51
|
Abstract
The human gut offers more than 200 m2 of mucosal surface, where direct interactions between the immune system and foreign antigens take place to eliminate pathogens or induce immune tolerance toward food antigens or normal gut flora. Therefore, mucosally administered antigens can induce tolerance under certain circumstances. In autoimmune diabetes, mucosal vaccination with autoantigens elicits some efficacy in restoring tolerance in mice, but it never succeeded in humans. Furthermore, in some instances autoimmunity can be precipitated upon oral or intranasal autoantigen administration. Therefore, it is difficult to predict the effect of mucosal vaccination on autoimmunity and much effort should be put into establishing better assays to reduce the risk for possible adverse events in humans and enable a rapid and smooth translation.
Collapse
Affiliation(s)
- Georgia Fousteri
- La Jolla Institute for Allergy and Immunology, Department of Developmental Immunology 3, 9420 Athena Circle, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
52
|
Zhou G, Levitsky HI. Natural regulatory T cells and de novo-induced regulatory T cells contribute independently to tumor-specific tolerance. THE JOURNAL OF IMMUNOLOGY 2007; 178:2155-62. [PMID: 17277120 DOI: 10.4049/jimmunol.178.4.2155] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thymus-derived, naturally occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells (nTregs) and Tregs induced in the periphery (iTregs) have both been implicated in regulating immune responses. However, the relationship between these populations in the same host, and their relative contribution to the overall Treg pool, has not been examined. Using a tumor-induced T cell tolerance model, we find that expansion of nTregs and de novo generation of iTregs both contribute to tumor-specific T cell tolerance. In this system in which the number of tumor-specific nTregs can be controlled, the efficiency of nTreg expansion significantly exceeds that of the induction of Tregs from uncommitted progenitors in the tumor-bearing host. However, pre-existing nTregs are neither required for the induction of Tregs nor measurably impact on the extent of their accumulation. Instead, induction of Ag-specific regulatory cells from naive cells is intrinsically influenced by the tumor microenvironment and the presence of tumor Ag.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD 21231, USA
| | | |
Collapse
|
53
|
Lair D, Degauque N, Miqueu P, Jovanovic V, Guillet M, Mérieau E, Moreau A, Soulillou JP, Brouard S. Functional compartmentalization following induction of long-term graft survival with pregraft donor-specific transfusion. Am J Transplant 2007; 7:538-49. [PMID: 17217443 DOI: 10.1111/j.1600-6143.2006.01660.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Long-term survival is achieved in rat recipients by pre-graft donor-specific blood transfusion. We characterized the immune compartments in long-term survivors and analyzed them for capacity to transfer tolerance and protect against chronic rejection. Splenocytes and spleen T cells from treated recipients transferred long-term graft survival to 100% of secondary recipients. In contrast, blood transferred graft survival to only 50% of recipients whereas blood T cells had no effect. An unaltered TCR repertoire, an increase in suppressive CD4+CD25+ T cells, a decrease in antidonor T-cell proliferative response and normal perforin-granzyme levels were the hallmarks of the spleen T cells. Blood T cells were characterized by a strongly altered CD8+ repertoire, normal CD4+CD25+ T cell number with unchanged antidonor T-cell proliferative response, an activated T-cell phenotype and an increase in perforin-granzyme levels. However, following the transfer of blood or spleen cells into secondary recipients, all grafts displayed chronic rejection. These findings provide evidence that distinct compartments play critical roles in DST recipients. Regulatory cells do not accumulate in blood, which appears to be a reservoir for cytotoxic T cells. Spleen T cells, which display a regulatory-like profile and transfer graft survival, are not able to prevent chronic rejection.
Collapse
Affiliation(s)
- D Lair
- INSERM U643, Nantes F-44000, France
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Fousteri G, Bresson D, von Herrath M. Rational development of antigen-specific therapies for type 1 diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 601:313-9. [PMID: 17713020 DOI: 10.1007/978-0-387-72005-0_34] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Administration of autoantigens, especially via the mucosal route, can induce tolerance under certain circumstances. In autoimmune diabetes, mucosal vaccination with autoantigens was frequently effective in restoring tolerance in mice but has not yet succeeded in humans. Furthermore, in some instances, autoimmunity can be precipitated upon autoantigen administration. We will here briefly discuss the underlying reasons and delineate which efforts should be made in the future to rationally translate antigen-specific immunotherapy, for example, by establishing better assays to reduce the risk for possible adverse events in humans.
Collapse
Affiliation(s)
- Georgia Fousteri
- La Jolla Institute for Allergy and Immunology, Department of Developmental Immunology, La Jolla, CA, USA
| | | | | |
Collapse
|
55
|
Abstract
Endowed with the ability to actively suppress an immune response, regulatory T cells (Tregs) hold the promise of halting ongoing pathogenic autoimmunity and restoring self-tolerance in patients suffering from autoimmune diseases. Through many in vitro and in vivo studies, we have learned that Tregs can function in the lymph nodes as well as in the peripheral tissues. In vivo, Tregs act through dendritic cells to limit autoreactive T-cell activation, thus preventing their differentiation and acquisition of effector functions. By limiting the supply of activated pathogenic cells, Tregs prevent or slow down the progression of autoimmune diseases. However, this protective mechanism appears insufficient in autoimmune individuals, likely because of a shortage of Tregs cells and/or the development and accumulation of Treg-resistant pathogenic T cells over the long disease course. Thus, restoration of self-tolerance in these patients will likely require purging of pathogenic T cells along with infusion of Tregs with increased ability to control ongoing tissue injury. In this review, we highlight advances in dissecting Treg function in vivo in autoimmune settings and summarize multiple studies that have overcome the limitations of the low abundance of Tregs and their hypoproliferative phenotype to develop Treg-based therapies.
Collapse
Affiliation(s)
- Qizhi Tang
- UCSF Diabetes Center, Department of Medicine and Department of Pathology, University of California, San Francisco, CA 94143-0540, USA
| | | |
Collapse
|
56
|
Abstract
Human liver allografts have a lower susceptibility to rejection than other organs. In addition, in some liver transplant recipients immunosuppressive drugs can be completely withdrawn, and these patients are considered as 'operationally' tolerant. Careful scrutiny of accumulated clinical experience indicates that elective immunosuppressive drug weaning is feasible in almost 20% of selected liver transplant recipients. This is associated with an incidence of 12% to 76% of acute cellular rejection, but these episodes are commonly mild and often resolve by return to baseline immunosuppression (IS), many times without the need to administer steroid boluses. Study of tolerance in liver transplantation (LT) has been hampered by confusion regarding the definitions of rejection and tolerance, and by the absence of prospective studies correlating results of immune monitoring assays and clinical outcome. Thus, we lack a clinically validated treatment-stopping rule capable of predicting the success of IS withdrawal and this procedure has to be performed on a 'trial and error' basis. The search for an accurate means to identify allograft tolerance among immunosuppressed recipients should become a priority in LT research. This information would provide a biological basis for guiding IS withdrawal protocols and for the implementation of tolerance-promoting strategies in LT.
Collapse
Affiliation(s)
- J Lerut
- Abdominal Transplant Unit, Université Catholique de Louvain, UCL-Brussels, Belgium.
| | | |
Collapse
|
57
|
Moore DJ, Markmann JF, Deng S. Avenues for immunomodulation and graft protection by gene therapy in transplantation. Transpl Int 2006; 19:435-45. [PMID: 16771864 DOI: 10.1111/j.1432-2277.2006.00314.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Organ transplantation represents the only definitive therapy for many causes of end-organ failure. However, the universal success of this therapy is limited by chronic allograft rejection, the side effects of chronic immunosuppressive therapy, and a severe shortage of donor organs. Presently, the success of solid-organ transplantation depends on the continuous administration of toxic and nonspecific immunosuppressive agents, therapies that present risks for opportunistic infection, malignancy, and a variety of agent-specific side effects. To promote the use of transplantation with limited risk of long-term sequelae, three dominant research challenges emerge: (i) elimination of the need for exogenous immunosuppression by immunological tolerance induction; (ii) prevention of chronic rejection/graft dysfunction; and (iii) expansion of available organs for transplantation. Gene therapy may provide significant advances and solutions in each of these areas. Rejection of the graft in the immediate post-transplant period has been attacked through the transfer of immunomodulatory molecules in addition to tolerance inducing approaches. Chronic graft rejection may be similarly addressed through permanent tolerance induction or alternatively through the introduction of molecules to resist chronic graft damage. Genetic manipulation of stem cells may ultimately produce transgenic animals to serve as tissue donors to overcome the limited donor organ supply. This review will highlight ongoing developments in the translation of gene therapy approaches to the challenges inherent in transplantation.
Collapse
Affiliation(s)
- Daniel J Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
58
|
Franzke A, Hunger JK, Dittmar KEJ, Ganser A, Buer J. Regulatory T-cells in the control of immunological diseases. Ann Hematol 2006; 85:747-58. [PMID: 16871392 DOI: 10.1007/s00277-006-0117-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Accepted: 03/30/2006] [Indexed: 12/22/2022]
Abstract
The immune system is challenged by randomly generated immune receptors that by chance can recognize self-antigens. Immunological tolerance functions as a fundamental concept in the control of a broad spectrum of immune responses not only to autoantigens but also to foreign antigens. During the past decade, CD4+ CD25+ regulatory T-cells (Tregs) have emerged as key players in the development of immunological tolerance. This review will present an update on the current knowledge about the phenotype, function, and clinical relevance of this regulatory T-cell population. The therapeutical potential of Tregs to specifically suppress immune responses in autoimmunity and transplantation and their inhibitory effects in anti-tumor immune responses will be discussed.
Collapse
Affiliation(s)
- A Franzke
- Department of Hematology, Hemostaseology and Oncology, Hannover Medical School, Hannover, Germany,
| | | | | | | | | |
Collapse
|
59
|
Negative costimulatory molecules: the proximal of regulatory T cells? Med Hypotheses 2006; 67:841-7. [PMID: 16762512 DOI: 10.1016/j.mehy.2006.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2006] [Accepted: 04/04/2006] [Indexed: 10/24/2022]
Abstract
Regulatory T cells (Tregs) are a central mechanism of immune regulation. It is well known that their regulatory effect is antigen-specific and depends on cell-cell contact. In addition, some immunological phenomenon such as linked suppression and iDC-induced tolerance are related with Tregs. But the surface markers, which reliably distinguish Treg from other T cell populations, and the regulatory mechanism still remain to be further revealed. Negative costimulatory molecule (NCM) family is one natural intrinsic mechanism that delivers the negative signal into cytoplasma to modulate immunoresponse and its expression can be induced not only on the immune cells but also on the parenchymal cells. Based on the present knowledge, we hypothesize NCMs are the specific surface markers to define Tregs. Tregs are one kind of activated T cells with high expression of NCM receptor and have the capability to induce NCM ligands expression on the membrane of APCs and the target cells of the activated cells. The NCM receptor-ligand complexes deliver negative signal into lymphocytes to regulate the immune response. This hypothesis remains to be fully elucidated.
Collapse
|
60
|
Becker C, Stoll S, Bopp T, Schmitt E, Jonuleit H. Regulatory T cells: present facts and future hopes. Med Microbiol Immunol 2006; 195:113-24. [PMID: 16715254 DOI: 10.1007/s00430-006-0017-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Indexed: 12/11/2022]
Abstract
Naturally occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells and several subsets of induced suppressor T cells are key players of the immune tolerance network and control the induction and effector phase of our immunological defense system. These T cell populations actively control the properties of other immune cells by suppressing their functional activity to prevent autoimmunity and transplant rejection but also influence the immune response to allergens as well as against tumor cells and pathogens. Even though we are far from completely understanding the molecular and cellular mechanisms that manage the different regulatory T cell populations, increasing evidence exists about their functional importance. The knowledge on their induction and activation opens the possibility for their selective manipulation in vivo as an attractive approach for an immunotherapy of unwanted immune responses. This review summarizes this knowledge and discusses the potential of regulatory T cells for novel immunointervention strategies in the future.
Collapse
Affiliation(s)
- Christian Becker
- Department of Dermatology, Johannes Gutenberg-University, Langenbeckstr. 1, 55101 Mainz, Germany
| | | | | | | | | |
Collapse
|
61
|
Veldman C, Nagel A, Hertl M. Type I Regulatory T Cells in Autoimmunity and Inflammatory Diseases. Int Arch Allergy Immunol 2006; 140:174-83. [PMID: 16601356 DOI: 10.1159/000092576] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Regulatory T cells exert a critical role in controlling autoimmunity and maintaining peripheral tolerance. The best described regulatory T cells are the naturally occurring CD4+CD25+ regulatory T cells, which have been shown to be continuously produced within the thymus. Other T-cell subsets bearing suppressive capacity have been reported, including T-helper-3 cells (Th3) and type 1 regulatory T (Tr1) cells. Tr1 cells have been shown to be induced upon antigen exposure under certain tolerogenic conditions and are characterized by the production of the immunosuppressive cytokines IL-10 and TGF-beta, while Th3 cells preferentially produce TGF-beta upon induction by intestinal tolerance. Recent progress has been made in the characterization of Tr1 cells in terms of isolation and induction, respectively. The present review provides an overview of the presence of Tr1 cells in inflammation, infection and neoplastic disorders. Moreover, the relationship between different regulatory T cell subsets and their transcriptional control is discussed. The recent development of methods allowing the ex vivo expansion of regulatory T cells may be the first step towards a cellular therapy with regulatory T cells to control T-cell-mediated pathology in inflammatory disorders.
Collapse
|
62
|
Tournoy KG, Hove C, Grooten J, Moerloose K, Brusselle GG, Joos GF. Animal models of allergen-induced tolerance in asthma: are T-regulatory-1 cells (Tr-1) the solution for T-helper-2 cells (Th-2) in asthma? Clin Exp Allergy 2006; 36:8-20. [PMID: 16393260 DOI: 10.1111/j.1365-2222.2005.02385.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Non-specific anti-inflammatory medication is actually the treatment of choice for controlling the T-helper type 2 (Th-2) cell-driven airway inflammation in asthma. The induction of counterbalancing Th-1 cell clones, long considered a promising approach for immunotherapy, has failed to fulfil its promise because of potentially detrimental side-effects. This is therefore probably not a valid option for the treatment of asthma. With the increasing awareness that active immune mechanisms exist to control inflammatory responses, interest rises to investigate whether these can be exploited to control allergen-induced airway disease. The induction of antigen-specific T cells with suppressive characteristics (regulatory T cells) is therefore a potentially interesting approach. These regulatory T cells mediate tolerance in healthy, non-atopic individuals and have the potential of becoming an effective means of preventing allergen-induced airway inflammation and possibly of suppressing ongoing allergic immune responses. Here we review the available knowledge about allergen-induced suppressive immunity obtained from animal models taking into account the different developmental stages of allergic airway disease.
Collapse
Affiliation(s)
- K G Tournoy
- Department of Respiratory Diseases, Ghent University Hospital, Ghent University, Flanders Interuniversity Institute for Biotechnology, Ghent, Belgium.
| | | | | | | | | | | |
Collapse
|
63
|
Wang YM, Zhang GY, Wang Y, Hu M, Wu H, Watson D, Hori S, Alexander IE, Harris DCH, Alexander SI. Foxp3-transduced polyclonal regulatory T cells protect against chronic renal injury from adriamycin. J Am Soc Nephrol 2006; 17:697-706. [PMID: 16467443 DOI: 10.1681/asn.2005090978] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Chronic proteinuric renal injury is a major cause of ESRD. Adriamycin nephropathy is a murine model of chronic proteinuric renal disease whereby chemical injury is followed by immune and structural changes that mimic human disease. Foxp3 is a gene that induces a regulatory T cell (Treg) phenotype. It was hypothesized that Foxp3-transduced Treg could protect against renal injury in Adriamycin nephropathy. CD4+ T cells were transduced with either a Foxp3-containing retrovirus or a control retrovirus. Foxp3-transduced T cells had a regulatory phenotype by functional and phenotypic assays. Adoptive transfer of Foxp3-transduced T cells protected against renal injury. Urinary protein excretion and serum creatinine were reduced (P<0.05), and there was significantly less glomerulosclerosis, tubular damage, and interstitial infiltrates (P<0.01). It is concluded that Foxp3-transduced Treg cells may have a therapeutic role in protecting against immune injury and disease progression in chronic proteinuric renal disease.
Collapse
Affiliation(s)
- Yuan Min Wang
- Centre for Kidney Research, Children's Hospital at Westmead, and Centre for Transplantation and Renal Research, University of Sydney at Westmead Millennium Institute, New South Wales 2145, Sydney, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Laffont S, Coudert JD, Garidou L, Delpy L, Wiedemann A, Demur C, Coureau C, Guéry JC. CD8+ T-cell-mediated killing of donor dendritic cells prevents alloreactive T helper type-2 responses in vivo. Blood 2006; 108:2257-64. [PMID: 16449531 DOI: 10.1182/blood-2005-10-4059] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Accumulating evidence indicates that, in absence of CD8+ T-cell activation, CD4+ T-cell-mediated allograft rejection is associated with a dominant Th2-cell response and eosinophil infiltrates. In this study, we analyzed the mechanisms by which CD8+ T cells regulate alloreactive CD4+ T-cell priming and differentiation into interleukin 4 (IL-4)-producing cells. We showed that interferon gamma (IFN-gamma) production by CD8+ T cells was dispensable for the inhibition of Th2-cell development, as well as tissue eosinophilia and type 2 cytokine production in the rejected grafts. Since we noticed that CD8+ T cells not only suppressed Th2 differentiation, but also down-modulated the overall priming of alloreactive CD4+ T cells, we evaluated whether CD8+ T cells act by limiting the accumulation of donor-derived dendritic cells (DCs) in lymph nodes. We found that indeed, alloreactive CD8+ T cells rapidly eliminated allogeneic DCs from T-cell areas of draining lymph nodes, through a perforin-dependent mechanism. Thus, our data demonstrate that cytotoxic T lymphocyte (CTL)-mediated clearance of allogeneic DCs is a negative feedback mechanism that limits the duration of alloantigen presentation in draining lymph nodes, thereby modulating the amplitude and polarization of the primary alloreactive CD4+ T-cell responses.
Collapse
Affiliation(s)
- Sophie Laffont
- INSERM U563, CHU Purpan, Place du Dr Baylac, 31 300 Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Buer J, Westendorf AM, Zeng AP, He F, Hansen W, Probst-Kepper M. Mechanisms of Central and Peripheral T-Cell Tolerance: An Update. Transfus Med Hemother 2005. [DOI: 10.1159/000089128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
66
|
Olcott AP, Tian J, Walker V, Dang H, Middleton B, Adorini L, Washburn L, Kaufman DL. Antigen-based therapies using ignored determinants of beta cell antigens can more effectively inhibit late-stage autoimmune disease in diabetes-prone mice. THE JOURNAL OF IMMUNOLOGY 2005; 175:1991-9. [PMID: 16034144 DOI: 10.4049/jimmunol.175.3.1991] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
As organ-specific autoimmune diseases do not become manifest until well-advanced, interventive therapies must inhibit late-stage disease processes. Using a panel of immunogenic peptides from various beta cell Ags, we evaluated the factors influencing the efficacy of Ag-based therapies in diabetes-prone NOD mice with advanced disease. The ability of the major beta cell autoantigen target determinants (TDs) to prime Th2 responses declined sharply between 6 and 12 wk of age, whereas the ability of immunogenic ignored determinants (IDs) of beta cell Ags to prime Th2 responses was unaffected by the disease process. The different patterns of TD and ID immunogenicity (even from the same beta cell Ag) may be due to the exhaustion of uncommitted TD-reactive, but not ID-reactive, T cell pools by recruitment into the autoimmune cascade. Therapeutic efficacy was associated with a peptide's immunogenicity and ability to promote Th2 spreading late in the disease process but not its affinity for I-Ag7 or its expression pattern (beta cell specific/nonspecific or rare/abundant). Characterization of some IDs revealed them to be "absolute" cryptic determinants. Such determinants have little impact on T cell selection, leaving large precursor T cell pools available for priming by synthetic peptides. Traditional Ag-based therapeutics using whole autoantigens or their TDs cannot prime responses to such determinants. These findings suggest a new strategy for designing more efficacious Ag-based therapeutics for late-stage autoimmune diseases.
Collapse
Affiliation(s)
- Angelica P Olcott
- Department of Molecular and Medical Pharmacology, University of California, School of Medicine, Los Angeles, CA 90095-1735, USA
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Barchet W, Price JD, Cella M, Colonna M, MacMillan SK, Cobb JP, Thompson PA, Murphy KM, Atkinson JP, Kemper C. Complement-induced regulatory T cells suppress T-cell responses but allow for dendritic-cell maturation. Blood 2005; 107:1497-504. [PMID: 16239430 PMCID: PMC1895395 DOI: 10.1182/blood-2005-07-2951] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Concurrent activation of the T-cell receptor (TCR) and complement regulator CD46 on human CD4+ T lymphocytes induces Tr1-like regulatory T cells that suppress through IL-10 secretion bystander T-cell proliferation. Here we show that, despite their IL-10 production, CD46-induced T-regulatory T cells (Tregs) do not suppress the activation/maturation of dendritic cells (DCs). DC maturation by complement/CD46-induced Tregs is mediated through simultaneous secretion of GM-CSF and soluble CD40L, factors favoring DC differentiation and reversing inhibitory effects of IL-10. Thus, CD46-induced Tregs produce a distinct cytokine profile that inhibits T-cell responses but leaves DC activation unimpaired. Such "DC-sparing" Tregs could be desirable at host/environment interfaces such as the gastrointestinal tract where their specific cytokine profile provides a mechanism that ensures unresponsiveness to commensal bacteria while maintaining reactivity to invading pathogens.
Collapse
Affiliation(s)
- Winfried Barchet
- Washington University School of Medicine, Department of Pathology and Immunology, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Abbott JR, Palmer GH, Kegerreis KA, Hetrick PF, Howard CJ, Hope JC, Brown WC. Rapid and long-term disappearance of CD4+ T lymphocyte responses specific for Anaplasma marginale major surface protein-2 (MSP2) in MSP2 vaccinates following challenge with live A. marginale. THE JOURNAL OF IMMUNOLOGY 2005; 174:6702-15. [PMID: 15905510 DOI: 10.4049/jimmunol.174.11.6702] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In humans and ruminants infected with Anaplasma, the major surface protein 2 (MSP2) is immunodominant. Numerous CD4(+) T cell epitopes in the hypervariable and conserved regions of MSP2 contribute to this immunodominance. Antigenic variation in MSP2 occurs throughout acute and persistent infection, and sequentially emerging variants are thought to be controlled by variant-specific Ab. This study tested the hypothesis that challenge of cattle with Anaplasma marginale expressing MSP2 variants to which the animals had been immunized, would stimulate variant epitope-specific recall CD4(+) T cell and IgG responses and organism clearance. MSP2-specific T lymphocyte responses, determined by IFN-gamma ELISPOT and proliferation assays, were strong before and for 3 wk postchallenge. Surprisingly, these responses became undetectable by the peak of rickettsemia, composed predominantly of organisms expressing the same MSP2 variants used for immunization. Immune responsiveness remained insignificant during subsequent persistent A. marginale infection up to 1 year. The suppressed response was specific for A. marginale, as responses to Clostridium vaccine Ag were consistently observed. CD4(+)CD25(+) T cells and cytokines IL-10 and TGF-beta1 did not increase after challenge. Furthermore, a suppressive effect of nonresponding cells was not observed. Lymphocyte proliferation and viability were lost in vitro in the presence of physiologically relevant numbers of A. marginale organisms. These results suggest that loss of memory T cell responses following A. marginale infection is due to a mechanism other than induction of T regulatory cells, such as peripheral deletion of MSP2-specific T cells.
Collapse
Affiliation(s)
- Jeffrey R Abbott
- Program in Vector-Borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, 99164, USA
| | | | | | | | | | | | | |
Collapse
|
69
|
Mirenda V, Millington O, Lechler RI, Scott D, Hernandez-Fuentes MP, Read J, Tan PH, George AJT, Garside P, Marelli-Berg FM. Tolerant T cells display impaired trafficking ability. Eur J Immunol 2005; 35:2146-56. [PMID: 15948215 DOI: 10.1002/eji.200425823] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Based on our previous observation that anergic T lymphocytes lose their migratory ability in vitro, we have proposed that anergic T cells are retained in the site where they have been generated to exert their regulatory function. In this study we have analyzed T lymphocyte trafficking and motility following the induction of tolerance in vivo. In a model of non-deletional negative vaccination to xenoantigens in which dendritic cells (DC) localize to specific lymphoid sites depending on the route of administration, tolerant T cells remained localized in the lymph nodes colonized by tolerogenic DC, while primed T cells could traffic efficiently. Using an oral tolerance model that enables the 'tracking' of ovalbumin-specific TCR-transgenic T cells, we confirmed that T cells lose the ability to migrate through syngeneic endothelial cell monolayers following tolerance induction in vivo. Finally, we show that tolerant T cells (both in vitro and ex vivo) can inhibit migration of responsive T cells in an antigen-independent manner. Thus, hyporesponsive T cells localize at the site of tolerance induction in vivo, where they exert their anti-inflammatory properties. In physiological terms, this effect is likely to render immunoregulation a more efficient and controllable event.
Collapse
Affiliation(s)
- Vincenzo Mirenda
- Department of Immunology, Division of Medicine, Imperial College London, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Abstract
Autoimmunity is a complex process that likely results from the summation of multiple defective tolerance mechanisms. The NOD mouse strain is an excellent model of autoimmune disease and an important tool for dissecting tolerance mechanisms. The strength of this mouse strain is that it develops spontaneous autoimmune diabetes, which shares many similarities to autoimmune or type 1a diabetes (T1D) in human subjects, including the presence of pancreas-specific autoantibodies, autoreactive CD4+ and CD8+ T cells, and genetic linkage to disease syntenic to that found in humans. During the past ten years, investigators have used a wide variety of tools to study these mice, including immunological reagents and transgenic and knockout strains; these tools have tremendously enhanced the study of the fundamental disease mechanisms. In addition, investigators have recently developed a number of therapeutic interventions in this animal model that have now been translated into human therapies. In this review, we summarize many of the important features of disease development and progression in the NOD strain, emphasizing the role of central and peripheral tolerance mechanisms that affect diabetes in these mice. The information gained from this highly relevant model of human disease will lead to potential therapies that may alter the development of the disease and its progression in patients with T1D.
Collapse
Affiliation(s)
- Mark S Anderson
- Diabetes Center, University of California, San Francisco, California 94143, USA.
| | | |
Collapse
|
71
|
Domenig C, Sanchez-Fueyo A, Kurtz J, Alexopoulos SP, Mariat C, Sykes M, Strom TB, Zheng XX. Roles of Deletion and Regulation in Creating Mixed Chimerism and Allograft Tolerance Using a Nonlymphoablative Irradiation-Free Protocol. THE JOURNAL OF IMMUNOLOGY 2005; 175:51-60. [PMID: 15972631 DOI: 10.4049/jimmunol.175.1.51] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The induction of mixed chimerism (MC) is a powerful and effective means to achieve transplantation tolerance in rodent models. Host conditioning with irradiation or cytotoxic drugs has been used in many protocols for chimeric induction across allogeneic barriers. The deletion of alloreactive T cell clones has been described as the main mechanism responsible for the induction of a stable MC. In this study, we demonstrate that a stable MC and skin allograft tolerance can be established across MHC barriers by a noncytotoxic, irradiation-free approach using costimulation blockade plus rapamycin treatment. By using an adoptive transfer model of skin allograft and using specific Vbeta TCR probes, we demonstrated that deletion of donor-reactive cytopathic T cell clones is indeed profound in tolerant hosts. Nonetheless, the challenge of tolerant mixed chimeras with 5 million mononuclear leukocytes (MNL) from naive syngeneic mice was neither able to abolish the stable MC nor to trigger skin allograft rejection, a hallmark of peripheral, not central tolerance. Furthermore, in an adoptive transfer model, MNLs harvested from tolerant hosts significantly inhibited the capacity of naive MNLs to reject same donor, but not third-party, skin allografts. Moreover, when we transplanted skin allografts from stable tolerant chimeras onto syngeneic immune-incompetent mice, graft-infiltrating T cells migrated from the graft site, expanded in the new host, and protected allografts from acute rejection by naive syngeneic MNLs. In this model, both deletional and immunoregulatory mechanisms are active during the induction and/or maintenance of allograft tolerance through creation of MC using a potentially clinically applicable regimen.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Bone Marrow Transplantation/immunology
- CD4-Positive T-Lymphocytes/immunology
- Chimera/immunology
- Clonal Deletion
- Graft Rejection/immunology
- Graft Rejection/prevention & control
- Immune Tolerance
- Immunosuppressive Agents/pharmacology
- Leukocytes, Mononuclear/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Models, Immunological
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Sirolimus/pharmacology
- Skin Transplantation/immunology
- Transplantation Immunology
- Transplantation, Homologous
- Transplantation, Isogeneic
Collapse
Affiliation(s)
- Christoph Domenig
- Transplantation Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Daniel V, Naujokat C, Sadeghi M, Wiesel M, Hergesell O, Opelz G. Association of Circulating Interleukin (IL)-12– and IL-10–Producing Dendritic Cells with Time Posttransplant, Dose of Immunosuppression, and Plasma Cytokines in Renal-Transplant Recipients. Transplantation 2005; 79:1498-506. [PMID: 15940038 DOI: 10.1097/01.tp.0000163470.83217.e6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Interleukin (IL)-12-producing dendritic cells (IL-12+DC) polarize T helper (Th) differentiation toward Th1, whereas IL-10+DC induce Th differentiation toward Th2. We investigated DC and plasma cytokine patterns early and late after transplantation. METHODS Twenty-five hospitalized renal-transplant recipients without acute rejection or infection early (<40 days) posttransplant, 32 symptom-free outpatients with long-term functioning transplants (2,762+/-2,423 days posttransplant), and 17 healthy controls were studied. The intracellular production of IL-12 and IL-10 in CD11c+ CD83+ CD40+ DC was measured in freshly obtained whole blood using four-color fluorescence flow cytometry. In addition, plasma cytokine levels were investigated. RESULTS Early and late posttransplant patients had significantly lower proportions of IL-12+DC (early: P=0.001; late: P=0.034) and lower ratios of IL-12+/IL-10+DC (early: P=0.0001; late: P<0.0001) than healthy controls. IL-10+DC (P=0.0004) and IL-12+DC (P=0.002) increased with time posttransplant in association with dose reductions of cyclosporine (IL-10+DC: P=0.003; IL-12+DC: P=0.005), methylprednisolone (IL-10+DC: P<0.0001; IL-12+DC: P=0.001) and mycophenolate mofetil (IL-10+DC: P<0.0001; IL-12+DC: P=0.004). Both IL-10+DC and IL-12+DC were associated with low plasma IL-10 (IL-10+DC: P=0.010; IL-12+DC: P=0.011) and high plasma IL-6 (IL-10+DC: P=0.001; IL-12+DC: P=0.009). IL-10+DC were also associated with high plasma levels of IL-3 (P=0.003), interferon (IFN)-gamma (P=0.014), and IL-2 (P=0.058). CONCLUSION IL-10+DC and IL-12+DC in peripheral blood are associated with time after transplantation and dosage of immunosuppression. IL-10+DC dominate late posttransplant in the presence of Th1 plasma cytokines (high IFN-gamma and IL-2), high IL-3, and low IL-10. These findings could be a reflection of immunoregulatory processes favoring long-term allograft acceptance.
Collapse
Affiliation(s)
- Volker Daniel
- Institute of Immunology, Department of Transplantation Immunology, University of Heidelberg, S-69120 Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
73
|
Abstract
The identification of suppressor T cells as important regulators of basic processes that are designed to maintain tolerance has opened an important area of potential clinical investigation in autoimmunity, graft-versus-host disease and transplantation. However, the field has been limited by an inability to define the antigenic specificities of these cells and by the small numbers of circulating regulatory T cells. Recently, new methods for expanding polyclonal and antigen-specific regulatory T cells have emerged. This article summarizes efforts to exploit regulatory T-cell therapy for the treatment of immunological diseases and poses the question of when and where regulatory T cells will first impact on clinical diseases.
Collapse
Affiliation(s)
- Jeffrey A Bluestone
- University of California at San Francisco Diabetes Center, University of California, San Francisco, Box 0540, 513 Parnassus Avenue, San Francisco, California 94143-0540, USA. jbluestdiabetes.ucsf.edu
| |
Collapse
|
74
|
Cai Y, Tang XD, Zhou PJ. A combination of anergic cells' adoptive transfer and rapamycin therapy prolongs cardiac allograft survival in mice. Scand J Immunol 2005; 61:266-73. [PMID: 15787744 DOI: 10.1111/j.1365-3083.2005.01555.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The in vivo immunoregulatory effect of anergic cells induced by blocking the costimulatory pathway was investigated in this study. Anergic cells were generated in vitro by mixed culture of murine splenic cells from BALB/c and C3H/HeJ under the blockade of anti-CD154 and anti-CD80 monoclonal antibodies, and the in vitro activity of anergic cells were observed. The 3.0 Gy gamma-irradiated BALB/c mice received cardic allografts from C3H/HeJ, and anergic cells were intravenously injected immediately after transplantation. Recipient mice injected with anergic cells also received rapamycin therapy (1 mg/kg/day) for 14 days. On day 7 after transplantation, the subsets of peripheral blood T lymphocytes, the pathology of grafts and the infiltration of lymphocytes in grafts were analysed. Untreated gamma-irradiated animals showed a graft median survival time (MST) of 9 days. Animals injected with anergic cells only or receiving rapamycin therapy alone showed MST of 11 and 17 days, respectively. MST of allograft in mice treated with control cells plus rapamycin therapy was 9 days. Animals injected with anergic cells plus rapamycin therapy, but receiving third-party allografts (C57BL/6J), showed an MST of 15 days. However, anergic cell injection plus rapamycin therapy prolonged allograft survival significantly (MST 28 days, P < 0.01). The rejection was mild and tissue architecture was preserved in recipient mice receiving anergic cell injection plus rapamycin therapy. Furthermore, anergic cells and rapamycin therapy decreased the percentage of peripheral blood CD4+ and CD8+ T cells (including CD25+, CD152+, CD154+ and CD28+ subsets) and greatly reduced the infiltrating lymphocytes in allografts (including CD3+, CD4+, CD8+ and CD25+ T cells). In conclusion, the treatment based on anergic cells' adoptive transfer plus rapamycin therapy demonstrated a significant prolongation of murine cardiac allograft survival in a donor antigen-specific manner. This therapeutic protocol alleviated allograft rejection to solid allograft in vivo.
Collapse
Affiliation(s)
- Y Cai
- Department of Renal Transplantation, the First Affiliated Hospital of Wenzhou Medical College, Wenzhou, China
| | | | | |
Collapse
|
75
|
Sánchez-Fueyo A. [Immunological tolerance and liver transplantation]. GASTROENTEROLOGIA Y HEPATOLOGIA 2005; 28:250-6. [PMID: 15811269 DOI: 10.1157/13073096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The induction of tolerance to allografts has traditionally been one of the basic aims of transplantation research. Multiple data obtained in experimental models indicate that the outcome of transplantation (rejection versus acceptance/tolerance) depends on the balance between allo-reactive cytopathic lymphocytes and immunoregulatory lymphocytes. Thus, most tolerance-inducing treatments aim to reduce the number of allo-aggressive lymphocytes and, at the same time, to increase the population of regulatory lymphocytes, which ensure graft viability once drug therapy has been withdrawn. Liver allografts are singular in that they are accepted without the need for treatment in most experimental models. Likewise, in humans, liver grafts also show a lower susceptibility to rejection than any other organ and immunosuppressive treatment can be completely eliminated in approximately 25% of recipients. Many mechanisms have been proposed to explain the tolerogenic properties of the liver. Notable among these are the effects derived from the large number of passing leukocytes present in the liver and its peculiar anatomy that maximizes contact among blood lymphocytes and liver cells with tolerogenic potential. Although there are many cases of tolerance in human allograft recipients, therapeutic strategies that would allow predictable tolerance induction and without a high risk of adverse affects are still lacking. Therefore, most studies in humans have traditionally aimed to minimize doses of immunosuppressive drugs rather than eliminate them. However, recent results in preclinical models and pilot studies indicate that therapeutic protocols for tolerance induction may become available in the not too distant future.
Collapse
Affiliation(s)
- A Sánchez-Fueyo
- Instituto de Enfermedades Digestivas, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain.
| |
Collapse
|
76
|
Schmidt-Weber CB, Blaser K. Regulation and role of transforming growth factor-beta in immune tolerance induction and inflammation. Curr Opin Immunol 2005; 16:709-16. [PMID: 15511662 DOI: 10.1016/j.coi.2004.09.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is known to mediate pleiotropic functions both inside and outside the immune system. Recent progress in this field underlines the role of TGF-beta in regulatory T (Treg) cells, where it participates in both suppression and differentiation. In addition, recent information highlights the role of TGF-beta in repair responses that lead to matrix deposition and tissue remodelling. Many chronic inflammatory diseases, such as asthma, profit from the suppression of specific immune responses by TGF-beta; however, TGF-beta-mediated tissue remodelling can be a serious complication in such diseases.
Collapse
Affiliation(s)
- Carsten B Schmidt-Weber
- Swiss Institute of Allergy and Asthma Research, Obere Strasse 22, CH-7270 Davos, Switzerland.
| | | |
Collapse
|
77
|
Hirata M, Hirata K, Hara T, Kawabuchi M, Fukuma T. Expression of TGF-?-like molecules in the life cycle of Schistosoma japonicum. Parasitol Res 2005; 95:367-73. [PMID: 15711848 DOI: 10.1007/s00436-004-1296-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2004] [Accepted: 11/23/2004] [Indexed: 11/28/2022]
Abstract
The transforming growth factor beta (TGF-beta) family controls an extremely wide range of biological activities, such as the growth and differentiation of cells, and immunological events against infectious agents. Although TGF-beta homologs appear to be widely present in metazoan animals, studies of parasite-derived molecules are relatively few. Using antibodies against anti-mouse TGF-beta1, -beta2, and -beta3, we show the expression of TGF-beta-like molecules in Schistosoma japonicum cercariae, schistosomula, eggs and adult worms. Intense immunoreactivity was found on the surface of free-living cercarial bodies. In transverse sections of cercariae, the molecules were localized in the tegument and subtegumental cells, and the number and distribution of producing cells significantly differed with each antibody. In the skin-migrating stage, the expression in the tegumental surface gradually decreased and became almost negative within 48 h of exposure. In adult worms and eggs, the reactivity was found in subtegumental cells and in cells of a tubular structure, respectively. In western blot analysis, the detection of conventional TGF-beta molecules failed. The expression of TGF-beta-like molecules was distinctly regulated at each developmental stage.
Collapse
Affiliation(s)
- M Hirata
- Department of Parasitology, Kurume University School of Medicine, 830-0011, Kurume, Japan.
| | | | | | | | | |
Collapse
|
78
|
Cardona PJ, Amat I, Gordillo S, Arcos V, Guirado E, Díaz J, Vilaplana C, Tapia G, Ausina V. Immunotherapy with fragmented Mycobacterium tuberculosis cells increases the effectiveness of chemotherapy against a chronical infection in a murine model of tuberculosis. Vaccine 2005; 23:1393-8. [PMID: 15661388 DOI: 10.1016/j.vaccine.2004.09.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2004] [Revised: 09/01/2004] [Accepted: 09/08/2004] [Indexed: 11/20/2022]
Abstract
Reduction of colony forming units by rifampicin-isoniazid therapy given 9-17 weeks post-infection was made more pronounced by immunotherapy with a vaccine made of fragmented Mycobacterium tuberculosis cells detoxified and liposomed (RUTI), given on weeks 17, 19 and 21 post-infection, in the murine model of tuberculosis in C57BL/6 and DBA/2 inbred strains. RUTI triggered a Th1/Th2 response, as demonstrated by the production of IgG1, IgG2a and IgG3 antibodies against a wide range of peptides. The histological analysis did not show neither eosinophilia nor necrosis, and granulomatous infiltration was only slightly increased in C57BL/6 mice when RUTI was administered intranasally.
Collapse
Affiliation(s)
- Pere-Joan Cardona
- Unitat de Tuberculosi Experimental, Department of Microbiology, Fundació Institut per a la Investigació en Ciències de la Salut "Germans Trias i Pujol", Universitat Autònoma de Barcelona, 08916 Badalona, Catalonia, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Granja C, Moliterno RA, Ferreira MS, Fonseca JA, Kalil J, Coelho V. T-cell autoreactivity to Hsp in human transplantation may involve both proinflammatory and regulatory functions. Hum Immunol 2005; 65:124-34. [PMID: 14969767 DOI: 10.1016/j.humimm.2003.10.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2003] [Revised: 09/24/2003] [Accepted: 10/06/2003] [Indexed: 10/26/2022]
Abstract
Heat shock proteins (Hsp) are moving from the category of basically intracellular chaperone molecules to important proteins in both innate and acquired immune responses, with great potential for clinical application as immunomodulators. Both proinflammatory and regulatory Hsp-reactive T cells have been described in animal models of autoimmune diseases. To investigate the role of autoreactivity to Hsp60 and Hsp70 in human transplantation, we analyzed, sequentially, peripheral blood mononuclear cell proliferation and cytokine production before and at different time points after renal transplantation, as well as the modulation of proliferation to Hsp in the presence of exogenous cytokines. Proliferation to Hsp60 and Hsp70 in the pretransplantation (pre-Tx) period was significantly associated with rejection episodes in the first months post-Tx. In contrast, IL-4 production was significantly associated with absence of rejection. Addition of exogenous IL-4 distinctly modulated the proliferative response to Hsp60; inhibiting proliferation in 83% of patients in the early post-Tx period (0-6 months), in which rejection episodes occurred, and inducing proliferation in 62.5% of patients in the later period (>12-24 months), when no rejection was observed. Characterization of autoreactive anti-Hsp60 regulatory T cells may permit new approaches to control the proinflammatory response to the graft, as well as aggressive autoimmunity.
Collapse
Affiliation(s)
- Clarissa Granja
- Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
80
|
Palucka AK, Laupeze B, Aspord C, Saito H, Jego G, Fay J, Paczesny S, Pascual V, Banchereau J. Immunotherapy via dendritic cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2005; 560:105-14. [PMID: 15932026 DOI: 10.1007/0-387-24180-9_14] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The immune system evolved to protect us from microbes. The antigen (Ag)-nonspecific innate immunity and Ag-specific adaptive immunity synergize to eradicate the invading pathogen through cells, such as dendritic cells (DCZ7) and lymphocytes, and through their effector proteins including antimicrobial peptides, complement, and antibodies. Its intrinsic complexity renders the immune system prone to dysfunction including cancer, autoimmunity, chronic inflammation and allergy. DCs are unique in their capacity to induce and regulate immune responses and are therefore attractive candidates for immunotherapy. However, DCs consist of distinct subsets with common as well as unique functions that lead to distinct types of immune responses. Therefore, understanding DC heterogeneity and their role in immunopathology is critical to design better strategies for immunotherapy. Indeed, what we learn from studying autoimmunity will help us induce strong vaccine specific immunity, either protective, as in the case of microbes, or therapeutic, as in the case of tumors.
Collapse
Affiliation(s)
- A Karolina Palucka
- Baylor Institute for Immunology Research, 3434 Live Oak, Dallas, TX 75204, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Liu J, Liu Z, Witkowski P, Vlad G, Manavalan JS, Scotto L, Kim-Schulze S, Cortesini R, Hardy MA, Suciu-Foca N. Rat CD8+ FOXP3+ T suppressor cells mediate tolerance to allogeneic heart transplants, inducing PIR-B in APC and rendering the graft invulnerable to rejection. Transpl Immunol 2004; 13:239-47. [PMID: 15589736 DOI: 10.1016/j.trim.2004.10.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Accepted: 10/26/2004] [Indexed: 11/28/2022]
Abstract
Human CD8+ FOXP3+ T suppressor cells (TS) were previously shown to induce the expression of the inhibitory receptors, Immunoglobulin-like transcript (ILT) 3 and ILT4 on dendritic and endothelial cells, rendering them tolerogenic to allogeneic T cells. We have demonstrated the importance of CD8+ TS in a rat model of heart allo-transplantation. Tolerance was induced in ACI recipients by multiple transfusions of UVB-irradiated blood from Lewis heart donors. CD8+ T cells from tolerant ACI rats expressed FOXP3, transferred tolerance to naive secondary hosts and induced the upregulation of the inhibitory receptor, paired immunoglobulin-like receptor (PIR)-B, an ILT4 orthologue, in Lewis dendritic cells (DC) and heart endothelial cells (EC). When long-term surviving Lewis heart allografts with PIR-B+ EC were retransplanted from a primary to a secondary ACI recipient they did not elicit rejection. This study focuses attention on the need to develop agents that act directly on graft EC in order to achieve tolerance.
Collapse
Affiliation(s)
- Jiawang Liu
- Columbia University, Department of Pathology, 630 West 168th Street-P and S 14-401, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
|
83
|
Affiliation(s)
- Alberto Sánchez-Fueyo
- Liver Unit, Institut de Malalties Digestives, Hospital Clínic de Barcelona, Institut d'Investigacions Biomédiques August PiSunyer, Universitat de Barcelona, Barcelona, Spain.
| | | |
Collapse
|
84
|
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
|
85
|
Bluestone JA, Tang Q. Therapeutic vaccination using CD4+CD25+ antigen-specific regulatory T cells. Proc Natl Acad Sci U S A 2004; 101 Suppl 2:14622-6. [PMID: 15322272 PMCID: PMC521996 DOI: 10.1073/pnas.0405234101] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Autoimmune disease results from the dysregulation of basic tolerogenic processes designed to control self/non-self-discrimination. Approaches to treat autoimmunity have focused historically on potent immunosuppressives that block the activation and expansion of antigen-specific T cells before they differentiate into pathogenic T cell responses. These therapies are very efficient in reducing clonal expansion and altering early signaling pathways. However, once the pathogenic responses are established (i.e., autoimmunity), the interventions are less effective on activated and differentiated T cell subsets (including memory T cells) or acting in the presence of an inflammatory milieu to abort immune responses at the target tissue and systemically. Moreover, the current immunotherapies require continuous use because they do not redirect the immune system to a state of tolerance. The continuous treatment leads to long-term toxicities and can profoundly suppress protective immune responses targeted at viruses, bacteria, and other pathogens. Over the past decade, there have been tremendous advances in our understanding of the basic processes that control immune tolerance. Among the most exciting has been the identification of a professional regulatory T cell subset that has shown enormous potential in suppressing pathologic immune responses in autoimmune diseases, transplantation, and graft vs. host disease. In this review, we summarize current efforts to induce and maintain tolerance in the autoimmune diabetes setting by using therapeutic vaccination with CD4(+)CD25(+) regulatory T cells. Emphasis will be placed on approaches to exploit regulatory T cells either directly or through the use of anti-CD3 immunotherapy.
Collapse
Affiliation(s)
- Jeffrey A Bluestone
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA 94143-0540, USA.
| | | |
Collapse
|
86
|
Abstract
Nickel is the most common contact allergen in humans. Until recently, many questions concerning tolerance mechanisms to nickel were unresolved. Besides human ex vivo, intervention and observation studies, the establishment of a reproducible mouse model has contributed to the analysis of these mechanisms. A more detailed understanding of the pathogenesis of nickel allergy and tolerance towards nickel by investigations in an animal model and in human studies is a prerequisite for developing specific prevention and therapy of nickel allergy. With this article, we provide a review of the investigations concerning nickel allergy and give perspectives towards oral tolerance induction to nickel in the animal model and in humans.
Collapse
Affiliation(s)
- S Artik
- Institut für umweltmedizinische Forschung an der Heinrich-Heine-Universität Düsseldorf gGmbH, Abteilung für Allergologie und Immunologie, Universitätshautklinik, Heinrich-Heine-Universität Düsseldorf
| | | | | |
Collapse
|
87
|
Anderson BE, McNiff JM, Matte C, Athanasiadis I, Shlomchik WD, Shlomchik MJ. Recipient CD4+ T cells that survive irradiation regulate chronic graft-versus-host disease. Blood 2004; 104:1565-73. [PMID: 15150080 DOI: 10.1182/blood-2004-01-0328] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Chronic graft-versus-host disease (cGVHD) is an increasingly common cause of morbidity and mortality in allogeneic stem cell transplantation (alloSCT). Relative to acute GVHD (aGVHD), much less is understood about cGVHD. Using the B10.D2 → BALB/c murine cGVHD model, which shares critical pathologic features with human cGVHD, we find that radiation-resistant host T cells regulate cGVHD. We initially observed that recipients lacking all lymphocytes developed accelerated and more severe cGVHD. Using genetically deficient recipients, we determined that αβ+CD4+ T cells were required to regulate cGVHD. Increased cGVHD severity was not due to the absence of T cells per se. Rather, the potency of regulation was proportional to host T-cell receptor (TCR) diversity. Only CD4+CD25+, and not CD4+CD25-, host T cells ameliorated cGVHD when added back, indicating that host T cells acted not via host-versus-graft activity or by reducing homeostatic proliferation but by an undefined regulatory mechanism. Thus, preparative regimens that spare host CD4+CD25+ T cells may reduce cGVHD. Donor CD4+CD25+ T cells also reduced cGVHD. Depletion of CD4+CD25+ cells from the inoculum exacerbated disease, whereas transplantation of additional CD4+CD25+ cells protected against severe cGVHD. Additional CD4+CD25+ cells also promoted healing of established lesions, suggesting that their effects persist during the evolution of cGVHD.
Collapse
Affiliation(s)
- Britt E Anderson
- Department of Laboratory Medicine, Yale University School of Medicine, 333 Cedar St, PO Box 208035, New Haven, CT 06520-8035, USA
| | | | | | | | | | | |
Collapse
|
88
|
Viguier M, Lemaître F, Verola O, Cho MS, Gorochov G, Dubertret L, Bachelez H, Kourilsky P, Ferradini L. Foxp3 expressing CD4+CD25(high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:1444-53. [PMID: 15240741 DOI: 10.4049/jimmunol.173.2.1444] [Citation(s) in RCA: 512] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dominant tolerance is mediated by regulatory T cells (T(reg)) that control harmful autoimmune T cells in the periphery. In this study, we investigate the implication of T(reg) in modulating infiltrating T lymphocytes in human metastatic melanoma. We found that CD4(+)CD25(high) T cells are overrepresented in metastatic lymph nodes (LNs) with a 2-fold increased frequency compared with both tumor-free LNs and autologous PBMCs. These cells express the Foxp3 transcription factor, display an activated phenotype, and display a polyclonal TCR Vbeta chain repertoire. They inhibit in vitro the proliferation and cytokine production of infiltrating CD4(+)CD25(-) and CD8(+) T cells (IL-2, IFN-gamma) through a cell-contact-dependent mechanism, thus behaving as T(reg). In some cases, the presence of T(reg) type 1/Th3-like lymphocytes could also be demonstrated. Thus, T(reg) are a major component of the immunosuppressive microenvironment of metastatic melanoma LNs. This could explain the poor clinical response of cancer patients under immunotherapeutic protocols, and provides a new basis for future immunotherapeutic strategies counteracting in vivo T(reg) to reinforce local antitumor immune responses.
Collapse
Affiliation(s)
- Manuelle Viguier
- Unité de Biologie Moléculaire du Gène, Institut National de la Santé et de la Recherche Médicale (INSERM) U277, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Abstract
Although central and peripheral tolerance are important for the regulation of human immune responses to self- and microbial antigens, an important role of suppressor CD4+ CD25+ T cells is suggested from the recent investigations of human autoimmune diseases and HIV. These new data provide increasing evidence that altered function of CD4+ CD25+ T cells may be an important factor in a wide range of human inflammatory and infectious diseases.
Collapse
|
90
|
Zheng XX, Sanchez-Fueyo A, Domenig C, Strom TB. The balance of deletion and regulation in allograft tolerance. Immunol Rev 2004; 196:75-84. [PMID: 14617199 DOI: 10.1046/j.1600-065x.2003.00089.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although the precise mechanisms involved in the establishment and maintenance of peripheral allograft tolerance are still not fully understood, it is now clear that acquisition of transplantation tolerance is an active, highly regulated, multistep process. According to the pool size model of allograft tolerance, the allograft outcome, rejection, or tolerance, often depends on the balance between cytopathic and regulatory T cells (Tregs). Although both deletion and regulation play important roles in allograft tolerance, our recent studies showed that the quantitative details for each mechanism differ from model to model. Therefore, we hypothesize that there is a delicate balance between deletion and regulation in allograft tolerance. In a model of allograft tolerance in which the deletional mechanism plays a dominant role, e.g. tolerance produced via creation of mixed chimeras, the regulatory mechanism, albeit sometimes present, is far less important. Whilst in a model in which the regulation mechanism plays a critical role, e.g. donor-specific transfusion plus MR1-induced allograft tolerance, a deletional mechanism lowers the threshold for effective Treg action.
Collapse
Affiliation(s)
- Xin Xiao Zheng
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
91
|
Tree TIM, Duinkerken G, Willemen S, de Vries RRP, Roep BO. HLA-DQ-regulated T-cell responses to islet cell autoantigens insulin and GAD65. Diabetes 2004; 53:1692-9. [PMID: 15220192 DOI: 10.2337/diabetes.53.7.1692] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
HLA-DQ is strongly associated with genetic predisposition to type 1 diabetes. It is assumed that HLA-DQ molecules exert their effects on the disease via the presentation of peptides from islet autoantigens to CD4(+) T-cells, but little information regarding HLA-DQ-restricted, islet antigen-specific, autoreactive T-cells is available. To investigate the role of HLA-DQ in the immune response to islet autoantigens, we measured T-cell proliferation to insulin and GAD65 in the presence and absence of monoclonal antibodies that block HLA-DQ-mediated antigen presentation in recent-onset type 1 diabetic patients and their siblings. Positive proliferative T-cell responses to GAD65 were observed in 60% of type 1 diabetic patients and 52% of siblings. This proliferation was significantly reduced in the presence of anti-DQ antibody, demonstrating the presence of primed, effector HLA-DQ-restricted T-cell responses to GAD65. Positive proliferative responses to insulin were observed in 25% of type 1 diabetic patients and 10% of siblings. However, blocking HLA-DQ-restricted T-cell responses led to a significant increase in proliferation to insulin, implying the presence of primed suppressive HLA-DQ-restricted T-cell responses to insulin. These results indicate that HLA-DQ acts as a restriction element for both proliferative and suppressor cells, with the relative balance of these cells dependent on the nature of the autoantigen.
Collapse
Affiliation(s)
- Timothy I M Tree
- Department of Immunology, Guy's, King's, and St. Thomas' School of Medicine, Denmark Hill Campus, Rayne Institute, London, UK
| | | | | | | | | |
Collapse
|
92
|
Manavalan JS, Kim-Schulze S, Scotto L, Naiyer AJ, Vlad G, Colombo PC, Marboe C, Mancini D, Cortesini R, Suciu-Foca N. Alloantigen specific CD8+CD28- FOXP3+ T suppressor cells induce ILT3+ ILT4+ tolerogenic endothelial cells, inhibiting alloreactivity. Int Immunol 2004; 16:1055-68. [PMID: 15226269 DOI: 10.1093/intimm/dxh107] [Citation(s) in RCA: 209] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Endothelial cells have been shown to activate T cell responses to alloantigens, triggering transplant rejection. However, they may also play a role in tolerance induction. Using RT-PCR we show here that alloantigen specific CD8(+)CD28(-) T suppressor cells generated in vitro are FOXP3 positive and interact with human endothelial cells. This interaction results in the induction of inhibitory receptors and down-regulation of costimulatory and adhesion molecules, thus rendering endothelial cells tolerogenic. In turn, tolerized endothelial cells elicit the differentiation of CD8(+)CD28(-) FOXP3(+) T suppressor cells. Taken together our data demonstrate a functional and phenotypic overlap between tolerogenic dendritic cells and endothelial cells. Furthermore, alloantigen specific CD8(+)CD28(-) FOXP3(+) T cells, which trigger the upregulation of inhibitory receptors in endothelial cells, are present in the circulation of heart allograft recipients in quiescence as demonstrated by flow cytometry, RT-PCR and luciferase transcription assays. Their detection facilitates the identification of patients who may benefit from partial or complete cessation of immunosuppressive therapy, a goal of obvious importance given the morbidity and mortality associated with chronic immunosuppression. Modulation of endothelial cells in favor of promoting tolerance may be important for long-term survival of organ allografts.
Collapse
Affiliation(s)
- John S Manavalan
- Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Abstract
We propose a model where autoimmunity can be viewed as a dynamic system driven by opposite vectors IFN-alpha/beta and TNF. These cytokines drive differentiation of distinct types of DCs, TNF-DCs, or IFN-DCs, which present different antigens leading to distinct autoimmune responses. When balanced, both cytokines synergize in protective immunity. When one of the cytokines prevails, autoimmunity occurs, Type I interferons (IFN-alpha/beta) playing a major role in systemic lupus erythematosus (SLE) and TNF playing a major role in rheumatoid arthritis. This model complements the Type 1/Type 2 paradigm. Therefore, immunity can be viewed as a dynamic system driven by two sets of opposite vectors: IFN-alpha/beta/TNF and IFN-gamma/IL-4.
Collapse
Affiliation(s)
- Jacques Banchereau
- Baylor Institute for Immunology Research, 3434 Live Oak, Dallas, TX 75204 USA
| | | | | |
Collapse
|
94
|
|
95
|
Nanji SA, Hancock WW, Anderson CC, Adams AB, Luo B, Schur CD, Pawlick RL, Wang L, Coyle AJ, Larsen CP, Shapiro AMJ. Multiple combination therapies involving blockade of ICOS/B7RP-1 costimulation facilitate long-term islet allograft survival. Am J Transplant 2004; 4:526-36. [PMID: 15023144 DOI: 10.1111/j.1600-6143.2004.00384.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In recent years a series of novel costimulatory molecules have been identified, including inducible costimulator (ICOS). In a fully major histocompatibility complex (MHC)-mismatched mouse model of islet transplantation, we demonstrate that while monotherapy with CTLA4-Ig, CD40 ligand monoclonal antibody (CD40L mAb) or rapamycin each improves islet allograft survival, graft rejection eventually develops. Immunohistologic analysis of rejected grafts revealed increased ICOS expression, suggesting a role for this costimulatory molecule as an alternate pathway for T-cell activation. The combination of a blocking anti-ICOS mAb with each of the above therapies resulted in significantly improved islet allograft survival, confirming the importance of ICOS signaling in islet allograft rejection. Mechanistic studies conducted in mice treated with anti-ICOS mAb and rapamycin demonstrated a lack of donor-specific immunological tolerance and an absence of regulatory T-cell activity. However, a dramatic effect was seen on acute anti-donor responses whereby anti-ICOS mAb and rapamycin significantly reduced the initial expansion and function of alloreactive T cells. These data demonstrate that blockade of the ICOS/B7RP-1 pathway has potential therapeutic benefit given its role in enhancing islet allograft survival and regulating acute alloresponses in vivo.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antigens, Differentiation, T-Lymphocyte/chemistry
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- B7-1 Antigen/chemistry
- B7-1 Antigen/metabolism
- CD4-Positive T-Lymphocytes/metabolism
- CD40 Ligand/chemistry
- CD8-Positive T-Lymphocytes/metabolism
- Cell Survival
- Graft Rejection
- Graft Survival
- Immunosuppressive Agents/pharmacology
- Inducible T-Cell Co-Stimulator Ligand
- Inducible T-Cell Co-Stimulator Protein
- Interferon-gamma/metabolism
- Islets of Langerhans Transplantation/methods
- Ligands
- Lymphocyte Activation
- Lymphocytes/cytology
- Lymphocytes/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Sirolimus/pharmacology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Time Factors
Collapse
Affiliation(s)
- Sulaiman A Nanji
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
|
97
|
Fujino M, Kitazawa Y, Kawasaki M, Funeshima N, Kimura H, Nakajima T, Saito H, Li XK. Differences in lymphocyte gene expression between tolerant and syngeneic liver grafted rats. Liver Transpl 2004; 10:379-91. [PMID: 15004765 DOI: 10.1002/lt.20082] [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: 02/07/2023]
Abstract
Induction of tolerance to allogeneic donor grafts is a clinically desirable goal in bone marrow and solid organ transplantation. We have taken the advantage of DNA microarray technology to investigate gene expression mechanism in regulatory cells. In the present study, using a tacrolimus (FK506) induced tolerance of the fully mismatched liver allograft rat model, we demonstrated that, in contrast with peripheral blood lymphocytes (PBLs) from syngeneic recipients, PBLs taken from tolerant recipients 100 days after transplantation were able to suppress the in vitro proliferation of allogeneic PBLs and to prolong the survival of second syngeneic recipients. We also compared messenger RNA profiles in PBLs from tolerant recipients with those from syngeneic recipients using a DNA microarray with probe sets corresponding to more than 8000 rat genes. There were 96 up-regulated and 103 down-regulated genes in the tolerant recipients. In the up-regulated group, there were 76 known genes and 20 expressed sequence tags (ESTs). In the down-regulated groups, there were 87 known genes and 16 ESTs. Our data indicated that FK506 treatment induced tolerance and expansion of regulatory cells and the DNA microarray technology was useful for this application and provided many informative insights into the mechanism of lymphocyte regulation.
Collapse
Affiliation(s)
- Masayuki Fujino
- Department of Innovative Surgery, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Tsai MK, Ho HN, Chien HF, Ou-Yang P, Lee CJ, Lee PH. The role of B7 ligands (CD80 and CD86) in CD152-mediated allograft tolerance: a crosscheck hypothesis. Transplantation 2004; 77:48-54. [PMID: 14724434 DOI: 10.1097/01.tp.0000107286.21985.ef] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The regulatory mechanism by which the B7 ligands (CD80 and CD86) direct the CD28/CD152 costimulatory pathways is unclear. This study investigated the role of CD80 and CD86 in a CD152-mediated allograft tolerance model. METHODS A low-responding cardiac transplant model (BALB/c-->B10.A) with possible long-term acceptance was used. Immunocytochemical and flow cytometric analyses of the graft-infiltrating cells were conducted to characterize this transplant model. The influence of anti-CD80 and anti-CD86 treatments on the proliferation and interleukin (IL)-2 productions of the tolerated splenocytes (SC) was analyzed. The role of CD80 and CD86 in the induction and maintenance of the graft acceptance in this transplant model were also tested. RESULTS B10.A mice could accept the BALA/c cardiac allografts (11/22), and an anti-CD152 antibody blocked the graft acceptance (10/10). Immunocytochemical and flow cytometric analyses showed that CD152+ cells were predominant among the CD4+ cells infiltrating the 100-day grafts of the B10.A recipients (B10.A-100). Either anti-CD80 or anti-CD86 treatment significantly enhanced polyclonal proliferation and IL-2 production of the B10.A-100 SC. Blockade of either CD80 or CD86 prohibited the tolerance transmitted by adoptive transfer, and anti-CD80 or anti-CD86 plus skin grafting undermined the established allograft tolerance. CONCLUSIONS Both CD80 and CD86 were essential for the induction and maintenance of the CD152-mediated allograft tolerance.
Collapse
Affiliation(s)
- Meng-Kun Tsai
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
99
|
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
|
100
|
Hori S, Takahashi T, Sakaguchi S. Control of autoimmunity by naturally arising regulatory CD4+ T cells. Adv Immunol 2004; 81:331-71. [PMID: 14711059 DOI: 10.1016/s0065-2776(03)81008-8] [Citation(s) in RCA: 218] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Naturally acquired immunological self-tolerance is not entirely accounted for by clonal deletion, anergy, and ignorance. It is now well established that the T cell-repertoire of healthy individuals harbors self-reactive lymphocytes with a potential to cause autoimmune disease and these lymphocytes are under dominant control by a unique subpopulation of CD4+ T cells now called regulatory T cells. Efforts to delineate these Treg cells naturally present in normal individuals have revealed that they are enriched in the CD25+ CD4+ population. The identification of the CD25 molecule as a useful marker for naturally arising CD4+ regulatory T cells has made it possible to investigate many key aspects of their immunobiology, including their antigen specificities and the cellular/molecular pathways involved in their development and their mechanisms of action. Furthermore, reduction or dysfunction of the CD25+ CD4+ regulatory T cell population can be responsible for certain autoimmune diseases in humans.
Collapse
Affiliation(s)
- Shohei Hori
- Laboratory of Immunopathology, Research Center for Allergy and Immunology, The Institute for Physical and Chemical Research (RIKEN), Yokohama 230-0045, Japan
| | | | | |
Collapse
|