1
|
Sanders JM, Jeyamogan S, Mathew JM, Leventhal JR. Foxp3+ regulatory T cell therapy for tolerance in autoimmunity and solid organ transplantation. Front Immunol 2022; 13:1055466. [PMID: 36466912 PMCID: PMC9714335 DOI: 10.3389/fimmu.2022.1055466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/02/2022] [Indexed: 08/03/2023] Open
Abstract
Regulatory T cells (Tregs) are critical for tolerance in humans. The exact mechanisms by which the loss of peripheral tolerance leads to the development of autoimmunity and the specific role Tregs play in allograft tolerance are not fully understood; however, this population of T cells presents a unique opportunity in the development of targeted therapeutics. In this review, we discuss the potential roles of Foxp3+ Tregs in the development of tolerance in transplantation and autoimmunity, and the available data regarding their use as a treatment modality.
Collapse
Affiliation(s)
- Jes M. Sanders
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Shareni Jeyamogan
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - James M. Mathew
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Joseph R. Leventhal
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
2
|
Rivillas-Reyes JF, Hernández-Durán EF, Morales-Manrique JL, Rivillas MP, Lozano-Marquez E, Lozano-Marquez H. Inducción de tolerancia por vía oral en trasplante de órganos y tejidos. Revisión de la Literatura. REVISTA DE LA FACULTAD DE MEDICINA 2020. [DOI: 10.15446/revfacmed.v68n1.72108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introducción. La tolerancia oral es la supresión de la respuesta inmune a antígenos administrados con anterioridad por vía oral; su inducción tiene el propósito de evitar el uso de fármacos inmunosupresores, los cuales, dado que son poco específicos a antígenos, vuelven al huésped más susceptible de contraer infecciones y desarrollar neoplasias.Objetivos. Realizar una revisión de la literatura sobre los referentes teóricos más relevantes de la inducción de a tolerancia oral en lo que respecta al trasplante de órganos y tejidos para demostrar que el uso de esta alternativa terapéutica es viable en pacientes trasplantados.Materiales y métodos. Se realizó una revisión de la literatura en PubMed, MEDLINE, LILACS y Embase mediante la siguiente estrategia de búsqueda: periodo de publicación: sin límites; idiomas: Inglés y Español; tipo de artículos: estudios caso-control, revisiones sistemáticas y de la literatura; términos de búsqueda: “T-Lymphocytes, Regulatory”, “Autoimmunity”, Immunosuppression”, “Immune system” and “Immune Tolerance”, y sus equivalentes en español.Resultados. La búsqueda inicial arrojó 719 registros, sin embargo solo 99 abordaban la inducción de la tolerancia oral. Una vez los registros duplicados y los artículos sin acceso a texto completo fueron removidos, se incluyeron 72 estudios en la revisión.Conclusiones. La administración oral de antígenos es una opción efectiva para inducir tolerancia inmunológica en pacientes trasplantados (modelos murinos), pues elimina los efectos adversos que conlleva la terapia inmunosupresora actualmente utilizada.
Collapse
|
3
|
Abstract
Long-term allograft survival is a major challenge facing solid organ transplantation. Recent studies have shown a negative correlation between infiltration of memory T cells and allograft survival. Furthermore, blockade of leukocyte activation increases acceptance of transplanted organs, including heart, liver, and kidney. Lung allografts are associated with high rates of rejection, and therapies that increase acceptance of other transplanted organs have not translated into the lung. In this issue of the JCI, Krupnick and colleagues demonstrate in a murine model that lung allograft acceptance requires infiltration of a specific T cell population into the graft. This study highlights the unique immunobiology of the lung and the complexity of lung transplant tolerance.
Collapse
|
4
|
Ge F, Gong W. Body weight difference between donor and recipient is an important affector of early graft function after renal transplantation. Transplant Proc 2013; 45:2171-5. [PMID: 23731895 DOI: 10.1016/j.transproceed.2012.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 12/03/2012] [Indexed: 10/26/2022]
Abstract
BACKGROUND Poor early graft function (EGF), a frequent complication of kidney transplantation, can be caused by many risk factors, including donor kidney and body weights. METHODS We studied the relationship to early graft function in a rat kidney transplantation model among 3 indices: ratio of graft to body weight; ratio of native kidney to body weight, and weight difference/body weight of the recipient. We categorized 2 groups based on contralateral nephrectomy at day 1 (G1) or day 3 (G2) after transplantation. EGF was evaluated by measuring serum creatinine levels at day 1 after bilateral nephrectomy. RESULTS The 3 indices, ie, weight difference/body weight of recipient (G1 and G2: P < .0001), ratio of native kidney to body weight (G1: P < .0001; G2: P = .0013), and ratio of graft to body weight (G1: P = .0064; G2: P = .014) strongly correlated with EGF regardless of the time of contralateral nephrectomy. CONCLUSIONS The index of weight difference/body weight of recipient sensitively and predominantly influenced EGF, which probably reflects the systemic metabolic profile.
Collapse
Affiliation(s)
- F Ge
- Department of General Surgery, Transplant Laboratory, 2(nd) Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, China
| | | |
Collapse
|
5
|
Vascularized composite allograft rejection is delayed by intrajejunal treatment with donor splenocytes without concomitant immunosuppressants. Clin Dev Immunol 2012; 2012:704063. [PMID: 23227087 PMCID: PMC3514848 DOI: 10.1155/2012/704063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 10/12/2012] [Indexed: 11/22/2022]
Abstract
Background. Mucosal or oral tolerance, an established method for inducing low-risk antigen-specific hyporesponsiveness, has not been investigated in vascularized composite allograft (VCA) research. We studied its effects on recipient immune responses and VCA rejection. Methods. Lewis rats (n = 12; TREATED) received seven daily intrajejunal treatments of 5 × 107 splenocytes from semiallogeneic Lewis-Brown-Norway rats (LBN) or vehicle (n = 11; SHAM). Recipients' immune responses were assessed by mixed lymphocyte reaction (MLR) against donor antigen and controls. Other Lewis (n = 8; TREATED/VCA) received LBN hindlimb VCA and daily intrajejunal treatments of 5 × 107 LBN splenocytes, or LBN VCA without treatment (n = 5; SHAM/VCA), until VCAs rejected. Recipients' immune responses were characterised and VCAs biopsied for histopathology. Immunosuppressants were not used.
Results. LBN-specific hyporesponsiveness was induced only in treated Lewis recipients. Treatment significantly reduced MLR alloreactivity, significantly reduced VCA rejection on histopathology, and significantly delayed clinical VCA rejection (P < 0.0005; TREATED/VCA mean 9.6 versus 6.0 days for SHAM/VCA). Treatment significantly increased immunosuppressive IL-10/IL-4/TGF-β production and significantly decreased proinflammatory IFN-γ/TNF-α. Conclusion. Jejunal exposure to antigen conferred donor specific hyporesponsiveness that delayed VCA rejection. This method may offer a low-risk adjunctive treatment option to help protect VCAs from rejection.
Collapse
|
6
|
Ettinger M, Gratz IK, Gruber C, Hauser-Kronberger C, Johnson TS, Mahnke K, Thalhamer J, Hintner H, Peckl-Schmid D, Bauer JW. Targeting of the hNC16A collagen domain to dendritic cells induces tolerance to human type XVII collagen. Exp Dermatol 2012; 21:395-8. [PMID: 22509840 DOI: 10.1111/j.1600-0625.2012.01474.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Antibodies, specific to murine DEC205, can be used to target antigens to dendritic cells. The immunodominant domain of human type XVII collagen, hNC16A, was fused to this antibody (DEC-hNC16A) and was administered as expression plasmid by gene gun transfection with the aim of inducing tolerance to human type XVII collagen in a skin transplantation model. Mice transfected with DEC-hNC16A were challenged with skin grafts from transgenic mice engineered to express human type XVII collagen. Graft survival was either prolonged or grafts were accepted infinitely (33% and 16%, respectively) upon treatment with DEC-hNC16A while 100% of grafts were rejected in untreated controls. Graft acceptance was associated with the absence of a CD4+ infiltrate and a dense CD8+ T-cell infiltrate and was not strictly dependent on antibody production. Our results show that DEC-hNC16A targets dendritic cells in vivo leading to prolonged survival of transgenic skin grafts. This indicates that DEC205-targeting may be used for the induction of tolerance to skin antigens, which would increase the chances of successful skin gene therapy of epidermolysis bullosa patients.
Collapse
|
7
|
Rogers NM, Kireta S, Coates PTH. Curcumin induces maturation-arrested dendritic cells that expand regulatory T cells in vitro and in vivo. Clin Exp Immunol 2011; 162:460-73. [PMID: 21070208 DOI: 10.1111/j.1365-2249.2010.04232.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Dendritic cells (DC) and regulatory T cells (T(regs) ) are vital to the development of transplant tolerance. Curcumin is a novel biological agent extracted from Curcuma longa (turmeric), with anti-inflammatory and anti-oxidant activity mediated via nuclear factor (NF)-κB inhibition. We investigated the immunomodulatory effects of curcumin on human monocyte-derived and murine DC. Human monocyte-derived DC (hu-Mo-DC) were generated in the presence (CurcDC) or absence (matDC) of 25 µM curcumin, and matured using lipopolysaccharide (1 µg/ml). DC phenotype and allostimulatory capacity was assessed. CD11c(+) DC were isolated from C57BL/6 mice, pretreated with curcumin and injected into BALB/c mice, followed by evaluation of in vivo T cell populations and alloproliferative response. Curcumin induced DC differentiation towards maturation-arrest. CurcDC demonstrated minimal CD83 expression (<2%), down-regulation of CD80 and CD86 (50% and 30%, respectively) and reduction (10%) in both major histocompatibility complex (MHC) class II and CD40 expression compared to matDC. CurcDC also displayed decreased RelB and interleukin (IL)-12 mRNA and protein expression. Functionally, CurcDC allostimulatory capacity was decreased by up to 60% (P < 0·001) and intracellular interferon (IFN-γ) expression in the responding T cell population were reduced by 50% (P < 0·05). T cell hyporesponsiveness was due to generation of CD4(+) CD25(hi) CD127(lo) forkhead box P3 (FoxP3)(+) T(regs) that exerted suppressive functions on naïve syngeneic T cells, although the effect was not antigen-specific. In mice, in vivo infusion of allogeneic CurcDC promoted development of FoxP3(+) T(regs) and reduced subsequent alloproliferative capacity. Curcumin arrests maturation of DC and induces a tolerogenic phenotype that subsequently promotes functional FoxP3(+) T(regs) in vitro and in vivo.
Collapse
Affiliation(s)
- N M Rogers
- Renal Transplant Immunology Laboratory, Hanson Institute, Adelaide, SA, Australia
| | | | | |
Collapse
|
8
|
Ménoret S, Guillonneau C, Bezié S, Caron L, Anegon I, Li XL. Phenotypic and functional characterization of CD8(+) T regulatory cells. Methods Mol Biol 2011; 677:63-83. [PMID: 20941603 DOI: 10.1007/978-1-60761-869-0_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Increasing evidence shows the presence and significance of CD8+ T regulatory cells (CD8+ Tregs) in both human and rodent transplant recipients, as well as in autoimmune disease models. We, hereafter, review all available data on the phenotypic and functional characterization of CD8+ Tregs, and we also provide detailed protocols to purify them and analyze their suppressive function. Different subsets of dendritic cells (DCs) and CD4+ effector T cells may modulate the suppression mediated by CD8+ Tregs. By analyzing the proliferation of CFSE-labeled naïve CD4+CD25- T cells in coculture MLR and transwell experiments, we explored the mutual modulation of CD8+ Tregs, DC subsets, and CD4+ T effector cells. The suppressive function of CD8+ Tregs was mediated by both cell-contact-dependent and -independent mechanisms.
Collapse
|
9
|
Antigen-primed splenic CD8+ T cells impede the development of oral antigen-induced allergic diarrhea. J Allergy Clin Immunol 2009; 123:889-94. [PMID: 19201014 DOI: 10.1016/j.jaci.2008.12.1115] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 12/22/2008] [Accepted: 12/26/2008] [Indexed: 11/22/2022]
Abstract
BACKGROUND Although CD4+ T-cell populations are thought to be involved in the pathophysiology of food allergy and oral tolerance, the role of CD8+ T cells remains uncertain. OBJECTIVE We analyzed regulatory effects of adoptively transferred CD8+ T cells on the development of allergic diarrhea in antigen-sensitized mice that had a significantly reduced number of conventional TCRalphabeta+ CD8+ T cells. METHODS Ovalbumin-specific T-cell receptor transgenic mice were systemically sensitized to ovalbumin. Splenic CD8+ T cells purified from ovalbumin-sensitized or nonsensitized wild-type mice or IL-10 knockout mice were adoptively transferred to ovalbumin-sensitized ovalbumin-specific T-cell receptor transgenic mice. Allergic diarrhea induced by oral administration of ovalbumin, ovalbumin-specific immunoglobulin production, and cytokine production in intestines and mesenteric lymph nodes were assessed. RESULTS Adoptive transfer of splenic CD8+ T cells from ovalbumin-primed mice, but not from nonprimed mice, suppressed the development of allergic diarrhea, which was associated with in vivo increased IL-10 mRNA expression and in vitro antigen-specific IL-10 production by mesenteric lymph node cells. Upregulation of serum ovalbumin-specific IgE was not suppressed by ovalbumin-primed CD8+ T-cell transfer. Although administration of IL-10 before ovalbumin challenge failed to alleviate allergic diarrhea, transfer of splenic CD8+ T cells from IL-10 knockout mice showed diminished preventive effects. CONCLUSION Systemic immunization with allergen simultaneously induces regulatory CD8+ T cells that can inhibit the development of allergic diarrhea. IL-10 production by regulatory CD8+ T cells appears to be partially involved in these inhibitory mechanisms.
Collapse
|
10
|
Friedline RH, Brown DS, Nguyen H, Kornfeld H, Lee J, Zhang Y, Appleby M, Der SD, Kang J, Chambers CA. CD4+ regulatory T cells require CTLA-4 for the maintenance of systemic tolerance. ACTA ACUST UNITED AC 2009; 206:421-34. [PMID: 19188497 PMCID: PMC2646578 DOI: 10.1084/jem.20081811] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cytotoxic T lymphocyte antigen-4 (CTLA-4) plays a critical role in negatively regulating T cell responses and has also been implicated in the development and function of natural FOXP3+ regulatory T cells. CTLA-4–deficient mice develop fatal, early onset lymphoproliferative disease. However, chimeric mice containing both CTLA-4–deficient and –sufficient bone marrow (BM)–derived cells do not develop disease, indicating that CTLA-4 can act in trans to maintain T cell self-tolerance. Using genetically mixed blastocyst and BM chimaeras as well as in vivo T cell transfer systems, we demonstrate that in vivo regulation of Ctla4−/− T cells in trans by CTLA-4–sufficient T cells is a reversible process that requires the persistent presence of FOXP3+ regulatory T cells with a diverse TCR repertoire. Based on gene expression studies, the regulatory T cells do not appear to act directly on T cells, suggesting they may instead modulate the stimulatory activities of antigen-presenting cells. These results demonstrate that CTLA-4 is absolutely required for FOXP3+ regulatory T cell function in vivo.
Collapse
Affiliation(s)
- Randall H Friedline
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Diagnostic Value of Regulatory T Cells: A New Facet of a Much Studied Cell Population. Transplantation 2008; 86:1485-91. [DOI: 10.1097/tp.0b013e31818f3d2a] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
12
|
Pomié C, Ménager-Marcq I, van Meerwijk JPM. Murine CD8+ regulatory T lymphocytes: the new era. Hum Immunol 2008; 69:708-14. [PMID: 18817827 DOI: 10.1016/j.humimm.2008.08.288] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 08/15/2008] [Accepted: 08/15/2008] [Indexed: 01/10/2023]
Abstract
Regulatory T lymphocytes unequivocally play a major role in the maintenance of immunologic homeostasis. The first descriptions of regulatory T lymphocytes concerned CD8(+) cells, but this field was brought into discredit when some of its central tenets turned out to be erroneous. CD4(+) regulatory T cells took over and, with the help of newly developed molecular tools, rapidly were phenotypically and functionally characterized. We now know that these cells control a large variety of immune responses. However some observations of in vitro or in vivo immune regulation could not be explained with CD4(+) regulatory T cell activity and depended on the action of a variety of CD8(+) T cell populations. In recent years, substantial progress has been made in the phenotypic and functional characterization of CD8(+) regulatory T cells. These cells play a role in the control of intestinal immunity, immunopathology, and autoimmunity, as well as in immune privilege of the eye, in oral tolerance, and in prevention of graft-versus-host disease and graft-rejection. The suppressor effector mechanisms used by these cells are in part shared with CD4(+) regulatory T cells and in part unique to this population. We here review the current literature on naturally occurring and experimentally induced murine CD8(+) regulatory T-cell populations.
Collapse
Affiliation(s)
- Céline Pomié
- Institut National de la Santé et de la Recherche Médicale, U563, Tolerance and Autoimmunity Section, Toulouse, France
| | | | | |
Collapse
|
13
|
Zhou J, Appleton SE, Stadnyk A, Lee TDG, Nashan BAP. CD8+γδ T regulatory cells mediate kidney allograft prolongation after oral exposure to alloantigen. Transpl Int 2008; 21:679-87. [DOI: 10.1111/j.1432-2277.2008.00669.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
14
|
Jordan J, Hirsch G, Lee T. C. sinensis ablates allograft vasculopathy when used as an adjuvant therapy with cyclosporin A. Transpl Immunol 2008; 19:159-66. [DOI: 10.1016/j.trim.2008.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 05/22/2008] [Accepted: 05/23/2008] [Indexed: 01/21/2023]
|
15
|
Jovanovic V, Lair D, Soulillou JP, Brouard S. Transfer of tolerance to heart and kidney allografts in the rat model. Transpl Int 2008; 21:199-206. [DOI: 10.1111/j.1432-2277.2007.00599.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
16
|
Yi H, Zhang J, Zhao Y. The effects of antibody treatment on regulatory CD4(+)CD25(+) T cells. Transpl Immunol 2007; 19:37-44. [PMID: 18346636 DOI: 10.1016/j.trim.2007.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 12/09/2007] [Accepted: 12/11/2007] [Indexed: 12/17/2022]
Abstract
Current therapeutic antibodies, at least some, possess the capacity to induce immune tolerance in experimental models with allo-grafts or autoimmune diseases. Clinical application of humanized or chimeric antibodies to treat graft rejection or autoimmune diseases is presently underway. It is now becoming clear that immune tolerance can be acquired in some cases due to the action of regulatory T cells (Tregs), especially CD4(+)CD25(+) Tregs. In addition to their inhibition on immune response, some antibodies could promote tolerance induction in organ transplantation and autoimmune diseases essentially through the induction of Tregs. In this manuscript, we review the recent progress on the effects of therapeutic antibodies on the development, phenotypic changes and functions of CD4(+)CD25(+) Tregs.
Collapse
Affiliation(s)
- Huanfa Yi
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | |
Collapse
|
17
|
Spadafora-Ferreira M, Caldas C, Faé KC, Marrero I, Monteiro SM, Lin-Wang HT, Socorro-Silva A, Fonseca SG, Fonseca JA, Kalil J, Coelho V. CD4+CD25+Foxp3+ indirect alloreactive T cells from renal transplant patients suppress both the direct and indirect pathways of allorecognition. Scand J Immunol 2007; 66:352-61. [PMID: 17635813 DOI: 10.1111/j.1365-3083.2007.01976.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alloreactive T cells recognize donor antigens by two routes: direct and indirect pathways of allorecognition. Although the direct pathway is reported to be dominant in allograft rejection, indirect allorecognition also plays an important role. Indirect alloreactivity is also observed in renal transplant patients irrespective of rejection. Previously we showed a predominance of interleukin (IL)-10 induced by indirect allorecognition of donor human leucocyte antigen (HLA)-DR peptides, suggesting the existence of indirect alloreactive T cells displaying regulatory activity. In the present work, our objective was to characterize these regulatory T cells. We detected indirect alloproliferation of peripheral blood mononuclear cells (PBMC) from renal transplant patients, induced by donor HLA-DR peptides, dependent on IL-4 or IL-10, suggesting regulatory activity as part of the alloreactive T-cell repertoire. PBMC-derived indirect alloreactive T-cell lines were established and produced both inflammatory and regulatory cytokines. We showed that two of these T-cell lines which were able to inhibit both direct and indirect alloproliferation of another T-cell line from the same patient presented a CD4(+)CD25(+)Foxp3(+) T-cell population. These data support the idea that indirect alloreactive T cells may also have regulatory activity and may contribute to the maintenance of the human renal allograft.
Collapse
Affiliation(s)
- M Spadafora-Ferreira
- Heart Institute (InCor), Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Guillonneau C, Hill M, Hubert FX, Chiffoleau E, Hervé C, Li XL, Heslan M, Usal C, Tesson L, Ménoret S, Saoudi A, Le Mauff B, Josien R, Cuturi MC, Anegon I. CD40Ig treatment results in allograft acceptance mediated by CD8CD45RC T cells, IFN-gamma, and indoleamine 2,3-dioxygenase. J Clin Invest 2007; 117:1096-106. [PMID: 17404623 PMCID: PMC1839240 DOI: 10.1172/jci28801] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2006] [Accepted: 01/16/2007] [Indexed: 12/12/2022] Open
Abstract
Treatment with CD40Ig results in indefinite allograft survival in a complete MHC-mismatched heart allograft model in the rat. Here we show that serial second, third, and fourth adoptive transfers of total splenocytes from CD40Ig-treated recipients into secondary recipients led to indefinite donor-specific allograft acceptance. Purification of splenocyte subpopulations from CD40Ig-treated recipients demonstrated that only the adoptively transferred CD8(+)CD45RC(low) subset resulted in donor-specific long-term survival, whereas CD8(+)CD45RC(low) T cells from naive animals did not. Accepted grafts displayed increased indoleamine 2,3-dioxygenase (IDO) expression restricted in the graft to ECs. Coculture of donor ECs with CD8(+)CD45RC(low) T cells purified from CD40Ig-treated animals resulted in donor-specific IDO expression dependent on IFN-gamma. Neutralization of IFN-gamma or IDO triggered acute allograft rejection in both CD40Ig-treated and adoptively transferred recipients. This study demonstrates for what we believe to be the first time that interference in CD40-CD40 ligand (CD40-CD40L) interactions induces allospecific CD8(+) Tregs that maintain allograft survival. CD8(+)CD45RC(low) T cells act through IFN-gamma production, which in turn induces IDO expression by graft ECs. Thus, donor alloantigen-specific CD8(+) Tregs may promote local graft immune privilege through IDO expression.
Collapse
Affiliation(s)
- Carole Guillonneau
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Marcelo Hill
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - François-Xavier Hubert
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Elise Chiffoleau
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Caroline Hervé
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Xian-Liang Li
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Michèle Heslan
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Claire Usal
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Laurent Tesson
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Séverine Ménoret
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Abdelhadi Saoudi
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Brigitte Le Mauff
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Régis Josien
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Maria Cristina Cuturi
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| | - Ignacio Anegon
- INSERM U643, Centre Hopitalier Universitaire de Nantes, Institut de Transplantation et de Recherche en Transplantation (ITERT), and Université de Nantes, Faculté de Médecine, Nantes, France.
INSERM U563, Département Immunologie, Toulouse, France
| |
Collapse
|
19
|
Abstract
T regulatory (Treg) cells have been studied for more than 30 years. Recently, changing technology and attitudes have led to new interest in T cell regulation of the immune responses. The eye is an immune-privileged site with unique mechanisms for the prevention of damaging immune inflammation. The eye fashions its Treg cells in novel ways to prevent immune inflammation locally and systemically. The purpose of this mini-review is to condense and summarize reports of Treg cells dependent on the eye in the context of the Treg literature in general.
Collapse
Affiliation(s)
- Joan Stein-Streilein
- Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | | |
Collapse
|
20
|
van Wijk F, Knippels L. Initiating mechanisms of food allergy: Oral tolerance versus allergic sensitization. Biomed Pharmacother 2006; 61:8-20. [PMID: 17218079 DOI: 10.1016/j.biopha.2006.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 08/21/2006] [Indexed: 12/12/2022] Open
Abstract
Immediately after birth the mucosa of the gastrointestinal tract, which represents the greatest body surface area exposed to the outside environment, is confronted with a large variety of foreign antigens. The immune system of the intestine now has to meet the task of discriminating between pathogens and harmless antigens, such as food proteins and commensal bacteria, and to respond accordingly. This important job is fulfilled by cells of the gut-associated lymphoid tissue, the largest immunologic organ in the body. Despite the large extent of food antigen exposure, only a small percentage of individuals experience adverse immunologic reactions to food. This is due to the fact that the normal immune response to dietary proteins is associated with the induction of oral tolerance, which refers to a state of active inhibition of immune responses to an antigen by means of prior exposure to that antigen via the oral route. Abrogation of oral tolerance or failure to induce oral tolerance may result in the development of food hypersensitivity. In the present review, factors that may play a role in the outcome of oral tolerance versus sensitization to food proteins are discussed.
Collapse
Affiliation(s)
- Femke van Wijk
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Department of Pediatric Immunology, Utrecht, The Netherlands
| | | |
Collapse
|
21
|
Abstract
Regulatory T cells (Tregs) are a specialized subpopulation of T cells that act to suppress activation of other immune cells and thereby maintain immune system homeostasis, self-tolerance as well as control excessive response to foreign antigens. The mere concept of Tregs was the subject of significant controversy among immunologists for many years owing to the paucity of reliable markers for defining these cells and the ambiguity of the nature and molecular basis of suppressive phenomena. However, recent advances in the molecular characterization of this cell population have firmly established their existence and their vital role in the vertebrate immune system. Of interest, accumulating evidence from both humans and experimental animal models has implicated the involvement of Tregs in the development of graft-versus-host disease (GVHD). The demonstration that Tregs could separate GVHD from graft-versus-tumor (GVT) activity suggests that their immunosuppressive potential could be manipulated to reduce GVHD without detrimental consequence on GVT effect. Although a variety of T lymphocytes with suppressive capabilities have been reported, the two best-characterized subsets are the naturally arising, intrathymic-generated Tregs (natural Tregs) and the peripherally generated, inducible Tregs (inducible Tregs). This review summarizes our current knowledge of the generation, function and regulation of these two populations of Tregs during an immune response. Their role in the development of GVHD and their therapeutic potential for the prevention and treatment of GVHD will also be described.
Collapse
Affiliation(s)
- N T Le
- Division of Cellular Therapy/Bone Marrow Transplantation, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
22
|
Wieërs G, Gras J, Bourdeaux C, Truong DQ, Latinne D, Reding R. Monitoring tolerance after human liver transplantation. Transpl Immunol 2006; 17:83-93. [PMID: 17306738 DOI: 10.1016/j.trim.2006.09.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 09/13/2006] [Indexed: 01/25/2023]
Abstract
The validation of reliable, non-invasive immunological assays evaluating anti-donor responsiveness in allograft recipients would provide a clinically relevant tool for the early detection of ongoing rejection process as well as for the identification of operational tolerance in the long term. A sequential approach towards immunological monitoring of allografts is proposed in this review: (i) investigations exploring the initial donor-recipient alloresponses, including the analysis of the cytokine network; (ii) investigations regarding graft acceptance and operational tolerance in long-term transplant patients, consisting in the analysis of regulatory T cells and of circulating precursors of dendritic cells, in the measurement of T cell alloreactivity as well as in the study of T cell receptor repertoires. Beside the conventional in vivo and in vitro immunological techniques, the potential applications of molecular imaging in transplantation also deserve further exploration, with particular respect to allograft immune monitoring. Enforced collaboration between transplant clinicians and immunologists will be required to develop the translational research protocols required for the development of immunological monitoring, within an international multicentric network.
Collapse
Affiliation(s)
- Grégoire Wieërs
- Pediatric Liver Transplant Program, Saint-Luc University Clinics, Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
23
|
Bharat A, Fields RC, Mohanakumar T. Regulatory T cell-mediated transplantation tolerance. Immunol Res 2006; 33:195-212. [PMID: 16461998 DOI: 10.1385/ir:33:3:195] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The existence of naturally occurring regulatory T cells in normal hosts and their pivotal role in maintaining both auto- and allo-tolerance have direct implications on the therapy of autoimmune disorders and for achieving immunosuppression-free allotransplantation. Among the various forms of regulatory T cells described, CD4(+)CD25(+) T cells have emerged as one of the most potent tolerogenic subsets. In this review, we discuss the molecular basis of development and function of these regulatory T cells and their potential role in the context of chronic lung allograft rejection.
Collapse
Affiliation(s)
- Ankit Bharat
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
24
|
Emmer PM, van der Vlag J, Adema GJ, Hilbrands LB. Dendritic Cells Activated by Lipopolysaccharide after Dexamethasone Treatment Induce Donor-Specific Allograft Hyporesponsiveness. Transplantation 2006; 81:1451-9. [PMID: 16732184 DOI: 10.1097/01.tp.0000208801.51222.bd] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Immature dendritic cells (imDC) can prolong allograft survival in murine transplantation models. Recent data indicate that semi-mature or alternatively activated DC (aaDC) may be even more tolerogenic. METHODS We compared the phenotype and regulatory capacity of: a) imDC, cultured in the presence of dexamethasone (DEX), b) mature DC (matDC), activated with LPS, and c) aaDC, activated with LPS after pretreatment with DEX. RESULTS As compared to imDC, aaDCs displayed a slight upregulation of CD40 while expression levels of MHC-II and CD86 remained low. The production of proinflammatory cytokines, in particular IL-12, by aaDC was much lower than by matDC while both produced similar amounts of the regulatory cytokine IL-10 leading to an increased IL-10/IL-12 ratio for aaDC. After infusion of donor type aaDCs, responder cells isolated from the recipient mice showed donor-specific hyporesponsiveness to restimulation by matDC. Infusion of matDC was immunogenic, while imDC induced partial hyporesponsiveness. Importantly, pretreatment with donor type aaDC (but not imDC) resulted in prolonged survival of a completely MHC-mismatched heart allograft. CONCLUSIONS Alternatively activated DC are more efficacious than the classical imDC in the regulation of the alloimmune response, which may be related to a distinct cytokine profile characterized by an increased IL-10/IL12 ratio.
Collapse
Affiliation(s)
- Peter M Emmer
- Department of Nephrology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, the Netherlands
| | | | | | | |
Collapse
|
25
|
Reding R, Gras J, Truong DQ, Wieërs G, Latinne D. The immunological monitoring of alloreactive responses in liver transplant recipients: a review. Liver Transpl 2006; 12:373-83. [PMID: 16498661 DOI: 10.1002/lt.20704] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The aim of this work is to review the current knowledge in the field of immunological monitoring of allogenic responsiveness in clinical liver transplantation. When compared to other solid-organ transplants, liver allografts are considered as immunologically privileged, and, accordingly, constitute a favorable setting to develop experimental as well as clinical strategies for minimization of immunosuppression and even induction of operational tolerance. The validation of simple, reliable, noninvasive assays exploring antidonor alloreactivity will constitute a crucial step toward implementing such approaches in the clinic. In contrast to research in rodents claiming the development of donor-specific tolerance in case of graft survivals of over 100 days without immunosuppression, it is impractical to confirm tolerance induction in this way in humans. Promising candidate assays include the detection of post-transplant immune deviation, of circulating precursors of dendritic cells subtypes, and of regulatory T cells. A conceptual framework for the development of tolerance assays in clinical liver transplantation is also proposed.
Collapse
Affiliation(s)
- Raymond Reding
- Pediatric Liver Transplant Program, Saint-Luc University Clinics, Université Catholique de Louvain, Brussels, Belgium.
| | | | | | | | | |
Collapse
|
26
|
Abstract
Multiple mechanisms of tolerance are induced by oral antigen. Low doses favor active suppression, whereas higher doses favor clonal anergy/deletion. Oral antigen induces T-helper 2 [interleukin (IL)-4/IL-10] and Th3 [transforming growth factor (TGF)-beta] T cells plus CD4+CD25+ regulatory cells and latency-associated peptide+ T cells. Induction of oral tolerance is enhanced by IL-4, IL-10, anti-IL-12, TGF-beta, cholera toxin B subunit, Flt-3 ligand, and anti-CD40 ligand. Oral (and nasal) antigen administration suppresses animal models of autoimmune diseases including experimental autoimmune encephalitis, uveitis, thyroiditis, myasthenia, arthritis, and diabetes in the non-obese diabetic (NOD) mouse, plus non-autoimmune diseases such as asthma, atherosclerosis, graft rejection, allergy, colitis, stroke, and models of Alzheimer's disease. Oral tolerance has been tested in human autoimmune diseases including multiple sclerosis (MS), arthritis, uveitis, and diabetes and in allergy, contact sensitivity to dinitrochlorobenzene (DNCB), and nickel allergy. Although positive results have been observed in phase II trials, no effect was observed in phase III trials of CII in rheumatoid arthritis or oral myelin and glatiramer acetate (GA) in MS. Large placebo effects were observed, and new trials of oral GA are underway. Oral insulin has recently been shown to delay onset of diabetes in at-risk populations, and confirmatory trials of oral insulin are being planned. Mucosal tolerance is an attractive approach for treatment of autoimmune and inflammatory diseases because of lack of toxicity, ease of administration over time, and antigen-specific mechanisms of action. The successful application of oral tolerance for the treatment of human diseases will depend on dose, developing immune markers to assess immunologic effects, route (nasal versus oral), formulation, mucosal adjuvants, combination therapy, and early therapy.
Collapse
Affiliation(s)
- Howard L. Weiner
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andre Pires da Cunha
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Henry Wu
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Beilke JN, Kuhl NR, Van Kaer L, Gill RG. NK cells promote islet allograft tolerance via a perforin-dependent mechanism. Nat Med 2005; 11:1059-65. [PMID: 16155578 DOI: 10.1038/nm1296] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Accepted: 08/08/2005] [Indexed: 02/02/2023]
Abstract
Although major histocompatibility complex (MHC) class II-restricted CD4 T cells are well appreciated for their contribution to peripheral tolerance to tissue allografts, little is known regarding MHC class I-dependent reactivity in this process. Here we show a crucial role for host MHC class I-dependent NK cell reactivity for allograft tolerance in mice induced through either costimulation blockade using CD154-specific antibody therapy or by targeting LFA-1 (also known as CD11a). Tolerance induction absolutely required host expression of MHC class I, but was independent of CD8 T cell-dependent immunity. Rather, tolerance required innate immunity involving NK1.1(+) cells, but was independent of CD1d-restricted NKT cells. Therefore, NK cells seem to be generally required for induction of tolerance to islet allografts. Additional studies indicate that CD154-specific antibody-induced allograft tolerance is perforin dependent. Notably, NK cells that are perforin competent are sufficient to restore allograft tolerance in perforin-deficient recipients. Together, these results show an obligatory role for NK cells, through perforin, for induction of tolerance to islet allografts.
Collapse
Affiliation(s)
- Joshua N Beilke
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, 1775 Ursula Street, Box B-140, Aurora, Colorado 80010, USA
| | | | | | | |
Collapse
|
28
|
Chen W, Zhou D, Torrealba JR, Waddell TK, Grant D, Zhang L. Donor Lymphocyte Infusion Induces Long-Term Donor-Specific Cardiac Xenograft Survival through Activation of Recipient Double-Negative Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:3409-16. [PMID: 16116235 DOI: 10.4049/jimmunol.175.5.3409] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Previous studies have shown that pretransplant donor lymphocyte infusion (DLI) can enhance xenograft survival. However, the mechanism by which DLI induces xenograft survival remains obscure. Using T cell subset-deficient mice as recipients we show that CD4+, but not CD8+, T cells are necessary to mediate the rejection of concordant cardiac xenografts. Adoptive transfer of naive CD4+ T cells induces rejection of accepted cardiac xenografts in CD4-/- mice. This rejection can be prevented by pretransplant DLI in the absence of any other treatment. Furthermore, we demonstrate that DLI activates alphabeta-TCR+CD3+CD4-CD8- double-negative (DN) regulatory T (Treg) cells in xenograft recipients, and that DLI-activated DN Treg cells can inhibit the proliferation of donor-specific xenoreactive CD4+ T cells in vitro. More importantly, adoptive transfer of DLI-activated DN Treg cells from xenograft recipients can suppress the proliferation of xenoreactive CD4+ T cells and their ability to produce IL-2 and IFN-gamma in vivo. Adoptive transfer of DLI-activated DN Treg cells also prevents CD4+ T cell-mediated cardiac xenograft rejection in an Ag-specific fashion. These data provide direct evidence that DLI can activate recipient DN Treg cells, which can induce donor-specific long-term cardiac xenograft survival by suppressing the proliferation and function of donor-specific CD4+ T cells in vivo.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Laboratory Medicine and Pathobiology, Multi Organ Transplantation Program, Toronto General Research Institute, University Health Network, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Tolerance has been defined as a lack of response to self but a more appropriate definition of tolerance is "any mechanism by which a potentially injurious immune response is prevented, suppressed, or shifted to a non-injurious class of immune response." Thus, tolerance is related to productive self-recognition, rather than blindness of the immune system to its auto-components. Oral tolerance, in this sense, is of unique immunologic importance, as it is a continuous natural immunologic event driven by exogenous antigen. Because of their privileged access to the internal milieu, antigens that are continuously in contact with the mucosa are a frontier between foreign and self-components. Thus, oral tolerance is an immunological mechanism that evolved to treat external agents that gain access to the body via a natural route as internal components that then become part of self. Given this, it would seem logical that autoimmune diseases caused by an inappropriate response to self-antigens might ultimately be treated by presenting such autoantigens to the mucosal surface where they can be dealt with in a non-injurious (noninflammatory) immunologic environment. Furthermore, mucosal tolerance as a treatment for autoimmune diseases is an attractive concept, as antigen-specific therapy is the most physiologic means to manipulate immune responses, and mucosal antigen is nontoxic and can be given on a chronic basis. The efficacy of mucosal tolerance has been clearly demonstrated in several animal models.
Collapse
Affiliation(s)
- Howard L Weiner
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115-5817, USA.
| |
Collapse
|
30
|
Dai Z, Nasr IW, Reel M, Deng S, Diggs L, Larsen CP, Rothstein DM, Lakkis FG. Impaired recall of CD8 memory T cells in immunologically privileged tissue. THE JOURNAL OF IMMUNOLOGY 2005; 174:1165-70. [PMID: 15661869 DOI: 10.4049/jimmunol.174.3.1165] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Foreign Ags that enter immunologically privileged sites such as the eye, brain, and testis persist for an extended period of time, whereas the same Ags are rapidly eliminated at conventional sites. Immune privilege, therefore, provides unwanted refuge for pathogens and tumor cells but is beneficial for the survival of allogeneic grafts. In this study, we asked whether memory T cells can eliminate foreign Ags deposited at an immunologically privileged site by studying CD8 memory T cell-mediated rejection of pancreatic islet allografts placed either in the testis (a privileged organ) or under the kidney capsule (a nonprivileged site) of diabetic mice. We found that CD8 memory T cells reject intratesticular grafts at a significantly slower rate than the rejection of intrarenal grafts. Delayed graft rejection in the testis was not due to reduced homing or proliferation of memory T cells but due to their increased apoptosis at that site. Apoptosis was mediated by the combined actions of two TNFR family members that are up-regulated on activated memory T cells, Fas, and CD30. Therefore, memory T cells survey immunologically privileged tissues but are subject to the immunosuppressive mechanisms present at these sites.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/transplantation
- Cell Movement/genetics
- Cell Movement/immunology
- Epitopes, T-Lymphocyte/immunology
- Graft Rejection/genetics
- Graft Rejection/immunology
- Graft Rejection/pathology
- Immunologic Memory
- Islets of Langerhans Transplantation/immunology
- Islets of Langerhans Transplantation/methods
- Islets of Langerhans Transplantation/pathology
- Ki-1 Antigen/physiology
- Kidney/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred MRL lpr
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Testis/cytology
- Testis/immunology
- Testis/pathology
- Transplantation, Heterotopic/immunology
- Transplantation, Heterotopic/methods
- Transplantation, Heterotopic/pathology
- fas Receptor/physiology
Collapse
Affiliation(s)
- Zhenhua Dai
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Delis S, Ciancio G, Burke GW, Garcia-Morales R, Miller J. Donor bone marrow transplantation: chimerism and tolerance. Transpl Immunol 2005; 13:105-15. [PMID: 15380541 DOI: 10.1016/j.trim.2004.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2004] [Indexed: 11/23/2022]
Abstract
Infusion of donor bone marrow (DBM)-derived cells continue to be tested in clinical protocols intended to induce specific immunologic tolerance. Central clonal deletion of donor-specific alloreactive cells associated with mixed chimerism reliably produced long-term graft tolerance. In this setting, depletion of recipient T cells by antilymphocyte antibodies and subsequent repopulation by donor hematopoietic cells after donor bone marrow infusion (DBMI) are prerequisites for tolerance induction. Major advances have been made in animal models and in pilot clinical trials and the key questions with the future perspectives are presented in this article.
Collapse
Affiliation(s)
- Spiros Delis
- Department of Surgery, Division of Kidney, Kidney/Pancreas Transplant, University of Miami School of Medicine, Miami, FL, USA
| | | | | | | | | |
Collapse
|
32
|
|
33
|
|
34
|
Abstract
The induction and maintenance of immune tolerance to transplanted tissues constitute an active process involving multiple mechanisms that work cooperatively to prevent graft rejection. These mechanisms are similar to inherent tolerance toward self antigens and have a requirement for active immunoregulation, largely T cell mediated, that promotes specific unresponsiveness to donor alloantigens. This review outlines our current understanding of the Treg subsets that contribute to allotolerance and the mechanisms by which these cells exert their effects as well as their potential for therapy.
Collapse
Affiliation(s)
- Patrick T Walsh
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6144, USA
| | | | | |
Collapse
|
35
|
Morris ES, MacDonald KPA, Rowe V, Johnson DH, Banovic T, Clouston AD, Hill GR. Donor treatment with pegylated G-CSF augments the generation of IL-10-producing regulatory T cells and promotes transplantation tolerance. Blood 2004; 103:3573-81. [PMID: 14726406 DOI: 10.1182/blood-2003-08-2864] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractWe investigated whether the protection from graft-versus-host disease (GVHD) afforded by donor treatment with granulocyte colony-stimulating factor (G-CSF) could be enhanced by dose escalation. Donor treatment with human G-CSF prevented GVHD in the B6 → B6D2F1 murine model in a dose-dependent fashion, and murine G-CSF provided equivalent protection from GVHD at 10-fold lower doses. Donor pretreatment with a single dose of pegylated G-CSF (peg-G-CSF) prevented GVHD to a significantly greater extent than standard G-CSF (survival, 75% versus 11%, P < .001). Donor T cells from peg-G-CSF-treated donors failed to proliferate to alloantigen and inhibited the responses of control T cells in an interleukin 10 (IL-10)-dependent fashion in vitro. T cells from peg-G-CSF-treated IL-10-/- donors induced lethal GVHD; T cells from peg-G-CSF-treated wild-type (wt) donors promoted long-term survival. Whereas T cells from peg-G-CSF wt donors were able to regulate GVHD induced by T cells from control-treated donors, T cells from G-CSF-treated wt donors and peg-G-CSF-treated IL-10-/- donors did not prevent mortality. Thus, peg-G-CSF is markedly superior to standard G-CSF for the prevention of GVHD following allogeneic stem cell transplantation (SCT), due to the generation of IL-10-producing regulatory T cells. These data support prospective clinical trials of peg-G-CSF-mobilized allogeneic blood SCT. (Blood. 2004;103:3573-3581)
Collapse
Affiliation(s)
- Edward S Morris
- Queensland Institute of Medical Research, Herston, Australia
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Regulatory T cells participate in immunologic homeostasis by active suppression of inappropriate immune responses. Regulatory T lymphocytes expressing CD4 and CD25 antigens and naturally present in the peripheral blood were the first to be phenotypically characterized. However, their small number and antigen nonspecific suppression has prompted efforts to identify and dissect antigen-specific regulatory T cells. In this review we discuss how antigen-specific regulatory T cells can be identified, the cellular and molecular mechanisms underlying their induction and activity, and the challenges facing their potential clinical application.
Collapse
Affiliation(s)
- Stephane Vigouroux
- Center for Cell and Gene Therapy and Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, Houston, 77030, USA.
| | | | | | | |
Collapse
|
37
|
Abstract
Autoimmune conditions caused by injurious immune responses against self-antigens can be ameliorated if the inappropriate responses to self-components that cause tissue injury can be modulated by regulatory cells or shut off via the induction of anergy or via deletion of pathogenic immune responses. Antigen encounter at the gut mucosa can lead to suppression of injurious immune responses to self-antigen via these mechanisms. This type of immunological event is termed oral tolerance. In this review, we examine the mechanisms behind the induction of oral tolerance and provide findings from its use as a form of treatment for autoimmune diseases.
Collapse
Affiliation(s)
- Henry Yim Wu
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
38
|
Dai Z, Li Q, Wang Y, Gao G, Diggs LS, Tellides G, Lakkis FG. CD4+CD25+ regulatory T cells suppress allograft rejection mediated by memory CD8+ T cells via a CD30-dependent mechanism. J Clin Invest 2004; 113:310-7. [PMID: 14722622 PMCID: PMC311434 DOI: 10.1172/jci19727] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2003] [Accepted: 11/11/2003] [Indexed: 01/07/2023] Open
Abstract
CD4(+)CD25(+) regulatory T (Treg) cells suppress naive T cell responses, prevent autoimmunity, and delay allograft rejection. It is not known, however, whether Treg cells suppress allograft rejection mediated by memory T cells, as the latter mount faster and stronger immune responses than their naive counterparts. Here we show that antigen-induced, but not naive, Treg cells suppress allograft rejection mediated by memory CD8(+) T cells. Suppression was allospecific, as Treg cells induced by third-party antigens did not delay allograft rejection. In vivo and in vitro analyses revealed that the apoptosis of allospecific memory CD8(+) T cells is significantly increased in the presence of antigen-induced Treg cells, while their proliferation remains unaffected. Importantly, neither suppression of allograft rejection nor enhanced apoptosis of memory CD8(+) T cells was observed when Treg cells lacked CD30 or when CD30 ligand-CD30 interaction was blocked with anti-CD30 ligand Ab. This study therefore provides direct evidence that pathogenic memory T cells are amenable to suppression in an antigen-specific manner and identifies CD30 as a molecule that is critical for the regulation of memory T cell responses.
Collapse
Affiliation(s)
- Zhenhua Dai
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven CT 06520, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Kataoka M, Margenthaler JA, Ku G, Flye MW. Development of infectious tolerance after donor-specific transfusion and rat heart transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:204-11. [PMID: 12816999 DOI: 10.4049/jimmunol.171.1.204] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Regulatory cells developed after donor-specific transfusion (DST)-induced acceptance of a LEW heart transplanted into a DA rat. Both DST and the cardiac transplant were necessary to generate the regulatory cells. This donor-specific tolerance can then be transferred into a new DA recipient by adoptive transfer of lymphocytes from the DST-treated long term survivor (LTS) in a dose-dependent manner. The effectiveness of tolerance did not diminish over five generations of adoptive transfer, thus supporting its infectious nature. Although both spleen and lymph node cells were equally effective, graft-infiltrating lymphocytes were more potent. A high level of indirect CTL activity and MLC proliferation were observed in lymphocytes from LTS. In vivo tracking of adoptively transferred CFSE-labeled splenocytes from LTS showed equivalent FACS proliferation and a higher percentage of graft-infiltrating lymphocytes 7 days after heart transplantation, compared with adoptively transferred naive splenocytes. Adoptive transfer of CD8(+)-depleted LTS splenocytes resulted in 100% subsequent LEW allograft acceptance; whereas CD4(+) depletion decreased acceptance to 40%, and depletion of both CD4 and CD8 resulted in 0% acceptance. When positively selected CD4(+) or CD8(+) cells were adoptively transferred, 100% or 62.5% of LEW cardiac allografts survived, respectively. In conclusion, DST alone promotes a donor-specific infectious tolerance of a heart graft that can be adoptively transferred to subsequent naive allograft recipients despite the undiminished in vitro immunological response to donor Ag. Although both CD4(+) and CD8(+) populations are responsible for the regulatory mechanism in DST-induced tolerance, the CD4(+) population appears to dominate.
Collapse
Affiliation(s)
- Masaaki Kataoka
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
40
|
Abstract
Self-tolerance is maintained by several mechanisms including deletion (via apoptosis) and regulation. Acquired tolerance to allogeneic tissues and organs exploits similar strategies. One key difference between alloantigens and peptide antigens is the enormous number of T cells that are alloreactive. Accumulating evidence suggests that in the face of this large mass of potentially graft-destructive T cells, tolerance requires an initial wave of deletion. This creates a more level playing field in which a smaller number of regulatory T cells can then act to maintain an established tolerant state. Deletion of alloreactive T cells by apoptosis actively promotes immunoregulation as well, by interfering with proinflammatory maturation of antigen presenting cells. This article reviews the immune response to alloantigens, the development and use of both necrotic and apoptotic means of cell death during the evolution of the immune response, and the likely role and mechanisms by which apoptosis promotes, and may even be required for, transplantation tolerance.
Collapse
Affiliation(s)
- Elise Chiffoleau
- Department of Medicine, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
41
|
Gutting BW, Bouzahzah F, Kong PL, Updyke LW, Amacher DE, Craft J. Oxazolone and diclofenac-induced popliteal lymph node assay reactions are attenuated in mice orally pretreated with the respective compound: potential role for the induction of regulatory mechanisms following enteric administration. Toxicol Appl Pharmacol 2003; 189:120-33. [PMID: 12781630 DOI: 10.1016/s0041-008x(03)00091-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The murine popliteal lymph node assay (PLNA) was examined as a preclinical assay with the potential to identify low-molecular-weight compounds (LMWCs) that are likely to be associated with immune-mediated drug hypersensitivity reactions (IDHRs) in humans. We hypothesized that the contact sensitizer oxazolone (OX) would cause a strong PLN reaction in naive mice and that the PLN reaction would be attenuated in mice orally pretreated with OX due to the induction of oral tolerance. In naive mice, OX induced a strong PLN reaction and caused dose-dependent increases in PLN size, weight, cellularity, percentage of CD4(+) PLN T cells, and percentage of PLN B cells, with a concomitant decrease in the percentage of CD8(+) PLN T cells. Next, the PLNA was conducted in mice gavaged three times with either OX or vehicle alone (olive oil). Mice pretreated with OX had suppressed PLN reactions following the footpad injection of OX (decrease in PLN size, weight, and cellularity), which was associated with an increase in the percentage of PLN CD8(+)T cells. In contrast, oral pretreatment with OX had no observable effect on the PLN reaction induced following footpad injection of the irrelevant hapten dinitrochlorobenzene (DNCB). Adoptive transfer studies were conducted to examine the mechanism of PLN hyporesponsiveness. It was found that either (1) unfractionated splenocytes or (2) purified CD8(+) splenocytes, but not (3) purified CD4(+) splenocytes isolated from mice gavaged with OX adoptively transferred PLN suppression to naive BALB/c mice. Because OX is not a pharmaceutical, we also examined the NSAID diclofenac (DF) (Voltaren). Like OX, DF caused dose-dependent increases in PLN size, weight, and cellularity in naive mice. Furthermore, like OX, the diclofenac-induced PLN reaction was attenuated in mice that had been orally pretreated three times with DF. However, splenocytes from mice orally treated with DF were not able to adoptively transfer PLN hyporesponsiveness. Collectively, these observations demonstrate that both OX and DF are potent immunostimulators in the PLNA. As importantly, these results demonstrate that the immunostimulating potential of OX and DF in the PLNA is significantly decreased in mice orally exposed to the respective drug, possibly due to the presence of a cellular mechanism of oral tolerance. For OX, the mechanism appears to involve, in part, CD8(+) T cells, whereas the mechanism(s) associated with PLN hyporesponsiveness using DF remain to be defined.
Collapse
Affiliation(s)
- Bradford W Gutting
- Yale University School of Medicine, Section of Rheumatology, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
The identification and characterization of regulatory T (T(Reg)) cells that can control immune responsiveness to alloantigens have opened up exciting opportunities for new therapies in transplantation. After exposure to alloantigens in vivo, alloantigen-specific immunoregulatory activity is enriched in a population of CD4+ T cells that express high levels of CD25. In vivo, common mechanisms seem to underpin the activity of CD4+CD25+ T(Reg) cells in both naive and manipulated hosts. However, the origin, allorecognition properties and molecular basis for the suppressive activity of CD4+CD25+ T(Reg) cells, as well as their relationship to other populations of regulatory cells that exist after transplantation, remain a matter of debate..
Collapse
Affiliation(s)
- Kathryn J Wood
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK.
| | | |
Collapse
|
43
|
Chen W, Ford MS, Young KJ, Cybulsky MI, Zhang L. Role of double-negative regulatory T cells in long-term cardiac xenograft survival. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:1846-53. [PMID: 12574350 DOI: 10.4049/jimmunol.170.4.1846] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A novel subset of CD3(+)CD4(-)CD8(-) (double negative; DN) regulatory T cells has recently been shown to induce donor-specific skin allograft acceptance following donor lymphocyte infusion (DLI). In this study, we investigated the effect of DLI on rat to mouse cardiac xenotransplant survival and the ability of DN T cells to regulate xenoreactive T cells. B6 mice were given either DLI from Lewis rats, a short course of depleting anti-CD4 mAb, both DLI and anti-CD4 treatment together, or left untreated. DLI alone did not prolong graft survival when compared with untreated controls. Although anti-CD4-depleting mAb alone significantly prolonged graft survival, grafts were eventually rejected by all recipients. However, the combination of DLI and anti-CD4 treatment induced permanent cardiac xenograft survival. We demonstrate that recipients given both DLI and anti-CD4 treatment had a significant increase in the total number of DN T cells in their spleens when compared with all other treatment groups. Furthermore, DN T cells harvested from the spleens of DLI plus anti-CD4-treated mice could dose-dependently inhibit the proliferation of syngeneic antidonor T cells. Suppression mediated by these DN T cells was specific for antidonor T cells as T cells stimulated by third-party Ags were not suppressed. These results demonstrate for the first time that a combination of pretransplant DLI and anti-CD4-depleting mAb can induce permanent survival of rat to mouse cardiac xenografts and that DN T regulatory cells play an important role in preventing long-term concordant xenograft rejection through the specific suppression of antidonor T cells.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens/metabolism
- Antigens, Surface
- CD3 Complex/biosynthesis
- CD4 Antigens/immunology
- CD4 Antigens/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Cell Division/genetics
- Cell Division/immunology
- Epitopes, T-Lymphocyte/immunology
- Graft Enhancement, Immunologic/methods
- Graft Survival/genetics
- Graft Survival/immunology
- Heart Transplantation/immunology
- Immune Tolerance/genetics
- Immunosuppression Therapy
- Injections, Intraperitoneal
- Interphase/immunology
- Lectins, C-Type
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Lymphocyte Transfusion
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- NK Cell Lectin-Like Receptor Subfamily B
- Proteins/metabolism
- Rats
- Rats, Inbred Lew
- Rats, Inbred WF
- Spleen/cytology
- Spleen/metabolism
- Spleen/transplantation
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Transplantation Conditioning
- Transplantation, Heterologous/immunology
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Laboratory Medicine and Pathobiology, Multi Organ Transplantation Program, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, M5G 2C4 Canada
| | | | | | | | | |
Collapse
|
44
|
Young KJ, Yang L, Phillips MJ, Zhang L. Donor-lymphocyte infusion induces transplantation tolerance by activating systemic and graft-infiltrating double-negative regulatory T cells. Blood 2002; 100:3408-14. [PMID: 12384444 DOI: 10.1182/blood-2002-01-0235] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Donor-lymphocyte infusion (DLI) before transplantation can lead to specific tolerance to allografts in mice, nonhuman primates, and humans. We and others have demonstrated a role for regulatory T cells in DLI-induced, donor-specific transplantation tolerance, but it is not known how regulatory T cells are activated and where they execute their function. In this study, we observed, in both transgenic and normal mice, that DLI before transplantation is required for activation of alphabeta-T-cell-receptor-positive, CD3(+)CD4(-)CD8(-) double-negative (DN) regulatory T cells in the periphery of recipient mice. More interestingly, DLI induced DN regulatory T cells to migrate preferentially to donor-specific allogeneic skin grafts and to form a majority of graft-infiltrating T cells in accepted skin allografts. Furthermore, both recipient-derived peripheral and graft-infiltrating DN T cells were able to suppress and kill antidonor CD8(+) T cells in an antigen-specific manner. These data indicate that DLI may induce donor-specific transplantation tolerance by activating recipient DN regulatory T cells in the periphery and by promoting migration of regulatory T cells to donor-specific allogeneic skin grafts. Our results also show that DN regulatory T cells can eliminate antidonor T cells both systemically and locally, a finding suggesting that graft-infiltrating T cells can be beneficial to graft survival.
Collapse
Affiliation(s)
- Kevin J Young
- Department of Laboratory Medicine, Multi Organ Transplantation Program, Toronto General Research Institute, University Health Network, University of Toronto, 101 College Street, Toronto, Ontario, Canada M5G 2C4
| | | | | | | |
Collapse
|
45
|
Abstract
Organ transplantation can be considered as replacement therapy for patients with end-stage organ failure. The percent of one-year allograft survival has increased due, among other factors, to a better understanding of the rejection process and new immunosuppressive drugs. Immunosuppressive therapy used in transplantation prevents activation and proliferation of alloreactive T lymphocytes, although not fully preventing chronic rejection. Recognition by recipient T cells of alloantigens expressed by donor tissues initiates immune destruction of allogeneic transplants. However, there is controversy concerning the relative contribution of CD4+ and CD8+ T cells to allograft rejection. Some animal models indicate that there is an absolute requirement for CD4+ T cells in allogeneic rejection, whereas in others CD4-depleted mice reject certain types of allografts. Moreover, there is evidence that CD8+ T cells are more resistant to immunotherapy and tolerance induction protocols. An intense focal infiltration of mainly CD8+CTLA4+ T lymphocytes during kidney rejection has been described in patients. This suggests that CD8+ T cells could escape from immunosuppression and participate in the rejection process. Our group is primarily interested in the immune mechanisms involved in allograft rejection. Thus, we believe that a better understanding of the role of CD8+ T cells in allograft rejection could indicate new targets for immunotherapy in transplantation. Therefore, the objective of the present review was to focus on the role of the CD8+ T cell population in the rejection of allogeneic tissue.
Collapse
Affiliation(s)
- V Bueno
- Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil.
| | | |
Collapse
|
46
|
Abstract
There has recently been an explosion of renewed interest in regulatory T cells, particularly those within the CD4(+)CD25(+) population. It is becoming increasingly apparent that these cells exist not only as naturally occurring cells that may contribute to the maintenance of self-tolerance, but they also have the potential to prevent rejection of allografts in experimental models. Such cells have now been identified in humans as well as in rodents.
Collapse
Affiliation(s)
- Mahzuz Karim
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, Headington, OX3 9DU, UK.
| | | | | |
Collapse
|
47
|
Niederkorn JY, Mayhew E. Phenotypic analysis of oral tolerance to alloantigens: evidence that the indirect pathway of antigen presentation is involved. Transplantation 2002; 73:1493-500. [PMID: 12023630 DOI: 10.1097/00007890-200205150-00021] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Oral administration of alloantigens induces down-regulation of Th1 immune responses and reduces the incidence of corneal graft rejection. This study examined the role of Th1 and Th2 cytokines, accessory cells, and lymphoid organs that are known to be instrumental in other forms of antigen-specific tolerance. METHODS Allogeneic dendritic cells (DC) were administered orally using a protocol that is known to reduce the incidence of corneal allograft rejection and prevent the generation of allospecific delayed-type hypersensitivity (DTH). Hosts included normal mice and gene knockout (KO) mice, including B cell-deficient (mu)MT, interleukin (IL)-4 KO, IL-10 KO, and interferon (IFN)-gamma KO mice. The requirement for either an intact spleen or thymus was also examined. Orally administered paraformaldehyde-fixed, UVB-treated, or sonicated allogeneic cells were tested to determine if dead cells were capable of inducing tolerance. RESULTS Studies on gene KO mice indicated that a Th1 cytokine (IFN-gamma) and a Th2 cytokine (IL-4) were needed for the development of oral tolerance to alloantigens. By contrast, IL-10 was not required. Although an intact spleen was necessary for the development of tolerance, removal of the thymus did not affect down-regulation of DTH. CONCLUSIONS Oral tolerance induced with allogeneic cells shares characteristics with antigen-specific unresponsiveness induced by other routes, yet there are some noteworthy differences. The capacity of killed or sonicated allogeneic cells to induce oral tolerance and enhance corneal graft survival indicates that oral tolerance to alloantigens can occur via the indirect pathway of alloantigen presentation. These results also emphasize the remarkable redundancy in the mechanisms that the immune system employs to produce antigen-specific unresponsiveness.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9057, USA.
| | | |
Collapse
|
48
|
Chiffoleau E, Bériou G, Dutartre P, Usal C, Soulillou JP, Cuturi MC. Role for thymic and splenic regulatory CD4+ T cells induced by donor dendritic cells in allograft tolerance by LF15-0195 treatment. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5058-69. [PMID: 11994458 DOI: 10.4049/jimmunol.168.10.5058] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A 20-day treatment with LF15-0195, a deoxyspergualine analogue, induced allograft tolerance in a fully MHC-mismatched heart allograft model in the rat. Long-term allografts displayed minimal cell infiltration with no signs of chronic rejection. CD4+ spleen T cells from tolerant LF15-0195-treated recipients were able to suppress in vitro proliferation of allogeneic CD4+ T cells and to transfer tolerance to second syngeneic recipients, demonstrating dominant suppression by regulatory cells. A significant increase in the percentage of CD4+CD25+ T cells was observed in the thymus and spleen from tolerant LF15-0195-treated recipient. In vitro direct stimulation with donor APCs demonstrated that CD4+ regulatory T cells proliferated weakly and expressed low levels of IFN-gamma, IL-10, and IL-2. CD4+CD25+ cell depletion increased IL-2 production by CD4+CD25- thymic cells, but not splenic cells. Moreover, tolerance was transferable with splenic and thymic CD4+CD25+ cells, but also in 50% of cases with splenic CD4+CD25- cells, demonstrating that CD25 can be a marker for regulatory cells in the thymus, but not in the periphery. In addition, we presented evidences that donor APCs were required to induce tolerance and to expand regulatory CD4+ T cells. This study demonstrates that LF15-0195 treatment induces donor APCs to expand powerful regulatory CD4+CD25+/- T cells present in both the central and peripheral compartments.
Collapse
Affiliation(s)
- Elise Chiffoleau
- Institut National de la Santé et de la Recherche Médicale Unité 437, 30 boulevard Jean Monnet, 44093 Nantes Cedex 01, France
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Corneal allografts are the oldest, most common, and most successful transplants performed on humans and animals. The cornea is endowed with a constellation of unique factors that contribute to its immune privilege and the low incidence of immune rejection. In spite of this immune privilege, 10 percent of first-time corneal grafts will undergo immune rejection. Several novel therapeutic strategies hold promise for modulating the alloimmune response by either promoting antigen-specific tolerance or redirecting the host's response from a Th1 pathway toward a Th2 pathway.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, U.T. Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9057, USA.
| |
Collapse
|