Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Biomed Eng ; 7(9): 1142-1155, 2023 09.
Article in English | MEDLINE | ID: mdl-37679570

ABSTRACT

Inducing antigen-specific tolerance during an established immune response typically requires non-specific immunosuppressive signalling molecules. Hence, standard treatments for autoimmunity trigger global immunosuppression. Here we show that established antigen-specific responses in effector T cells and memory T cells can be suppressed by a polymer glycosylated with N-acetylgalactosamine (pGal) and conjugated to the antigen via a self-immolative linker that allows for the dissociation of the antigen on endocytosis and its presentation in the immunoregulatory environment. We show that pGal-antigen therapy induces antigen-specific tolerance in a mouse model of experimental autoimmune encephalomyelitis (with programmed cell-death-1 and the co-inhibitory ligand CD276 driving the tolerogenic responses), as well as the suppression of antigen-specific responses to vaccination against a DNA-based simian immunodeficiency virus in non-human primates. Our findings show that pGal-antigen therapy invokes mechanisms of immune tolerance to resolve antigen-specific inflammatory T-cell responses and suggest that the therapy may be applicable across autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Immune Tolerance , Animals , Mice , Autoimmunity , Glycosylation , Acetylgalactosamine , Encephalomyelitis, Autoimmune, Experimental/therapy
2.
Front Immunol ; 9: 2007, 2018.
Article in English | MEDLINE | ID: mdl-30233592

ABSTRACT

Hematopoietic stem/progenitor cells (HSPC) are characterized by their unique capacities of self-renewal and multi-differentiation potential. This second property makes them able to adapt their differentiation profile depending on the local environment they reach. Taking advantage of an animal model of peritonitis, induced by injection of the TLR-2 ligand, zymosan, we sought to study the relationship between bone marrow-derived hematopoietic stem/progenitor cells (BM-HSPCs) and innate lymphoid cells (ILCs) regarding their emergence and differentiation at the site of inflammation. Our results demonstrate that the strength of the inflammatory signals affects the capacity of BM-derived HSPCs to migrate and give rise in situ to ILCs. Both low- and high-dose of zymosan injections trigger the appearance of mature ILCs in the peritoneal cavity where the inflammation occurs. Herein, we show that only in low-dose injected mice, the recovered ILCs are dependent on an in situ differentiation of BM-derived HSPCs and/or ILC2 precursors (ILC2P) wherein high-dose, the stronger inflammatory environment seems to be able to induce the emergence of ILCs independently of BM-derived HSPCs. We suggest that a relationship between HSPCs and ILCs seems to be affected by the strength of the inflammatory stimuli opening new perspectives in the manipulation of these early hematopoietic cells.


Subject(s)
Hematopoietic Stem Cells/immunology , Lymphocytes/immunology , Peritonitis/immunology , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Movement , Cell Self Renewal , Cells, Cultured , Disease Models, Animal , Female , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Signal Transduction , Stem Cell Niche , Zymosan
3.
Cancer Discov ; 7(7): 750-765, 2017 07.
Article in English | MEDLINE | ID: mdl-28274958

ABSTRACT

Several kinase inhibitors that target aberrant signaling pathways in tumor cells have been deployed in cancer therapy. However, their impact on the tumor immune microenvironment remains poorly understood. The tyrosine kinase inhibitor cabozantinib showed striking responses in cancer clinical trial patients across several malignancies. Here, we show that cabozantinib rapidly eradicates invasive, poorly differentiated PTEN/p53-deficient murine prostate cancer. This was associated with enhanced release of neutrophil chemotactic factors from tumor cells, including CXCL12 and HMGB1, resulting in robust infiltration of neutrophils into the tumor. Critically, cabozantinib-induced tumor clearance in mice was abolished by antibody-mediated granulocyte depletion or HMGB1 neutralization or blockade of neutrophil chemotaxis with the CXCR4 inhibitor plerixafor. Collectively, these data demonstrate that cabozantinib triggers a neutrophil-mediated anticancer innate immune response, resulting in tumor clearance.Significance: This study is the first to demonstrate that a tyrosine kinase inhibitor can activate neutrophil-mediated antitumor innate immunity, resulting in invasive cancer clearance. Cancer Discov; 7(7); 750-65. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 653.


Subject(s)
Anilides/administration & dosage , Chemokine CXCL12/antagonists & inhibitors , HMGB1 Protein/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/drug therapy , Pyridines/administration & dosage , Tumor Suppressor Protein p53/genetics , Animals , Benzylamines , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/genetics , Cyclams , HMGB1 Protein/genetics , Heterocyclic Compounds/administration & dosage , Humans , Immunity, Innate/drug effects , Male , Mice , Neutrophils/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Tumor Microenvironment/genetics
4.
PLoS One ; 11(3): e0150792, 2016.
Article in English | MEDLINE | ID: mdl-26943809

ABSTRACT

The innate immune system critically shapes diabetogenic adaptive immunity during type 1 diabetes (T1D) pathogenesis. While the role of tissue-infiltrating monocyte-derived macrophages in T1D is well established, the role of their tissue-resident counterparts remains undefined. We now demonstrate that islet resident macrophages (IRMs) from non-autoimmune mice have an immunoregulatory phenotype and powerfully induce FoxP3+ Tregs in vitro. The immunoregulatory phenotype and function of IRMs is compromised by TLR4 activation in vitro. Moreover, as T1D approaches in NOD mice, the immunoregulatory phenotype of IRMs is diminished as is their relative abundance compared to immunostimulatory DCs. Our findings suggest that maintenance of IRM abundance and their immunoregulatory phenotype may constitute a novel therapeutic strategy to prevent and/or cure T1D.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/pathology , Macrophages/immunology , Animals , Antigens, CD/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Forkhead Transcription Factors/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Phenotype , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 4/metabolism
5.
J Clin Invest ; 124(8): 3443-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24983317

ABSTRACT

Macrophages characterized as M2 and M2-like regulate immune responses associated with immune suppression and healing; however, the relationship of this macrophage subset to CD169+ tissue-resident macrophages and their contribution to shaping alloimmune responses is unknown. Here we identified a population of M2-like tissue-resident macrophages that express high levels of the phosphatidylserine receptor TIM-4 and CD169 (TIM-4hiCD169+). Labeling and tracking of TIM-4hiCD169+ macrophages in mice revealed that this population is a major subset of tissue-resident macrophages, homes to draining LNs following oxidative stress, exhibits an immunoregulatory and hypostimulatory phenotype that is maintained after migration to secondary lymphoid organs, favors preferential induction of antigen-stimulated Tregs, and is highly susceptible to apoptosis. Moreover, CD169+ tissue-resident macrophages were resistant to oxidative stress-induced apoptosis in mice lacking TIM-4. Compared with heart allografts from WT mice, Tim4-/- heart allografts survived much longer and were more easily tolerized by non-immunosuppressed recipients. Furthermore, Tim4-/- allograft survival was associated with the infiltration of Tregs into the graft. Together, our data provide evidence that M2-like TIM-4hiCD169+ tissue-resident macrophages are immunoregulatory and promote engraftment of cardiac allografts, but their influence is diminished by TIM-4-dependent programmed cell death.


Subject(s)
Macrophages/immunology , Macrophages/physiology , Membrane Proteins/metabolism , Allografts , Animals , Apoptosis , Cell Differentiation , Cell Movement , Cell Proliferation , Graft Survival , Heart Transplantation , Lymphocyte Culture Test, Mixed , Macrophages/cytology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidative Stress , Sialic Acid Binding Ig-like Lectin 1/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Transplantation Tolerance
6.
Immunology ; 138(4): 322-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23190301

ABSTRACT

Blockade of co-stimulatory signals to T cells is extremely effective for the induction of transplantation tolerance in immunologically naive rodents. However, infections and inflammation compromise the efficacy of co-stimulation blockade regimens for the induction of tolerance, thereby stimulating the rejection of allografts. Previous studies have shown that stimulation of innate immunity abrogates tolerance induction by preventing the deletion of alloreactive CD8(+) T cells that normally occurs during co-stimulation blockade. Although inflammation prevents the deletion of alloreactive T cells during co-stimulation blockade, it is not known if this resistance to cell death is the result of a mechanism intrinsic to the T cell. Here, we used syngeneic bone marrow chimeric mice that contain a trace population of T-cell receptor transgenic alloreactive CD8(+) T cells to investigate the early apoptotic signature and activation status of alloreactive T cells following exposure to inflammatory signals during co-stimulation blockade with an antibody specific for CD154. Our findings revealed that the presence of bacterial lipopolysaccharide during co-stimulation blockade enhanced the early activation of alloreactive CD8(+) T cells, as indicated by the up-regulation of CD25 and CD69, suppressed Fas ligand expression, and prevented apoptotic cell death. However, alloreactive CD8(+) T cells from lipopolysaccharide-treated mice remained sensitive to Fas-mediated apoptosis in vitro. These findings suggest that alloreactive T cells rescued from deletion during co-stimulation blockade by inflammation are still sensitive to pro-apoptotic signals and that stimulating these apoptotic pathways during co-stimulation blockade may augment the induction of tolerance.


Subject(s)
Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , Fas Ligand Protein/immunology , Toll-Like Receptors/immunology , fas Receptor/immunology , Animals , Antibodies/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Apoptosis/immunology , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/genetics , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Fas Ligand Protein/genetics , Gene Expression Regulation/drug effects , Immune Tolerance/drug effects , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , fas Receptor/genetics
7.
J Clin Invest ; 122(7): 2395-404, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22684103

ABSTRACT

Tregs play a pivotal role in inducing and maintaining donor-specific transplant tolerance. The T cell immunoglobulin and mucin domain-3 protein (TIM-3) is expressed on many fully activated effector T cells. Along with program death 1 (PD-1), TIM-3 is used as a marker for exhausted effector T cells, and interaction with its ligand, galectin-9, leads to selective death of TIM-3+ cells. We report herein the presence of a galectin-9-sensitive CD4+FoxP3+TIM-3+ population of T cells, which arose from CD4+FoxP3+TIM-3- proliferating T cells in vitro and in vivo and were often PD-1+. These cells became very prominent among graft-infiltrating Tregs during allograft response. The frequency and number of TIM-3+ Tregs peaked at the time of graft rejection and declined thereafter. Moreover, these cells also arise in a tolerance-promoting donor-specific transfusion model, representing a pool of proliferating, donor-specific Tregs. Compared with TIM-3- Tregs, TIM-3+ Tregs, which are often PD-1+ as well, exhibited higher in vitro effector function and more robust expression of CD25, CD39, CD73, CTLA-4, IL-10, and TGF-ß but not galectin-9. However, these TIM-3+ Tregs did not flourish when passively transferred to newly transplanted hosts. These data suggest that a heretofore unrecognized graft-infiltrating, short-lived subset of Tregs can restrain rejection.


Subject(s)
Forkhead Transcription Factors/metabolism , Graft Rejection/metabolism , Programmed Cell Death 1 Receptor/metabolism , Receptors, Virus/metabolism , Skin Transplantation/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Apoptosis , CD4 Antigens/metabolism , Cell Proliferation , Cells, Cultured , Forkhead Transcription Factors/genetics , Galectins/physiology , Graft Rejection/pathology , Hepatitis A Virus Cellular Receptor 2 , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Receptors, Virus/genetics , Skin Transplantation/pathology , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/physiology , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology
9.
J Immunol ; 182(9): 5547-59, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380803

ABSTRACT

Activation of TLR4 by administration of LPS shortens the survival of skin allografts in mice treated with costimulation blockade through a CD8 T cell-dependent, MyD88-dependent, and type I IFN receptor-dependent pathway. The effect of TLR activation on the establishment of allogeneic hematopoietic chimerism in mice treated with costimulation blockade is not known. Using a costimulation blockade protocol based on a donor-specific transfusion (DST) and a short course of anti-CD154 mAb, we show that LPS administration at the time of DST matures host alloantigen-presenting dendritic cells, prevents the establishment of mixed allogeneic hematopoietic chimerism, and shortens survival of donor-specific skin allografts. LPS mediates its effects via a mechanism that involves both CD4(+) and CD8(+) T cells and results from signaling through either the MyD88 or the type I IFN receptor pathways. We also document that timing of LPS administration is critical, as injection of LPS 24 h before treatment with DST and anti-CD154 mAb does not prevent hematopoietic engraftment but administration the day after bone marrow transplantation does. We conclude that TLR4 activation prevents the induction of mixed allogeneic hematopoietic chimerism through type I IFN receptor and MyD88-dependent signaling, which leads to the up-regulation of costimulatory molecules on host APCs and the generation of alloreactive T cells. These data suggest that distinct but overlapping cellular and molecular mechanisms control the ability of TLR agonists to block tolerance induction to hematopoietic and skin allografts in mice treated with costimulation blockade.


Subject(s)
Chimerism , Hematopoietic Stem Cell Transplantation , Immunosuppression Therapy , Isoantigens/genetics , Lipopolysaccharides/administration & dosage , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Toll-Like Receptors/agonists , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carrier Proteins/administration & dosage , Graft Survival/genetics , Graft Survival/immunology , Immunosuppression Therapy/methods , Interferon Type I/biosynthesis , Interferon Type I/metabolism , Interferon Type I/physiology , Isoantigens/immunology , Isoantigens/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , Poly I-C/administration & dosage , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Skin Transplantation/immunology , Toll-Like Receptors/administration & dosage , Toll-Like Receptors/metabolism
10.
Clin Dev Immunol ; 2008: 742810, 2008.
Article in English | MEDLINE | ID: mdl-18815618

ABSTRACT

Transplantation of allogeneic organs has proven to be an effective therapeutic for a large variety of disease states, but the chronic immunosuppression that is required for organ allograft survival increases the risk for infection and neoplasia and has direct organ toxicity. The establishment of transplantation tolerance, which obviates the need for chronic immunosuppression, is the ultimate goal in the field of transplantation. Many experimental approaches have been developed in animal models that permit long-term allograft survival in the absence of chronic immunosuppression. These approaches function by inducing peripheral or central tolerance to the allograft. Emerging as some of the most promising approaches for the induction of tolerance are protocols based on costimulation blockade. However, as these protocols move into the clinic, there is recognition that little is known as to their safety and efficacy when confronted with environmental perturbants such as virus infection. In animal models, it has been reported that virus infection can prevent the induction of tolerance by costimulation blockade and, in at least one experimental protocol, can lead to significant morbidity and mortality. In this review, we discuss how viruses modulate the induction and maintenance of transplantation tolerance.


Subject(s)
Transplantation Tolerance/immunology , Virus Diseases/immunology , Animals , Graft Rejection/immunology , Graft Rejection/virology , Humans , Immunity, Innate/immunology , Immunosuppression Therapy/methods , Signal Transduction/immunology , Transplantation Immunology
11.
Cell Stress Chaperones ; 13(4): 421-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18418732

ABSTRACT

T cell receptor (TCR) ligation (signal one) in the presence of co-stimulation (signal two) results in downstream signals that increase protein production enabling naïve T cells to fully activate and gain effector function. Enhanced production of proteins by a cell requires an increase in endoplasmic reticulum (ER) chaperone expression, which is accomplished through activation of a cellular mechanism known as the ER stress response. The ER stress response is initiated during the cascade of events that occur for the activation of many cells; however, this process has not been comprehensively studied for T cell function. In this study, we used primary T cells and mice circulating TCR transgenic CD8(+) T cells to investigate ER chaperone expression in which TCR signaling was initiated in the presence or absence of co-stimulation. In the presence of both signals, in vitro and in vivo analyses demonstrated induction of the ER stress response, as evidenced by elevated expression of GRP78 and other ER chaperones. Unexpectedly, ER chaperones were also increased in T cells exposed only to signal one, a treatment known to cause T cells to enter the 'nonresponsive' states of anergy and tolerance. Treatment of T cells with an inhibitor to protein kinase C (PKC), a serine/threonine protein kinase found downstream of TCR signaling, indicated PKC is involved in the induction of the ER stress response during the T cell activation process, thus revealing a previously unknown role for this signaling protein in T cells. Collectively, these data suggest that induction of the ER stress response through PKC signaling is an important component for the preparation of a T cell response to antigen.


Subject(s)
Endoplasmic Reticulum/enzymology , Lymphocyte Activation/immunology , Protein Kinase C/metabolism , Signal Transduction , Stress, Physiological , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Animals , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/drug effects , Heat-Shock Proteins/metabolism , Immune Tolerance/drug effects , Interleukin-2/biosynthesis , Ionomycin/pharmacology , Lymphocyte Activation/drug effects , Male , Mice , Models, Biological , Molecular Chaperones/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Thapsigargin/pharmacology , Up-Regulation/drug effects
12.
J Immunol ; 179(10): 6620-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982052

ABSTRACT

TLR activation of innate immunity prevents the induction of transplantation tolerance and shortens skin allograft survival in mice treated with costimulation blockade. The mechanism by which TLR signaling mediates this effect has not been clear. We now report that administration of the TLR agonists LPS (TLR4) or polyinosinic:polycytidylic acid (TLR3) to mice treated with costimulation blockade prevents alloreactive CD8(+) T cell deletion, primes alloreactive CTLs, and shortens allograft survival. The TLR4- and MyD88-dependent pathways are required for LPS to shorten allograft survival, whereas polyinosinic:polycytidylic acid mediates its effects through a TLR3-independent pathway. These effects are all mediated by signaling through the type 1 IFN (IFN-alphabeta) receptor. Administration of IFN-beta recapitulates the detrimental effects of TLR agonists on transplantation tolerance. We conclude that the type 1 IFN generated as part of an innate immune response to TLR activation can in turn activate adaptive immune responses that abrogate transplantation tolerance. Blocking of type 1 IFN-dependent pathways in patients may improve allograft survival in the presence of exogenous TLR ligands.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Innate , Interferon Type I/immunology , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Skin Transplantation/immunology , Transplantation Tolerance , Animals , Graft Survival/drug effects , Graft Survival/immunology , Immunity, Innate/drug effects , Interferon Inducers/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Poly I-C/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Transplantation Tolerance/drug effects , Transplantation, Homologous
13.
Blood ; 109(2): 819-26, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16973964

ABSTRACT

Allograft transplantation requires chronic immunosuppression, but there is no effective strategy to evaluate the long-term maintenance of immunosuppression other than assessment of graft function. The ability to monitor naive alloreactive T cells would provide an alternative guide for drug therapy at early, preclinical stages of graft rejection and for evaluating tolerance-inducing protocols. To detect and quantify naive alloreactive T cells directly ex vivo, we used the unique ability of naive T cells to rapidly produce TNF-alpha but not IFN-gamma. Naive alloreactive T cells were identified by the production of TNF-alpha after a 5-hour in vitro stimulation with alloantigen and were distinguished from effector/memory alloreactive T cells by the inability to produce IFN-gamma. Moreover, naive alloreactive T cells were not detected in mice tolerized against specific alloantigens. The frequency of TNF-alpha-producing cells was predictive for rejection in an in vivo cytotoxicity assay and correlated with skin allograft rejection. Naive alloreactive T cells were also detected in humans, suggesting clinical relevance. We conclude that rapid production of TNF-alpha can be used to quantify naive alloreactive T cells, that it is abrogated after the induction of tolerance, and that it is a potential tool to predict allograft rejection.


Subject(s)
Graft Rejection/immunology , T-Lymphocytes/immunology , Transplantation Tolerance , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/blood , Leukocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Skin Transplantation/immunology , T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/blood
14.
J Immunol ; 176(3): 1561-70, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16424185

ABSTRACT

Costimulation blockade protocols are effective in prolonging allograft survival in animal models and are entering clinical trials, but how environmental perturbants affect graft survival remains largely unstudied. We used a costimulation blockade protocol consisting of a donor-specific transfusion and anti-CD154 mAb to address this question. We observed that lymphocytic choriomeningitis virus infection at the time of donor-specific transfusion and anti-CD154 mAb shortens allograft survival. Lymphocytic choriomeningitis virus 1) activates innate immunity, 2) induces allo-cross-reactive T cells, and 3) generates virus-specific responses, all of which may adversely affect allograft survival. To investigate the role of innate immunity, mice given costimulation blockade and skin allografts were coinjected with TLR2 (Pam3Cys), TLR3 (polyinosinic:polycytidylic acid), TLR4 (LPS), or TLR9 (CpG) agonists. Costimulation blockade prolonged skin allograft survival that was shortened after coinjection by TLR agonists. To investigate underlying mechanisms, we used "synchimeric" mice which circulate trace populations of anti-H2b transgenic alloreactive CD8+ T cells. In synchimeric mice treated with costimulation blockade, coadministration of all four TLR agonists prevented deletion of alloreactive CD8+ T cells and shortened skin allograft survival. These alloreactive CD8+ T cells 1) expressed the proliferation marker Ki-67, 2) up-regulated CD44, and 3) failed to undergo apoptosis. B6.TNFR2-/- and B6.IL-12R-/- mice treated with costimulation blockade plus LPS also exhibited short skin allograft survival whereas similarly treated B6.CD8alpha-/- and TLR4-/- mice exhibited prolonged allograft survival. We conclude that TLR signaling abrogates the effects of costimulation blockade by preventing alloreactive CD8+ T cell apoptosis through a mechanism not dependent on TNFR2 or IL-12R signaling.


Subject(s)
Graft Enhancement, Immunologic , Graft Rejection/prevention & control , Growth Inhibitors/administration & dosage , Skin Transplantation/immunology , Toll-Like Receptors/agonists , Animals , Antibodies, Monoclonal/administration & dosage , Apoptosis/immunology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Clonal Deletion/drug effects , Female , Graft Rejection/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Radiation Chimera , Receptors, Interleukin/metabolism , Receptors, Interleukin-12 , Receptors, Tumor Necrosis Factor, Type II/metabolism , Toll-Like Receptor 4/metabolism , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...