Journal List > J Korean Soc Transplant > v.31(3) > 1034540

Lee: Immunologic Mechanism of Ischemia Reperfusion Injury in Transplantation

Abstract

Ischemia-reperfusion injury (IRI) is an inevitable consequence of organ transplantation that has major consequences for graft-and patient survival. During transplantation procedures, allografts are exposed to various periods of complete ischemia. Ischemic insult starts with brain death, and its associated hemodynamic disturbances continue during donor organ procurement, cold preservation, and implantation. Ischemia-reperfusion injury, which is a risk factor for acute graft injury, delayed graft function, and acute and chronic rejection, is triggered following reperfusion. Along the cascade of pathogenic events that accompany ischemic insults and cause IRI, there has been an appreciation for various immune mechanisms within the allograft itself. The pathophysiological events associated with IRI originate in signals derived from pattern recognition receptors (PRRs) expressed in the donor organ. Danger associated molecular patterns (DAMP) released from injured cells serve as ligands for PRRs expressed on many cells in the donor organ. Activation of PRR signaling in the donor organ leads to production of proinflammatory cytokines and activates the innate immune system, triggering adaptive immune responses as well as cell death signaling, ultimately worsening the initial ischemic injury. Accordingly, deciphering the inflammatory pathway of innate immunity in IRI may provide a good therapeutic target to block acute sterile inflammation caused by tissue damage.

Figures and Tables

Fig. 1

Schematic vies of innate inflammatory response. Abbreviations: DAMPs, Danger associated molecular patterns; TLRs, Toll-like receptors; TRAF6, TNF receptor-associated factor 6; MyD88, Myeloid differentiation primary response 88; TIRAP, Toll-interleukin 1 receptor (TIR) domain containing adaptor protein; TRAM, TRIF-related adaptor molecule; TRIF, TIR domain containing adaptor protein inducing interferon; IRAK1, Interleukin 1- receptor-associated kinase 1; TBK1, TANK binding kinase 1; IKK, Inhibitor of nuclear factor kappa-B kinase; NFkB, Nuclear factor kappa B; MAP3, MAP3 kinase; IFR3, Interferon regulatory factor 3.

jkstn-31-99-g001
Fig. 2

ROS, cell stress/death, emission of DAMP axis initiating inflammation. Abbreviations: ROS, Reactive oxygen species; DAMP, Danger associated molecular pattern; PRR, pattern recognition receptor.

jkstn-31-99-g002
Fig. 3

An example of model summarizing the role of the tubular epithelial cell/NK cell/neutrophil axis in kidney IRI. Injury to TECs following IRI (step 1) promotes release of HMGB1 (step 2). This molecule stimulates TECs to produce CCR5 chemokines through TLR2 activation (step 3) in an autocrine fashion. CCR5 chemokines in turn induce NK cell recruitment (step 5). Infiltrated NK cells use their cell surface molecule CD137 to stimulate CD137L on the surface of TECs (step 6). CD137L signaling results in the production of additional signaling molecules, CXCL1 and CXCL2, in TECs (step 7). Once infiltrated (step 8), neutrophils participate in active tissue destruction (step 9). Abbreviations: CCR5, chemokine receptors 5; CD137L, CD137 ligand; CXCL1, CXC chemokine ligand 1; CXCR2, C-X-C chemokine receptor type 2; HMGB1, High mobility group box-1 protein; IR, ischemia-reperfusion; NK cells, Natural killer cells; TECs, tubular epithelial cells; TLR2, toll like receptor 2. Reprinted from Fig. 6 of reference [80].

jkstn-31-99-g003
Table 1

Type of pattern recognition receptor (PRR) and their ligand

jkstn-31-99-i001

Abbreviations: iE-DAP, D-gamma-Glu-mDAP; LPS, Lipopolysaccharides; MDP, muramyl dipeptide; NOD, nucleotide-binding oligomerization domain; RLR, RIG-I-like receptors SAP, serum amyloid P.

Table 2

A list of prominent damage-associated molecular patterns (DAMPs) associated with cell stress/cell death and/or tissue injury

jkstn-31-99-i002

Abbreviations: AIM2, absent in melanoma 2; CD, cluster of differentiation; cGAS, cyclic GMP-AMP synthase; cytDNA, cytosolic DNA; FEEL-1, fasciclin epidermal growth factor-like/common lymphatic endothelial and vascular endothelial receptor-1; mtDNA, mitochondrial DNA; NKG2D, natural-killer group 2, member D; NLRP3, NLR family, pyrin domain-containing protein 3; P2X7, purinergic P2X7 receptor; P2Y2, purinergic P2Y2 receptor; RAGE, receptor for advanced glycation end products; RIG-I, retinoic acid inducible gene I; SREC-1, scavenger receptor class f member 1; TIM, transmembrane immunoglobulin and mucin domain; TLR, toll-like receptor.

Notes

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government. (Ministry of Education) (NRF-2015R1D1A3A01020086).

References

1. Johnson KJ, Weinberg JM. Postischemic renal injury due to oxygen radicals. Curr Opin Nephrol Hypertens. 1993; 2:625–635.
crossref
2. Perico N, Cattaneo D, Sayegh MH, Remuzzi G. Delayed graft function in kidney transplantation. Lancet. 2004; 364:1814–1827.
crossref
3. Kosieradzki M, Rowinski W. Ischemia/reperfusion injury in kidney transplantation: mechanisms and prevention. Transplant Proc. 2008; 40:3279–3288.
crossref
4. Ali S, Sheerin NS. Biomarkers of acute injury: predicting the long-term outcome after transplantation. Kidney Int. 2013; 84:1072–1074.
crossref
5. Gjertson DW, Cecka JM. Living unrelated donor kidney transplantation. Kidney Int. 2000; 58:491–499.
crossref
6. Sapir-Pichhadze R, Young A, Joseph Kim S. Living donor age and kidney transplant outcomes: an assessment of risk across the age continuum. Transpl Int. 2013; 26:493–501.
crossref
7. Damman J, Daha MR, van Son WJ, Leuvenink HG, Ploeg RJ, Seelen MA. Crosstalk between complement and Toll-like receptor activation in relation to donor brain death and renal ischemia-reperfusion injury. Am J Transplant. 2011; 11:660–669.
crossref
8. Westendorp WH, Leuvenink HG, Ploeg RJ. Brain death induced renal injury. Curr Opin Organ Transplant. 2011; 16:151–156.
crossref
9. Fuquay R, Renner B, Kulik L, McCullough JW, Amura C, Strassheim D, et al. Renal ischemia-reperfusion injury amplifies the humoral immune response. J Am Soc Nephrol. 2013; 24:1063–1072.
crossref
10. Pratt JR, Basheer SA, Sacks SH. Local synthesis of complement component C3 regulates acute renal transplant rejection. Nat Med. 2002; 8:582–587.
crossref
11. Menke J, Sollinger D, Schamberger B, Heemann U, Lutz J. The effect of ischemia/reperfusion on the kidney graft. Curr Opin Organ Transplant. 2014; 19:395–400.
crossref
12. Ponticelli C. Ischaemia-reperfusion injury: a major protagonist in kidney transplantation. Nephrol Dial Transplant. 2014; 29:1134–1140.
crossref
13. Ortiz F, Paavonen T, Tornroth T, Koskinen P, Finne P, Salmela K, et al. Predictors of renal allograft histologic damage progression. J Am Soc Nephrol. 2005; 16:817–824.
crossref
14. Denecke C, Tullius SG. Innate and adaptive immune responses subsequent to ischemia-reperfusion injury in the kidney. Prog Urol. 2014; 24:Suppl 1. S13–S19.
crossref
15. Cavaille-Coll M, Bala S, Velidedeoglu E, Hernandez A, Archdeacon P, Gonzalez G, et al. Summary of FDA workshop on ischemia reperfusion injury in kidney transplantation. Am J Transplant. 2013; 13:1134–1148.
crossref
16. Gueler F, Gwinner W, Schwarz A, Haller H. Long-term effects of acute ischemia and reperfusion injury. Kidney Int. 2004; 66:523–527.
crossref
17. Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010; 10:826–837.
crossref
18. Iyer SS, Pulskens WP, Sadler JJ, Butter LM, Teske GJ, Ulland TK, et al. Necrotic cells trigger a sterile inflammatory response through the Nlrp3 inflammasome. Proc Natl Acad Sci U S A. 2009; 106:20388–20393.
crossref
19. McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CC, et al. Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science. 2010; 330:362–366. Erratum in Science. 2011 Mar 25;331 (6024):1517.
crossref
20. Wahl LM, Shankavaram U, Zhang Y. Role of macrophages in vascular tissue remodelling. Transpl Immunol. 1997; 5:173–176.
crossref
21. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003; 21:335–376.
crossref
22. Callahan MK, Halleck MS, Krahling S, Henderson AJ, Williamson P, Schlegel RA. Phosphatidylserine expression and phagocytosis of apoptotic thymocytes during differentiation of monocytic cells. J Leukoc Biol. 2003; 74:846–856.
crossref
23. Lavelle EC, Murphy C, O'Neill LA, Creagh EM. The role of TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis. Mucosal Immunol. 2010; 3:17–28.
crossref
24. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010; 140:805–820.
crossref
25. Schroder K, Tschopp J. The inflammasomes. Cell. 2010; 140:821–832.
crossref
26. Chouchani ET, Pell VR, James AM, Work LM, Saeb-Parsy K, Frezza C, et al. A unifying mechanism for mitochondrial superoxide production during ischemia-reperfusion injury. Cell Metab. 2016; 23:254–263.
crossref
27. Zweier JL, Flaherty JT, Weisfeldt ML. Direct measurement of free radical generation following reperfusion of ischemic myocardium. Proc Natl Acad Sci U S A. 1987; 84:1404–1407.
crossref
28. Montezano AC, Touyz RM. Reactive oxygen species and endothelial function: role of nitric oxide synthase uncoupling and Nox family nicotinamide adenine dinucleotide phosphate oxidases. Basic Clin Pharmacol Toxicol. 2012; 110:87–94.
crossref
29. Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 2012; 298:229–317.
crossref
30. Linkermann A, Hackl MJ, Kunzendorf U, Walczak H, Krautwald S, Jevnikar AM. Necroptosis in immunity and ischemia-reperfusion injury. Am J Transplant. 2013; 13:2797–2804.
crossref
31. Silke J, Rickard JA, Gerlic M. Erratum: the diverse role of RIP kinases in necroptosis and inflammation. Nat Immunol. 2015; 16:889.
crossref
32. Galluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, et al. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ. 2015; 22:58–73.
33. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. N Engl J Med. 2009; 361:1570–1583.
crossref
34. Chekeni FB, Elliott MR, Sandilos JK, Walk SF, Kinchen JM, Lazarowski ER, et al. Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis. Nature. 2010; 467:863–867.
crossref
35. Wickman GR, Julian L, Mardilovich K, Schumacher S, Munro J, Rath N, et al. Blebs produced by actin-myosin contraction during apoptosis release damage-associated molecular pattern proteins before secondary necrosis occurs. Cell Death Differ. 2013; 20:1293–1305.
crossref
36. Mulay SR, Linkermann A, Anders HJ. Necroinflammation in Kidney Disease. J Am Soc Nephrol. 2016; 27:27–39.
crossref
37. Tabas I, Ron D. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol. 2011; 13:184–190.
crossref
38. Finka A, Sharma SK, Goloubinoff P. Multi-layered molecular mechanisms of polypeptide holding, unfolding and disaggregation by HSP70/HSP110 chaperones. Front Mol Biosci. 2015; 2:29.
crossref
39. Hetz C, Chevet E, Oakes SA. Proteostasis control by the unfolded protein response. Nat Cell Biol. 2015; 17:829–838.
crossref
40. Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 2014; 3:e02523.
crossref
41. Ciccia A, Elledge SJ. The DNA damage response: making it safe to play with knives. Mol Cell. 2010; 40:179–204.
crossref
42. Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature. 2009; 461:1071–1078.
crossref
43. Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007; 81:1–5.
crossref
44. Rubartelli A, Lotze MT. Inside, outside, upside down: damage-associated molecular-pattern molecules (DAMPs) and redox. Trends Immunol. 2007; 28:429–436.
crossref
45. Gallo PM, Gallucci S. The dendritic cell response to classic, emerging, and homeostatic danger signals. Implications for autoimmunity. Front Immunol. 2013; 4:138.
crossref
46. Zhou JQ, Qiu T, Zhang L, Chen ZB, Wang ZS, Ma XX, et al. Allopurinol preconditioning attenuates renal ischemia/reperfusion injury by inhibiting HMGB1 expression in a rat model. Acta Cir Bras. 2016; 31:176–182.
crossref
47. Sugihara M, Sadamori H, Nishibori M, Sato Y, Tazawa H, Shinoura S, et al. Anti-high mobility group box 1 monoclonal antibody improves ischemia/reperfusion injury and mode of liver regeneration after partial hepatectomy. Am J Surg. 2016; 211:179–188.
crossref
48. Venereau E, Ceriotti C, Bianchi ME. DAMPs from cell death to new life. Front Immunol. 2015; 6:422.
49. Tsung A, Tohme S, Billiar TR. High-mobility group box-1 in sterile inflammation. J Intern Med. 2014; 276:425–443.
crossref
50. Manfredi AA, Capobianco A, Bianchi ME, Rovere-Querini P. Regulation of dendritic- and T-cell fate by injury-associated endogenous signals. Crit Rev Immunol. 2009; 29:69–86.
crossref
51. Chalasani G, Li Q, Konieczny BT, Smith-Diggs L, Wrobel B, Dai Z, et al. The allograft defines the type of rejection (acute versus chronic) in the face of an established effector immune response. J Immunol. 2004; 172:7813–7820.
crossref
52. Nace G, Evankovich J, Eid R, Tsung A. Dendritic cells and damage-associated molecular patterns: endogenous danger signals linking innate and adaptive immunity. J Innate Immun. 2012; 4:6–15.
crossref
53. Garg AD, Krysko DV, Verfaillie T, Kaczmarek A, Ferreira GB, Marysael T, et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J. 2012; 31:1062–1079.
crossref
54. Colombaro V, Jadot I, Decleves AE, Voisin V, Giordano L, Habsch I, et al. Lack of hyaluronidases exacerbates renal post-ischemic injury, inflammation, and fibrosis. Kidney Int. 2015; 88:61–71.
crossref
55. Venner JM, Famulski KS, Badr D, Hidalgo LG, Chang J, Halloran PF. Molecular landscape of T cell-mediated rejection in human kidney transplants: prominence of CTLA4 and PD ligands. Am J Transplant. 2014; 14:2565–2576.
crossref
56. Otterbein LE, Fan Z, Koulmanda M, Thronley T, Strom TB. Innate immunity for better or worse govern the allograft response. Curr Opin Organ Transplant. 2015; 20:8–12.
crossref
57. Zhai Y, Petrowsky H, Hong JC, Busuttil RW, Kupiec-Weglinski JW. Ischaemia-reperfusion injury in liver transplantation: from bench to bedside. Nat Rev Gastroenterol Hepatol. 2013; 10:79–89.
crossref
58. Kruger B, Krick S, Dhillon N, Lerner SM, Ames S, Bromberg JS, et al. Donor Toll-like receptor 4 contributes to ischemia and reperfusion injury following human kidney transplantation. Proc Natl Acad Sci U S A. 2009; 106:3390–3395.
crossref
59. Velasquez-Lopera MM, Correa LA, Garcia LF. Human spleen contains different subsets of dendritic cells and regulatory T lymphocytes. Clin Exp Immunol. 2008; 154:107–114.
crossref
60. Chen L, Xu D, Gao Y, Cui X, Du Z, Ding Q, et al. Effect of donor JNK signal transduction inhibition on transplant outcome in brain dead rat model. Inflammation. 2012; 35:122–129.
crossref
61. Rostron AJ, Cork DM, Avlonitis VS, Fisher AJ, Dark JH, Kirby JA. Contribution of Toll-like receptor activation to lung damage after donor brain death. Transplantation. 2010; 90:732–739.
crossref
62. Devarajan P. Update on mechanisms of ischemic acute kidney injury. J Am Soc Nephrol. 2006; 17:1503–1520.
crossref
63. Thurman JM, Royer PA, Ljubanovic D, Dursun B, Lenderink AM, Edelstein CL, et al. Treatment with an inhibitory monoclonal antibody to mouse factor B protects mice from induction of apoptosis and renal ischemia/reperfusion injury. J Am Soc Nephrol. 2006; 17:707–715.
crossref
64. Kelly KJ, Williams WW Jr, Colvin RB, Meehan SM, Springer TA, Gutierrez-Ramos JC, et al. Intercellular adhesion molecule-1-deficient mice are protected against ischemic renal injury. J Clin Invest. 1996; 97:1056–1063.
crossref
65. Kelly KJ, Williams WW Jr, Colvin RB, Bonventre JV. Antibody to intercellular adhesion molecule 1 protects the kidney against ischemic injury. Proc Natl Acad Sci U S A. 1994; 91:812–816.
crossref
66. Haug CE, Colvin RB, Delmonico FL, Auchincloss H Jr, Tolkoff-Rubin N, Preffer FI, et al. A phase I trial of immunosuppression with anti-ICAM-1 (CD54) mAb in renal allograft recipients. Transplantation. 1993; 55:766–772. discussion 772-3.
crossref
67. Salmela K, Wramner L, Ekberg H, Hauser I, Bentdal O, Lins LE, et al. A randomized multicenter trial of the anti-ICAM-1 monoclonal antibody (enlimomab) for the prevention of acute rejection and delayed onset of graft function in cadaveric renal transplantation: a report of the European Anti-ICAM-1 Renal Transplant Study Group. Transplantation. 1999; 67:729–736.
crossref
68. Kwan T, Wu H, Chadban SJ. Macrophages in renal transplantation: roles and therapeutic implications. Cell Immunol. 2014; 291:58–64.
crossref
69. Tsung A, Sahai R, Tanaka H, Nakao A, Fink MP, Lotze MT, et al. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med. 2005; 201:1135–1143.
crossref
70. Messmer D, Yang H, Telusma G, Knoll F, Li J, Messmer B, et al. High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol. 2004; 173:307–313.
crossref
71. Kielar ML, John R, Bennett M, Richardson JA, Shelton JM, Chen L, et al. Maladaptive role of IL-6 in ischemic acute renal failure. J Am Soc Nephrol. 2005; 16:3315–3325.
crossref
72. Donnahoo KK, Meng X, Ayala A, Cain MP, Harken AH, Meldrum DR. Early kidney TNF-alpha expression mediates neutrophil infiltration and injury after renal ischemia-reperfusion. Am J Physiol. 1999; 277:R922–R929.
73. Wu H, Craft ML, Wang P, Wyburn KR, Chen G, Ma J, et al. IL-18 contributes to renal damage after ischemia-reperfusion. J Am Soc Nephrol. 2008; 19:2331–2341.
crossref
74. Dong X, Swaminathan S, Bachman LA, Croatt AJ, Nath KA, Griffin MD. Resident dendritic cells are the predominant TNF-secreting cell in early renal ischemia-reperfusion injury. Kidney Int. 2007; 71:619–628.
crossref
75. Alikhan MA, Jones CV, Williams TM, Beckhouse AG, Fletcher AL, Kett MM, et al. Colony-stimulating factor-1 promotes kidney growth and repair via alteration of macrophage responses. Am J Pathol. 2011; 179:1243–1256.
crossref
76. Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998; 101:890–898.
crossref
77. Huen SC, Cantley LG. Macrophage-mediated injury and repair after ischemic kidney injury. Pediatr Nephrol. 2015; 30:199–209.
crossref
78. Filardy AA, Pires DR, Nunes MP, Takiya CM, Freire-de-Lima CG, Ribeiro-Gomes FL, et al. Proinflammatory clearance of apoptotic neutrophils induces an IL-12(low) IL-10(high) regulatory phenotype in macrophages. J Immunol. 2010; 185:2044–2050.
crossref
79. Zhang ZX, Shek K, Wang S, Huang X, Lau A, Yin Z, et al. Osteopontin expressed in tubular epithelial cells regulates NK cell-mediated kidney ischemia reperfusion injury. J Immunol. 2010; 185:967–973.
crossref
80. Kim HJ, Lee JS, Kim A, Koo S, Cha HJ, Han JA, et al. TLR2 signaling in tubular epithelial cells regulates NK cell recruitment in kidney ischemia-reperfusion injury. J Immunol. 2013; 191:2657–2664.
crossref
81. Kim HJ, Lee JS, Kim JD, Cha HJ, Kim A, Lee SK, et al. Reverse signaling through the costimulatory ligand CD137L in epithelial cells is essential for natural killer cell-mediated acute tissue inflammation. Proc Natl Acad Sci U S A. 2012; 109:E13–E22.
crossref
82. Lemley KV, Kriz W. Anatomy of the renal interstitium. Kidney Int. 1991; 39:370–381.
crossref
83. Soos TJ, Sims TN, Barisoni L, Lin K, Littman DR, Dustin ML, et al. CX3CR1+ interstitial dendritic cells form a contiguous network throughout the entire kidney. Kidney Int. 2006; 70:591–596.
crossref
84. Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003; 3:745–756.
crossref
85. Loverre A, Capobianco C, Stallone G, Infante B, Schena A, Ditonno P, et al. Ischemia-reperfusion injury-induced abnormal dendritic cell traffic in the transplanted kidney with delayed graft function. Kidney Int. 2007; 72:994–1003.
crossref
86. Diao H, Kon S, Iwabuchi K, Kimura C, Morimoto J, Ito D, et al. Osteopontin as a mediator of NKT cell function in T cell-mediated liver diseases. Immunity. 2004; 21:539–550.
crossref
87. Sharma AK, LaPar DJ, Zhao Y, Li L, Lau CL, Kron IL, et al. Natural killer T cell-derived IL-17 mediates lung ischemia-reperfusion injury. Am J Respir Crit Care Med. 2011; 183:1539–1549.
crossref
88. Lappas CM, Day YJ, Marshall MA, Engelhard VH, Linden J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation. J Exp Med. 2006; 203:2639–2648.
crossref
89. Li L, Huang L, Sung SS, Lobo PI, Brown MG, Gregg RK, et al. NKT cell activation mediates neutrophil IFN-gamma production and renal ischemia-reperfusion injury. J Immunol. 2007; 178:5899–5911.
crossref
90. Wang S, Diao H, Guan Q, Cruikshank WW, Delovitch TL, Jevnikar AM, et al. Decreased renal ischemia-reperfusion injury by IL-16 inactivation. Kidney Int. 2008; 73:318–326.
crossref
91. Burne MJ, Daniels F, El Ghandour A, Mauiyyedi S, Colvin RB, O'Donnell MP, et al. Identification of the CD4(+) T cell as a major pathogenic factor in ischemic acute renal failure. J Clin Invest. 2001; 108:1283–1290.
crossref
92. De Greef KE, Ysebaert DK, Dauwe S, Persy V, Vercauteren SR, Mey D, et al. Anti-B7-1 blocks mononuclear cell adherence in vasa recta after ischemia. Kidney Int. 2001; 60:1415–1427.
crossref
93. Takada M, Chandraker A, Nadeau KC, Sayegh MH, Tilney NL. The role of the B7 costimulatory pathway in experimental cold ischemia/reperfusion injury. J Clin Invest. 1997; 100:1199–1203.
crossref
94. Shen X, Wang Y, Gao F, Ren F, Busuttil RW, Kupiec-Weglinski JW, et al. CD4 T cells promote tissue inflammation via CD40 signaling without de novo activation in a murine model of liver ischemia/reperfusion injury. Hepatology. 2009; 50:1537–1546.
crossref
95. Satpute SR, Park JM, Jang HR, Agreda P, Liu M, Gandolfo MT, et al. The role for T cell repertoire/antigen-specific interactions in experimental kidney ischemia reperfusion injury. J Immunol. 2009; 183:984–992.
crossref
96. Ascon DB, Lopez-Briones S, Liu M, Ascon M, Savransky V, Colvin RB, et al. Phenotypic and functional characterization of kidney-infiltrating lymphocytes in renal ischemia reperfusion injury. J Immunol. 2006; 177:3380–3387.
crossref
97. Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, et al. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med. 2009; 15:192–199.
crossref
98. Nadig SN, Wieckiewicz J, Wu DC, Warnecke G, Zhang W, Luo S, et al. In vivo prevention of transplant arteriosclerosis by ex vivo-expanded human regulatory T cells. Nat Med. 2010; 16:809–813.
crossref
99. Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med. 2007; 13:1299–1307.
crossref
100. Chen G, Chen S, Chen X. Role of complement and perspectives for intervention in transplantation. Immunobiology. 2013; 218:817–827.
crossref
101. van der Touw W, Cravedi P, Kwan WH, Paz-Artal E, Merad M, Heeger PS. Cutting edge: receptors for C3a and C5a modulate stability of alloantigen-reactive induced regulatory T cells. J Immunol. 2013; 190:5921–5925.
crossref
102. Thurman JM, Ljubanovic D, Royer PA, Kraus DM, Molina H, Barry NP, et al. Altered renal tubular expression of the complement inhibitor Crry permits complement activation after ischemia/reperfusion. J Clin Invest. 2006; 116:357–368.
crossref
103. Zhang M, Alicot EM, Chiu I, Li J, Verna N, Vorup-Jensen T, et al. Identification of the target self-antigens in reperfusion injury. J Exp Med. 2006; 203:141–152.
crossref
104. Kulik L, Fleming SD, Moratz C, Reuter JW, Novikov A, Chen K, et al. Pathogenic natural antibodies recognizing annexin IV are required to develop intestinal ischemia-reperfusion injury. J Immunol. 2009; 182:5363–5373.
crossref
105. Singbartl K, Ley K. Protection from ischemia-reperfusion induced severe acute renal failure by blocking E-selectin. Crit Care Med. 2000; 28:2507–2514.
crossref
106. Takada M, Nadeau KC, Shaw GD, Marquette KA, Tilney NL. The cytokine-adhesion molecule cascade in ischemia/reperfusion injury of the rat kidney. Inhibition by a soluble P-selectin ligand. J Clin Invest. 1997; 99:2682–2690.
crossref
107. Sutton TA, Fisher CJ, Molitoris BA. Microvascular endothelial injury and dysfunction during ischemic acute renal failure. Kidney Int. 2002; 62:1539–1549.
crossref
108. Garcia JG. Concepts in microvascular endothelial barrier regulation in health and disease. Microvasc Res. 2009; 77:1–3.
crossref
109. Oh DJ, Dursun B, He Z, Lu L, Hoke TS, Ljubanovic D, et al. Fractalkine receptor (CX3CR1) inhibition is protective against ischemic acute renal failure in mice. Am J Physiol Renal Physiol. 2008; 294:F264–F271.
crossref
110. Wu H, Chen G, Wyburn KR, Yin J, Bertolino P, Eris JM, et al. TLR4 activation mediates kidney ischemia/reperfusion injury. J Clin Invest. 2007; 117:2847–2859.
crossref
111. Wolfs TG, Buurman WA, van Schadewijk A, de Vries B, Daemen MA, Hiemstra PS, et al. In vivo expression of Toll-like receptor 2 and 4 by renal epithelial cells: IFN-gamma and TNF-alpha mediated up-regulation during inflammation. J Immunol. 2002; 168:1286–1293.
crossref
112. Kim BS, Lim SW, Li C, Kim JS, Sun BK, Ahn KO, et al. Ischemia-reperfusion injury activates innate immunity in rat kidneys. Transplantation. 2005; 79:1370–1377.
crossref
113. Leemans JC, Stokman G, Claessen N, Rouschop KM, Teske GJ, Kirschning CJ, et al. Renal-associated TLR2 mediates ischemia/reperfusion injury in the kidney. J Clin Invest. 2005; 115:2894–2903.
crossref
TOOLS
Similar articles