دورية أكاديمية

Innate Allorecognition in Transplantation: Ancient Mechanisms With Modern Impact.

التفاصيل البيبلوغرافية
العنوان: Innate Allorecognition in Transplantation: Ancient Mechanisms With Modern Impact.
المؤلفون: Gui Z; Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA., Al Moussawy M, Sanders SM, Abou-Daya KI
المصدر: Transplantation [Transplantation] 2024 Jul 01; Vol. 108 (7), pp. 1524-1531. Date of Electronic Publication: 2023 Dec 05.
نوع المنشور: Journal Article; Review
اللغة: English
بيانات الدورية: Publisher: Lippincott Williams & Wilkins Country of Publication: United States NLM ID: 0132144 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1534-6080 (Electronic) Linking ISSN: 00411337 NLM ISO Abbreviation: Transplantation Subsets: MEDLINE
أسماء مطبوعة: Publication: Hagerstown, MD : Lippincott Williams & Wilkins
Original Publication: Baltimore, Williams & Wilkins.
مواضيع طبية MeSH: Immunity, Innate* , Graft Rejection*/immunology , Graft Rejection*/prevention & control , Organ Transplantation*/history , Immunologic Memory*, Humans ; Animals ; Graft Survival/immunology ; Adaptive Immunity ; Transplantation Immunology ; Transplantation Tolerance
مستخلص: Through the effective targeting of the adaptive immune system, solid organ transplantation became a life-saving therapy for organ failure. However, beyond 1 y of transplantation, there is little improvement in transplant outcomes. The adaptive immune response requires the activation of the innate immune system. There are no modalities for the specific targeting of the innate immune system involvement in transplant rejection. However, the recent discovery of innate allorecognition and innate immune memory presents novel targets in transplantation that will increase our understanding of organ rejection and might aid in improving transplant outcomes. In this review, we look at the latest developments in the study of innate allorecognition and innate immune memory in transplantation.
Competing Interests: The authors declare no conflicts of interest.
(Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
References: Hariharan S, Israni AK, Danovitch G. Long-term survival after kidney transplantation. N Engl J Med. 2021;385:729–743.
Black CK, Termanini KM, Aguirre O, et al. Solid organ transplantation in the 21(st) century. Ann Transl Med. 2018;6:409.
Thabut G, Mal H. Outcomes after lung transplantation. J Thorac Dis. 2017;9:2684–2691.
Moayedi Y, Fan CPS, Cherikh WS, et al. Survival outcomes after heart transplantation: does recipient sex matter? Circ Heart Fail. 2019;12:e006218.
Rana A, Ackah RL, Webb GJ, et al. No gains in long-term survival after liver transplantation over the past three decades. Ann Surg. 2019;269:20–27.
Lakkis FG, Lechler RI. Origin and biology of the allogeneic response. Cold Spring Harb Perspect Med. 2013;3:a014993.
Zecher D, Li Q, Williams AL, et al. Innate immunity alone is not sufficient for chronic rejection but predisposes healed allografts to T cell-mediated pathology. Transpl Immunol. 2011;26:113–118.
Todd JL, Palmer SM. Danger signals in regulating the immune response to solid organ transplantation. J Clin Invest. 2017;127:2464–2472.
Lima K, Ribas GT, Riella LV, et al. Inhibitory innate receptors and their potential role in transplantation. Transplant Rev (Orlando). 2023;37:100776.
Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous activators of dendritic cells. Nat Med. 1999;5:1249–1255.
Felix NJ, Donermeyer DL, Horvath S, et al. Alloreactive T cells respond specifically to multiple distinct peptide-MHC complexes. Nat Immunol. 2007;8:388–397.
Sykes M, Wood K, Sachs DH. Transplantation immunology. In: Paul WE, ed. Fundamental Immunology. 6th ed. Lippincott Williams & Wilkins Publishers; 2008:1426–1488.
Goldstein DR, Tesar BM, Akira S, et al. Critical role of the Toll-like receptor signal adaptor protein MyD88 in acute allograft rejection. J Clin Invest. 2003;111:1571–1578.
Tesar BM, Zhang J, Li Q, et al. TH1 immune responses to fully MHC mismatched allografts are diminished in the absence of MyD88, a toll-like receptor signal adaptor protein. Am J Transplant. 2004;4:1429–1439.
Hutton MJ, Westwell-Roper C, Soukhatcheva G, et al. Islet allograft rejection is independent of toll-like receptor signaling in mice. Transplantation. 2009;88:1075–1080.
McKay D, Shigeoka A, Rubinstein M, et al. Simultaneous deletion of MyD88 and Trif delays major histocompatibility and minor antigen mismatch allograft rejection. Eur J Immunol. 2006;36:1994–2002.
Oberbarnscheidt MH, Obhrai JS, Williams AL, et al. Type I interferons are not critical for skin allograft rejection or the generation of donor-specific CD8+ memory T cells. Am J Transplant. 2010;10:162–167.
Li H, Matte-Martone C, Tan HS, et al. Graft-versus-host disease is independent of innate signaling pathways triggered by pathogens in host hematopoietic cells. J Immunol. 2011;186:230–241.
Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045.
Oberbarnscheidt MH, Lakkis FG. Innate allorecognition. Immunol Rev. 2014;258:145–149.
Lakkis FG, Li XC. Innate allorecognition by monocytic cells and its role in graft rejection. Am J Transplant. 2018;18:289–292.
Panzer SE. Macrophages in transplantation: a matter of plasticity, polarization, and diversity. Transplantation. 2022;106:257–267.
Abou-Daya KI, Oberbarnscheidt MH. Innate allorecognition in transplantation. J Heart Lung Transplant. 2021;40:557–561.
Dai H, Friday AJ, Abou-Daya KI, et al. Donor SIRPa polymorphism modulates the innate immune response to allogeneic grafts. Sci Immunol. 2017;2:eaam6202.
Dai H, Lan P, Zhao D, et al. PIRs mediate innate myeloid cell memory to nonself MHC molecules. Science. 2020;368:1122–1127.
Uehara S, Chase CM, Kitchens WH, et al. NK cells can trigger allograft vasculopathy: the role of hybrid resistance in solid organ allografts. J Immunol. 2005;175:3424–3430.
Zecher D, Li Q, Williams AL, et al. Innate immunity alone is not sufficient for chronic rejection but predisposes healed allografts to T cell-mediated pathology. Transpl Immunol. 2012;26:113–118.
Hirayasu K, Arase H. Functional and genetic diversity of leukocyte immunoglobulin-like receptor and implication for disease associations. J Hum Genet. 2015;60:703–708.
Garcia-Sanchez C, Casillas-Abundis MA, Pinelli DF, et al. Impact of SIRPalpha polymorphism on transplant outcomes in HLA-identical living donor kidney transplantation. Clin Transplant. 2021;35:e14406.
Oberbarnscheidt MH, Zecher D, Lakkis FG. The innate immune system in transplantation. Semin Immunol. 2011;23:264–272.
Chong AS, Alegre ML. The impact of infection and tissue damage in solid-organ transplantation. Nat Rev Immunol. 2012;12:459–471.
Lei YM, Chen L, Wang Y, et al. The composition of the microbiota modulates allograft rejection. J Clin Invest. 2016;126:2736–2744.
Rosengarten RD, Nicotra ML. Model systems of invertebrate allorecognition. Curr Biol. 2011;21:R82–R92.
Nicotra ML, Powell AE, Rosengarten RD, et al. A hypervariable invertebrate allodeterminant. Curr Biol. 2009;19:583–589.
Rosa SF, Powell AE, Rosengarten RD, et al. Hydractinia allodeterminant alr1 resides in an immunoglobulin superfamily-like gene complex. Curr Biol. 2010;20:1122–1127.
Huene AL, Sanders SM, Ma Z, et al. A family of unusual immunoglobulin superfamily genes in an invertebrate histocompatibility complex. Proc Natl Acad Sci U S A. 2022;119:e2207374119.
Janeway CA Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol. 1989;54(Pt 1):1–13.
Broz P, Monack DM. Newly described pattern recognition receptors team up against intracellular pathogens. Nat Rev Immunol. 2013;13:551–565.
Medzhitov R, PrestonHurlburt P, Janeway CA. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388:394–397.
Poltorak A, He X, Smirnova I, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282:2085–2088.
Klein J, Sato A. The HLA system first of two parts. N Engl J Med. 2000;343:702–709.
Zinkernagel RM, Doherty PC. Immunological surveillance against altered self components by sensitised T lymphocytes in lymphocytic choriomeningitis. Nature. 1974;251:547–548.
Bjorkman PJ, Saper MA, Samraoui B, et al. Structure of the human class I histocompatibility antigen HLA-A2. Nature. 1987;329:506–512.
Zecher D, van Rooijen N, Rothstein D, et al. An innate response to allogeneic nonself mediated by monocytes. J Immunol. 2009;183:7810–7816.
Liu W, Xiao X, Demirci G, et al. Innate NK cells and macrophages recognize and reject allogeneic nonself in vivo via different mechanisms. J Immunol. 2012;188:2703–2711.
Chen W, Ghobrial RM, Li XC. The evolving roles of memory immune cells in transplantation. Transplantation. 2015;99:2029–2037.
Oberbarnscheidt MH, Zeng Q, Li Q, et al. Non-self recognition by monocytes initiates allograft rejection. J Clin Invest. 2014;124:3579–3589.
Chow KV, Delconte RB, Huntington ND, et al. Innate allorecognition results in rapid accumulation of monocyte-derived dendritic cells. J Immunol. 2016;197:2000–2008.
Barclay AN, Van den Berg TK. The interaction between signal regulatory protein alpha (SIRPalpha) and CD47: structure, function, and therapeutic target. Ann Rev Immunol. 2014;32:25–50.
Yamauchi T, Takenaka K, Urata S, et al. Polymorphic Sirpa is the genetic determinant for NOD-based mouse lines to achieve efficient human cell engraftment. Blood. 2013;121:1316–1325.
Wong AS, Mortin-Toth S, Sung M, et al. Polymorphism in the innate immune receptor SIRPalpha controls CD47 binding and autoimmunity in the nonobese diabetic mouse. J Immunol. 2014;193:4833–4844.
Xie MM, Dai B, Hackney JA, et al. An agonistic anti-signal regulatory protein alpha antibody for chronic inflammatory diseases. Cell Rep Med. 2023;101130.
Charmetant X, Bachelet T, Dechanet-Merville J, et al. Innate (and innate-like) lymphoid cells: emerging immune subsets with multiple roles along transplant life. Transplantation. 2021;105:e322–e336.
Lanier LL. NK cell recognition. Annu Rev Immunol. 2005;23:225–274.
Hankey KG, Drachenberg CB, Papadimitriou JC, et al. MIC expression in renal and pancreatic allografts. Transplantation. 2002;73:304–306.
Alexander KL, Ford ML. The entangled world of memory T cells and implications in transplantation. Transplantation. 2024;108:137–147.
Espinosa JR, Samy KP, Kirk AD. Memory T cells in organ transplantation: progress and challenges. Nat Rev Nephrol. 2016;12:339–347.
Ford ML, Larsen CP. Overcoming the memory barrier in tolerance induction: molecular mimicry and functional heterogeneity among pathogen-specific T-cell populations. Curr Opin Organ Transplant. 2010;15:405–410.
Macedo C, Orkis EA, Popescu I, et al. Contribution of naïve and memory T-cell populations to the human alloimmune response. Am J Transplant. 2009;9:2057–2066.
O’Leary JG, Goodarzi M, Drayton DL, et al. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat Immunol. 2006;7:507–516.
Paust S, Gill HS, Wang BZ, et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat Immunol. 2010;11:1127–1135.
Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer cells. Nature. 2009;457:557–561.
Cooper MA, Elliott JM, Keyel PA, et al. Cytokine-induced memory-like natural killer cells. Proc Natl Acad Sci USA. 2009;106:1915–1919.
Quintin J, Saeed S, Martens JH, et al. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe. 2012;12:223–232.
Nabekura T, Lanier LL. Antigen-specific expansion and differentiation of natural killer cells by alloantigen stimulation. J Exp Med. 2014;211:2455–2465.
Weavers H, Evans IR, Martin P, et al. Corpse engulfment generates a molecular memory that primes the macrophage inflammatory response. Cell. 2016;165:1658–1671.
Hole CR, Wager CML, Castro-Lopez N, et al. Induction of memory-like dendritic cell responses in vivo. Nat Commun. 2019;10:2955.
O’Sullivan TE, Sun JC, Lanier LL. Natural killer cell memory. Immunity. 2015;43:634–645.
Zhao D, Abou-Daya KI, Dai H, et al. Innate allorecognition and memory in transplantation. Front Immunol. 2020;11:918.
Kubagawa H, Chen CC, Ho LH, et al. Biochemical nature and cellular distribution of the paired immunoglobulin-like receptors, PIR-A and PIR-B. J Exp Med. 1999;189:309–318.
Hughes AD, Zhao D, Dai H, et al. Cross-dressed dendritic cells sustain effector T cell responses in islet and kidney allografts. J Clin Invest. 2020;130:287–294.
Patel AA, Zhang Y, Fullerton JN, et al. The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J Exp Med. 2017;214:1913–1923.
Yona S, Kim KW, Wolf Y, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 2013;38:79–91.
Ruckert T, Lareau CA, Mashreghi MF, et al. Clonal expansion and epigenetic inheritance of long-lasting NK cell memory. Nat Immunol. 2022;23:1551–1563.
Netea MG, Quintin J, van der Meer JW. Trained immunity: a memory for innate host defense. Cell Host Microbe. 2011;9:355–361.
Netea MG. Training innate immunity: the changing concept of immunological memory in innate host defence. Eur J Clin Invest. 2013;43:881–884.
Netea MG, Joosten LA, Latz E, et al. Trained immunity: a program of innate immune memory in health and disease. Science. 2016;352:aaf1098.
Gardiner CM, Mills KH. The cells that mediate innate immune memory and their functional significance in inflammatory and infectious diseases. Semin Immunol. 2016;28:343–350.
Pradeu T, Du Pasquier L. Immunological memory: what’s in a name? Immunol Rev. 2018;283:7–20.
Divangahi M, Aaby P, Khader SA, et al. Trained immunity, tolerance, priming and differentiation: distinct immunological processes. Nat Immunol. 2021;22:2–6.
Cunningham KT, Mills KHG. Trained innate immunity in hematopoietic stem cell and solid organ transplantation. Transplantation. 2021;105:1666–1676.
Dominguez-Andres J, Netea MG. The specifics of innate immune memory. Science. 2020;368:1052–1053.
Ochando J, Mulder WJM, Madsen JC, et al. Trained immunity - basic concepts and contributions to immunopathology. Nat Rev Nephrol. 2023;19:23–37.
Zhuang Q, Liu Q, Divito SJ, et al. Graft-infiltrating host dendritic cells play a key role in organ transplant rejection. Nat Commun. 2016;7:12623.
Cherukuri A, Dai H, Oberbarnscheidt M, et al. Genomic mismatch at the Sirpa locus and long-term kidney allograft outcomes. Am J Transplant. 2023;23.
Zhao D, Abou-Daya K, Dai H, et al. Genomic mismatch at the SIRPA locus and long-term kidney allograft outcomes. In: International Transplantation Science Meeting . Transplantation. 2023.
Friday A, Dai H, Williams A, et al. Identification of human SIRPa diversity that could regulate innate allorecognition [abstract]. Am J Transplant. 2017;17(Suppl 3).
Hudson LE, Allen RL. Leukocyte Ig-like receptors - a model for MHC class I disease associations. Front Immunol. 2016;7:281.
Kubagawa H, Burrows PD, Cooper MD. A novel pair of immunoglobulin-like receptors expressed by B cells and myeloid cells. Proc Natl Acad Sci U S A. 1997;94:5261–5266.
Takai T. Paired immunoglobulin-like receptors and their MHC class I recognition. Immunology. 2005;115:433–440.
Tun T, Kubagawa Y, Dennis G, et al. Genomic structure of mouse PIR-A6, an activating member of the paired immunoglobulin-like receptor gene family. Tissue Antigens. 2003;61:220–230.
Nakamura A, Kobayashi E, Takai T. Exacerbated graft-versus-host disease in Pirb-/- mice. Nat Immunol. 2004;5:623–629.
van der Touw W, Chen HM, Pan PY, et al. LILRB receptor-mediated regulation of myeloid cell maturation and function. Cancer Immunol Immunother. 2017;66:1079–1087.
Burshtyn DN, Morcos C. The expanding spectrum of ligands for leukocyte Ig-like receptors. J Immunol. 2016;196:947–955.
Lamarthee B, Callemeyn J, Van Herck Y, et al. Transcriptional and spatial profiling of the kidney allograft unravels a central role for FcyRIII+ innate immune cells in rejection. Nat Commun. 2023;14:4359.
Koenig A, Chen CC, Marcais A, et al. Missing self triggers NK cell-mediated chronic vascular rejection of solid organ transplants. Nat Commun. 2019;10:5350.
Koenig A, Mezaache S, Callemeyn J, et al. Missing self-induced activation of NK cells combines with non-complement-fixing donor-specific antibodies to accelerate kidney transplant loss in chronic antibody-mediated rejection. J Am Soc Nephrol. 2021;32:479–494.
Callemeyn J, Senev A, Coemans M, et al. Missing self-induced microvascular rejection of kidney allografts: a population-based study. J Am Soc Nephrol. 2021;32:2070–2082.
van Bergen JA, Thompson GW, Haasnoot JI, et al. KIR-ligand mismatches are associated with reduced long-term graft survival in HLA-compatible kidney transplantation. Am J Transplant 2011;11:1959–1964.
Zhang Y, Yan AW, Boelen L, et al. KIR-HLA interactions extend human CD8+ T cell lifespan in vivo. J Clin Invest. 2023;133:e169496.
معلومات مُعتمدة: 2020 Stuart K. Patrick Grant for Transplant Innovation Thomas E. Starzl Transplantation Institute
تواريخ الأحداث: Date Created: 20231205 Date Completed: 20240625 Latest Revision: 20240625
رمز التحديث: 20240626
مُعرف محوري في PubMed: PMC11188633
DOI: 10.1097/TP.0000000000004847
PMID: 38049941
قاعدة البيانات: MEDLINE
الوصف
تدمد:1534-6080
DOI:10.1097/TP.0000000000004847