Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
2.
Diabetes ; 71(3): 483-496, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35007324

ABSTRACT

The induction of antigen (Ag)-specific tolerance and replacement of islet ß-cells are major ongoing goals for the treatment of type 1 diabetes (T1D). Our group previously showed that a hybrid insulin peptide (2.5HIP) is a critical autoantigen for diabetogenic CD4+ T cells in the NOD mouse model. In this study, we investigated whether induction of Ag-specific tolerance using 2.5HIP-coupled tolerogenic nanoparticles (NPs) could protect diabetic NOD mice from disease recurrence upon syngeneic islet transplantation. Islet graft survival was significantly prolonged in mice treated with 2.5HIP NPs, but not NPs containing the insulin B chain peptide 9-23. Protection in 2.5HIP NP-treated mice was attributed both to the simultaneous induction of anergy in 2.5HIP-specific effector T cells and the expansion of Foxp3+ regulatory T cells specific for the same Ag. Notably, our results indicate that effector function of graft-infiltrating CD4+ and CD8+ T cells specific for other ß-cell epitopes was significantly impaired, suggesting a novel mechanism of therapeutically induced linked suppression. This work establishes that tolerance induction with an HIP can delay recurrent autoimmunity in NOD mice, which could inform the development of an Ag-specific therapy for T1D.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Graft Survival/drug effects , Insulin/administration & dosage , Islets of Langerhans Transplantation/methods , Peptide Fragments/administration & dosage , Animals , Autoantigens/immunology , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/prevention & control , Female , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Nanoparticles/administration & dosage , Recurrence
3.
Sci Rep ; 11(1): 16182, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376755

ABSTRACT

Acute kidney injury (AKI) after transplantation of human deceased donor kidneys is associated with upregulation of tubular toll like receptor 4 (TLR4), but whether TLR4 is required for AKI is unknown. We hypothesized that TLR4 knockout mice (TLR4KO) subjected to cold ischemia followed by kidney transplant (CI + Txp) would be protected from AKI. C57Bl/6J wild type or TLR4KO kidneys were subjected to CI + Txp into wild type recipients. Tubular cell apoptosis, tubular injury and cast formation were significantly improved in recipients of TLR4KO kidneys. TLR4KO kidneys also demonstrated significantly decreased expression of the effector caspase 8. Brush border injury scores and serum creatinine were not different in recipients of TLR4KO versus wild type kidneys. Phosphorylated RIP3 and MLKL through which TLR4 signals programmed necrosis were expressed in both recipient groups. In addition, TNF-α and TNFR1 expression were significantly increased in recipient serum and TLR4KO kidneys respectively after CI + Txp, suggesting continued activation of programmed necrosis despite TLR4 deletion. Our results suggest that TLR4 deletion decreases apoptosis via inhibition of the death receptor pathway and decreases tubular injury and cast formation.


Subject(s)
Acute Kidney Injury/prevention & control , Apoptosis , Cold Ischemia/adverse effects , Kidney Transplantation/adverse effects , Necrosis , Toll-Like Receptor 4/physiology , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Tissue Donors/supply & distribution
4.
Mol Metab ; 53: 101323, 2021 11.
Article in English | MEDLINE | ID: mdl-34416394

ABSTRACT

BACKGROUND: The pancreatic ß cell, as the sole source of the vital hormone insulin, has been under intensive study for more than a century. Given the potential of newly created insulin-producing cells as a treatment or even cure of type 1 diabetes (T1D) and possibly in severe cases of type 2 diabetes (T2D), multiple academic and commercial laboratories are working to derive surrogate glucose-responsive, insulin-producing cells. SCOPE OF REVIEW: The recent development of advanced phenotyping technologies, including molecular, epigenomic, histological, or functional, have greatly improved our understanding of the critical properties of human ß cells. Using this information, here we summarize the salient features of normal, fully functional adult human ß cells, and propose minimal criteria for what should rightfully be termed 'ß cells' as opposed to insulin-producing but not fully-functional surrogates that we propose should be referred to as 'ß-like' cells or insulin-producing cells. MAJOR CONCLUSIONS: Clear criteria can be established to differentiate fully functional, mature ß cells from 'ß-like' surrogates. In addition, we outline important knowledge gaps that must be addressed to enable a greater understanding of the ß cell.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans
5.
Am J Transplant ; 21(11): 3550-3560, 2021 11.
Article in English | MEDLINE | ID: mdl-34014614

ABSTRACT

Chronic rejection is among the most pressing clinical challenges in solid organ transplantation. Interestingly, in a mouse model of heterotopic heart transplantation, antibody-dependent, natural killer (NK) cell-mediated chronic cardiac allograft vasculopathy occurs in some donor-recipient strain combinations, but not others. In this study, we sought to identify the mechanism underlying this unexplained phenomenon. Cardiac allografts from major histocompatibility complex (MHC) mismatched donors were transplanted into immune-deficient C57Bl/6.rag-/- recipients, followed by administration of a monoclonal antibody against the donor MHC class I antigen. We found marked allograft vasculopathy in hearts from C3H donors, but near-complete protection of BALB/c allografts from injury. We found no difference in recipient NK cell phenotype or intrinsic responsiveness to activating signals between recipients of C3H versus BALB/c allografts. However, cardiac endothelial cells from C3H allografts showed an approximately twofold higher expression of Rae-1, an activating ligand of the NK cell receptor natural killer group 2D (NKG2D). Importantly, the administration of a neutralizing antibody against NKG2D abrogated the development of allograft vasculopathy in recipients of C3H allografts, even in the presence of donor-specific antibodies. Therefore, the activating NK cell receptor NKG2D is necessary in this model of chronic cardiac allograft vasculopathy, and strain-dependent expression of NK activating ligands correlates with the development of this disease.


Subject(s)
Heart Transplantation , NK Cell Lectin-Like Receptor Subfamily K , Animals , Antibodies, Monoclonal , Endothelial Cells , Graft Rejection/etiology , Heart Transplantation/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Receptors, Natural Killer Cell
6.
Cell Signal ; 85: 110039, 2021 09.
Article in English | MEDLINE | ID: mdl-33991613

ABSTRACT

BACKGROUND: Caspase-1 knockout mice (Casp1KO) are protected from Acute Kidney Injury (AKI) after warm ischemia/reperfusion injury in non-transplant models. Since Caspase-1 plays a central role as an inflammatory response initiator, we hypothesized that Casp1KO mice would be protected from AKI following transplant. METHODS: Renal tubular cells (RTECs) were subjected to cold storage and rewarming (CS/REW). C57Bl/6 J wild type or Casp1KO kidneys were subjected to CI for 30 min and then transplanted into wild type recipients (CI + Txp). The recipients underwent bilateral native nephrectomy at the time of transplant. Serum creatinine (sCr) was measured 24 h after native nephrectomy to assess transplant function. RESULTS: We found that RTECs subjected to CS/REW had significantly increased expression of the Caspase-1 and inflammasome protein NLRP1. Wild type kidneys subjected to CI + Txp into wild type recipients also demonstrated significantly increased Caspase-1 and NLRP1 protein expression compared to kidneys transplanted from Casp1KO donors into wild type recipients. Caspase-1 deletion results in significantly decreased RTEC apoptosis in transplanted Casp1KO vs WT kidneys. Surprisingly, however, renal function, ATN scores including brush border injury, cast formation and tubular simplification were similar in both groups and not significantly different. CONCLUSIONS: Our data suggest that other triggers of inflammation and programmed necrosis may need to be inhibited in addition to attenuating Caspase-1 to fully prevent AKI after kidney transplant. Importantly, requirements may be distinct for AKI induced by transplantation as opposed to other transient models such as the clamp model of AKI.


Subject(s)
Acute Kidney Injury , Apoptosis , Caspase 1/deficiency , Reperfusion Injury , Animals , Caspase 1/metabolism , Kidney/metabolism , Mice , Mice, Inbred C57BL , Necrosis , Reperfusion Injury/metabolism
7.
Front Immunol ; 11: 580483, 2020.
Article in English | MEDLINE | ID: mdl-33117387

ABSTRACT

Memory T lymphocytes constitute a significant problem in tissue and organ transplantation due their contribution to early rejection and their relative resistance to tolerance-promoting therapies. Memory cells generated by environmental antigen exposure, as with T cells in general, harbor a high frequency of T cell receptors (TCR) spontaneously cross-reacting with allogeneic major histocompatibility complex (MHC) molecules. This phenomenon, known as 'heterologous' immunity, is thought to be a key barrier to transplant tolerance induction since such memory cells can potentially react directly with essentially any prospective allograft. In this review, we describe two additional concepts that expand this commonly held view of how memory cells contribute to transplant immunity and tolerance disruption. Firstly, autoimmunity is an additional response that can comprise an endogenously generated form of heterologous alloimmunity. However, unlike heterologous immunity generated as a byproduct of indiscriminate antigen sensitization, autoimmunity can generate T cells that have the unusual potential to interact with the graft either through the recognition of graft-bearing autoantigens or by their cross-reactive (heterologous) alloimmune specificity to MHC molecules. Moreover, we describe an additional pathway, independent of significant heterologous immunity, whereby immune memory to vaccine- or pathogen-induced antigens also may impair tolerance induction. This latter form of immune recognition indirectly disrupts tolerance by the licensing of naïve alloreactive T cells by vaccine/pathogen directed memory cells recognizing the same antigen-presenting cell in vivo. Thus, there appear to be recognition pathways beyond typical heterologous immunity through which memory T cells can directly or indirectly impact allograft immunity and tolerance.


Subject(s)
Graft Rejection/immunology , Organ Transplantation , T-Lymphocytes/immunology , Animals , Humans , Immunity, Heterologous , Immunologic Memory , Transplantation Tolerance , Transplantation, Homologous , Vaccination
8.
Am J Transplant ; 20(10): 2652-2668, 2020 10.
Article in English | MEDLINE | ID: mdl-32342639

ABSTRACT

The purpose of the STAR 2019 Working Group was to build on findings from the initial STAR report to further clarify the expectations, limitations, perceptions, and utility of alloimmune assays that are currently in use or in development for risk assessment in the setting of organ transplantation. The goal was to determine the precision and clinical feasibility/utility of such assays in evaluating both memory and primary alloimmune risks. The process included a critical review of biologically driven, state-of-the-art, clinical diagnostics literature by experts in the field and an open public forum in a face-to-face meeting to promote broader engagement of the American Society of Transplantation and American Society of Histocompatibility and Immunogenetics membership. This report summarizes the literature review and the workshop discussions. Specifically, it highlights (1) available assays to evaluate the attributes of HLA antibodies and their utility both as clinical diagnostics and as research tools to evaluate the effector mechanisms driving rejection; (2) potential assays to assess the presence of alloimmune T and B cell memory; and (3) progress in the development of HLA molecular mismatch computational scores as a potential prognostic biomarker for primary alloimmunity and its application in research trial design.


Subject(s)
Isoantibodies , Kidney Transplantation , Graft Rejection/diagnosis , Graft Rejection/etiology , Group Processes , HLA Antigens , Histocompatibility
9.
Curr Opin Organ Transplant ; 24(6): 694-698, 2019 12.
Article in English | MEDLINE | ID: mdl-31599762

ABSTRACT

PURPOSE OF REVIEW: To summarize recent findings linking donor-specific antibodies with innate immunity resulting in chronic allograft rejection. RECENT FINDINGS: Studies in recent years highlight the significance of donor-specific antibodies (DSA) in both acute and chronic allograft rejection. Since chronic rejection is the leading cause of graft failure, this review centers on the contribution of three areas of innate immunity of particular recent focus: complement, NK cells, and macrophages. Recent advances indicate the diverse roles that complement components play both in directly initiating allograft injury and indirectly by contributing to enhanced alloreactivity. NK cells also have emerged as an additional innate response that directly links DSA with chronic graft injury. Finally, recent studies identify alternatively activated macrophages as an additional arm of innate immunity contributing to chronic allograft rejection. SUMMARY: Chronic allograft rejection involves a significant contribution of DSA and differing pathways of the innate immune system. However, key issues remain unresolved. First, it is not always clear which of these varied sources of innate immunity contributing to chronic rejection may be antibody dependent. Moreover, it is not yet clear if these innate pathways represent independent routes that contribute to chronic rejection or rather act in concert to mediate allograft injury.


Subject(s)
Allografts/immunology , Graft Rejection/immunology , Immunity, Innate/immunology , Humans
10.
Cell Immunol ; 332: 101-110, 2018 10.
Article in English | MEDLINE | ID: mdl-30103941

ABSTRACT

The induction of tolerance to transplanted organs is a major objective in transplantation immunology research. Lymphocyte function-associated antigen-1 (LFA-1) interactions have been identified as a key component of the T-cell activation process that may be interrupted to lead to allograft tolerance. In mice, αLFA-1 mAb is a potent monotherapy that leads to the induction of donor-specific transferable tolerance. By interrogating important adaptive and innate immunity pathways, we demonstrate that the induction of tolerance relies on CD8+T-cells. We further demonstrate that αLFA-1 induced tolerance is associated with CD8+CD28-T-cells with a suppressor phenotype, and that while CD8 cells are present, the effector T-cell response is abrogated. A recent publication has shown that CD8+CD28- cells are not diminished by cyclosporine or rapamycin, therefore CD8+CD28- cells represent a clinically relevant population. To our knowledge, this is the first time that a mechanism for αLFA-1 induced tolerance has been described.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Graft Survival/immunology , Immune Tolerance/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Transplantation Tolerance/immunology , Animals , Antibodies, Monoclonal/immunology , CD28 Antigens/immunology , Cyclosporine/pharmacology , Female , Graft Rejection/drug therapy , Graft Rejection/immunology , Graft Survival/drug effects , Immune Tolerance/drug effects , Immunity, Innate/drug effects , Immunity, Innate/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Sirolimus/pharmacology , Transplantation Tolerance/drug effects , Transplantation, Homologous/methods
11.
Am J Transplant ; 18(7): 1604-1614, 2018 07.
Article in English | MEDLINE | ID: mdl-29603613

ABSTRACT

The presence of preexisting (memory) or de novo donor-specific HLA antibodies (DSAs) is a known barrier to successful long-term organ transplantation. Yet, despite the fact that laboratory tools and our understanding of histocompatibility have advanced significantly in recent years, the criteria to define presence of a DSA and assign a level of risk for a given DSA vary markedly between centers. A collaborative effort between the American Society for Histocompatibility and Immunogenetics and the American Society of Transplantation provided the logistical support for generating a dedicated multidisciplinary working group, which included experts in histocompatibility as well as kidney, liver, heart, and lung transplantation. The goals were to perform a critical review of biologically driven, state-of-the-art, clinical diagnostics literature and to provide clinical practice recommendations based on expert assessment of quality and strength of evidence. The results of the Sensitization in Transplantation: Assessment of Risk (STAR) meeting are summarized here, providing recommendations on the definition and utilization of HLA diagnostic testing, and a framework for clinical assessment of risk for a memory or a primary alloimmune response. The definitions, recommendations, risk framework, and highlighted gaps in knowledge are intended to spur research that will inform the next STAR Working Group meeting in 2019.


Subject(s)
Graft Survival/immunology , HLA Antigens/immunology , Histocompatibility/immunology , Isoantibodies/immunology , Organ Transplantation , Practice Guidelines as Topic/standards , Risk Assessment/methods , Tissue Donors , Humans , Research Report
12.
J Immunol ; 200(4): 1504-1512, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29311365

ABSTRACT

The autoimmune condition is a primary obstacle to inducing tolerance in type 1 diabetes patients receiving allogeneic pancreas transplants. It is unknown how autoreactive T cells that recognize self-MHC molecules contribute to MHC-disparate allograft rejection. In this report, we show the presence and accumulation of dual-reactive, that is autoreactive and alloreactive, T cells in C3H islet allografts that were transplanted into autoimmune diabetic NOD mice. Using high-throughput sequencing, we discovered that T cells prevalent in allografts share identical TCRs with autoreactive T cells present in pancreatic islets. T cells expressing TCRs that are enriched in allograft lesions recognized C3H MHC molecules, and five of six cell lines expressing these TCRs were also reactive to NOD islet cells. These results reveal the presence of autoreactive T cells that mediate cross-reactive alloreactivity, and indicate a requirement for regulating such dual-reactive T cells in tissue replacement therapies given to autoimmune individuals.


Subject(s)
Allografts/immunology , Autoantigens/immunology , Graft Rejection/immunology , Islets of Langerhans Transplantation/immunology , Animals , Diabetes Mellitus, Type 1/surgery , Mice , Mice, Inbred NOD , Receptors, Antigen, T-Cell/immunology
14.
Diabetes ; 66(4): 981-986, 2017 04.
Article in English | MEDLINE | ID: mdl-28069641

ABSTRACT

Diabetes is prevalent among solid organ transplant recipients and is universal among islet transplant recipients. Whereas diabetes is often considered to result in an immune-compromised state, the impact of chronic hyperglycemia on host alloimmunity is not clear. Potential immune-modifying effects of obesity, autoimmunity, or diabetogenic agents like streptozotocin may confound understanding alloimmunity in experimental models of diabetes. Therefore, we sought to determine the role of chronic hyperglycemia due to insulinopenia on alloimmunity using the nonautoimmune, spontaneously diabetic H-2b-expressing C57BL/6 Ins2Akita mice (Akita). Akita mice harbor a mutated Ins2 allele that dominantly suppresses insulin secretion, resulting in lifelong diabetes. We used BALB/c donors (H-2d) to assess alloimmunization and islet transplantation outcomes in Akita recipients. Surprisingly, chronic hyperglycemia had little effect on primary T-cell reactivity after alloimmunization. Moreover, Akita mice readily rejected islet allografts, and chronic hyperglycemia had no impact on the magnitude or quality of intragraft T-cell responses. In contrast, allospecific IgM and IgG were significantly decreased in Akita mice after alloimmunization. Thus, whereas diabetes influences host immune defense, hyperglycemia itself does not cause generalized alloimmune impairment. Our data suggest that immune compromise in diabetes due to hyperglycemia may not apply to cellular rejection of transplants.


Subject(s)
Diabetes Mellitus/immunology , Graft Rejection/immunology , Hyperglycemia/immunology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , T-Lymphocytes/immunology , Animals , Diabetes Mellitus/surgery , Insulin/genetics , Islets of Langerhans Transplantation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Transplantation, Homologous
15.
Diabetes ; 65(5): 1310-6, 2016 05.
Article in English | MEDLINE | ID: mdl-26718498

ABSTRACT

There is an ongoing need to develop strategic combinations of therapeutic agents to prevent type 1 diabetes (T1D) or to preserve islet ß-cell mass in new-onset disease. Although clinical trials using candidate therapeutics are commonly based on preclinical studies, concern is growing regarding the reproducibility as well as the potential clinical translation of reported results using animal models of human disorders. In response, the National Institutes of Health Immune Tolerance Network and JDRF established a multicenter consortium of academic institutions designed to assess the efficacy and intergroup reproducibility of clinically applicable immunotherapies for reversing new-onset disease in the NOD mouse model of T1D. Predicated on prior studies, this consortium conducted coordinated, prospective studies, using joint standard operating procedures, fixed criteria for study entry, and common reagents, to optimize combined anti-CD3 treatment plus interleukin-1 (IL-1) blockade to reverse new-onset disease in NOD mice. We did not find that IL-1 blockade with anti-IL-1ß monoclonal antibody or IL-1trap provided additional benefit for reversing new-onset disease compared with anti-CD3 treatment alone. These results demonstrate the value of larger, multicenter preclinical studies for vetting and prioritizing therapeutics for future clinical use.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Autoimmune Diseases/drug therapy , CD3 Complex/chemistry , Diabetes Mellitus, Type 1/drug therapy , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Interleukin-1beta/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Biomedical Research/methods , CD3 Complex/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Drug Administration Schedule , Drug Therapy, Combination , Female , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/therapeutic use , Immunotherapy/methods , Insulin Secretion , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Interleukin-1 Receptor Accessory Protein/metabolism , Interleukin-1beta/metabolism , Mice, Inbred NOD , Multicenter Studies as Topic , Pilot Projects , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Receptors, Interleukin-1 Type I/metabolism , Recombinant Fusion Proteins/therapeutic use , Reproducibility of Results , Research Design , Specific Pathogen-Free Organisms , United States
16.
Curr Opin Organ Transplant ; 21(1): 40-4, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26575853

ABSTRACT

PURPOSE OF REVIEW: The T cell-dependent recognition of allogeneic tissues and organs is complicated by the fact that both donor and host antigen-presenting cells can present donor antigens to host T cells. As such, these pathways result in T cells that can be restricted to either donor ('direct') or host ('indirect') major histocompatibility complex (MHC). These pathways are well recognized, but how these distinct patterns actually dictate allograft recognition is less clear. Thus, the purpose of the review is to summarize results from preclinical animal models in an attempt to clarify the distinct forms of allograft rejection dictated by these recognition pathways. RECENT FINDINGS: CD4 and CD8 donor MHC-restricted T cells are sufficient to reject allografts by a T-cell receptor-mediated direct ('cognate') interaction using a defined array of effector molecules. Conversely, 'noncognate' host MHC-restricted CD4 T cells must interact with intermediate host-type antigen-presenting cells and so greatly amplify the response by triggering antibody and inflammatory responses. SUMMARY: Importantly, 'cognate' CD4 and CD8 T cells have strikingly similar requirements for rejection, suggesting that this effector mechanism is dictated by the nature of allograft recognition rather than by T-cell subset. Conversely, 'noncognate' allograft recognition drives an increasingly appreciated role for inciting innate immunity in mediating allograft injury.


Subject(s)
Graft Rejection , Allografts , Animals , Antigen-Presenting Cells/immunology , Humans , T-Lymphocytes/immunology , Transplantation, Homologous
17.
J Immunol ; 195(1): 46-50, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26034174

ABSTRACT

In transplantation, a major obstacle for graft acceptance in MHC-matched individuals is the mismatch of minor histocompatibility Ags. Minor histocompatibility Ags are peptides derived from polymorphic proteins that can be presented by APCs on MHC molecules. The APC subtype uniquely responsible for the rejection of minor Ag-mismatched grafts has not yet been identified. In this study, we examined graft rejection in three mouse models: 1) mismatch of male-specific minor Ags, 2) mismatch of minor Ags distinct from male-specific minor Ags, and 3) skin transplant. This study demonstrates that in the absence of pathogen-associated molecular patterns, Batf3-dependent dendritic cells elicit the rejection of cells and grafts expressing mismatched minor Ags. The implication of our findings in clinical transplantation may be significant, as minor Ag reactivity has been implicated in the pathogenesis of multiple allograft tissues.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Dendritic Cells/immunology , Gene Expression Regulation, Developmental , Graft Rejection , Minor Histocompatibility Antigens/immunology , Repressor Proteins/immunology , Skin Transplantation , Adoptive Transfer , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Dendritic Cells/cytology , Female , Histocompatibility Testing , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Mice , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Repressor Proteins/genetics , Signal Transduction , Spleen/cytology , Spleen/immunology , Transplantation, Homologous
18.
Cell Immunol ; 293(1): 30-3, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25497973

ABSTRACT

Fas Ligand limits inflammatory injury and permits allograft survival by inducing apoptosis of Fas-bearing lymphocytes. Previous studies have shown that the CD4(+) T-cell is both sufficient and required for murine cardiac allograft rejection. Here, utilizing a transgenic mouse that over-expresses Fas Ligand specifically on cardiomyocytes as heart donors, we sought to determine if Fas Ligand on graft parenchymal cells could resist CD4(+) T-cell mediated rejection. When transplanted into fully immunocompetent BALB/c recipients Fas Ligand transgenic hearts were acutely rejected. However, when transplanted into CD4(+) T-cell reconstituted BALB/c-rag(-/-) recipients, Fas Ligand hearts demonstrated long-term survival. These results indicate that Fas Ligand over-expression on cardiomyocytes can indeed resist CD4(+) T-cell mediated cardiac rejection and suggests contact dependence between Fas Ligand expressing graft parenchymal cells and the effector CD4(+) T-cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fas Ligand Protein/immunology , Gene Expression/immunology , Graft Rejection/prevention & control , Graft Survival/genetics , Heart Transplantation , Animals , CD4-Positive T-Lymphocytes/cytology , Fas Ligand Protein/genetics , Female , Gene Deletion , Genes, RAG-1/immunology , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/immunology , Mice , Mice, Transgenic , Myocardium/cytology , Myocardium/immunology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/immunology , Transplantation, Heterotopic , Transplantation, Homologous
19.
J Immunol ; 191(4): 1686-91, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851694

ABSTRACT

An ongoing dilemma faced during an immune response is generating an effective, often proinflammatory response to eliminate pathogens and/or infected cells while also minimizing collateral damage to adjacent noninfected tissues. The factors limiting bystander cell injury during an Ag-specific immune response in vivo are largely unknown. In this study, using an in vivo model of islet transplants in TCR transgenic mice, we show that both CD4 and CD8 T cells do have the capacity to inflict adjacent tissue damage and that this injury is greatly enhanced in sensitized hosts. CD4 T cell-mediated killing of specific and bystander cells occurred via different mechanisms. Unlike specific target cell killing, CD4-mediated bystander injury required tissue Fas expression and was inhibited with anti-IFN-γ Ab treatment in vivo. Moreover, bystander cell injury was not entirely nonspecific but rather required, in naive recipients, that the MHC allele expressed by the bystanders was self. Importantly, the coinhibitor programmed death-1 plays an important role in restraining bystander cell injury mediated either by defined TCR transgenic T cells or by polyclonal T cell populations. Thus, the differential requirements for specific versus bystander cell injury suggest that there are opportunities for inhibiting immune pathology without compromising Ag-specific immunity in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Islets of Langerhans Transplantation/immunology , T-Lymphocyte Subsets/immunology , Animals , Bystander Effect/immunology , Cytotoxicity, Immunologic , Diabetes Mellitus, Experimental/surgery , Female , H-2 Antigens/immunology , Immunization , Inflammation , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/physiology , Islets of Langerhans Transplantation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Ovalbumin/immunology , Programmed Cell Death 1 Receptor , fas Receptor/immunology
20.
PLoS One ; 8(2): e54712, 2013.
Article in English | MEDLINE | ID: mdl-23405091

ABSTRACT

A recent type 1 diabetes (T1D) clinical trial of rituximab (a B cell-depleting anti-CD20 antibody) achieved some therapeutic benefit in preserving C-peptide for a period of approximately nine months in patients with recently diagnosed diabetes. Our previous data in the NOD mouse demonstrated that co-administration of antigen (insulin) with anti-CD3 antibody (a T cell-directed immunomodulator) offers better protection than either entity alone, indicating that novel combination therapies that include a T1D-related autoantigen are possible. To accelerate the identification and development of novel combination therapies that can be advanced into the clinic, we have evaluated the combination of a mouse anti-CD20 antibody with either oral insulin or a proinsulin-expressing DNA vaccine. Anti-CD20 alone, given once or on 4 consecutive days, produced transient B cell depletion but did not prevent or reverse T1D in the NOD mouse. Oral insulin alone (twice weekly for 6 weeks) was also ineffective, while proinsulin DNA (weekly for up to 12 weeks) showed a trend toward modest efficacy. Combination of anti-CD20 with oral insulin was ineffective in reversing diabetes in NOD mice whose glycemia was controlled with SC insulin pellets; these experiments were performed in three independent labs. Combination of anti-CD20 with proinsulin DNA was also ineffective in diabetes reversal, but did show modest efficacy in diabetes prevention (p = 0.04). In the prevention studies, anti-CD20 plus proinsulin resulted in modest increases in Tregs in pancreatic lymph nodes and elevated levels of proinsulin-specific CD4+ T-cells that produced IL-4. Thus, combination therapy with anti-CD20 and either oral insulin or proinsulin does not protect hyperglycemic NOD mice, but the combination with proinsulin offers limited efficacy in T1D prevention, potentially by augmentation of proinsulin-specific IL-4 production.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , B-Lymphocytes/drug effects , Insulin/administration & dosage , Proinsulin/administration & dosage , Proinsulin/genetics , Vaccines, DNA/administration & dosage , Administration, Oral , Animals , Antibodies, Monoclonal/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Drug Therapy, Combination/methods , Female , Hyperglycemia/drug therapy , Hyperglycemia/immunology , Insulin/genetics , Insulin/immunology , Interleukin-4/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/drug effects , Pancreas/immunology , Proinsulin/immunology , Spleen/drug effects , Spleen/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...