101
|
Goodman WA, Cooper KD, McCormick TS. Regulation generation: the suppressive functions of human regulatory T cells. Crit Rev Immunol 2012; 32:65-79. [PMID: 22428855 PMCID: PMC3413266 DOI: 10.1615/critrevimmunol.v32.i1.40] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Proper regulation of immune homeostasis is necessary to limit inflammation and prevent autoimmune and chronic inflammatory diseases. Many autoimmune diseases, such as psoriasis, are driven by vicious cycles of activated T cells that are unable to be suppressed by regulatory T cells. Effective suppression of auto-reactive T cells by regulatory T cells (Treg) is critical for the prevention of spontaneous autoimmune disease. Psoriatic Treg cells have been observed to a defect in their capacity to regulate, which clearly contributes to psoriasis pathogenesis. A challenge for translational research is the development of novel therapeutic interventions for autoimmune diseases that will result in durable remissions. Understanding the mechanism(s) of dysregulated T cell responses in autoimmune disease will allow for the development of future therapeutic strategies that may be employed to specifically target pathogenic, proinflammatory cells. Several reports have demonstrated a pathogenic role for Thl and Thl7 cells in psoriasis as well as other autoimmune diseases. Similarly, several laboratories have independently demonstrated functional defects in regulatory T cells isolated from patients with numerous divergent autoimmune diseases. One primary challenge of research in autoimmune diseases is therefore to restore the balance between chronic T cell activation and impairment of Treg suppressor mechanisms. To this end, it is critical to develop an understanding of the many suppressive mechanisms employed by Treg cells in hopes of developing more targeted therapeutic strategies for Treg-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Wendy A Goodman
- Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA.
| | | | | |
Collapse
|
102
|
Nayak S, Sarkar D, Perrin GQ, Moghimi B, Hoffman BE, Zhou S, Byrne BJ, Herzog RW. Prevention and Reversal of Antibody Responses Against Factor IX in Gene Therapy for Hemophilia B. Front Microbiol 2011; 2:244. [PMID: 22279442 PMCID: PMC3260742 DOI: 10.3389/fmicb.2011.00244] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 11/22/2011] [Indexed: 11/13/2022] Open
Abstract
Intramuscular (IM) administration of an adeno-associated viral (AAV) vector represents a simple and safe method of gene transfer for treatment of the X-linked bleeding disorder hemophilia B (factor IX, F.IX, deficiency). However, the approach is hampered by an increased risk of immune responses against F.IX. Previously, we demonstrated that the drug cocktail of immune suppressants rapamycin, IL-10, and a specific peptide (encoding a dominant CD4+ T cell epitope) caused an induction of regulatory T cells (Treg) with a concomitant apoptosis of antigen-specific effector T cells (Nayak et al., 2009). This protocol was effective in preventing inhibitory antibody formation against human F.IX (hF.IX) in muscle gene transfer to C3H/HeJ hemophilia B mice (with targeted F9 gene deletion). Here, we show that this protocol can also be used to reverse inhibitor formation. IM injection of AAV1–hF.IX vector resulted in inhibitors of on average 8–10 BU within 1 month. Subsequent treatment with the tolerogenic cocktail accomplished a rapid reduction of hF.IX-specific antibodies to <2 BU, which lasted for >4.5 months. Systemic hF.IX expression increased from undetectable to >200 ng/ml, and coagulation times improved. In addition, we developed an alternative prophylactic protocol against inhibitor formation that did not require knowledge of T cell epitopes, consisting of daily oral administration of rapamycin for 1-month combined with frequent, low-dose intravenous injection of hF.IX protein. Experiments in T cell receptor transgenic mice showed that the route and dosing schedule of drug administration substantially affected Treg induction. When combined with intravenous antigen administration, oral delivery of rapamycin had to be performed daily in order to induce Treg, which were suppressive and phenotypically comparable to natural Treg.
Collapse
Affiliation(s)
- Sushrusha Nayak
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida Gainesville, FL, USA
| | | | | | | | | | | | | | | |
Collapse
|
103
|
Yolcu ES, Zhao H, Bandura-Morgan L, Lacelle C, Woodward KB, Askenasy N, Shirwan H. Pancreatic islets engineered with SA-FasL protein establish robust localized tolerance by inducing regulatory T cells in mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:5901-9. [PMID: 22068235 PMCID: PMC3232043 DOI: 10.4049/jimmunol.1003266] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allogeneic islet transplantation is an important therapeutic approach for the treatment of type 1 diabetes. Clinical application of this approach, however, is severely curtailed by allograft rejection primarily initiated by pathogenic effector T cells regardless of chronic use of immunosuppression. Given the role of Fas-mediated signaling in regulating effector T cell responses, we tested if pancreatic islets can be engineered ex vivo to display on their surface an apoptotic form of Fas ligand protein chimeric with streptavidin (SA-FasL) and whether such engineered islets induce tolerance in allogeneic hosts. Islets were modified with biotin following efficient engineering with SA-FasL protein that persisted on the surface of islets for >1 wk in vitro. SA-FasL-engineered islet grafts established euglycemia in chemically diabetic syngeneic mice indefinitely, demonstrating functionality and lack of acute toxicity. Most importantly, the transplantation of SA-FasL-engineered BALB/c islet grafts in conjunction with a short course of rapamycin treatment resulted in robust localized tolerance in 100% of C57BL/6 recipients. Tolerance was initiated and maintained by CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells, as their depletion early during tolerance induction or late after established tolerance resulted in prompt graft rejection. Furthermore, Treg cells sorted from graft-draining lymph nodes, but not spleen, of long-term graft recipients prevented the rejection of unmodified allogeneic islets in an adoptive transfer model, further confirming the Treg role in established tolerance. Engineering islets ex vivo in a rapid and efficient manner to display on their surface immunomodulatory proteins represents a novel, safe, and clinically applicable approach with important implications for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Esma S Yolcu
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, KY 40202
| | - Hong Zhao
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, KY 40202
| | - Laura Bandura-Morgan
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, KY 40202
| | - Chantale Lacelle
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, KY 40202
| | - Kyle B Woodward
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, KY 40202
| | - Nadir Askenasy
- Frankel Laboratory of Experimental Bone Marrow Transplantation, Department of Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Israel
| | - Haval Shirwan
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, KY 40202
| |
Collapse
|
104
|
Duarte J, Caridade M, Graca L. CD4-blockade can induce protection from peanut-induced anaphylaxis. Front Immunol 2011; 2:56. [PMID: 22566846 PMCID: PMC3341953 DOI: 10.3389/fimmu.2011.00056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 10/04/2011] [Indexed: 11/13/2022] Open
Abstract
Monoclonal antibodies (mAb) have been shown effective in inducing immune tolerance in a range of animal models of autoimmunity, allergy, and transplantation. We investigated whether CD4-blockade, effective in inducing transplantation tolerance, could prevent systemic immune responses leading to anaphylaxis. We found that treatment with a non-depleting anti-CD4 mAb could prevent peanut-induced anaphylaxis following subsequent systemic exposure to crude peanut extract (CPE). Furthermore, the effect of CD4-blockade did not interfere with overall immune competence, as anti-CD4 treated mice remained fully competent to respond to unrelated antigens. Protection from anaphylaxis correlated with increased frequency of Foxp3⁺ regulatory T cells (Treg), and was abrogated following Treg depletion. Taken together our data suggest that activation of T cells by CPE in presence of CD4-blockade leads to Treg expansion that can prevent peanut-induced anaphylaxis.
Collapse
Affiliation(s)
- Joana Duarte
- Instituto de Medicina Molecular, University of Lisbon Lisbon, Portugal
| | | | | |
Collapse
|
105
|
Fan H, Cao P, Game DS, Dazzi F, Liu Z, Jiang S. Regulatory T cell therapy for the induction of clinical organ transplantation tolerance. Semin Immunol 2011; 23:453-61. [PMID: 21920772 DOI: 10.1016/j.smim.2011.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The pursuit of transplantation tolerance is the holygrail in clinical organ transplantation. It has been established that regulatory T cells (Tregs) can confer donor-specific tolerance in mouse models of transplantation. However, this is crucially dependent on the strain combination, the organ transplanted and most importantly, the ratio of Tregs to alloreactive effector T cells. The ex vivo expansion of Tregs is one solution to increase the number of alloantigen specific cells capable of suppressing the alloresponse. Indeed, ex vivo expanded, alloantigen specific murine Tregs are shown to preferentially migrate to, and proliferate in, the graft and draining lymph node. In human transplantation it has been proposed that depletion of the majority of direct pathway alloreactive T cells will be required to tip the balance in favour of regulation. Ex vivo expansion of alloantigen specific, indirect pathway human Tregs, which can cross regulate the residual direct pathway has been established. Rapid expansion of these cells is possible, whilst they retain antigen specificity, suppressive properties and favourable homing markers. Furthermore, considerable progress has been made to define which immunosuppressive drugs favour the expansion and function of Tregs. Currently a series of clinical trials of adoptive Treg therapy in combination with depletion of alloreactive T cells and short term immunosuppression are underway for human transplantation with the aim of minimizing immunosuppressive drugs and completely withdrawal.
Collapse
Affiliation(s)
- Huimin Fan
- Shanghai East Hospital of Tongji University, Shanghai 200120, China
| | | | | | | | | | | |
Collapse
|
106
|
Dodd-o JM, Lendermon EA, Miller HL, Zhong Q, John ER, Jungraithmayr WM, D'Alessio FR, McDyer JF. CD154 blockade abrogates allospecific responses and enhances CD4(+) regulatory T-cells in mouse orthotopic lung transplant. Am J Transplant 2011; 11:1815-24. [PMID: 21827610 PMCID: PMC3827913 DOI: 10.1111/j.1600-6143.2011.03623.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Acute cellular rejection (ACR) is a common and important clinical complication following lung transplantation. While there is a clinical need for the development of novel therapies to prevent ACR, the regulation of allospecific effector T-cells in this process remains incompletely understood. Using the MHC-mismatched mouse orthotopic lung transplant model, we investigated the short-term role of anti-CD154 mAb therapy alone on allograft pathology and alloimmune T-cell effector responses. Untreated C57BL/6 recipients of BALB/c left lung allografts had high-grade rejection and diminished CD4(+) : CD8(+) graft ratios, marked by predominantly CD8(+) >CD4(+) IFN-γ(+) allospecific effector responses at day 10, compared to isograft controls. Anti-CD154 mAb therapy strikingly abrogated both CD8(+) and CD4(+) alloeffector responses and significantly increased lung allograft CD4(+) : CD8(+) ratios. Examination of graft CD4(+) T-cells revealed significantly increased frequencies of CD4(+) CD25(+) Foxp3(+) regulatory T-cells in the lung allografts of anti-CD154-treated mice and was associated with significant attenuation of ACR compared to untreated controls. Together, these data show that CD154/CD40 costimulation blockade alone is sufficient to abrogate allospecific effector T-cell responses and significantly shifts the lung allograft toward an environment predominated by CD4(+) T regulatory cells in association with an attenuation of ACR.
Collapse
Affiliation(s)
- J M Dodd-o
- Department of Anesthesiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
107
|
Cippà PE, Kraus AK, Edenhofer I, Segerer S, Chen J, Hausmann M, Liu Y, Guimezanes A, Bardwell PD, Wüthrich RP, Fehr T. The BH3-mimetic ABT-737 inhibits allogeneic immune responses. Transpl Int 2011; 24:722-32. [DOI: 10.1111/j.1432-2277.2011.01272.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
108
|
Wood KJ, Bushell A, Jones ND. Immunologic unresponsiveness to alloantigen in vivo: a role for regulatory T cells. Immunol Rev 2011; 241:119-32. [PMID: 21488894 DOI: 10.1111/j.1600-065x.2011.01013.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Exposure to alloantigen in vivo or in vitro induces alloantigen reactive regulatory T cells that can control transplant rejection. The mechanisms that underpin the activity of alloantigen reactive regulatory T cells in vivo are common with those of regulatory T cells that prevent autoimmunity. The identification and characterization of regulatory T cells that control rejection and contribute to the induction of immunologic unresponsiveness to alloantigens in vivo has opened up exciting opportunities for new therapies in transplantation. Findings from laboratory studies are informing the design of clinical protocols using regulatory T cells as a cellular therapy.
Collapse
Affiliation(s)
- Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| | | | | |
Collapse
|
109
|
Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/ B7 family. Immunol Rev 2011; 241:180-205. [PMID: 21488898 PMCID: PMC3077803 DOI: 10.1111/j.1600-065x.2011.01011.x] [Citation(s) in RCA: 306] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Positive and negative costimulation by members of the CD28 family is critical for the development of productive immune responses against foreign pathogens and their proper termination to prevent inflammation-induced tissue damage. In addition, costimulatory signals are critical for the establishment and maintenance of peripheral tolerance. This paradigm has been established in many animal models and has led to the development of immunotherapies targeting costimulation pathways for the treatment of cancer, autoimmune disease, and allograft rejection. During the last decade, the complexity of the biology of costimulatory pathways has greatly increased due to the realization that costimulation does not affect only effector T cells but also influences regulatory T cells and antigen-presenting cells. Thus, costimulation controls T-cell tolerance through both intrinsic and extrinsic pathways. In this review, we discuss the influence of costimulation on intrinsic and extrinsic pathways of peripheral tolerance, with emphasis on members of the CD28 family, CD28, cytotoxic T-lymphocyte antigen-4 (CTLA-4), and programmed death-1 (PD-1), as well as the downstream cytokine interleukin-1 (IL-2).
Collapse
Affiliation(s)
- Hélène Bour-Jordan
- UCSF Diabetes Center, University of California at San Francisco, San Francisco, CA 94143-0400, USA
| | | | | | | | | | | |
Collapse
|
110
|
Abstract
Immune responses during infection, injury, and cancer proceed in the presence of tissue injury and cell death. Consequently, the system must deal with its own dead cells while it determines the appropriate response to the invader. As apoptotic cells are known to induce immune tolerance and necrotic cells can be potent stimulators of immunity, this decision becomes more complex. The key to understanding the immunologic choices made during cell death is to examine the mechanisms of tolerance induction by dying cells and then relate them to the mechanisms of immunity. Ideally, immunogenic cell death should be directed toward tumor cells and infected cells, whereas tolerogenic cell death should be associated with preventing unwanted immune responses to self. In this review, we discuss how the decision is made by focusing on the biochemical process of cell death and how its key components can influence both tolerance and immunity.
Collapse
Affiliation(s)
- Thomas A Ferguson
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
111
|
Bozulic LD, Wen Y, Xu H, Ildstad ST. Evidence that FoxP3+ regulatory T cells may play a role in promoting long-term acceptance of composite tissue allotransplants. Transplantation 2011; 91:908-15. [PMID: 21304439 PMCID: PMC3592205 DOI: 10.1097/tp.0b013e31820fafb4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND FoxP3/CD4/CD25 regulatory T cells (Treg) play an important role in maintaining peripheral tolerance and are potent suppressors of T-cell activation. In this study, we evaluated the role of Treg in peripheral tolerance to composite tissue allografts (CTA). METHODS Mixed allogeneic chimeric rats were prepared by preconditioning recipients with anti-αβ-T-cell receptor monoclonal antibody followed by total body irradiation. Animals received T-cell-depleted August Copenhagen Irish bone marrow cells followed by antilymphocyte serum and FK-506. A modified osteomyocutaneous hindlimb flap composed of bone and all limb tissue components was placed in animals with chimerism greater than or equal to 1% on day 28. Recipients with CTA surviving more than or equal to 6 months were evaluated for Treg. Skin samples from tolerant long-term allogeneic transplanted, syngeneic transplanted, rejected, and naïve animals were immunostained with fluorochrome-conjugated anti-FoxP3 and anti-CD4 monoclonal antibody and visualized under a laser confocal microscope. RESULTS Significant CD4/FoxP3 Treg infiltrates were observed in tolerant donor-allograft skin samples. No graft infiltrating FoxP3 cells were observed in rejector, naïve, or skin from syngeneic CTA. In parallel experiments, mixed leukocyte reaction assays were performed to investigate the suppressor function of Treg cells. Splenocytes from tolerant, rejected, and naïve rats were sorted by flow cytometry for CD4/CD25 T cells. Treg demonstrated similar suppressive levels between the three groups. CONCLUSIONS These data suggest that Treg may play an important role in maintenance of tolerance and promoting graft acceptance in long-term CTA acceptors and may explain the favorable outcomes observed in clinical CTA recipients.
Collapse
Affiliation(s)
- Larry D. Bozulic
- Regenerex, LLC, 333 East Main Street, Suite 400, Louisville, KY 40202
| | - Yujie Wen
- Institute for Cellular Therapeutics, University of Louisville, 570 S. Preston Street, Suite 404, Louisville, KY 40202-1760
| | - Hong Xu
- Institute for Cellular Therapeutics, University of Louisville, 570 S. Preston Street, Suite 404, Louisville, KY 40202-1760
| | - Suzanne T. Ildstad
- Institute for Cellular Therapeutics, University of Louisville, 570 S. Preston Street, Suite 404, Louisville, KY 40202-1760
| |
Collapse
|
112
|
von Rossum A, Krall R, Escalante NK, Choy JC. Inflammatory cytokines determine the susceptibility of human CD8 T cells to Fas-mediated activation-induced cell death through modulation of FasL and c-FLIP(S) expression. J Biol Chem 2011; 286:21137-44. [PMID: 21518761 DOI: 10.1074/jbc.m110.197657] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The nature of inflammatory signals determines the outcome of T cell responses. However, little is known about how inflammatory cytokines provided to human CD8 T cells during activation affects their susceptibility to post-activation cell death. We have examined and compared the effects of the inflammatory cytokine IL-12, as well as the combination of IL-1, IL-6, and IL-23 (IL-1/6/23) on the susceptibility of primary human CD8 T cells to post-activation cell death. Human CD8 T cells activated in the presence of IL-1/6/23 underwent significantly less cell death after activation as compared with those activated in IL-12. This was due to reduced susceptibility to Fas-mediated activation-induced cell death (AICD). Mechanistically, the reduced level of cell death in CD8 T cells activated in IL-1/6/23 was a result of a low level of FasL expression and high level of c-FLIP(S) expression. When the effect of IL-1, IL-6, and IL-23 individually was examined, IL-1 or IL-6 alone was sufficient to inhibit CD8 T cell death that occurs after activation in IL-12. IL-1, but not IL-6, inhibited expression of FasL, whereas IL-6, but not IL-1, increased c-FLIP(S) expression. Our findings show that the presence of IL-1 and/or IL-6 during activation of human CD8 T cells attenuates Fas-mediated AICD, whereas IL-12 increases the susceptibility of activated CD8 T cells to this form of cell death.
Collapse
Affiliation(s)
- Anna von Rossum
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A1S6
| | | | | | | |
Collapse
|
113
|
El Essawy B, Putheti P, Gao W, Strom TB. Rapamycin generates graft-homing murine suppressor CD8(+) T cells that confer donor-specific graft protection. Cell Transplant 2011; 20:1759-69. [PMID: 21439133 DOI: 10.3727/096368911x566244] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
It has been reported that rapamycin (RPM) can induce de novo conversion of the conventional CD4(+)Foxp3(-) T cells into CD4(+)Foxp3(+) regulatory T cells (iTregs) in transplantation setting. It is not clear whether RPM can similarly generate suppressor CD8(+) T cells to facilitate graft acceptance. In this study, we investigated the ability of short-term RPM treatment in promoting long-term acceptance (LTA) of MHC-mismatched skin allografts by generating a CD8(+) suppressor T-cell population. We found that CD4 knockout (KO) mice (in C57BL/6 background, H-2(b)) can promptly reject DBA/2 (H-2(d)) skin allografts with mean survival time (MST) being 13 days (p < 0.01). However, a short course RPM treatment in these animals induced LTA with graft MST longer than 100 days. Adoptive transfer of CD8(+) T cells from LTA group into recombination-activating gene 1 (Rag-1)-deficient mice provided donor-specific protection of DBA/2 skin grafts against cotransferred conventional CD8(+) T cells. Functionally active immunoregulatory CD8(+) T cells also resided in donor skin allografts. Eighteen percent of CD8(+) suppressor T cells expressed CD28 as measured by flow cytometry, and produced reduced levels of IFN-γ, IL-2, and IL-10 in comparison to CD8(+) effector T cells as measured by ELISA. It is unlikely that CD8(+) suppressor T cells mediated graft protection via IL-10, as IL-10/Fc fusion protein impaired RPM-induced LTA in CD4 KO mice. Our data supported the notion that RPM-induced suppressor CD8(+) T cells home to the allograft and exert donor-specific graft protection.
Collapse
Affiliation(s)
- Basset El Essawy
- Department of Medicine, Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
114
|
Abstract
Every liver transplant (LT) center has had patients who either self-discontinue immunosuppressive (IS) therapy or are deliberately withdrawn due to a research protocol or clinical concern (ie, lymphoproliferative disorder [LPD], overwhelming infection). This is understandable because maintenance IS therapy, particularly calcineurin inhibitors (CNI), is associated with significant cost, side effects, and considerable long-term morbidity and mortality. Detrimental effects of IS therapy include increased risk of cardiovascular disease, metabolic syndrome, bone loss, opportunistic and community-acquired infections, and malignancy. In fact, LT recipients have among the highest rates of chronic kidney disease and associated mortality among all nonkidney solid organ recipients. This mortality is only ameliorated by undergoing a curative kidney transplant, usurping costs and valuable organ resources. The search for improved treatment algorithms includes trial and error CNI dose minimization, the use of alternative IS agents (antimetabolites, mammalian target of rapamycin [mTOR] inhibitors), or even complete CNI withdrawal. Yet those who are successful in achieving such operational tolerance (no immunosuppression and normal allograft function) are considered lucky. The vast majority of recipients will fail this approach, develop acute rejection or immune-mediated hepatitis, and require resumption of IS therapy. As such, withdrawal of IS following LT is not standard-of-care, leaving clinicians to currently maintain transplant patients on IS therapy for life. Nonetheless, the long-term complications of all IS therapies highlight the need for strategies to promote immunologic or operational tolerance. Clinically applicable biomarker assays signifying the potential for tolerance as well as tolerogenic IS conditioning are invariably needed if systematic, controlled rather than "hit or miss" approaches to withdrawal are considered. This review will provide an overview of the basic mechanisms of tolerance, particularly in relation to LT, data from previous IS withdrawal protocols and biomarker studies in tolerant recipients, and a discussion on the prospect of increasing the clinical feasibility and success of withdrawal.
Collapse
Affiliation(s)
- Josh Levitsky
- Division of Hepatology and Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
115
|
Francis RS, Feng G, Tha-In T, Lyons IS, Wood KJ, Bushell A. Induction of transplantation tolerance converts potential effector T cells into graft-protective regulatory T cells. Eur J Immunol 2011; 41:726-38. [PMID: 21243638 PMCID: PMC3175037 DOI: 10.1002/eji.201040509] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 09/28/2010] [Accepted: 11/29/2010] [Indexed: 12/14/2022]
Abstract
Naturally occurring FOXP3(+) CD4(+) Treg have a crucial role in self-tolerance. The ability to generate similar populations against alloantigens offers the possibility of preventing transplant rejection without indefinite global immunosuppression. Exposure of mice to donor alloantigens combined with anti-CD4 antibody induces operational tolerance to cardiac allografts, and generates Treg that prevent skin and islet allograft rejection in adoptive transfer models. If protocols that generate Treg in vivo are to be developed in the clinical setting it will be important to know the origin of the Treg population and the mechanisms responsible for their generation. In this study, we demonstrate that graft-protective Treg arise in vivo both from naturally occurring FOXP3(+) CD4(+) Treg and from non-regulatory FOXP3(-) CD4(+) cells. Importantly, tolerance induction also inhibits CD4(+) effector cell priming and T cells from tolerant mice have impaired effector function in vitro. Thus, adaptive tolerance induction shapes the immune response to alloantigen by converting potential effector cells into graft-protective Treg and by expanding alloreactive naturally occurring Treg. In relation to clinical tolerance induction, the data indicate that while the generation of alloreactive Treg may be critical for long-term allograft survival without chronic immunosuppression, successful protocols will also require strategies that target potential effector cells.
Collapse
Affiliation(s)
- Ross S Francis
- Transplant Research Immunology Group, Nuffield Department of Surgery, University of Oxford John Radcliffe Hospital, Oxford, UK
| | | | | | | | | | | |
Collapse
|
116
|
Peters JH, Koenen HJPM, Hilbrands LB, Joosten I. Immunotherapy with regulatory T cells in transplantation. Immunotherapy 2011; 1:855-71. [PMID: 20636028 DOI: 10.2217/imt.09.45] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Regulatory T cell (Treg)-based immunotherapy is of great interest to induce tolerance in clinical transplantation settings. In fact, the first clinical trials of Treg infusion after stem cell transplantation have recently begun. However, many important issues regarding human Treg immunotherapy are still to be resolved. In this review, we provide a short update on Tregs and elaborate on various strategies for Treg-based immunotherapy. First, infusion of ex vivo-selected naturally occurring Tregs is addressed, with emphasis on Treg isolation, expansion, antigen specificity, homing and stability. Next, the potential of ex vivo-induced Treg transfusion strategies is discussed. Finally, therapies aimed at in vivo increase of Treg numbers or function are addressed. In addition, we summarize the current knowledge on effects of immunosuppressive drugs on Tregs. In the following years, we expect exciting new data regarding the clinical application of Treg immunotherapy in transplantation to be released.
Collapse
Affiliation(s)
- Jorieke H Peters
- Department of Blood Transfusion & Transplantation Immunology, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
117
|
Muller YD, Seebach JD, Bühler LH, Pascual M, Golshayan D. Transplantation tolerance: Clinical potential of regulatory T cells. SELF/NONSELF 2011; 2:26-34. [PMID: 21776332 PMCID: PMC3136901 DOI: 10.4161/self.2.1.15422] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 03/09/2011] [Indexed: 01/12/2023]
Abstract
The major challenge in transplantation medicine remains long-term allograft acceptance, with preserved allograft function under minimal chronic immunosuppression. To safely achieve the goal of sustained donor-specific T and B cell non-responsiveness, research efforts are now focusing on therapies based on cell subsets with regulatory properties. In particular the transfusion of human regulatory T cells (Treg) is currently being evaluated in phase I/II clinical trials for the treatment of graft versus host disease following hematopoietic stem cell transplantation, and is also under consideration for solid organ transplantation. The purpose of this review is to recapitulate current knowledge on naturally occurring as well as induced human Treg, with emphasis on their specific phenotype, suppressive function and how these cells can be manipulated in vitro and/or in vivo for therapeutic purposes in transplantation medicine. We highlight the potential but also possible limitations of Treg-based strategies to promote long-term allograft survival. It is evident that the bench-to-beside translation of these protocols still requires further understanding of Treg biology. Nevertheless, current data already suggest that Treg therapy alone will not be sufficient and needs to be combined with other immunomodulatory approaches in order to induce allograft tolerance.
Collapse
Affiliation(s)
- Yannick D Muller
- Surgical Research Unit; Department of Surgery; University Hospital Geneva
- Service of Clinical Immunology and Allergology; Department of Internal Medicine; University Hospital Geneva
| | - Jörg D Seebach
- Service of Clinical Immunology and Allergology; Department of Internal Medicine; University Hospital Geneva
| | - Leo H Bühler
- Surgical Research Unit; Department of Surgery; University Hospital Geneva
| | - Manuel Pascual
- Transplantation Centre and Transplantation Immunopathology Laboratory; Departments of Medicine and Surgery; Centre Hospitalier Universitaire Vaudois and University of Lausanne; Switzerland
| | - Dela Golshayan
- Transplantation Centre and Transplantation Immunopathology Laboratory; Departments of Medicine and Surgery; Centre Hospitalier Universitaire Vaudois and University of Lausanne; Switzerland
| |
Collapse
|
118
|
SÁNCHEZ–FUEYO ALBERTO, STROM TERRYB. Immunologic basis of graft rejection and tolerance following transplantation of liver or other solid organs. Gastroenterology 2011; 140:51-64. [PMID: 21073873 PMCID: PMC3866688 DOI: 10.1053/j.gastro.2010.10.059] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/24/2010] [Accepted: 10/26/2010] [Indexed: 12/13/2022]
Abstract
Transplantation of organs between genetically different individuals of the same species causes a T cell-mediated immune response that, if left unchecked, results in rejection and graft destruction. The potency of the alloimmune response is determined by the antigenic disparity that usually exists between donors and recipients and by intragraft expression of proinflammatory cytokines in the early period after transplantation. Studies in animal models have identified many molecules that, when targeted, inhibit T-cell activation. In addition, some of these studies have shown that certain immunologic interventions induce transplantation tolerance, a state in which the allograft is specifically accepted without the need for chronic immunosuppression. Tolerance is an important aspect of liver transplantation, because livers have a unique microenvironment that promotes tolerance rather than immunity. In contrast to the progress achieved in inducing tolerance in animal models, patients who receive transplanted organs still require nonspecific immunosuppressant drugs. The development of calcineurin inhibitors has reduced the acute rejection rate and improved short-term, but not long-term, graft survival. However, long-term use of immunosuppressive drugs leads to nephrotoxicity and metabolic disorders, as well as manifestations of overimmunosuppression such as opportunistic infections and cancers. The status of pharmacologic immunosuppression in the clinic is therefore not ideal. We review recently developed therapeutic strategies to promote tolerance to transplanted livers and other organs and diagnostic tools that might be used to identify patients most likely to accept or reject allografts.
Collapse
Affiliation(s)
- ALBERTO SÁNCHEZ–FUEYO
- Liver Transplant Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain
| | - TERRY B. STROM
- Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
119
|
Mechanism Analysis of Long-Term Graft Survival by Monocarboxylate Transporter-1 Inhibition. Transplantation 2010; 90:1299-306. [DOI: 10.1097/tp.0b013e3181ff8818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
120
|
Verbinnen B, Van Gool SW, Ceuppens JL. Blocking costimulatory pathways: prospects for inducing transplantation tolerance. Immunotherapy 2010; 2:497-509. [PMID: 20636004 DOI: 10.2217/imt.10.31] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tolerance induction to alloantigens is a major challenge in transplant immunology. Whereas conventional immunosuppression inhibits the immune system in a nonspecific way, thereby also undermining an appropriate immune response towards potentially harmful infectious organisms, tolerance in a transplantation setting is restricted to alloantigens, while protective immunity is preserved. Moreover, tolerance implies an immunological status that is preserved after withdrawal of the tolerance-inducing therapy. Among the most promising strategies to induce immunological tolerance are costimulation blockade and establishment of mixed chimerism. Despite significant advances, we still know little about the mechanisms responsible for such tolerance. In this article, we discuss tolerance induction to transplantation antigens by costimulation blockade.
Collapse
Affiliation(s)
- Bert Verbinnen
- University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | | | | |
Collapse
|
121
|
Anam K, Amare MF, Zins SR, Davis TA. Infusion of Lin- bone marrow cells results in multilineage macrochimerism and skin allograft tolerance in minimally conditioned recipient mice. Transpl Immunol 2010; 24:69-75. [PMID: 20950686 DOI: 10.1016/j.trim.2010.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 10/06/2010] [Indexed: 01/08/2023]
Abstract
Donor-specific immunological tolerance using high doses of donor bone marrow cells (BMC) has been demonstrated in mixed chimerism-based tolerance induction protocols; however, the development of graft versus host disease (GVHD) remains a risk. In the present study, we demonstrate that the infusion of low numbers of donor Lin(-) bone marrow cells (Lin(-) BMC) 7 days post allograft transplantation facilitates high level macrochimerism induction and graft tolerance. Full-thickness BALB/c skin allografts were transplanted onto C57BL/6 mice. Mice were treated with anti-CD4 and anti-CD8 mAbs on day 0, +2, +5, +7 and +14 along with low dose busulfan on day +5. A low dose of highly purified Lin(-) BMC from BALB/c donor mice was infused on day +7. Chimerism and clonal cell deletion were evaluated using flow cytometry. Donor-specific tolerance was tested by donor and third-party skin grafting and mixed leukocyte reaction (MLR). Lin(-) BMC infusion with minimal immunosuppression led to stable, mixed, multilineage macrochimerism and long-term allograft survival (>300 days). Mixed donor-recipient macrochimerism was observed. Donor-reactive T cells were clonally deleted and a 130% increase in CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) was observed in the spleen. Tolerant mice subsequently accepted second donor, but not third-party (C3H), skin grafts and recipient splenocytes failed to react with allogeneic donor cells indicating donor-specific immunological tolerance was achieved. We conclude that the infusion of donor Lin(-) BMC without cytoreductive recipient conditioning can induce indefinite survival of skin allografts via mechanisms involving the establishment of a multilineage macrochimeric state principally through clonal deletion of alloreactive T cells and peripherally induced CD4(+)Foxp3(+) Tregs.
Collapse
Affiliation(s)
- Khairul Anam
- Regenerative Medicine Department, Operational and Undersea Medicine Directorate at the Naval Medical Research Center, Silver Spring, MD 20910-7500, USA
| | | | | | | |
Collapse
|
122
|
GVHD after haploidentical transplantation: a novel, MHC-defined rhesus macaque model identifies CD28- CD8+ T cells as a reservoir of breakthrough T-cell proliferation during costimulation blockade and sirolimus-based immunosuppression. Blood 2010; 116:5403-18. [PMID: 20833977 DOI: 10.1182/blood-2010-06-289272] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We have developed a major histocompatibility complex-defined primate model of graft-versus-host disease (GVHD) and have determined the effect that CD28/CD40-directed costimulation blockade and sirolimus have on this disease. Severe GVHD developed after haploidentical transplantation without prophylaxis, characterized by rapid clinical decline and widespread T-cell infiltration and organ damage. Mechanistic analysis showed activation and possible counter-regulation, with rapid T-cell expansion and accumulation of CD8(+) and CD4(+) granzyme B(+) effector cells and FoxP3(pos)/CD27(high)/CD25(pos)/CD127(low) CD4(+) T cells. CD8(+) cells down-regulated CD127 and BCl-2 and up-regulated Ki-67, consistent with a highly activated, proliferative profile. A cytokine storm also occurred, with GVHD-specific secretion of interleukin-1 receptor antagonist (IL-1Ra), IL-18, and CCL4. Costimulation Blockade and Sirolimus (CoBS) resulted in striking protection against GVHD. At the 30-day primary endpoint, CoBS-treated recipients showed 100% survival compared with no survival in untreated recipients. CoBS treatment resulted in survival, increasing from 11.6 to 62 days (P < .01) with blunting of T-cell expansion and activation. Some CoBS-treated animals did eventually develop GVHD, with both clinical and histopathologic evidence of smoldering disease. The reservoir of CoBS-resistant breakthrough immune activation included secretion of interferon-γ, IL-2, monocyte chemotactic protein-1, and IL-12/IL-23 and proliferation of cytotoxic T-lymphocyte-associated antigen 4 immunoglobulin-resistant CD28(-) CD8(+) T cells, suggesting adjuvant treatments targeting this subpopulation will be needed for full disease control.
Collapse
|
123
|
|
124
|
Garcia MR, Ledgerwood L, Yang Y, Xu J, Lal G, Burrell B, Ma G, Hashimoto D, Li Y, Boros P, Grisotto M, van Rooijen N, Matesanz R, Tacke F, Ginhoux F, Ding Y, Chen SH, Randolph G, Merad M, Bromberg JS, Ochando JC. Monocytic suppressive cells mediate cardiovascular transplantation tolerance in mice. J Clin Invest 2010; 120:2486-96. [PMID: 20551515 PMCID: PMC2898596 DOI: 10.1172/jci41628] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 05/05/2010] [Indexed: 12/24/2022] Open
Abstract
One of the main unresolved questions in solid organ transplantation is how to establish indefinite graft survival that is free from long-term treatment with immunosuppressive drugs and chronic rejection (i.e., the establishment of tolerance). The failure to achieve this goal may be related to the difficulty in identifying the phenotype and function of the cell subsets that participate in the induction of tolerance. To address this issue, we investigated the suppressive roles of recipient myeloid cells that may be manipulated to induce tolerance to transplanted hearts in mice. Using depleting mAbs, clodronate-loaded liposomes, and transgenic mice specific for depletion of CD11c+, CD11b+, or CD115+ cells, we identified a tolerogenic role for CD11b+CD115+Gr1+ monocytes during the induction of tolerance by costimulatory blockade with CD40L-specific mAb. Early after transplantation, Gr1+ monocytes migrated from the bone marrow into the transplanted organ, where they prevented the initiation of adaptive immune responses that lead to allograft rejection and participated in the development of Tregs. Our results suggest that mobilization of bone marrow CD11b+CD115+Gr1+ monocytes under sterile inflammatory conditions mediates the induction of indefinite allograft survival. We propose that manipulating the common bone marrow monocyte progenitor could be a useful clinical therapeutic approach for inducing transplantation tolerance.
Collapse
Affiliation(s)
- Mercedes Rodriguez Garcia
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Levi Ledgerwood
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Yu Yang
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Jiangnan Xu
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Girdhari Lal
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Bryna Burrell
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Ge Ma
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Daigo Hashimoto
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Yansui Li
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Peter Boros
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Marcos Grisotto
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Nico van Rooijen
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Rafael Matesanz
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Frank Tacke
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Florent Ginhoux
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Yaozhong Ding
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Shu-Hsia Chen
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Gwendalyn Randolph
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Miriam Merad
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Jonathan S. Bromberg
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| | - Jordi C. Ochando
- Inmunología de Trasplantes, Centro Nacional de
Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
Department of Otolaryngology-Head and Neck Surgery, UC Davis Medical
Center, Sacramento, California, USA. Department of Gene and Cell
Medicine and Department of Surgery, Mount Sinai School of Medicine, New
York, New York, USA. Centro Universitário do
Maranhão UNICEUMA, Sao Luis, Brazil. Department of Molecular
Cell Biology, VUMC, Amsterdam, The Netherlands. Organización
Nacional de Trasplantes, Madrid, Spain. Medical Clinic III, University
Hospital Aachen, Aachen University (RWTH), Aachen, Germany. Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR),
Singapore
| |
Collapse
|
125
|
Wang T, Ahmed EB, Chen L, Xu J, Tao J, Wang CR, Alegre ML, Chong AS. Infection with the intracellular bacterium, Listeria monocytogenes, overrides established tolerance in a mouse cardiac allograft model. Am J Transplant 2010; 10:1524-33. [PMID: 20642679 PMCID: PMC4060596 DOI: 10.1111/j.1600-6143.2010.03066.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Infections and TLR signals at the time of transplantation have been shown to prevent the induction of tolerance, but their effect on allografts after tolerance has been established is unclear. We here report that infection with Listeria monocytogenes precipitated the loss of tolerance and the MyD88- and T cell-dependent rejection of accepted cardiac allografts in mice. This loss of tolerance was associated with increases in the numbers of graft-infiltrating macrophages and dendritic cells, as well as CD4(+)FoxP3(-) and CD8(+) T cells. Rejection was also associated with increased numbers of graft-infiltrating alloreactive as well as Listeria-reactive IFNgamma-producing T cells. Rejection of the established grafts required both IL-6 and IFNss, cytokines produced during acute Listeria infection. However, IL-6 and IFNss alone, even when present at higher concentrations than during Listeria infection, were insufficient to break tolerance, while the combination of IL-6 and IFNss was sufficient to break tolerance. These and in vitro observations that IL-6 but not IFNss enhanced T cell proliferation while IFNss but not IL-6 enhanced IFNgamma production support a hypothesis that these cytokines play nonredundant roles. In conclusion, these studies demonstrate that the proinflammatory effects of infections can induce the loss of tolerance and acute rejection of accepted allografts.
Collapse
Affiliation(s)
- Tongmin Wang
- The Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Emily B. Ahmed
- The Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Luqiu Chen
- Section of Rheumatology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Jing Xu
- The Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Jing Tao
- The Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Chyung-Ru Wang
- Department of Microbiology/Immunology, Northwestern University, Chicago, Illinois, USA
| | - Maria-Luisa Alegre
- Section of Rheumatology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Anita S. Chong
- The Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
126
|
Posadas A, Lisse J, Sarkar S. Abatacept in the treatment of rheumatoid arthritis. Expert Rev Clin Immunol 2010; 5:9-17. [PMID: 20476895 DOI: 10.1586/1744666x.5.1.9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory arthritis affecting 1% of the population. The immunologic dysfunction underlying this immune disorder is complex and intricate with the involvement of various immune cells as well as cytokines and surface molecules. While inhibition of TNF-alpha has changed the outlook of patients with this disorder, it regulates only one aspect of the inflammatory cascade associated with RA. This is corroborated by experience in the clinic, where a significant proportion of the patients do not have clinical benefit with such therapies. Furthermore, a number of patients experience blunting of the initial therapeutic benefits of TNF-alpha-targeted therapies. Thus, a different approach to regulate the immune dysfunction associated with RA is necessary. T cells are considered important in the pathogenesis of RA and abatacept, a fusion protein, was developed to abolish the activation of the T cell by blocking its interaction with the antigen-presenting cell. Abatacept has demonstrated promising clinical improvements in patients with RA. Although clinical experience with this new drug is limited and its mechanism of action remains to be understood, the data on the safety profile are reassuring.
Collapse
Affiliation(s)
- Augusto Posadas
- Section of Rheumatology, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | | | | |
Collapse
|
127
|
Fan Z, Spencer JA, Lu Y, Pitsillides CM, Singh G, Kim P, Yun SH, Toxavidis V, Strom TB, Lin CP, Koulmanda M. In vivo tracking of 'color-coded' effector, natural and induced regulatory T cells in the allograft response. Nat Med 2010; 16:718-22. [PMID: 20495571 PMCID: PMC2894571 DOI: 10.1038/nm.2155] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 01/01/2010] [Indexed: 12/11/2022]
Abstract
Here we present methods to longitudinally track islet allograft-infiltrating T cells in live mice by endoscopic confocal microscopy and to analyze circulating T cells by in vivo flow cytometry. We developed a new reporter mouse whose T cell subsets express distinct, 'color-coded' proteins enabling in vivo detection and identification of effector T cells (T(eff) cells) and discrimination between natural and induced regulatory T cells (nT(reg) and iT(reg) cells). Using these tools, we observed marked differences in the T cell response in recipients receiving tolerance-inducing therapy (CD154-specific monoclonal antibody plus rapamycin) compared to untreated controls. These results establish real-time cell tracking as a powerful means to probe the dynamic cellular interplay mediating immunologic rejection or transplant tolerance.
Collapse
Affiliation(s)
- Zhigang Fan
- Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Sharma KG, Radha R, Pao A, Amet N, Baden L, Jordan SC, Toyoda M. Mycophenolic acid and intravenous immunoglobulin exert an additive effect on cell proliferation and apoptosis in the mixed lymphocyte reaction. Transpl Immunol 2010; 23:117-20. [PMID: 20450974 DOI: 10.1016/j.trim.2010.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 04/20/2010] [Accepted: 04/26/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Intravenous immunoglobulin (IVIG) has known immunomodulatory effects in autoimmune diseases and transplantation and is commonly used in desensitization protocols and for treatment of antibody-mediated rejection (AMR). IVIG inhibits the MLR and induces apoptosis in immune cells. Mycophenolate mofetil inhibits immune cell proliferation and is an effective immunsuppressive agent. Here, we examined the possible synergistic effects of combined MMF and IVIG on cell proliferation and apoptosis induction in the MLR. METHODS Two-way MLRs were performed with mycophenolic acid (MPA), IVIG and both in combination. Cell proliferation and apoptosis were detected by 3H-thymidine incorporation and Annexin flow cytometry, respectively. RESULTS IVIG (1-10mg/ml) or MPA (0.01-0.25 microg/ml) alone inhibited cell proliferation in the MLR in a dose-dependent manner. MPA at 0.01-0.03 microg/ml showed minimal inhibition, but the addition of 5 and 10mg/ml IVIG increased inhibition significantly (p<0.05) to 43% and 64%, respectively. Annexin V positive cell number was significantly higher in IVIG (5mg/ml) treated CD19+ cells (68+/-13% vs. 43+/-12%, p=0.001) compared to untreated cells and to a lesser degree in CD3+ cells (29+/-7% vs. 25+/-10 %, p=0.02). MPA (0.25-10 microg/ml) alone neither induced nor inhibited apoptosis. Addition of MPA had no effect on apoptosis induced by IVIG. CONCLUSION 1) Combining low concentrations of IVIG (5-10 mg/ml) and MPA (0.01-0.03 microg/ml)has an additive effect on inhibition of cell proliferation in the MLR. 2) MPA alone neither induces nor inhibits apoptosis in T or B cells in the MLR, and has no effect on apoptosis induced by IVIG. These in vitro observations may have implications for modification of therapeutic approaches to protocols utilizing IVIG for desensitization and immune modulation.
Collapse
Affiliation(s)
- Kavita G Sharma
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Cedars-Sinai Medical Center/UCLA School of Medicine, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | | | | | | | | | | | | |
Collapse
|
129
|
Muller YD, Mai G, Morel P, Serre-Beinier V, Gonelle-Gispert C, Yung GP, Ehirchiou D, Wyss JC, Bigenzahn S, Irla M, Heusser C, Golshayan D, Seebach JD, Wekerle T, Bühler LH. Anti-CD154 mAb and rapamycin induce T regulatory cell mediated tolerance in rat-to-mouse islet transplantation. PLoS One 2010; 5:e10352. [PMID: 20436684 PMCID: PMC2859949 DOI: 10.1371/journal.pone.0010352] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 03/31/2010] [Indexed: 12/16/2022] Open
Abstract
Background Anti-CD154 (MR1) monoclonal antibody (mAb) and rapamycin (RAPA) treatment both improve survival of rat-to-mouse islet xenograft. The present study investigated the effect of combined RAPA/MR1 treatment on rat-to-mouse islet xenograft survival and analyzed the role of CD4+CD25+Foxp3+ T regulatory cells (Treg) in the induction and maintenance of the ensuing tolerance. Methodology/Principal Findings C57BL/6 mice were treated with MR1/RAPA and received additional monoclonal anti-IL2 mAb or anti CD25 mAb either early (0–28 d) or late (100–128 d) post-transplantation. Treg were characterised in the blood, spleen, draining lymph nodes and within the graft of tolerant and rejecting mice by flow cytometry and immunohistochemistry. Fourteen days of RAPA/MR1 combination therapy allowed indefinite islet graft survival in >80% of the mice. Additional administration of anti-IL-2 mAb or depleting anti-CD25 mAb at the time of transplantation resulted in rejection (100% and 89% respectively), whereas administration at 100 days post transplantation lead to lower rejection rates (25% and 40% respectively). Tolerant mice showed an increase of Treg within the graft and in draining lymph nodes early post transplantation, whereas 100 days post transplantation no significant increase of Treg was observed. Rejecting mice showed a transient increase of Treg in the xenograft and secondary lymphoid organs, which disappeared within 7 days after rejection. Conclusions/Significances These results suggest a critical role for Treg in the induction phase of tolerance early after islet xenotransplantation. These encouraging data support the need of developing further Treg therapy for overcoming the species barrier in xenotransplantation.
Collapse
Affiliation(s)
- Yannick D Muller
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Deenick EK, Po L, Chapatte L, Murakami K, Lu YC, Elford AR, Saibil SD, Ruland J, Gerondakis S, Mak TW, Ohashi PS. c-Rel phenocopies PKCtheta but not Bcl-10 in regulating CD8+ T-cell activation versus tolerance. Eur J Immunol 2010; 40:867-77. [PMID: 19950170 DOI: 10.1002/eji.200939445] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elucidating the signaling events that promote T-cell tolerance versus activation provides important insights for manipulating immunity in vivo. Previous studies have suggested that the absence of PKCtheta results in the induction of anergy and that the balance between the induction of the transcription factors NFAT, AP1 and NF-kappaB plays a key role in determining whether T-cell anergy or activation is induced. Here, we examine whether Bcl-10 and specific family members of NF-kappaB act downstream of PKCtheta to alter CD8(+) T-cell activation and/or anergy. We showed that T cells from mice deficient in c-Rel but not NF-kappaB1 (p50) have increased susceptibility to the induction of anergy, similar to T cells from PKCtheta-deficient mice. Surprisingly T cells from Bcl-10-deficient mice showed a strikingly different phenotype to the PKCtheta-deficient T cells, with a severe block in TCR-mediated activation. Furthermore, we have also shown that survival signals downstream of NF-kappaB, are uncoupled from signals that mediate T-cell anergy. These results suggest that c-Rel plays a critical role downstream of PKCtheta in controlling CD8(+) T-cell anergy induction.
Collapse
Affiliation(s)
- Elissa K Deenick
- Campbell Family Institute, Ontario Cancer Institute, University of Toronto, Toronto, ON, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Lange CM, Tran TYV, Farnik H, Jungblut S, Born T, Wagner TO, Hirche TO. Increased frequency of regulatory T Cells and selection of highly potent CD62L+ cells during treatment of human lung transplant recipients with rapamycin. Transpl Int 2010; 23:266-76. [DOI: 10.1111/j.1432-2277.2009.00973.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
132
|
Ray WZ, Kasukurthi R, Papp EM, Moore AM, Yee A, Hunter DA, Solowski NL, Mohanakumar T, Mackinnon SE, Tung TH. The role of T helper cell differentiation in promoting nerve allograft survival with costimulation blockade. J Neurosurg 2010; 112:386-93. [PMID: 19663546 PMCID: PMC2956431 DOI: 10.3171/2009.7.jns09187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Peripheral nerve allografts provide a temporary scaffold for host nerve regeneration and allow for the repair of significant segmental nerve injuries. Despite this potential, nerve allograft transplantation requires temporary systemic immunosuppression. Characterization of the immunological mechanisms involved in the induction of immune hyporesponsiveness to prevent nerve allograft rejection will help provide a basis for optimizing immunomodulation regimens or manipulating donor nerve allografts to minimize or eliminate the need for global immunosuppression. METHODS The authors used C57Bl/6 mice and STAT4 and STAT6 gene BALB/c knockout mice. A nonvascularized nerve allograft was used to reconstruct a 1-cm sciatic nerve gap in the murine model. A triple costimulatory blockade of the CD40, CD28/B7, and inducible costimulatory (ICOS) pathways was used. Quantitative assessment was performed at 3 weeks with nerve histomorphometry, walking track analysis, and the enzyme-linked immunospot assay. RESULTS The STAT6 -/- mice received 3 doses of costimulation-blocking antibodies and had axonal regeneration equivalent to nerve isografts, while treated STAT4 -/- mice demonstrated moderate axonal regeneration but inferior to the T helper cell Type 2-deficient animals. Enzyme-linked immunospot assay analysis demonstrated a minimal immune response in both STAT4 -/- and STAT6 -/- mice treated with a costimulatory blockade. CONCLUSIONS The authors' findings suggest that Type 1 T helper cells may play a more significant role in costimulatory blockade-induced immune hyporesponsiveness in the nerve allograft model, and that Type 2 T helper differentation may represent a potential target for directed immunosuppression.
Collapse
Affiliation(s)
- Wilson Z. Ray
- Department of Neurological Surgery, St. Louis, Missouri
| | | | - Esther M. Papp
- Washington University School of Medicine, St. Louis, Missouri
| | - Amy M. Moore
- Division of Plastic and Reconstructive Surgery, St. Louis, Missouri
| | - Andrew Yee
- Division of Plastic and Reconstructive Surgery, St. Louis, Missouri
| | - Daniel A. Hunter
- Division of Plastic and Reconstructive Surgery, St. Louis, Missouri
| | | | | | | | - Thomas H. Tung
- Division of Plastic and Reconstructive Surgery, St. Louis, Missouri
| |
Collapse
|
133
|
Zhang S, Dai H, Wan N, Moore Y, Dai Z. Manipulating IL-2 availability amid presentation of donor MHC antigens suppresses murine alloimmune responses by inducing regulatory T cells. PLoS One 2010; 5:e8756. [PMID: 20090908 PMCID: PMC2807454 DOI: 10.1371/journal.pone.0008756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 12/18/2009] [Indexed: 12/04/2022] Open
Abstract
Background Major histocompatibility complex (MHC) antigens are important for alloimmune responses as well as immune tolerance. Previous studies have shown that presentation of donor MHC antigens by donor-specific transfusion prior to or upon transplantation promotes transplant tolerance induced by other agents. However, it is unclear whether presentation of donor MHC antigens by DNA vaccination induces long-term allograft survival. Methodology/Principal Findings We investigated whether presentation of MHC class-II and/or class-I donor antigens by DNA vaccination suppresses alloimmune responses and promotes long-term allograft acceptance. We initially found that presentation of both MHC donor antigens by DNA vaccination itself prior to transplantation fails to significantly prolong islet allograft survival in otherwise untreated mice. However, islet allograft survival was significantly prolonged when MHC class-II DNA vaccination was accompanied with IL-2 administration (MHCII + IL-2) while MHC class-I DNA vaccination was followed by IL-2 and subsequent neutralizing anti-IL-2 treatments (MHCI + IL-2/anti-IL-2). Especially, this protocol promoted long-term allograft survival in the majority of recipients (57%) when combined with low doses of rapamycin post-transplantation. Importantly, MHCII + IL-2 induced FoxP3+ Treg cells in both spleens and grafts and suppressed graft-infiltrating CD4+ cell proliferation, whereas MHCI + IL-2/anti-IL-2 mainly inhibited graft-infiltrating CD8+ cell proliferation and donor-specific CTL activity. The combined protocol plus rapamycin treatment further reduced both CD4+ and CD8+ T cell proliferation as well as donor-specific CTL activity but spared FoxP3+ Treg cells. Depleting CD25+ Treg cells or adoptive transfer of pre-sensitized CD8+ T cells abolished this long-term allograft survival. Conclusions/Significance Manipulating IL-2 availability during presentation of MHC class-II and class-I donor antigens by DNA vaccination pre-transplantation induces Treg cells, suppresses alloimmune responses and promotes long-term allograft survival.
Collapse
Affiliation(s)
- Shuzi Zhang
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Hehua Dai
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Ni Wan
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Yolonda Moore
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Zhenhua Dai
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
- * E-mail:
| |
Collapse
|
134
|
Park YH, Koo SK, Kim Y, Kim HM, Joe IY, Park CS, Kim SC, Han DJ, Lim DG. Effect of in vitroexpanded CD4(+)CD25(+)Foxp3(+) regulatory T cell therapy combined with lymphodepletion in murine skin allotransplantation. Clin Immunol 2009; 135:43-54. [PMID: 20006940 DOI: 10.1016/j.clim.2009.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 11/18/2009] [Accepted: 11/19/2009] [Indexed: 01/27/2023]
Abstract
A promising approach for preventing allograft rejection involves shifting the balance between cytopathic and regulatory T cells to dominance of the latter cell type. Nonspecific lymphodepletion was conducted by administration of depleting anti-CD4 and anti-CD8 antibodies to reduce effector T cells and adoptive transfer of ex vivo-expanded host Treg cells by stimulation with donor dendritic cells to augment the Treg cell compartment. Evaluation of an MHC-mismatched skin allograft model revealed that combined therapy with these two protocols consistently induced modest prolongation of allograft survival, although all skin grafts were eventually rejected. The administration of IL-2/anti-IL-2 complexes significantly improved the efficacy of combination therapy via promoting the expansion of adoptively transferred Treg cells as well as endogenous recipient Treg cells. We conclude that Treg cell therapy combined with lymphodepletion is of practical benefit for the control of allograft rejection, but requires supplementary measures to promote immune tolerance.
Collapse
Affiliation(s)
- Youn-Hee Park
- Asan Institute for Life Sciences, Ulsan University College of Medicine, Seoul 138-736, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Raimondi G, Sumpter TL, Matta BM, Pillai M, Corbitt N, Vodovotz Y, Wang Z, Thomson AW. Mammalian target of rapamycin inhibition and alloantigen-specific regulatory T cells synergize to promote long-term graft survival in immunocompetent recipients. THE JOURNAL OF IMMUNOLOGY 2009; 184:624-36. [PMID: 20007530 DOI: 10.4049/jimmunol.0900936] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Minimization of immunosuppression and donor-specific tolerance to MHC-mismatched organ grafts are important clinical goals. The therapeutic potential of regulatory T cells (Tregs) has been demonstrated, but conditions for optimizing their in vivo function posttransplant in nonlymphocyte-depleted hosts remain undefined. In this study, we address mechanisms through which inhibition of the mammalian target of rapamycin (Rapa) synergizes with alloantigen-specific Treg (AAsTreg) to permit long-term, donor-specific heart graft survival in immunocompetent hosts. Crucially, immature allogeneic dendritic cells allowed AAsTreg selection in vitro, with minimal expansion of unwanted (Th17) cells. The rendered Treg potently inhibited T cell proliferation in an Ag-specific manner. However, these AAsTreg remained unable to control T cells stimulated by allogeneic mature dendritic cells, a phenomenon dependent on the release of proinflammatory cytokines. In vivo, Rapa administration reduced danger-associated IL-6 production, T cell proliferation, and graft infiltration. Based on these observations, AAsTreg were administered posttransplant (day 7) in combination with a short course of Rapa and rendered >80% long-term (>150 d) graft survival, a result superior to that achieved with polyclonal Treg. Moreover, graft protection was alloantigen-specific. Significantly, long-term graft survival was associated with alloreactive T cell anergy. These findings delineate combination of transient mammalian target of Rapa inhibition with appropriate AAsTreg selection as an effective approach to promote long-term organ graft survival.
Collapse
Affiliation(s)
- Giorgio Raimondi
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | |
Collapse
|
136
|
Choy JC, Pober JS. Generation of NO by bystander human CD8 T cells augments allogeneic responses by inhibiting cytokine deprivation-induced cell death. Am J Transplant 2009; 9:2281-91. [PMID: 19663890 PMCID: PMC3505447 DOI: 10.1111/j.1600-6143.2009.02771.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Nitric oxide (NO), generated by inducible NO synthase (iNOS) in bystander human CD8 T cells, augments the accumulation of allogeneically activated human CD8 T cells in vitro and in vivo. Here, we report that iNOS-derived NO does not affect T-cell proliferation but rather inhibits cell death of activated human CD8 T cells after activation by allogeneic endothelial cells in culture. Exogenous NO did not affect activation-induced cell death of human CD8 T cells but specifically reduced death of activated T cells due to cytokine deprivation. NO-mediated inhibition of T-cell death did not involve cGMP signaling, and NO did not affect the expression of Bcl-2-related proteins known to regulate cytokine deprivation-induced cell death. However, NO inhibited the activity of caspases activated as a consequence of cytokine deprivation in activated T cells. This protective effect correlated with S-nitrosylation of caspases and was phenocopied by z-VAD.fmk and z-LEHD.fmk, pharmacological inhibitors of caspases. In summary, our findings indicate that NO augments the accumulation of activated human T cells principally by inhibiting cytokine deprivation-induced cell death through S-nitrosylation of caspases.
Collapse
Affiliation(s)
- Jonathan C. Choy
- Section of Human and Translational Immunology, Yale University School of Medicine, New Haven, CT
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Jordan S. Pober
- Section of Human and Translational Immunology, Yale University School of Medicine, New Haven, CT
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
- Department of Pathology, Yale University School of Medicine, New Haven, CT
- Department of Dermatology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
137
|
Nayak S, Cao O, Hoffman BE, Cooper M, Zhou S, Atkinson MA, Herzog RW. Prophylactic immune tolerance induced by changing the ratio of antigen-specific effector to regulatory T cells. J Thromb Haemost 2009; 7:1523-32. [PMID: 19583824 PMCID: PMC2981868 DOI: 10.1111/j.1538-7836.2009.03548.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Gene and protein replacement therapies for inherited protein deficiencies such as hemophilia or lysosomal storage disorders are limited by deleterious immune responses directed against their respective therapeutic proteins. Therefore, the development of protocols preventing such responses is key to providing successful long-term therapy. OBJECTIVES We sought to develop a protocol, utilizing a drug/peptide cocktail, that would effectively shift the antigen-specific CD4+ T-cell population, tipping the balance from effector T cells (Teffs) towards regulatory T cells (Tregs). METHODS Treg-deficient (DO11.10-tg Rag2(-/-)) BALB/c mice were used to screen for an optimal protocol addressing the aforementioned goal and to study the mechanisms underlying in vivo changes in T-cell populations. Muscle-directed gene transfer to hemophilia B mice was also performed in order to test the optimal protocol in a therapeutically relevant setting. RESULTS Specific antigen administration (4-week repeated dosing) combined with rapamycin and interleukin-10 led to substantial reductions in Teffs, via activation-induced cell death, and induced CD4+CD25+FoxP3+ Tregs to a large extent in multiple organs. The proportion of apoptotic T cells also increased over time, whereas Teffs and Tregs were differentially affected. When applied to a model of protein deficiency (gene therapy for hemophilia B), the protocol successfully prevented inhibitor formation, whereas non-specific immunosuppression was only marginally effective. CONCLUSIONS It is feasible to provide a short-term, prophylactic protocol allowing for the induction of immune tolerance. This protocol may provide a marked advance in efforts seeking to improve clinical outcomes in disorders involving therapeutic protein replacement.
Collapse
Affiliation(s)
- S Nayak
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | |
Collapse
|
138
|
Graves SS, Stone D, Loretz C, Peterson L, McCune JS, Mielcarek M, Storb R. Establishment of long-term tolerance to SRBC in dogs by recombinant canine CTLA4-Ig. Transplantation 2009; 88:317-22. [PMID: 19667932 PMCID: PMC2753482 DOI: 10.1097/tp.0b013e3181ae3285] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Blockade of the CD28 costimulatory molecule by recombinant human cytotoxic T lymphocyte (CTL)-associated antigen (CTLA4)-Ig or CD40-CD154 interaction with the monoclonal antibody 5C8 together with donor-specific transfusion led to enhanced engraftment in the canine model of dog leukocyte antigen (DLA)-identical marrow transplantation after 1 Gy total body irradiation. To reduce or eliminate total body irradiation conditioning regimens, we have sought to develop canine specific reagents. METHODS We have created a fusion protein of the extracellular domain of canine (c) CTLA-4 linked to the hinge-CH2-CH3 domains of canine IgG1 in a pcDNA3.1+ vector. Chinese hamster ovarian cells were cotransfected with CTLA4-Ig vector and a dihydrofolate reductase-containing vector. Stable, high producing clones were generated. RESULTS Cell binding and mixed leukocyte reactions indicated no significant differences in activity between cCTLA4-Ig and human CTLA4-Ig. Mixed leukocyte reaction data indicated that combinations of cCTLA4-Ig and the monoclonal antibody 5C8 were superior in blocking H-thymidine uptake compared to either reagent alone. In dogs, the circulating half-life of cCTLA4-Ig was approximately 7 days with no immune response against the fusion protein. Finally, two injections of cCTLA4-Ig effectively tolerized two dogs against eight consecutive challenges with sheep red blood cells, given over 330 days as indicated by a complete block of IgG antibody production. Tolerance was broken in one of the two dogs when a ninth injection of sheep red blood cell was given subcutaneously in incomplete Freund's adjuvant. CONCLUSION cCTLA4-Ig is an effective nonimmunogenic blocking reagent of the CD28 costimulatory pathway in dogs and is a promising reagent for studies of tolerance induction in hematopoietic cell transplantation in the canine model.
Collapse
Affiliation(s)
- Scott S Graves
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109-1024, USA.
| | | | | | | | | | | | | |
Collapse
|
139
|
Increased Interleukin-10 Production Without Expansion of CD4+CD25+ T-Regulatory Cells in Early Stable Renal Transplant Patients on Calcineurin Inhibitors. Transplantation 2009; 88:435-41. [DOI: 10.1097/tp.0b013e3181af20fd] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
140
|
|
141
|
Mancuso ME, Graca L, Auerswald G, Santagostino E. Haemophilia care in children--benefits of early prophylaxis for inhibitor prevention. Haemophilia 2009; 15 Suppl 1:8-14. [PMID: 19125935 DOI: 10.1111/j.1365-2516.2008.01947.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Haemophilia therapy is aimed at treating and preventing bleeding episodes and related complications and clinical studies have shown that regular prophylaxis, started at an early age, is able to reduce physical impairment from haemophilic arthropathy. Today, the development of anti-Factor VIII (FVIII) inhibitors is the most serious treatment-related complication of haemophilia therapy and a number of genetic and environmental risk factors have been identified in the past years. Clinical data show that early start of prophylaxis and the avoidance of intensive treatment periods may protect patients from inhibitor development. The mechanisms are not completely understood; yet, recent experimental data suggest that pro-inflammatory or 'danger signals' may be involved in inducing tolerance vs. an effector immune response. So, exposure to a factor concentrate by itself may not be enough to trigger an immune response, while an intensive exposure to FVIII in the presence of such 'danger signals' can activate antigen-presenting cells, up-regulating co-stimulatory signals for T lymphocytes and ultimately enhancing antibody production. The 'optimal' regimen for primary prophylaxis is still not identified and barriers to prophylaxis implementation remain relevant. Key issues include the optimal age at prophylaxis onset, the optimal dosage/schedule, the proper clinical and laboratory monitoring and patients' compliance. Practical approaches to early prophylaxis as implemented in the haemophilia centres in Milan and Bremen are discussed in this respect.
Collapse
Affiliation(s)
- M E Mancuso
- Angelo Bianchi Bonomi Hemophilia & Thrombosis Center, IRCCS Maggiore Hospital, Mangiagalli & Regina Elena Foundation, University of Milan, Milan, Italy.
| | | | | | | |
Collapse
|
142
|
Gilson CR, Milas Z, Gangappa S, Hollenbaugh D, Pearson TC, Ford ML, Larsen CP. Anti-CD40 monoclonal antibody synergizes with CTLA4-Ig in promoting long-term graft survival in murine models of transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:1625-35. [PMID: 19592649 PMCID: PMC2828346 DOI: 10.4049/jimmunol.0900339] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Blockade of the CD40/CD154 signaling pathway using anti-CD154 Abs has shown promise in attenuating the alloimmune response and promoting long-term graft survival in murine model systems, although side effects observed in humans have hampered its progression through clinical trials. Appropriately designed anti-CD40 Abs may provide a suitable alternative. We investigated two isoforms of a novel monoclonal rat anti-mouse CD40 Ab (7E1) for characteristics and effects mirroring those of anti-CD154: 7E1-G1 (an IgG1 isotype); and 7E1-G2b (an IgG2b isotype). In vitro proliferation assays to measure the agonist properties of the two anti-CD40 Abs revealed similar responses when plate bound. However, when present as a soluble stimulus, 7E1-G1 but not 7E1-G2b led to proliferation. 7E1-G2b was as effective as anti-CD154 when administered in vivo in concert with CTLA4-Ig in promoting both allogeneic bone marrow chimerism and skin graft survival, whereas 7E1-G1 was not. The protection observed with 7E1-G2b was not due to depletion of CD40-bearing APCs. These data suggest that an appropriately designed anti-CD40 Ab can promote graft survival as well as anti-CD154, making 7E1-G2b an attractive substitute in mouse models of costimulation blockade-based tolerance regimens.
Collapse
Affiliation(s)
- Christopher R Gilson
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - Zvonimir Milas
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - Shivaprakash Gangappa
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - Diane Hollenbaugh
- Bristol-Myers Squibb Pharmaceutical Research Institute, Princeton, NJ 08543
| | - Thomas C. Pearson
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - Mandy L. Ford
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - Christian P. Larsen
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322
| |
Collapse
|
143
|
Zheng H, Du G, Zhao R, Tang B, Zhu N. Enhanced allogeneic skin-graft survival using sCD95L, sCD152, interleukin-10 and transforming growth factor-β in combination, and comparison with ciclosporin. Clin Exp Dermatol 2009; 34:721-7. [DOI: 10.1111/j.1365-2230.2009.03237.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
144
|
Turka LA, Lechler RI. Towards the identification of biomarkers of transplantation tolerance. Nat Rev Immunol 2009; 9:521-6. [PMID: 19483711 DOI: 10.1038/nri2568] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although transplantation has been a standard medical practice for decades, the marked morbidity from the use of immunosuppressive drugs and poor long-term graft survival remain important limitations in the field. Achieving tolerance to transplanted organs should solve both problems, but has been an elusive goal. Recent advances in the human immunological toolbox have rekindled interest in studying the small number of transplant recipients who become tolerant to their grafts over time. The development of biomarkers of transplantation tolerance holds promise to improve the care of organ allograft recipients, to provide surrogate end points of tolerance induction strategies and to advance our understanding of the human immune response to both self and foreign antigens.
Collapse
Affiliation(s)
- Laurence A Turka
- Department of Medicine, University of Pennsylvania, Philadelphia 19104, USA.
| | | |
Collapse
|
145
|
Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev 2009; 229:152-72. [PMID: 19426221 DOI: 10.1111/j.1600-065x.2009.00782.x] [Citation(s) in RCA: 1124] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SUMMARY During the generation of a successful adaptive immune response, multiple molecular signals are required. A primary signal is the binding of cognate antigen to an antigen receptor expressed by T and B lymphocytes. Multiple secondary signals involve the engagement of costimulatory molecules expressed by T and B lymphocytes with their respective ligands. Because of its essential role in immunity, one of the best characterized of the costimulatory molecules is the receptor CD40. This receptor, a member of the tumor necrosis factor receptor family, is expressed by B cells, professional antigen-presenting cells, as well as non-immune cells and tumors. CD40 binds its ligand CD40L, which is transiently expressed on T cells and other non-immune cells under inflammatory conditions. A wide spectrum of molecular and cellular processes is regulated by CD40 engagement including the initiation and progression of cellular and humoral adaptive immunity. In this review, we describe the downstream signaling pathways initiated by CD40 and overview how CD40 engagement or antagonism modulates humoral and cellular immunity. Lastly, we discuss the role of CD40 as a target in harnessing anti-tumor immunity. This review underscores the essential role CD40 plays in adaptive immunity.
Collapse
Affiliation(s)
- Raul Elgueta
- Department of Microbiology and Immunology, Dartmouth Medical School and The Norris Cotton Cancer Center, Lebanon, NH 03756, USA
| | | | | | | | | | | |
Collapse
|
146
|
Perruche S, Zhang P, Maruyama T, Bluestone JA, Saas P, Chen W. Lethal effect of CD3-specific antibody in mice deficient in TGF-beta1 by uncontrolled flu-like syndrome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:953-61. [PMID: 19561097 PMCID: PMC2842991 DOI: 10.4049/jimmunol.0804076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD3-specific Ab therapy results in a transient, self-limiting, cytokine-associated, flu-like syndrome in experimental animals and in patients, but the underlying mechanism for this spontaneous resolution remains elusive. By using an in vivo model of CD3-specific Ab-induced flu-like syndrome, we show in this paper that a single injection of sublethal dose of the Ab killed all TGF-beta1(-/-) mice. The death of TGF-beta1(-/-) mice was associated with occurrence of this uncontrolled flu-like syndrome, as demonstrated by a sustained storm of systemic inflammatory TNF and IFN-gamma cytokines. We present evidence that deficiency of professional phagocytes to produce TGF-beta1 after apoptotic T cell clearance may be responsible, together with hypersensitivity of T cells to both activation and apoptosis, for the uncontrolled inflammation. These findings indicate a key role for TGF-beta1 and phagocytes in protecting the recipients from lethal inflammation and resolving the flu-like syndrome after CD3-specific Ab treatment. The study may also provide a novel molecular mechanism explaining the early death in TGF-beta1(-/-) mice.
Collapse
Affiliation(s)
- Sylvain Perruche
- Mucosal Immunology Unit, Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Pin Zhang
- Mucosal Immunology Unit, Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Takashi Maruyama
- Mucosal Immunology Unit, Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | | | - Philippe Saas
- INSERM UMR645, University of Franche-Comte, Establissement Francais du Sana de Bourgogne Franche Comte, Institut Federatif de Recherche 133, Besançon, France
| | - WanJun Chen
- Mucosal Immunology Unit, Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
147
|
Wang R, Gan C, Gao W, He W, Wang X, Peng Y, Zhuo J, Tan J, Peng X, Wu J, Luo G. A novel recombinant immunotoxin with the smallest ribosome-inactivating protein Luffin P1: T-cell cytotoxicity and prolongation of allograft survival. J Cell Mol Med 2009; 14:578-86. [PMID: 19583807 PMCID: PMC3823457 DOI: 10.1111/j.1582-4934.2009.00840.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
In the creation of stable tolerance to MHC-incompatible allografts, reducing the large mass of donor-reactive cells via apoptosis is often required. Apoptosis induction by immunotoxins targeting surface molecules specifically presented on donor-reactive cytopathic T effector (Teff) cells is a promising strategy. Traditionally, the toxin moieties are bacterial exotoxins or plant-derived ribosome-inactivating proteins (RIPs) with large molecular size and strong immunogenicity, hence causing the problems of tissue penetration, host immune reaction and quick clearance. We have identified a novel class of small molecule RIPs (<10 kD) from the seeds of the plant Luffa cylindrica. The smallest member of this family, Luffin P1, has a molecular weight of 5226.8 Da, yet possessing a highly potent inhibitory activity on cell-free protein synthesis with IC50 of 0.88 nM. We now report a recombinant hIL-2-Luffin P1 immunotoxin, which strongly inhibited T-cell proliferation in mixed lymphocyte reaction and ConA response with IC50 of 1.8–10 nM. In vivo, hIL-2-Luffin P1 significantly prolonged the survival of major MHC-mismatched skin and kidney allografts in animal models. Thus, we demonstrate for the first time the efficacy of the smallest immunotoxin that could be further combined with other pharmacological and immunological reagents for synergistic control of pathogenic lymphocytes in immune-mediated diseases.
Collapse
Affiliation(s)
- Rupeng Wang
- Chongqing Key Lab for Diseases Proteomics, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
|
149
|
Okazaki M, Sugimoto S, Lai J, Kornfeld CG, Hotchkiss RS, Richardson SB, Li W, Kreisel FH, Huang HJ, Patterson GA, Krupnick AS, Gelman AE, Kreisel D. Costimulatory blockade-mediated lung allograft acceptance is abrogated by overexpression of Bcl-2 in the recipient. Transplant Proc 2009; 41:385-7. [PMID: 19249562 DOI: 10.1016/j.transproceed.2008.10.068] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 10/06/2008] [Indexed: 11/29/2022]
Abstract
Lung allografts are considered to be more immunogenic than other solid organs. Little is known about the effectiveness of immunosuppressive regimens after lung transplantation. Herein, we describe a novel model of murine vascularized orthotopic lung transplantation we used to study the effects of costimulatory blockade on lung rejection. Transplants were performed in the Balb --> B6 strain combination. Recipients were either not immunosuppressed or received perioperative CD40/CD40L and CD28/B7 costimulatory blockade. Nonimmunosupressed Balb/c --> B6 lung transplants had severe acute rejection 7 days after transplantation and CD8(+) T cells outnumbered CD4(+) T cells within the allografts. Alternatively, B6 recipients that received perioperative costimulatory blockade had minimal inflammation and there were nearly equal numbers of CD8(+) and CD4(+) T cells in these grafts. Approximately one third of graft-infiltrating CD4(+) T cells expressed Foxp3. CD4(+) T cells isolated from these grafts induced apoptosis of alloreactive CD8(+) T cells that were stimulated with donor splenocytes in vitro. In contrast with wild-type B6 recipient mice, we observed severe rejection of Balb/c lungs 7 days after transplantation into Bcl-2 transgenic B6 recipients that had received costimulatory blockade. CD8(+) T cells outnumbered CD4(+) T cells in these immunosuppressed Bcl-2 transgenic recipients and, compared with immunosuppressed wild-type B6 recipients, a lower percentage of graft-infiltrating CD4(+) T cells expressed Foxp3, and a higher percentage of graft-infiltrating CD8(+) T cells expressed intereferon-gamma. Thus, our results show that perioperative blockade of the CD40/CD40L and CD28/B7 costimulatory pathways markedly ameliorates acute rejection of lung allografts in wild type but not Bcl-2 transgenic recipients.
Collapse
Affiliation(s)
- M Okazaki
- Department of Surgery, Washington University in St. Louis, St. Louis, Missourri 63110-1013, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Molinero LL, Wang Y, Zhou P, Yagita H, Alegre ML. Fas mediates cardiac allograft acceptance in mice with impaired T-cell-intrinsic NF-kappaB signaling. Transpl Int 2009; 22:845-52. [PMID: 19351347 DOI: 10.1111/j.1432-2277.2009.00875.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The transcription factor NF-kappaB is critical for T-cell activation and survival. We have shown that mice expressing a T-cell-restricted NF-kappaB superrepressor (IkappaBalphaDeltaN-Tg) permanently accept heart but not skin allografts. Overexpression of the prosurvival factor Bcl-x(L) in T cells restored heart rejection, suggesting that graft acceptance in IkappaBalphaDeltaN-Tg mice was attributable to deletion of alloreactive T cells.In vitro, the increased death of IkappaBalphaDeltaN-Tg T cells upon TCR stimulation when compared with wildtype T cells was mostly because of Fas/FasL interaction. Similarly, Fas played a key role in cardiac allograft acceptance by IkappaBalphaDeltaN-Tg mice as both genetic and antibody-mediated inhibition of Fas-signaling restored cardiac allograft rejection. Rejection correlated with graft infiltration by T cells and splenic production of IFN-gamma upon allostimulation. These results indicate that T-cell inhibition of NF-kappaB results in cardiac allograft acceptance because of increased susceptibility to Fas-mediated cell death.
Collapse
Affiliation(s)
- Luciana Lorena Molinero
- Department of Medicine, Section of Rheumatology, The University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA.
| | | | | | | | | |
Collapse
|