Impact of irradiation and immunosuppressive agents on immune system homeostasis in rhesus macaques
C. Meyer
Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, USA
Search for more papers by this authorJ. Walker
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorJ. Dewane
Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, USA
Search for more papers by this authorF. Engelmann
Division of Biomedical Sciences, University of California–Riverside, Riverside, CA, USA
Search for more papers by this authorW. Laub
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorS. Pillai
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorCharles. R. Thomas Jr
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorCorresponding Author
I. Messaoudi
Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, USA
Division of Biomedical Sciences, University of California–Riverside, Riverside, CA, USA
Correspondence: I. Messaoudi, University of California–Riverside, School of Medicine, 900 University Avenue, Riverside, CA 92521, USA. E-mail: [email protected]Search for more papers by this authorC. Meyer
Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, USA
Search for more papers by this authorJ. Walker
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorJ. Dewane
Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, USA
Search for more papers by this authorF. Engelmann
Division of Biomedical Sciences, University of California–Riverside, Riverside, CA, USA
Search for more papers by this authorW. Laub
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorS. Pillai
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorCharles. R. Thomas Jr
Department of Radiation Medicine, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
Search for more papers by this authorCorresponding Author
I. Messaoudi
Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, USA
Division of Biomedical Sciences, University of California–Riverside, Riverside, CA, USA
Correspondence: I. Messaoudi, University of California–Riverside, School of Medicine, 900 University Avenue, Riverside, CA 92521, USA. E-mail: [email protected]Search for more papers by this authorSummary
In this study we examined the effects of non-myeloablative total body irradiation (TBI) in combination with immunosuppressive chemotherapy on immune homeostasis in rhesus macaques. Our results show that the administration of cyclosporin A or tacrolimus without radiotherapy did not result in lymphopenia. The addition of TBI to the regimen resulted in lymphopenia as well as alterations in the memory/naive ratio following reconstitution of lymphocyte populations. Dendritic cell (DC) numbers in whole blood were largely unaffected, while the monocyte population was altered by immunosuppressive treatment. Irradiation also resulted in increased levels of circulating cytokines and chemokines that correlated with T cell proliferative bursts and with the shift towards memory T cells. We also report that anti-thymocyte globulin (ATG) treatment and CD3 immunotoxin administration resulted in a selective and rapid depletion of naive CD4 and CD8 T cells and increased frequency of memory T cells. We also examined the impact of these treatments on reactivation of latent simian varicella virus (SVV) infection as a model of varicella zoster virus (VZV) infection of humans. None of the treatments resulted in overt SVV reactivation; however, select animals had transient increases in SVV-specific T cell responses following immunosuppression, suggestive of subclinical reactivation. Overall, we provide detailed observations into immune modulation by TBI and chemotherapeutic agents in rhesus macaques, an important research model of human disease.
Supporting Information
Additional Supporting information may be found in the online version of this article at the publisher's Web site:
Filename | Description |
---|---|
cei12646-sup-0001-suppinfo01.eps318.2 KB |
Fig. S1. Simian varicella virus (SVV) infection, viral load and immunoglobulin (Ig)G titre. All animals in (a) cohort 1, (b) cohort 2 and (c) cohort 3 were infected intrabronchially with SVV at day 0. SVV DNA viral load was measured in whole blood (closed square) and bronchoalveolar lavage cells (BAL, open circle) by quantitative polymerase chain reaction (PCR) using primers and probe specific for SVV ORF21. Average copy number per 100 ng of DNA. (b) SVV-specific IgG antibody end-point titres were measured by standard enzyme-linked immunosorbent assay (ELISA) in (b) cohort 1 (open triangle), (d) cohort 2 (closed triangle) and (f) cohort 3 (closed square) rhesus macaques (RMs). Average ± standard error of the mean (s.e.m.). Solid line indicates limit of detection. Dashed line indicates onset or change in immunosuppressive treatment; total body irradiation (TBI), cyclosporin A (CsA), tacrolimus (TAC), prednisone (pred), anti-thymocyte globulin (ATG), anti-CD3 immunotoxin (CD3-IT) and Janus activated kinase inhibitor (JAK-Inh). |
cei12646-sup-0002-suppinfo02.eps337.5 KB |
Fig. 2. The effect of immunosuppression on multiple factors in the plasma. Levels of chemokines, cytokines and growth factors in plasma were determined using multiplex technology in individual rhesus macaques (RMs) from (a) cohort 1, (b) cohort 2 and (c) cohort 3. Dashed line indicates onset or change in immunosuppressive treatment; total body irradiation (TBI), cyclosporin A (CsA), tacrolimus (TAC), prednisone (pred), anti-thymocyte globulin (ATG), anti-CD3 immunotoxin (CD3-IT) and Janus activated kinase inhibitor (JAK-Inh). |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1 Dainiak N. Hematologic consequences of exposure to ionizing radiation. Exp Hematol 2002; 30: 513–28.
- 2 Barrett J, Childs R. Non-myeloablative stem cell transplants. Br J Haematol 2000; 111: 6–17.
- 3 Baron F, Maris MB, Sandmaier BM et al. Graft-versus-tumor effects after allogeneic hematopoietic cell transplantation with nonmyeloablative conditioning. J Clin Oncol 2005; 23: 1993–2003.
- 4 Hegenbart U, Niederwieser D, Sandmaier BM et al. Treatment for acute myelogenous leukemia by low-dose, total-body, irradiation-based conditioning and hematopoietic cell transplantation from related and unrelated donors. J Clin Oncol 2006; 24: 444–53.
- 5 Laport GG, Sandmaier BM, Storer BE et al. Reduced-intensity conditioning followed by allogeneic hematopoietic cell transplantation for adult patients with myelodysplastic syndrome and myeloproliferative disorders. Biol Blood Marrow Transplant 2008; 14: 246–55.
- 6 Tomblyn M, Brunstein C, Burns LJ et al. Similar and promising outcomes in lymphoma patients treated with myeloablative or nonmyeloablative conditioning and allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2008; 14: 538–45.
- 7 McSweeney PA, Niederwieser D, Shizuru JA et al. Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects. Blood 2001; 97: 3390–400.
- 8 Flanagan WM, Corthesy B, Bram RJ, Crabtree GR. Nuclear association of a T-cell transcription factor blocked by FK-506 and cyclosporin A. Nature 1991; 352: 803–7.
- 9 Liu J, Farmer JD Jr, Lane WS, Friedman J, Weissman I, Schreiber SL. Calcineurin is a common target of cyclophilin–cyclosporin A and FKBP–FK506 complexes. Cell 1991; 66: 807–15.
- 10 O'Keefe SJ, Tamura J, Kincaid RL, Tocci MJ, O'Neill EA. FK-506- and CsA-sensitive activation of the interleukin-2 promoter by calcineurin. Nature 1992; 357: 692–4.
- 11 Shevach EM. The effects of cyclosporin A on the immune system. Annu Rev Immunol 1985; 3: 397–423.
- 12 Almawi WY, Lipman ML, Stevens AC, Zanker B, Hadro ET, Strom TB. Abrogation of glucocorticoid-mediated inhibition of T cell proliferation by the synergistic action of IL-1, IL-6, and IFN-gamma. J Immunol 1991; 146: 3523–7.
- 13 Genestier L, Fournel S, Flacher M, Assossou O, Revillard JP, Bonnefoy-Berard N. Induction of Fas (Apo-1, CD95)-mediated apoptosis of activated lymphocytes by polyclonal antithymocyte globulins. Blood 1998; 91: 2360–8.
- 14 Michallet MC, Preville X, Flacher M, Fournel S, Genestier L, Revillard JP. Functional antibodies to leukocyte adhesion molecules in antithymocyte globulins. Transplantation 2003; 75: 657–62.
- 15 Zand MS, Vo T, Huggins J et al. Polyclonal rabbit antithymocyte globulin triggers B-cell and plasma cell apoptosis by multiple pathways. Transplantation 2005; 79: 1507–15.
- 16 Starzl TE, Marchioro TL, Hutchinson DE, Porter KA, Cerilli GJ, Brettschneider L. The clinical use of antilymphocyte globulin in renal homotransplantation. Transplantation 1967; 5 (Suppl.): 1100–5.
- 17 Preville X, Flacher M, LeMauff B et al. Mechanisms involved in antithymocyte globulin immunosuppressive activity in a nonhuman primate model. Transplantation 2001; 71: 460–8.
- 18 Srinivas TR, Meier-Kriesche HU. Minimizing immunosuppression, an alternative approach to reducing side effects: objectives and interim result. Clin J Am Soc Nephrol 2008; 3 (Suppl. 2): S101–16.
- 19 Kim GB, Wang Z, Liu YY et al. A fold-back single-chain diabody format enhances the bioactivity of an anti-monkey CD3 recombinant diphtheria toxin-based immunotoxin. Protein Eng Des Sel 2007; 20: 425–32.
- 20 Woo JH, Lee YJ, Neville DM, Frankel AE. Pharmacology of anti-CD3 diphtheria immunotoxin in CD3 positive T-cell lymphoma trials. Methods Mol Biol 2010; 651: 157–75.
- 21 Frankel AE, Zuckero SL, Mankin AA et al. Anti-CD3 recombinant diphtheria immunotoxin therapy of cutaneous T cell lymphoma. Curr Drug Targets 2009; 10: 104–9.
- 22 Fechner JH Jr, Dong Y, Hong X et al. Graft survival in a rhesus renal transplant model after immunotoxin-mediated T-cell depletion is enhanced by mycophenolate and steroids. Transplantation 2001; 72: 581–7.
- 23 Matar AJ, Pathiraja V, Wang Z et al. Effect of pre-existing anti-diphtheria toxin antibodies on T cell depletion levels following diphtheria toxin-based recombinant anti-monkey CD3 immunotoxin treatment. Transplant Immunol 2012; 27: 52–4.
- 24 Page EK, Page AJ, Kwun J et al. Enhanced de novo alloantibody and antibody-mediated injury in rhesus macaques. Am J Transplant 2012; 12: 2395–405.
- 25 Wamala I, Matar AJ, Farkash E, Wang Z, Huang CA, Sachs DH. Recombinant anti-monkey CD3 immunotoxin depletes peripheral lymph node T lymphocytes more effectively than rabbit anti-thymocyte globulin in naive baboons. Transplant Immunol 2013; 29: 60–3.
- 26 Ghoreschi K, Jesson MI, Li X et al. Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J Immunol 2011; 186: 4234–43.
- 27 Karaman MW, Herrgard S, Treiber DK et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol 2008; 26: 127–32.
- 28 Meyer DM, Jesson MI, Li X et al. Anti-inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP-690,550, in rat adjuvant-induced arthritis. J Inflamm (Lond) 2010; 7: 41.
- 29 Paniagua R, Si MS, Flores MG et al. Effects of JAK3 inhibition with CP-690,550 on immune cell populations and their functions in nonhuman primate recipients of kidney allografts. Transplantation 2005; 80: 1283–92.
- 30 Borie DC, Changelian PS, Larson MJ et al. Immunosuppression by the JAK3 inhibitor CP-690,550 delays rejection and significantly prolongs kidney allograft survival in nonhuman primates. Transplantation 2005; 79: 791–801.
- 31 Busque S, Leventhal J, Brennan DC et al. Calcineurin-inhibitor-free immunosuppression based on the JAK inhibitor CP-690,550: a pilot study in de novo kidney allograft recipients. Am J Transplant 2009; 9: 1936–45.
- 32 Vincenti F, Tedesco Silva H, Busque S et al. Randomized phase 2b trial of tofacitinib (CP-690,550) in de novo kidney transplant patients: efficacy, renal function and safety at 1 year. Am J Transplant 2012; 12: 2446–56.
- 33 Burmester GR, Blanco R, Charles-Schoeman C et al. Tofacitinib (CP-690,550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial. Lancet 2013; 381: 451–60.
- 34 Fleischmann R, Kremer J, Cush J et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med 2012; 367: 495–507.
- 35 Papp KA, Menter A, Strober B et al. Efficacy and safety of tofacitinib, an oral Janus kinase inhibitor, in the treatment of psoriasis: a Phase 2b randomized placebo-controlled dose-ranging study. Br J Dermatol 2012; 167: 668–77.
- 36 Sandborn WJ, Ghosh S, Panes J et al. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med 2012; 367: 616–24.
- 37 Messaoudi I, Barron A, Wellish M et al. Simian varicella virus infection of rhesus macaques recapitulates essential features of varicella zoster virus infection in humans. PLOS Pathog 2009; 5: e1000657.
- 38 Donahue JG, Choo PW, Manson JE, Platt R. The incidence of herpes zoster. Archives Inter Med 1995; 155: 1605–9.
- 39
Guinee VF,
Guido JJ,
Pfalzgraf KA et al. The incidence of herpes zoster in patients with Hodgkin's disease. An analysis of prognostic factors. Cancer 1985; 56: 642–8.
10.1002/1097-0142(19850801)56:3<642::AID-CNCR2820560334>3.0.CO;2-N CAS PubMed Web of Science® Google Scholar
- 40
Mandal BK. Herpes zoster in the immunocompromized populations. Indian J Dermatol 2006; 51: 235–43.
10.4103/0019-5154.30285 Google Scholar
- 41 Ragozzino MW, Melton LJ III, Kurland LT, Chu CP, Perry HO. Population-based study of herpes zoster and its sequelae. Medicine 1982; 61: 310–6.
- 42 Rifkind D. The activation of varicella-zoster virus infections by immunosuppressive therapy. J Lab Clin Med 1966; 68: 463–74.
- 43 Gourishankar S, McDermid JC, Jhangri GS, Preiksaitis JK. Herpes zoster infection following solid organ transplantation: incidence, risk factors and outcomes in the current immunosuppressive era. Am J Transplant 2004; 4: 108–15.
- 44 Winthrop KL, Yamanaka H, Valdez H et al. Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis. Arthritis Rheumatol 2014; 66: 2675–84.
- 45 Rijkelijkhuizen JK, Haanstra KG, Wubben J et al. T-cell-specific immunosuppression results in more than 53 days survival of porcine islets of Langerhans in the monkey. Transplantation 2003; 76: 1359–68.
- 46 Jurewicz WA. Tacrolimus versus cyclosporin immunosuppression: long-term outcome in renal transplantation. Nephrol Dial Transplant 2003; 18 (Suppl. 1): i7–11.
- 47 Vincenti F, Laskow DA, Neylan JF, Mendez R, Matas AJ. One-year follow-up of an open-label trial of FK506 for primary kidney transplantation. A report of the U.S. Multicenter FK506 Kidney Transplant Group. Transplantation 1996; 61: 1576–81.
- 48 Kindt MV, Kemp R, Allen HL, Jensen RD, Patrick DH. Tacrolimus toxicity in rhesus monkey: model for clinical side effects. Transplant Proc 1999; 31: 3393–6.
- 49 Luznik L, O'Donnell PV, Symons HJ et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant 2008; 14: 641–50.
- 50 Laukkanen MO, Kuramoto K, Calmels B et al. Low-dose total body irradiation causes clonal fluctuation of primate hematopoietic stem and progenitor cells. Blood 2005; 105: 1010–5.
- 51 Huhn RD, Tisdale JF, Agricola B, Metzger ME, Donahue RE, Dunbar CE. Retroviral marking and transplantation of rhesus hematopoietic cells by nonmyeloablative conditioning. Human Gene Ther 1999; 10: 1783–90.
- 52 Rosenzweig M, MacVittie TJ, Harper D et al. Efficient and durable gene marking of hematopoietic progenitor cells in nonhuman primates after nonablative conditioning. Blood 1999; 94: 2271–86.
- 53 Kang EM, Hanazano Y, Frare P et al. Persistent low-level engraftment of rhesus peripheral blood progenitor cells transduced with the fanconi anemia C gene after conditioning with low-dose irradiation. Mol Ther 2001; 3: 911–9.
- 54 Brennan DC, Flavin K, Lowell JA et al. A randomized, double-blinded comparison of thymoglobulin versus atgam for induction immunosuppressive therapy in adult renal transplant recipients. Transplantation 1999; 67: 1011–8.
- 55 Gaber AO, First MR, Tesi RJ et al. Results of the double-blind, randomized, multicenter, phase III clinical trial of thymoglobulin versus atgam in the treatment of acute graft rejection episodes after renal transplantation. Transplantation 1998; 66: 29–37.
- 56 Haberthur K, Engelmann F, Park B et al. CD4 T cell immunity is critical for the control of simian varicella virus infection in a nonhuman primate model of VZV infection. PLOS Pathog 2011; 7: e1002367.
- 57 Meyer C, Dewane J, Haberthur K et al. Bacterial artificial chromosome derived simian varicella virus is pathogenic in vivo. Virol J 2013; 10: 278.
- 58 Meyer C, Engelmann F, Arnold N et al. Abortive intrabronchial infection of rhesus macaques with varicella-zoster virus provides partial protection against simian varicella virus challenge. J Virol 2015; 89: 1781–93.
- 59 Meyer C, Kerns A, Haberthur K et al. Attenuation of the adaptive immune response in rhesus macaques infected with simian varicella virus lacking open reading frame 61. J Virol 2013; 87: 2151–63.
- 60 Gray WL, Zhou F, Noffke J, Tischer BK. Cloning the simian varicella virus genome in E. coli as an infectious bacterial artificial chromosome. Archives Virol 2011; 156: 739–46.
- 61 Pitcher CJ, Hagen SI, Walker JM et al. Development and homeostasis of T cell memory in rhesus macaque. J Immunol 2002 ;168: 29–43.
- 62 Aldebert D, Diallo M, Niang M et al. Differences in circulating dendritic cell subtypes in peripheral, placental and cord blood in African pregnant women. J Reprod Immunol 2007; 73: 11–9.
- 63 Diallo M, Aldebert D, Moreau JC, Ndiaye M, Jambou R. Decrease of lymphoid dendritic cells in blood from malaria-infected pregnant women. Int J Parasitol 2008; 38: 1557–65.
- 64 Dombrecht EJ, Aerts NE, Schuerwegh AJ et al. Influence of anti-tumor necrosis factor therapy (Adalimumab) on regulatory T cells and dendritic cells in rheumatoid arthritis. Clin Exp Rheumatol 2006; 24: 31–7.
- 65 Efron PA, Matsumoto T, McAuliffe PF et al. Major hepatectomy induces phenotypic changes in circulating dendritic cells and monocytes. J Clin Immunol 2009; 29: 568–81.
- 66 Hagendorens MM, Ebo DG, Schuerwegh AJ et al. Differences in circulating dendritic cell subtypes in cord blood and peripheral blood of healthy and allergic children. Clin Exp Allergy 2003; 33: 633–9.
- 67 McAuliffe PF, Efron PA, Scumpia PO et al. Varying blood monocyte and dendritic cell responses after laparoscopic versus open gastric bypass surgery. Obes Surg 2005; 15: 1424–31.
- 68 Forssmann U, Uguccioni M, Loetscher P et al. Eotaxin-2, a novel CC chemokine that is selective for the chemokine receptor CCR3, and acts like eotaxin on human eosinophil and basophil leukocytes. J Exp Med 1997; 185: 2171–6.
- 69 Griffiths-Johnson DA, Collins PD, Rossi AG, Jose PJ, Williams TJ. The chemokine, eotaxin, activates guinea-pig eosinophils in vitro and causes their accumulation into the lung in vivo. Biochem Biophys Res Commun 1993; 197: 1167–72.
- 70 Sallusto F, Mackay CR, Lanzavecchia A. Selective expression of the eotaxin receptor CCR3 by human T helper 2 cells. Science 1997; 277: 2005–7.
- 71 Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res 2009; 29: 313–26.
- 72 Schall TJ, Bacon K, Toy KJ, Goeddel DV. Selective attraction of monocytes and T lymphocytes of the memory phenotype by cytokine RANTES. Nature 1990; 347: 669–71.
- 73 Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to interferon-γ. Annu Rev Immunol 1997; 15: 749–95.
- 74 Scott P. IL-12: initiation cytokine for cell-mediated immunity. Science 1993; 260: 496–7.
- 75 Fehniger TA, Caligiuri MA. Interleukin 15: biology and relevance to human disease. Blood 2001; 97: 14–32.
- 76 MacVittie TJ, Bennett AW, Cohen MV, Farese AM, Higgins A, Hankey KG. Immune cell reconstitution after exposure to potentially lethal doses of radiation in the nonhuman primate. Health Phys 2014; 106: 84–96.
- 77
Chambers KA,
Harrington NP,
Ross WM,
Filion LG. Relative alterations in blood mononuclear cell populations reflect radiation injury in mice. Cytometry 1998 ;31: 45–52.
10.1002/(SICI)1097-0320(19980101)31:1<45::AID-CYTO6>3.0.CO;2-I PubMed Web of Science® Google Scholar
- 78 Garg S, Boerma M, Wang J et al. Influence of sublethal total-body irradiation on immune cell populations in the intestinal mucosa. Radiat Res 2010; 173: 469–78.
- 79 Harrington NP, Chambers KA, Ross WM, Filion LG. Radiation damage and immune suppression in splenic mononuclear cell populations. Clin Exp Immunol 1997; 107: 417–24.
- 80 Pecaut MJ, Nelson GA, Gridley DS. Dose and dose rate effects of whole-body gamma-irradiation: I. Lymphocytes and lymphoid organs. In Vivo 2001; 15: 195–208.
- 81 Crompton NE, Ozsahin M. A versatile and rapid assay of radiosensitivity of peripheral blood leukocytes based on DNA and surface-marker assessment of cytotoxicity. Radiat Res 1997; 147: 55–60.
- 82 Kajioka EH, Andres ML, Li J et al. Acute effects of whole-body proton irradiation on the immune system of the mouse. Radiat Res 2000; 153: 587–94.
- 83 Seki H, Iwai K, Kanegane H et al. Differential protective action of cytokines on radiation-induced apoptosis of peripheral lymphocyte subpopulations. Cell Immunol 1995; 163: 30–6.
- 84 Asquith M, Haberthur K, Brown M et al. Age-dependent changes in innate immune phenotype and function in rhesus macaques (Macaca mulatta). Pathobiol Aging Age Relat Dis 2012; 2:[S.l.], June 2012. Available at: http://www.pathobiologyofaging.net/index.php/pba/article/view/18052
- 85 Tchao NK, Turka LA. Lymphodepletion and homeostatic proliferation: implications for transplantation. Am J Transplant 2012; 12: 1079–90.
- 86 Sasaki H, Kodama K, Yamada M. A review of forty-five years study of Hiroshima and Nagasaki atomic bomb survivors. Aging. J Radiat Res 1991; 32 (Suppl.): 310–26.
- 87 Chernyshov VP, Vykhovanets EV, Slukvin, II, Antipkin YG, Vasyuk AN, Strauss KW. Analysis of blood lymphocyte subsets in children living on territory that received high amounts of fallout from Chernobyl accident. Clin Immunol Immunopathol 1997; 84: 122–8.
- 88 Castermans E, Hannon M, Dutrieux J et al. Thymic recovery after allogeneic hematopoietic cell transplantation with non-myeloablative conditioning is limited to patients younger than 60 years of age. Haematologica 2011; 96: 298–306.
- 89 Yakoub-Agha I, Saule P, Magro L et al. Immune reconstitution following myeloablative allogeneic hematopoietic stem cell transplantation: the impact of expanding CD28negative CD8+ T cells on relapse. Biol Blood Marrow Transplant 2009; 15: 496–504.
- 90 Ostrand-Rosenberg S, Sinha P, Beury DW, Clements VK. Cross-talk between myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells enhances tumor-induced immune suppression. Semin Cancer Biol 2012; 22: 275–81.
- 91 Ferrer IR, Hester J, Bushell A, Wood KJ. Induction of transplantation tolerance through regulatory cells: from mice to men. Immunol Rev 2014; 258: 102–16.
- 92 Hallahan DE, Spriggs DR, Beckett MA, Kufe DW, Weichselbaum RR. Increased tumor necrosis factor alpha mRNA after cellular exposure to ionizing radiation. Proc Natl Acad Sci USA 1989; 86: 10104–7.
- 93 Ishihara H, Tanaka I, Nemoto K et al. Immediate-early, transient induction of the interleukin-1 beta gene in mouse spleen macrophages by ionizing radiation. J Radiat Res 1995; 36: 112–24.
- 94 Nemoto K, Ishihara H, Tanaka I et al. Expression of IL-1 beta mRNA in mice after whole body X-irradiation. J Radiat Res 1995; 36: 125–33.
- 95 Stoecklein VM, Osuka A, Ishikawa S, Lederer MR, Wanke-Jellinek L, Lederer JA. Radiation exposure induces inflammasome pathway activation in immune cells. J Immunol 2015 ;194: 1178–89.
- 96 Pearl JP, Parris J, Hale DA et al. Immunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletion. Am J Transplant 2005; 5: 465–74.
- 97 Changelian PS, Flanagan ME, Ball DJ et al. Prevention of organ allograft rejection by a specific Janus kinase 3 inhibitor. Science 2003; 302: 875–8.
- 98 Conklyn M, Andresen C, Changelian P, Kudlacz E. The JAK3 inhibitor CP-690550 selectively reduces NK and CD8+ cell numbers in cynomolgus monkey blood following chronic oral dosing. J Leukocyte Biol 2004; 76: 1248–55.