubiquitin ligases, glycosyltransferases, and tissue transglutaminase 2.17 Ischemia-reperfusion injury induces the expression of peptidyl-arginine deiminases, which convert arginine residues to citrulline,25 whereas protein cysteinylation (disulfide bond-formation with free cysteine) occurs as a consequence of acute inflammation.26 Irradiation of tumors increases neoantigen presentation and subsequently broadens the repertoire of responding T cells,27,28 suggesting that irradiation of normal tissues may also give rise to neoepitopes. Finally, oxidative stress is associated with dysregulation of protein translation14,15 promoting presentation of cryptic peptides.
The immunogenicity of PTM, spliced, and cryptic peptide neoepitopes is increasingly well documented. Deamidation of asparagine yields both aspartyl and isoaspartyl residues. Isoaspartyl modifications are linked to increased immunogenicity of self, tumor, and viral vector capsid antigens.29-31 Acetylation and citrullination feature prominently in the pathogenesis of autoimmune disorders,18,32,33 whereas cysteinylation and disulfide bond rearrangements have been implicated in both alloreactivity and autoreactivity.34,35 Tumor antigens derived from alternative ORFs elicit CD8+ T-cell responses,36,37 and an H-2Kb-restricted self-peptide derived from an upstream ORF of α-tubulin is recognized by alloreactive T cells.38 Immunogenic spliced peptides have been isolated from human renal cancer,39 melanoma cells,40 acute myeloid leukemia,41 and murine cells infected with Listeria monocytogenes,42 whereas hybrid epitopes formed by the splicing of insulin peptides with fragments of other islet proteins are recognized by autoreactive T cells from human subjects with type 1 diabetes.43 The altered self-peptide concept of de novo autoreactivity in transplantation does not exclude the contributions of dysbiosis or Treg loss to autoimmune components of GvHD or rejection; rather, these factors may act synergistically to promote autoreactivity.
Although it has been well recognized that the graft microenvironment influences transplant outcomes, this has been viewed primarily as a consequence of innate immunity through mechanisms such as the promotion of leukocyte infiltration, upregulation of costimulatory ligand expression on antigen-presenting cell, and interference with Treg functions. Recently, intrarenal B-cell populations from kidneys undergoing chronic rejection were shown to contain cells recognizing tissue-specific autoantigens (rather than alloantigens), some of which were only present in inflamed kidneys.44 Given that these antibodies were isotype-switched and extensively mutated, a role for CD4+ T cells that recognize inflammation-related neoepitopes presented by B cells and promote autoantibody production through the provision of T cell help seems likely.
Based on our hypothesis, we would predict that transplanted organs procured from deceased donors would be more likely to trigger autoreactivity than those obtained through living donation. Localized tissue injury can also result in systemic inflammation,45,46 in turn setting up the conditions under which de novo autoimmunity could occur in organs other than the graft. Similarly, we anticipate that organs exposed to environmental antigens would harbor a tissue immunopeptidome incorporating more altered self-peptides with a greater propensity for recognition by autoreactive T cells. In part, this may relate to augmentation of graft inflammation due to innate signaling by microbial products, although this does not explain all the relevant findings. The half-life of male to female C57BL/6 skin grafts colonized by commensal bacteria in otherwise germ-free recipient mice is shortened compared with that of sterile grafts to germ-free recipients.47 Pattern-recognition receptor signaling alone is not sufficient to drive the acceleration of graft rejection resulting from Staphylococcus epidermidis colonization of donor skin,47 but metabolic or biochemical alterations in colonized skin might change the immunopeptidome with a resultant autoreactive response that hastens graft destruction. Mice primed against S epidermidis or Staphylococcus aureus mounted a brisk immune response against sex-matched syngeneic skin grafts colonized by these commensals, resulting in moderate graft damage in the absence of alloreactivity.48 In parallel to a memory response against the commensal organisms per se, autoreactive memory against induced/altered self-peptides could explain these findings. There may also be a role for molecules like MR1 that present bacterial metabolites.49
Discovery of immunogenic altered self-epitopes expressed by host target tissues and transplantable organs is feasible. Novel immunopeptidomic workflows enable the unbiased capture of the full spectrum of immunogenic peptides. Cryptic epitopes can be revealed using methods such as immunopeptidogenomics, whereby whole transcriptome data are translated in 3 frames and used to create a database of potential proteins resulting from use of alternative ORFs,16,50-53 whereas further recently established pipelines are suitable for the detection of spliced24,54 and other PTM55 peptides. These pipelines can be used in combination with cellular assays for the identification of immunogenic epitopes that are recognized by alloreactive and autoreactive CD8+ T cells,56 thus providing a means of testing the hypothesis proposed herein (Figure S1, SDC, https://links.lww.com/TP/C668).
Dissection of the relative contributions of altered self-peptide antigens, dysbiosis, and Treg loss to GvHD could be attempted using available experimental approaches. If T cells specific for matched MHC complexed with altered self-peptides were depleted (eg, using toxin-coupled57,58 or column-bound tetramers), any resulting amelioration of GvHD severity would suggest a functional role for those pMHC ligands, distinct from the effects of dysbiosis or Treg depletion. This could be compared with the effects of fecal microbiota transfers or regulatory T-cell transfer, although it should be noted that those interventions may also change the peptide landscape by reducing inflammation in the host tissues. Other predictions flowing from the hypothesis are also amenable to empirical testing. Mouse models of donation after brain death or circulatory death versus living donation could be combined with haploidentical (F1 to parent) organ transplantation, with examination of the tissue immunopeptidome and T-cell responses in the different groups. Broad-spectrum antibiotic treatment of donors or recipients or manipulation of extra-intestinal microbiota alone could be accompanied by the assessment of changes to the immunopeptidome and to auto and alloreactive T-cell responses.
New workflows to interrogate the full diversity of the immunopeptidome, along with a robust methodology for assessing pMHC recognition by alloreactive or autoreactive T cells, allow us to ask whether a proinflammatory or metabolically altered microenvironment causes modifications of the pMHC epitopes at the heart of allorecognition and altered self-recognition, thus rendering donor organs or host GvHD target tissues more immunogenic. Recognizing the unknown contributors to rejection and GvHD will allow us to develop strategies to address these.
REFERENCES
1. Hood AF, Vogelsang GB, Black LP, et al. Acute graft-vs-host disease. Development following autologous and syngeneic bone marrow transplantation. Arch Dermatol. 1987;123:745–750.
2. Sica S, Chiusolo P, Salutari P, et al. Autologous graft-versus-host disease after CD34+-purified autologous peripheral blood progenitor cell transplantation. J Hematother Stem Cell Res. 2000;9:375–379.
3. Hinterberger W, Rowlings PA, Hinterberger-Fischer M, et al. Results of transplanting bone marrow from genetically identical twins into patients with aplastic anemia. Ann Intern Med. 1997;126:116–122.
4. Divito SJ, Aasebo AT, Matos TR, et al. Peripheral host T cells survive hematopoietic stem cell transplantation and promote graft-versus-host disease. J Clin Invest. 2020;130:4624–4636.
5. Rafei H, Jenq RR. Microbiome-intestine cross talk during acute graft-versus-host disease. Blood. 2020;136(4):401–409.
6. Raposo B, Merky P, Lundqvist C, et al. T cells specific for post-translational modifications escape intrathymic tolerance induction. Nat Commun. 2018;9:353.
7. Watts RP, Thom O, Fraser JF. Inflammatory signalling associated with brain dead organ donation: from brain injury to brain stem death and posttransplant ischaemia reperfusion injury. J Transplant. 2013;2013:521369.
8. Manara AR, Murphy PG, O’Callaghan G. Donation after circulatory death. Br J Anaesth. 2012;108(Suppl 1):i108–i121.
9. Faridi P, Woods K, Ostrouska S, et al. Spliced peptides and cytokine-driven changes in the immunopeptidome of melanoma. Cancer Immunol Res. 2020;8:1322–1334.
10. von Stechow L, Olsen JV. Proteomics insights into DNA damage response and translating this knowledge to clinical strategies. Proteomics. 2017;17(3-4):1600018.
11. Rookyard AW, Paulech J, Thyssen S, et al. A global profile of reversible and irreversible cysteine redox post-translational modifications during myocardial ischemia/reperfusion injury and antioxidant intervention. Antioxid Redox Signal. 2021;34:11–31.
12. Nanda SK, Vollmer S, Perez-Oliva AB. Posttranslational regulation of inflammasomes, its potential as biomarkers and in the identification of novel drugs targets. Front Cell Dev Biol. 2022;10:887533.
13. Baskal S, Post A, Kremer D, et al. Urinary excretion of amino acids and their advanced glycation end-products (AGEs) in adult kidney transplant recipients with emphasis on lysine: furosine excretion is associated with cardiovascular and all-cause mortality. Amino Acids. 2021;53:1679–1693.
14. Gerashchenko MV, Lobanov AV, Gladyshev VN. Genome-wide ribosome profiling reveals complex translational regulation in response to oxidative stress. Proc Natl Acad Sci USA. 2012;109:17394–17399.
15. Orr MW, Mao Y, Storz G, et al. Alternative ORFs and small ORFs: shedding light on the dark proteome. Nucleic Acids Res. 2020;48:1029–1042.
16. Scull KE, Pandey K, Ramarathinam SH, et al. Immunopeptidogenomics: harnessing RNA-Seq to illuminate the dark immunopeptidome. Mol Cell Proteomics. 2021;20:100143.
17. Petersen J, Purcell AW, Rossjohn J. Post-translationally modified T cell epitopes: immune recognition and immunotherapy. J Mol Med (Berl). 2009;87:1045–1051.
18. Scally SW, Petersen J, Law SC, et al. A molecular basis for the association of the HLA-DRB1 locus, citrullination, and rheumatoid arthritis. J Exp Med. 2013;210:2569–2582.
19. Rousseau A, Bertolotti A. Regulation of proteasome assembly and activity in health and disease. Nat Rev Mol Cell Biol. 2018;19(11):697–712.
20. Groettrup M, Kirk CJ, Basler M. Proteasomes in immune cells: more than peptide producers? Nat Rev Immunol. 2010;10:73–78.
21. Yao L, Zhou L, Xuan Y, et al. The proteasome activator REGgamma counteracts immunoproteasome expression and autoimmunity. J Autoimmun. 2019;103:102282.
22. Muchamuel T, Basler M, Aujay MA, et al. A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nat Med. 2009;15:781–787.
23. Boisgerault F, Anosova NG, Tam RC, et al. Induction of T-cell response to cryptic MHC determinants during allograft rejection. Hum Immunol. 2000;61:1352–1362.
24. Faridi P, Li C, Ramarathinam SH, et al. A subset of HLA-I peptides are not genomically templated: Evidence for cis- and trans-spliced peptide ligands. Sci Immunol. 2018;3:eaar3947.
25. Raup-Konsavage WM, Wang Y, Wang WW, et al. Neutrophil peptidyl arginine deiminase-4 has a pivotal role in ischemia/reperfusion-induced acute kidney injury. Kidney Int. 2018;93:365–374.
26. Moreno ML, Escobar J, Izquierdo-Alvarez A, et al. Disulfide stress: a novel type of oxidative stress in acute pancreatitis. Free Radic Biol Med. 2014;70:265–277.
27. Rudqvist NP, Pilones KA, Lhuillier C, et al. Radiotherapy and CTLA-4 blockade shape the TCR repertoire of tumor-infiltrating T cells. Cancer Immunol Res. 2018;6:139–150.
28. Lhuillier C, Rudqvist NP, Elemento O, et al. Radiation therapy and anti-tumor immunity: exposing immunogenic mutations to the immune system. Genome Med. 2019;11:40.
29. Mamula MJ, Gee RJ, Elliott JI, et al. Isoaspartyl post-translational modification triggers autoimmune responses to self-proteins. J Biol Chem. 1999;274:22321–22327.
30. Bing SJ, Justesen S, Wu WW, et al. Differential T cell immune responses to deamidated adeno-associated virus vector. Mol Ther Methods Clin Dev. 2022;24:255–267.
31. Chen W, Ede NJ, Jackson DC, et al. CTL recognition of an altered peptide associated with asparagine bond rearrangement. Implications for immunity and vaccine design. J Immunol. 1996;157:1000–1005.
32. Lehmann PV, Forsthuber T, Miller A, et al. Spreading of T-cell autoimmunity to cryptic determinants of an autoantigen. Nature. 1992;358:155–157.
33. Law SC, Street S, Yu CH, et al. T-cell autoreactivity to citrullinated autoantigenic peptides in rheumatoid arthritis patients carrying HLA-DRB1 shared epitope alleles. Arthritis Res Ther. 2012;14:R118.
34. Meadows L, Wang W, den Haan JM, et al. The HLA-A*0201-restricted H-Y antigen contains a posttranslationally modified cysteine that significantly affects T cell recognition. Immunity. 1997;6:273–281.
35. Mannering SI, Harrison LC, Williamson NA, et al. The insulin A-chain epitope recognized by human T cells is posttranslationally modified. J Exp Med. 2005;202:1191–1197.
36. Wang RF, Parkhurst MR, Kawakami Y, et al. Utilization of an alternative open reading frame of a normal gene in generating a novel human cancer antigen. J Exp Med. 1996;183:1131–1140.
37. Weinzierl AO, Maurer D, Altenberend F, et al. A cryptic vascular endothelial growth factor T-cell epitope: identification and characterization by mass spectrometry and T-cell assays. Cancer Res. 2008;68:2447–2454.
38. Malarkannan S, Afkarian M, Shastri N. A rare cryptic translation product is presented by Kb major histocompatibility complex class I molecule to alloreactive T cells. J Exp Med. 1995;182:1739–1750.
39. Hanada K, Yewdell JW, Yang JC. Immune recognition of a human renal cancer antigen through post-translational protein splicing. Nature. 2004;427:252–256.
40. Vigneron N, Stroobant V, Chapiro J, et al. An antigenic peptide produced by peptide splicing in the proteasome. Science. 2004;304:587–590.
41. Warren EH, Vigneron NJ, Gavin MA, et al. An antigen produced by splicing of noncontiguous peptides in the reverse order. Science. 2006;313:1444–1447.
42. Platteel ACM, Mishto M, Textoris-Taube K, et al. CD8+T cells of Listeria monocytogenes-infected mice recognize both linear and spliced proteasome products. Eur J Immunol. 2016;46:1109–1118.
43. Tran MT, Faridi P, Lim JJ, et al. T cell receptor recognition of hybrid insulin peptides bound to HLA-DQ8. Nat Commun. 2021;12:5110.
44. Asano Y, Daccache J, Jain D, et al. Innate-like self-reactive B cells infiltrate human renal allografts during transplant rejection. Nat Commun. 2021;12:4372.
45. Grigoryev DN, Liu M, Hassoun HT, et al. The local and systemic inflammatory transcriptome after acute kidney injury. J Am Soc Nephrol. 2008;19:547–558.
46. Fang L, Moore XL, Dart AM, et al. Systemic inflammatory response following acute myocardial infarction. J Geriatr Cardiol. 2015;12:305–312.
47. Lei YM, Sepulveda M, Chen L, et al. Skin-restricted commensal colonization accelerates skin graft rejection. JCI Insight. 2019;5:e127569.
48. Pirozzolo I, Sepulveda M, Chen L, et al. Host-versus-commensal immune responses participate in the rejection of colonized solid organ transplants. J Clin Invest. 2022;132:e153403.
49. Kjer-Nielsen L, Patel O, Corbett AJ, et al. MR1 presents microbial vitamin B metabolites to MAIT cells. Nature. 2012;491:717–723.
50. Marcu A, Schlosser A, Keupp A, et al. Natural and cryptic peptides dominate the immunopeptidome of atypical teratoid rhabdoid tumors. J ImmunoTher Cancer. 2021;9:e003404.
51. Wang TY, Liu Q, Ren Y, et al. A pan-cancer transcriptome analysis of exitron splicing identifies novel cancer driver genes and neoepitopes. Mol Cell. 2021;81:2246–2260.e12.
52. Ruiz Cuevas MV, Hardy MP, Holly J, et al. Most non-canonical proteins uniquely populate the proteome or immunopeptidome. Cell Rep. 2021;34:108815.
53. Ouspenskaia T, Law T, Clauser KR, et al. Unannotated proteins expand the MHC-I-restricted immunopeptidome in cancer. Nat Biotechnol. 2022;40:209–217.
54. Yewdell JW. MHC class I immunopeptidome: past, present, and future. Mol Cell Proteomics. 2022;21:100230.
55. Kacen A, Javitt A, Kramer MP, et al. Uncovering the modified immunopeptidome reveals insights into the principles of PTM-driven antigenicity. Biorxiv. Preprint posted online April 15, 2021.
56. Son ET, Faridi P, Paul-Heng M, et al. The self-peptide repertoire plays a critical role in transplant tolerance induction. J Clin Invest. 2021;131:e146771.
57. Vincent BG, Young EF, Buntzman AS, et al. Toxin-coupled MHC class I tetramers can specifically ablate autoreactive CD8+ T cells and delay diabetes in nonobese diabetic mice. J Immunol. 2010;184:4196–4204.
58. Hess SM, Young EF, Miller KR, et al. Deletion of naive T cells recognizing the minor histocompatibility antigen HY with toxin-coupled peptide-MHC class I tetramers inhibits cognate CTL responses and alters immunodominance. Transpl Immunol. 2013;29:138–145.