Browse > Article
http://dx.doi.org/10.3904/kjim.2018.095

Clinical significance of Th17 cells in kidney transplantation  

Chung, Byung Ha (Convergent Research Consortium for Immunologic Disease,Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea)
Yang, Chul Woo (Convergent Research Consortium for Immunologic Disease,Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea)
Cho, Mi-La (Convergent Research Consortium for Immunologic Disease,Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea)
Publication Information
The Korean journal of internal medicine / v.33, no.5, 2018 , pp. 860-866 More about this Journal
Abstract
Transplantation research has focused on cytotoxic T-cell and plasma cell/ B-cell-targeted strategies, but little attention has been paid to the role of T helper 17 (Th17) cells in allograft dysfunction. However, accumulating evidence suggests that Th17 cells contribute to the development of acute and chronic allograft injury after transplantation of various organs, including the kidney. This review summarizes recent reports on the role of Th17 cells in kidney transplantation. Means of improving allograft outcomes by targeting the Th17 pathway are also suggested.
Keywords
Allograft rejection; Chronic allograft dysfunction; Kidney transplantation; Mammalian target of rapamycin; Th17 cells;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Yurchenko E, Shio MT, Huang TC, et al. Inflammation-driven reprogramming of CD4+ Foxp3+ regulatory T cells into pathogenic Th1/Th17 T effectors is abrogated by mTOR inhibition in vivo. PLoS One 2012;7:e35572.   DOI
2 Li Y, Shi Y, Huang Z, et al. CNI induced Th17/Treg imbalance and susceptibility to renal dysfunction in renal transplantation. Int Immunopharmacol 2011;11:2033-2038.   DOI
3 Kim KW, Chung BH, Kim BM, Cho ML, Yang CW. The effect of mammalian target of rapamycin inhibition on T helper type 17 and regulatory T cell differentiation in vitro and in vivo in kidney transplant recipients. Immunology 2015;144:68-78.   DOI
4 Mora JR, Iwata M, von Andrian UH. Vitamin effects on the immune system: vitamins A and D take centre stage. Nat Rev Immunol 2008;8:685-698.   DOI
5 Peelen E, Knippenberg S, Muris AH, et al. Effects of vitamin D on the peripheral adaptive immune system: a review. Autoimmun Rev 2011;10:733-743.   DOI
6 Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C. Vitamin D: modulator of the immune system. Curr Opin Pharmacol 2010;10:482-496.   DOI
7 Smolders J, Menheere P, Thewissen M, et al. Regulatory T cell function correlates with serum 25-hydroxyvitamin D, but not with 1,25-dihydroxyvitamin D, parathyroid hormone and calcium levels in patients with relapsing remitting multiple sclerosis. J Steroid Biochem Mol Biol 2010;121:243-246.   DOI
8 Park H, Li Z, Yang XO, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol 2005;6:1133-1141.   DOI
9 Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 2005;6:1123-1132.   DOI
10 Wynn TA. T(H)-17: a giant step from T(H)1 and T(H)2. Nat Immunol 2005;6:1069-1070.   DOI
11 Loong CC, Lin CY, Lui WY. Expression of interleukin-17 as a predictive parameter in acute renal allograft rejection. Transplant Proc 2000;32:1773.   DOI
12 Hsieh HG, Loong CC, Lui WY, Chen A, Lin CY. IL-17 expression as a possible predictive parameter for subclinical renal allograft rejection. Transpl Int 2001;14:287-298.   DOI
13 Mitchell P, Afzali B, Lombardi G, Lechler RI. The T helper 17-regulatory T cell axis in transplant rejection and tolerance. Curr Opin Organ Transplant 2009;14:326-331.   DOI
14 Calvo-Turrubiartes M, Romano-Moreno S, Garcia-Hernandez M, et al. Quantitative analysis of regulatory T cells in kidney graft recipients: a relationship with calcineurin inhibitor level. Transpl Immunol 2009;21:43-49.   DOI
15 Warrens AN. Pharmacological control of the immune response in renal transplantation. BJU Int 2002;90:784-791.   DOI
16 Crispim JC, Grespan R, Martelli-Palomino G, et al. Interleukin-17 and kidney allograft outcome. Transplant Proc 2009;41:1562-1564.   DOI
17 Datta Mitra A, Raychaudhuri SP, Abria CJ, et al. $1{\alpha}$,25-Dihydroxyvitamin-D3-3-bromoacetate regulates AKT/mTOR signaling cascades: a therapeutic agent for psoriasis. J Invest Dermatol 2013;133:1556-1564.   DOI
18 Ranganathan P, Khalatbari S, Yalavarthi S, Marder W, Brook R, Kaplan MJ. Vitamin D deficiency, interleukin 17, and vascular function in rheumatoid arthritis. J Rheumatol 2013;40:1529-1534.   DOI
19 Lisse TS, Hewison M. Vitamin D: a new player in the world of mTOR signaling. Cell Cycle 2011;10:1888-1889.   DOI
20 Lisse TS, Liu T, Irmler M, et al. Gene targeting by the vitamin D response element binding protein reveals a role for vitamin D in osteoblast mTOR signaling. FASEB J 2011;25:937-947.   DOI
21 Chung BH, Kim BM, Doh KC, et al. Suppressive effect of $1{\alpha}$,25-dihydroxyvitamin D3 on Th17-immune responses in kidney transplant recipients with tacrolimus-based immunosuppression. Transplantation 2017;101:1711-1719.   DOI
22 Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B. Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era. Am J Transplant 2004;4:378-383.   DOI
23 Guerra G, Srinivas TR, Meier-Kriesche HU. Calcineurin inhibitor-free immunosuppression in kidney transplantation. Transpl Int 2007;20:813-827.   DOI
24 Deteix C, Attuil-Audenis V, Duthey A, et al. Intragraft Th17 infiltrate promotes lymphoid neogenesis and hastens clinical chronic rejection. J Immunol 2010;184:5344-5351.   DOI
25 Loong CC, Hsieh HG, Lui WY, Chen A, Lin CY. Evidence for the early involvement of interleukin 17 in human and experimental renal allograft rejection. J Pathol 2002;197:322-332.   DOI
26 Van Kooten C, Boonstra JG, Paape ME, et al. Interleukin-17 activates human renal epithelial cells in vitro and is expressed during renal allograft rejection. J Am Soc Nephrol 1998;9:1526-1534.
27 Halloran PF. Immunosuppressive drugs for kidney transplantation. N Engl J Med 2004;351:2715-2729.   DOI
28 Liu Z, Fan H, Jiang S. CD4(+) T-cell subsets in transplantation. Immunol Rev 2013;252:183-191.   DOI
29 Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 2006;24:179-189.   DOI
30 Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006;441:235-238.   DOI
31 Ivanov II, McKenzie BS, Zhou L, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006;126:1121-1133.   DOI
32 Kuchroo VK, Awasthi A. Emerging new roles of Th17 cells. Eur J Immunol 2012;42:2211-2214.   DOI
33 Rentenaar RJ, van Diepen FN, Meijer RT, et al. Immune responsiveness in renal transplant recipients: mycophenolic acid severely depresses humoral immunity in vivo. Kidney Int 2002;62:319-328.   DOI
34 Takatsuki M, Uemoto S, Inomata Y, et al. Analysis of alloreactivity and intragraft cytokine profiles in living donor liver transplant recipients with graft acceptance. Transpl Immunol 2001;8:279-286.   DOI
35 Weimer R, Melk A, Daniel V, Friemann S, Padberg W, Opelz G. Switch from cyclosporine A to tacrolimus in renal transplant recipients: impact on Th1, Th2, and monokine responses. Hum Immunol 2000;61:884-897.   DOI
36 Syrjala SO, Keranen MA, Tuuminen R, et al. Increased Th17 rather than Th1 alloimmune response is associated with cardiac allograft vasculopathy after hypothermic preservation in the rat. J Heart Lung Transplant 2010;29:1047-1057.   DOI
37 Chung BH, Kim KW, Kim BM, et al. Dysregulation of Th17 cells during the early post-transplant period in patients under calcineurin inhibitor based immunosuppression. PLoS One 2012;7:e42011.   DOI
38 Chung BH, Oh HJ, Piao SG, et al. Clinical significance of the ratio between FOXP3 positive regulatory T cell and interleukin-17 secreting cell in renal allograft biopsies with acute T-cell-mediated rejection. Immunology 2012;136:344-351.   DOI
39 Singh SP, Zhang HH, Foley JF, Hedrick MN, Farber JM. Human T cells that are able to produce IL-17 express the chemokine receptor CCR6. J Immunol 2008;180:214-221.   DOI
40 Wilson NJ, Boniface K, Chan JR, et al. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol 2007;8:950-957.   DOI
41 Merville P, Pouteil-Noble C, Wijdenes J, Potaux L, Touraine JL, Banchereau J. Cells infiltrating rejected human kidney allografts secrete IFN-gamma, IL-6, and IL-10, and are modulated by IL-2 and IL-4. Transplant Proc 1993;25(1 Pt 1):111-113.
42 Merville P, Pouteil-Noble C, Wijdenes J, Potaux L, Touraine JL, Banchereau J. Detection of single cells secreting IFN-gamma, IL-6, and IL-10 in irreversibly rejected human kidney allografts, and their modulation by IL-2 and IL-4. Transplantation 1993;55:639-646.   DOI
43 Pavlakis M, Strehlau J, Lipman M, Shapiro M, Maslinski W, Strom TB. Intragraft IL-15 transcripts are increased in human renal allograft rejection. Transplantation 1996;62:543-545.   DOI
44 Solez K, Colvin RB, Racusen LC, et al. Banff 07 classification of renal allograft pathology: updates and future directions. Am J Transplant 2008;8:753-760.   DOI
45 Delgoffe GM, Pollizzi KN, Waickman AT, et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol 2011;12:295-303.
46 Zeiser R, Leveson-Gower DB, Zambricki EA, et al. Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells. Blood 2008;111:453-462.   DOI
47 Flechner SM, Kurian SM, Solez K, et al. De novo kidney transplantation without use of calcineurin inhibitors preserves renal structure and function at two years. Am J Transplant 2004;4:1776-1785.   DOI
48 Larson TS, Dean PG, Stegall MD, et al. Complete avoidance of calcineurin inhibitors in renal transplantation: a randomized trial comparing sirolimus and tacrolimus. Am J Transplant 2006;6:514-522.   DOI
49 Hackstein H. Rapamycin and dendritic cells: keep on movin'. Transplantation 2006;82:739-740.
50 Chung BH, Oh HJ, Piao SG, et al. Higher infiltration by Th17 cells compared with regulatory T cells is associated with severe acute T-cell-mediated graft rejection. Exp Mol Med 2011;43:630-637.   DOI
51 Van Voorhis M, Fechner JH, Zhang X, Mezrich JD. The aryl hydrocarbon receptor: a novel target for immunomodulation in organ transplantation. Transplantation 2013;95:983-990.   DOI
52 Stegall MD, Chedid MF, Cornell LD. The role of complement in antibody-mediated rejection in kidney transplantation. Nat Rev Nephrol 2012;8:670-678.   DOI
53 Mauiyyedi S, Pelle PD, Saidman S, et al. Chronic humoral rejection: identification of antibody-mediated chronic renal allograft rejection by C4d deposits in peritubular capillaries. J Am Soc Nephrol 2001;12:574-582.
54 Delgoffe GM, Kole TP, Zheng Y, et al. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 2009;30:832-844.   DOI
55 Chi H. Regulation and function of mTOR signalling in T cell fate decisions. Nat Rev Immunol 20120;12:325-338.   DOI
56 Gore-Hyer E, Shegogue D, Markiewicz M, et al. TGF-beta and CTGF have overlapping and distinct fibrogenic effects on human renal cells. Am J Physiol Renal Physiol 2002;283:F707-F716.   DOI
57 LeBleu VS, Taduri G, O'Connell J, et al. Origin and function of myofibroblasts in kidney fibrosis. Nat Med 2013;19:1047-1053.   DOI
58 Chung BH, Kim KW, Kim BM, Doh KC, Cho ML, Yang CW. Increase of Th17 cell phenotype in kidney transplant recipients with chronic allograft dysfunction. PLoS One 2015;10:e0145258.   DOI
59 Chung BH, Kim KW, Sun IO, et al. Increased interleukin-17 producing effector memory T cells in the end-stage renal disease patients. Immunol Lett 2012;141:181-189.   DOI
60 Healy DG, Watson RW, O'Keane C, et al. Neutrophil transendothelial migration potential predicts rejection severity in human cardiac transplantation. Eur J Cardiothorac Surg 2006;29:760-766.   DOI
61 Vanaudenaerde BM, Dupont LJ, Wuyts WA, et al. The role of interleukin-17 during acute rejection after lung transplantation. Eur Respir J 2006;27:779-787.   DOI