Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Clin Immunol ; 245: 109165, 2022 12.
Article in English | MEDLINE | ID: mdl-36257528

ABSTRACT

The receptor for advanced glycation endproducts (RAGE) is involved in multiple inflammatory processes. RAGE participates in adaptive and innate immune responses but its role in human immune cell responses has not been directly tested in vivo. We treated humanized mice (NSG) with the small molecule antagonist of RAGE, azeliragon, (AZ), and measured effects on xenogeneic (B6) skin graft rejection. AZ delayed the median time to xenograft rejection (22 vs 56 days, P = 0.0001). PD-1 expression on CD4+ and CD8+ T cells was lower following AZ therapy. Transcriptome studies showed inhibition of pathways in splenocytes with AZ including IL-23, IL-17A and IL-1ß signaling. The serum levels of IL-1ß and IL-17A in AZ treated mice were reduced in mice that did not reject skin grafts. The RAGE antagonist prevented xenograft rejection by human immune cells in a murine model. A RAGE antagonist may be a useful inhibitor of adaptive human immune responses.


Subject(s)
Graft Rejection , Interleukin-17 , Humans , Mice , Animals , Receptor for Advanced Glycation End Products/metabolism , Heterografts , Transplantation, Heterologous , Mice, Inbred C57BL
2.
JCI Insight ; 7(17)2022 09 08.
Article in English | MEDLINE | ID: mdl-35925682

ABSTRACT

Checkpoint inhibitors (CPIs) targeting programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have revolutionized cancer treatment but can trigger autoimmune complications, including CPI-induced diabetes mellitus (CPI-DM), which occurs preferentially with PD-1 blockade. We found evidence of pancreatic inflammation in patients with CPI-DM with shrinkage of pancreases, increased pancreatic enzymes, and in a case from a patient who died with CPI-DM, peri-islet lymphocytic infiltration. In the NOD mouse model, anti-PD-L1 but not anti-CTLA-4 induced diabetes rapidly. RNA sequencing revealed that cytolytic IFN-γ+CD8+ T cells infiltrated islets with anti-PD-L1. Changes in ß cells were predominantly driven by IFN-γ and TNF-α and included induction of a potentially novel ß cell population with transcriptional changes suggesting dedifferentiation. IFN-γ increased checkpoint ligand expression and activated apoptosis pathways in human ß cells in vitro. Treatment with anti-IFN-γ and anti-TNF-α prevented CPI-DM in anti-PD-L1-treated NOD mice. CPIs targeting the PD-1/PD-L1 pathway resulted in transcriptional changes in ß cells and immune infiltrates that may lead to the development of diabetes. Inhibition of inflammatory cytokines can prevent CPI-DM, suggesting a strategy for clinical application to prevent this complication.


Subject(s)
Diabetes Mellitus , Programmed Cell Death 1 Receptor , Animals , Humans , Inflammation Mediators , Mice , Mice, Inbred NOD , Tumor Necrosis Factor Inhibitors
3.
Nat Commun ; 12(1): 5074, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34417463

ABSTRACT

ß cells may participate and contribute to their own demise during Type 1 diabetes (T1D). Here we report a role of their expression of Tet2 in regulating immune killing. Tet2 is induced in murine and human ß cells with inflammation but its expression is reduced in surviving ß cells. Tet2-KO mice that receive WT bone marrow transplants develop insulitis but not diabetes and islet infiltrates do not eliminate ß cells even though immune cells from the mice can transfer diabetes to NOD/scid recipients. Tet2-KO recipients are protected from transfer of disease by diabetogenic immune cells.Tet2-KO ß cells show reduced expression of IFNγ-induced inflammatory genes that are needed to activate diabetogenic T cells. Here we show that Tet2 regulates pathologic interactions between ß cells and immune cells and controls damaging inflammatory pathways. Our data suggests that eliminating TET2 in ß cells may reduce activating pathologic immune cells and killing of ß cells.


Subject(s)
DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 1/pathology , Inflammation/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Proto-Oncogene Proteins/metabolism , Animals , Base Sequence , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Dioxygenases , Disease Progression , Female , Humans , Immunity , Inflammation/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , T-Lymphocytes/immunology , Transcription, Genetic
4.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093268

ABSTRACT

The microbiome affects development and activity of the immune system, and may modulate immune therapies, but there is little direct information about this control in vivo. We studied how the microbiome affects regulation of human immune cells in humanized mice. When humanized mice were treated with a cocktail of 4 antibiotics, there was an increase in the frequency of effector T cells in the gut wall, circulating levels of IFN-γ, and appearance of anti-nuclear antibodies. Teplizumab, a non-FcR-binding anti-CD3ε antibody, no longer delayed xenograft rejection. An increase in CD8+ central memory cells and IL-10, markers of efficacy of teplizumab, were not induced. IL-10 levels were only decreased when the mice were treated with all 4 but not individual antibiotics. Antibiotic treatment affected CD11b+CD11c+ cells, which produced less IL-10 and IL-27, and showed increased expression of CD86 and activation of T cells when cocultured with T cells and teplizumab. Soluble products in the pellets appeared to be responsible for the reduced IL-27 expression in DCs. Similar changes in IL-10 induction were seen when human peripheral blood mononuclear cells were cultured with human stool samples. We conclude that changes in the microbiome may impact the efficacy of immunosuppressive medications by altering immune regulatory pathways.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Adaptive Immunity/immunology , Animals , Antibodies, Antinuclear , Antibodies, Monoclonal, Humanized/pharmacology , Autoimmune Diseases/immunology , Autoimmune Diseases/microbiology , B7-2 Antigen/metabolism , CD11b Antigen , CD11c Antigen , CD3 Complex , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Gastrointestinal Tract/microbiology , Graft Rejection/immunology , Humans , Immunosuppressive Agents/pharmacology , Immunotherapy , Interferon-gamma , Interleukin-10/metabolism , Interleukin-27/metabolism , Mice , Mice, Knockout , Mucous Membrane/immunology , STAT5 Transcription Factor/metabolism , Skin Transplantation , T-Lymphocytes/immunology , Transplantation, Heterologous
5.
Clin Immunol ; 183: 240-246, 2017 10.
Article in English | MEDLINE | ID: mdl-28739191

ABSTRACT

Oral administration of biologics may be a feasible approach for immune therapy that improves drug safety and potentiates mechanisms of tolerance at mucosal barriers. We tested the ability of a fully human non-FcR binding anti-CD3 mAb, foralumab, to prevent skin xenograft rejection in mice with human immune systems. At an intragastric dose of 15µg, the drug could transit through the small bowel. Serum absorption and binding of lymphoid cells was seen and proliferative responses of splenic CD8+ T cells to mitogen were reduced. Five consecutive daily doses, then weekly dosing led to indefinite graft acceptance without depletion of peripheral T cells. Proliferative and cytokine responses to activation of splenocytes with PHA were reduced. The serum levels of IL-10 but not TNF were increased 6days after application of the skin graft. Oral treatment with anti-CD3 mAb may represent a feasible approach for immune modulation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD3 Complex/immunology , CD3 Complex/metabolism , Graft Rejection/prevention & control , Skin Transplantation , Administration, Oral , Animals , Cell Proliferation , Humans , Mice , T-Lymphocytes
6.
Cell Metab ; 25(3): 727-738, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28190773

ABSTRACT

Type 1 diabetes (T1D) is a chronic autoimmune disease that involves immune-mediated destruction of ß cells. How ß cells respond to immune attack is unknown. We identified a population of ß cells during the progression of T1D in non-obese diabetic (NOD) mice that survives immune attack. This population develops from normal ß cells confronted with islet infiltrates. Pathways involving cell movement, growth and proliferation, immune responses, and cell death and survival are activated in these cells. There is reduced expression of ß cell identity genes and diabetes antigens and increased immune inhibitory markers and stemness genes. This new subpopulation is resistant to killing when diabetes is precipitated with cyclophosphamide. Human ß cells show similar changes when cultured with immune cells. These changes may account for the chronicity of the disease and the long-term survival of ß cells in some patients.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Progression , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Animals , Antibodies, Monoclonal/pharmacology , CD3 Complex/metabolism , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Female , Humans , Immunotherapy , Insulin-Secreting Cells/drug effects , Lymphocytes/drug effects , Lymphocytes/pathology , Mice, Inbred NOD , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Transcription, Genetic/drug effects , Transcriptome/drug effects , Transcriptome/genetics
7.
J Biol Chem ; 291(21): 11230-40, 2016 May 20.
Article in English | MEDLINE | ID: mdl-26994137

ABSTRACT

Type 1 diabetes mellitus is caused by the killing of insulin-producing ß cells by CD8+T cells. The disease progression, which is chronic, does not follow a course like responses to conventional antigens such as viruses, but accelerates as glucose tolerance deteriorates. To identify the unique features of the autoimmune effectors that may explain this behavior, we analyzed diabetogenic CD8+ T cells that recognize a peptide from the diabetes antigen IGRP (NRP-V7-reactive) in prediabetic NOD mice and compared them to others that shared their phenotype (CD44(+)CD62L(lo)PD-1(+)CXCR3(+)) but negative for diabetes antigen tetramers and to LCMV (lymphocytic choriomeningitis)-reactive CD8+ T cells. There was an increase in the frequency of the NRP-V7-reactive cells coinciding with the time of glucose intolerance. The T cells persisted in hyperglycemic NOD mice maintained with an insulin pellet despite destruction of ß cells. We compared gene expression in the three groups of cells compared with the other two subsets of cells, and the NRP-V7-reactive cells exhibited gene expression of memory precursor effector cells. They had reduced cellular proliferation and were less dependent on oxidative phosphorylation. When prediabetic NOD mice were treated with 2-deoxyglucose to block aerobic glycolysis, there was a reduction in the diabetes antigen versus other cells of similar phenotype and loss of lymphoid cells infiltrating the islets. In addition, treatment of NOD mice with 2-deoxyglucose resulted in improved ß cell granularity. These findings identify a link between metabolic disturbances and autoreactive T cells that promotes development of autoimmune diabetes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Animals , Insulin , Islets of Langerhans/immunology , Mice, Inbred NOD , Mice, Transgenic , Prediabetic State/immunology
8.
Diabetologia ; 59(5): 1021-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26910463

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes is caused by the immunological destruction of pancreatic beta cells. Preclinical and clinical data indicate that there are changes in beta cell function at different stages of the disease, but the fate of beta cells has not been closely studied. We studied how immune factors affect the function and epigenetics of beta cells during disease progression and identified possible triggers of these changes. METHODS: We studied FACS sorted beta cells and infiltrating lymphocytes from NOD mouse and human islets. Gene expression was measured by quantitative real-time RT-PCR (qRT-PCR) and methylation of the insulin genes was investigated by high-throughput and Sanger sequencing. To understand the role of DNA methyltransferases, Dnmt3a was knocked down with small interfering RNA (siRNA). The effects of cytokines on methylation and expression of the insulin gene were studied in humans and mice. RESULTS: During disease progression in NOD mice, there was an inverse relationship between the proportion of infiltrating lymphocytes and the beta cell mass. In beta cells, methylation marks in the Ins1 and Ins2 genes changed over time. Insulin gene expression appears to be most closely regulated by the methylation of Ins1 exon 2 and Ins2 exon 1. Cytokine transcription increased with age in NOD mice, and these cytokines could induce methylation marks in the insulin DNA by inducing methyltransferases. Similar changes were induced by cytokines in human beta cells in vitro. CONCLUSIONS/INTERPRETATION: Epigenetic modification of DNA by methylation in response to immunological stressors may be a mechanism that affects insulin gene expression during the progression of type 1 diabetes.


Subject(s)
DNA Methylation/genetics , DNA/genetics , Insulin-Secreting Cells/metabolism , Insulin/genetics , Adult , Animals , Cytokines , Diabetes Mellitus, Type 1 , Female , Humans , Mice , Mice, Inbred NOD
9.
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
10.
J Clin Invest ; 125(11): 4212-22, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26524592

ABSTRACT

FOXP3+ Tregs are central for the maintenance of self-tolerance and can be defective in autoimmunity. In multiple sclerosis and type-1 diabetes, dysfunctional self-tolerance is partially mediated by a population of IFNγ-secreting Tregs. It was previously reported that increased NaCl concentrations promote the induction of proinflammatory Th17 cells and that high-salt diets exacerbate experimental models of autoimmunity. Here, we have shown that increasing NaCl, either in vitro or in murine models via diet, markedly impairs Treg function. NaCl increased IFNγ secretion in Tregs, and reducing IFNγ - either by neutralization with anti-IFNγ antibodies or shRNA-mediated knockdown - restored suppressive activity in Tregs. The heightened IFNγ secretion and loss of Treg function were mediated by the serum/glucocorticoid-regulated kinase (SGK1). A high-salt diet also impaired human Treg function and was associated with the induction of IFNγ-secreting Tregs in a xenogeneic graft-versus-host disease model and in adoptive transfer models of experimental colitis. Our results demonstrate a putative role for an environmental factor that promotes autoimmunity by inducing proinflammatory responses in CD4 effector cells and Treg pathways.


Subject(s)
Autoimmunity/drug effects , Sodium Chloride, Dietary/adverse effects , Sodium Chloride/pharmacology , T-Lymphocytes, Regulatory/drug effects , Adoptive Transfer , Animals , Antibodies, Neutralizing/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Colitis/immunology , Cytokines/biosynthesis , Cytokines/genetics , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Genes, Reporter , Graft vs Host Disease/immunology , Heterografts , Humans , Immediate-Early Proteins/physiology , Inflammation , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/transplantation , Male , Mice , Protein Serine-Threonine Kinases/physiology , RNA Interference , RNA, Small Interfering/genetics , Sodium Chloride, Dietary/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
11.
J Clin Invest ; 124(10): 4603-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25202978

ABSTRACT

Regulatory T cells (Tregs), which express CD4 and FOXP3, are critical for modulating the immune response and promoting immune tolerance. Consequently, methods to expand Tregs for therapeutic use are of great interest. While transfer of Tregs after massive ex vivo expansion can be achieved, in vivo expansion of Tregs would be more practical. Here, we demonstrate that targeting the CD45 tyrosine phosphatase with a tolerogenic anti-CD45RB mAb acutely increases Treg numbers in WT mice, even in absence of exogenous antigen. Treg expansion occurred through substantial augmentation of homeostatic proliferation in the preexisting Treg population. Moreover, anti-CD45RB specifically increased Treg proliferation in response to cognate antigen. Compared with conventional T cells, Tregs differentially regulate their conjugation with DCs. Therefore, we determined whether CD45 ligation could alter interactions between Tregs and DCs. Live imaging showed that CD45 ligation specifically reduced Treg motility in an integrin-dependent manner, resulting in enhanced interactions between Tregs and DCs in vivo. Increased conjugate formation, in turn, augmented nuclear translocation of nuclear factor of activated T cells (NFAT) and Treg proliferation. Together, these results demonstrate that Treg peripheral homeostasis can be specifically modulated in vivo to promote Treg expansion and tolerance by increasing conjugation between Tregs and DCs.


Subject(s)
Dendritic Cells/cytology , Leukocyte Common Antigens/metabolism , T-Lymphocytes, Regulatory/cytology , Animals , Cell Proliferation , Female , Forkhead Transcription Factors/metabolism , Homeostasis , Immune Tolerance , Interleukin-2/metabolism , Ligands , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/metabolism
12.
J Immunol ; 193(2): 587-96, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24943216

ABSTRACT

Immune-deficient mice, reconstituted with human stem cells, have been used to analyze human immune responses in vivo. Although they have been used to study immune responses to xenografts, allografts, and pathogens, there have not been models of autoimmune disease in which the mechanisms of the pathologic process can be analyzed. We have found that reconstituted "humanized" mice treated with anti-CTLA-4 Ab (ipilimumab) develop autoimmune disease characterized by hepatitis, adrenalitis, sialitis, anti-nuclear Abs, and weight loss. Induction of autoimmunity involved activation of T cells and cytokine production, and increased infiltration of APCs. When anti-CTLA-4 mAb-treated mice were cotreated with anti-CD3 mAb (teplizumab), hepatitis and anti-nuclear Abs were no longer seen and weight loss did not occur. The anti-CD3 blocked proliferation and activation of T cells, release of IFN-γ and TNF, macrophage infiltration, and release of IP-10 that was induced with anti-CTLA-4 mAb. We also found increased levels of T regulatory cells (CD25(+)CD127(-)) in the spleen and mesenteric lymph nodes in the mice treated with both Abs and greater constitutive phosphorylation of STAT5 in T regulatory cells in spleen cells compared with mice treated with anti-CTLA-4 mAb alone. We describe a model of human autoimmune disease in vivo. Humanized mice may be useful for understanding the mechanisms of biologics that are used in patients. Hepatitis, lymphadenopathy, and other inflammatory sequelae are adverse effects of ipilimumab treatment in humans, and this study may provide insights into this pathogenesis and the effects of immunologics on autoimmunity.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Autoimmune Diseases/therapy , Disease Models, Animal , Stem Cell Transplantation/methods , T-Lymphocytes/immunology , Adrenal Glands/drug effects , Adrenal Glands/immunology , Adrenal Glands/metabolism , Animals , Antibodies, Monoclonal/toxicity , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Cytokines/blood , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Ipilimumab , Liver/drug effects , Liver/immunology , Liver/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phosphorylation/drug effects , Phosphorylation/immunology , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transplantation, Heterologous , Weight Loss/drug effects , Weight Loss/immunology
13.
Diabetes ; 62(5): 1676-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23423576

ABSTRACT

Type 1 diabetes (T1D) results from immune-mediated destruction of insulin-producing ß-cells. The killing of ß-cells is not currently measurable; ß-cell functional studies routinely used are affected by environmental factors such as glucose and cannot distinguish death from dysfunction. Moreover, it is not known whether immune therapies affect killing. We developed an assay to identify ß-cell death by measuring relative levels of unmethylated INS DNA in serum and used it to measure ß-cell death in a clinical trial of teplizumab. We studied 43 patients with recent-onset T1D, 13 nondiabetic subjects, and 37 patients with T1D treated with FcR nonbinding anti-CD3 monoclonal antibody (teplizumab) or placebo. Patients with recent-onset T1D had higher rates of ß-cell death versus nondiabetic control subjects, but patients with long-standing T1D had lower levels. When patients with recent-onset T1D were treated with teplizumab, ß-cell function was preserved (P < 0.05) and the rates of ß-cell were reduced significantly (P < 0.05). We conclude that there are higher rates of ß-cell death in patients with recent-onset T1D compared with nondiabetic subjects. Improvement in C-peptide responses with immune intervention is associated with decreased ß-cell death.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Cytotoxicity, Immunologic/drug effects , Diabetes Mellitus, Type 1/therapy , Immunologic Factors/therapeutic use , Immunotherapy , Insulin-Secreting Cells/drug effects , Adult , C-Peptide/blood , CD3 Complex/chemistry , Cytotoxicity Tests, Immunologic , DNA Methylation , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Female , Humans , Hypoglycemic Agents/therapeutic use , Insulin/genetics , Insulin/metabolism , Insulin/therapeutic use , Insulin Secretion , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Male , Postprandial Period , Young Adult
14.
Sci Transl Med ; 4(118): 118ra12, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22277969

ABSTRACT

The development and optimization of immune therapies in patients has been hampered by the lack of preclinical models in which their effects on human immune cells can be studied. As a result, observations that have been made in preclinical studies have suggested mechanisms of drug action in murine models that have not been confirmed in clinical studies. Here, we used a humanized mouse reconstituted with human hematopoietic stem cells to study the mechanism of action of teplizumab, an Fc receptor nonbinding humanized monoclonal antibody to CD3 being tested in clinical trials for the treatment of patients with type 1 diabetes mellitus. In this model, human gut-tropic CCR6(+) T cells exited the circulation and secondary lymph organs and migrated to the small intestine. These cells then produced interleukin-10 (IL-10), a regulatory cytokine, in quantities that could be detected in the peripheral circulation. Blocking T cell migration to the small intestine with natalizumab, which prevents cellular adhesion by inhibiting α(4) integrin binding, abolished the treatment effects of teplizumab. Moreover, IL-10 expression by CD4(+)CD25(high)CCR6(+)FoxP3 cells returning to the peripheral circulation was increased in patients with type 1 diabetes treated with teplizumab. These findings demonstrate that humanized mice may be used to identify novel immunologic mechanisms that occur in patients treated with immunomodulators.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , CD3 Complex/immunology , Cell Movement/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/metabolism , Gastrointestinal Tract/drug effects , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Interleukin-10/metabolism , Intestine, Small/cytology , Intestine, Small/drug effects , Intestine, Small/immunology , L-Selectin/metabolism , Mice , Mucous Membrane/cytology , Mucous Membrane/drug effects , Mucous Membrane/immunology , Natalizumab , Oligonucleotide Array Sequence Analysis , Receptors, CCR6/metabolism
15.
J Immunol ; 186(5): 2809-18, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270412

ABSTRACT

Rapamycin (Rapa), an immunosuppressive drug that acts through mammalian target of Rapa inhibition, broadly synergizes with tolerogenic agents in animal models of transplantation and autoimmunity. Rapa preferentially inhibits conventional CD4(+) Foxp3(-) T cells (Tconv) and promotes outgrowth of CD4(+)Foxp3(+) regulatory T cells (Treg) during in vitro expansion. Moreover, Rapa is widely perceived as augmenting both expansion and conversion of Treg in vivo. However, most quantitative studies were performed in lymphopenic hosts or in graft-versus-host disease models. We show in this study that in replete wild-type mice, Rapa significantly inhibits both homeostatic and alloantigen-induced proliferation of Treg, and promotes their apoptosis. Together, these lead to significant Treg depletion. Tconv undergo depletion to a similar degree, resulting in no change in the percent of Treg among CD4 cells. Moreover, in this setting, there was no evidence of conversion of Tconv into Treg. However, after withdrawal of Rapa, Treg recover Ag-induced proliferation more quickly than Tconv, leading to recovery to baseline numbers and an increase in the percent of Treg compared with Tconv. These findings suggest that the effects of Rapa on Treg survival, homeostasis, and induction, depend heavily on the cellular milieu and degree of activation. In vivo, the resistance of Treg to mammalian target of Rapa inhibition is relative and results from lymphopenic and graft-versus-host disease models cannot be directly extrapolated to settings more typical of solid organ transplantation or autoimmunity. Moreover, these results have important implications for the timing of Rapa therapy with tolerogenic agents designed to increase the number of Treg in vivo.


Subject(s)
Cell Proliferation , Homeostasis/immunology , Isoantigens/physiology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/physiology , Animals , Cell Survival/drug effects , Cell Survival/immunology , Gene Knock-In Techniques , Homeostasis/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocyte Depletion , Lymphoid Tissue/cytology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/drug effects , Sirolimus/administration & dosage , Skin Transplantation/immunology , Skin Transplantation/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/transplantation
16.
Ann N Y Acad Sci ; 1150: 217-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19120299

ABSTRACT

Type 1 diabetes is an autoimmune disease characterized by T cell-mediated destruction of pancreatic islet beta cells. Pancreatic islet transplantation with long-term immunosuppressive drug treatment is an accepted therapeutic option for patients with type 1 diabetes suffering from disabling hypoglycemia on insulin treatment. Here we investigated the replacement of immunosuppressive drug treatment with immune tolerance establishment induced by temporary B cell-depletion therapy for islet transplantation. The result suggested that the combined therapy of B cell depletion and syngeneic islet transplantation may reverse the disease in hCD20/NOD mice.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Diabetes Mellitus, Type 1/prevention & control , Graft Survival/drug effects , Islets of Langerhans Transplantation/methods , Animals , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Cell Separation , Diabetes Mellitus, Type 1/immunology , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Male , Mice , Mice, Inbred NOD , Secondary Prevention , Transplantation, Isogeneic
17.
J Immunol ; 176(4): 2292-8, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16455985

ABSTRACT

The role of CTLA-4 in tolerance is primarily inferred from knockout and blocking studies. Anti-CD45RB mediates allograft tolerance in mice by inducing CTLA-4 expression on CD4 cells, providing a novel opportunity to determine how therapeutic enhancement of CTLA-4 promotes tolerance. We now show that induced CTLA-4 expression normally resolves by day 17. Although thymectomy prolongs enhanced CTLA-4 expression, long-term engraftment is unaffected. To address the temporal relationship between increased CTLA-4 expression and engraftment, transplantation was delayed for various times after anti-CD45RB treatment. Delaying transplantation for 7 days (when CTLA-4 expression had peaked but treatment mAb was no longer detectable), resulted in long-term engraftment comparable to transplantation with no delay (day 0). Delaying transplantation from 10 to 18 days led to a progressively poorer outcome as CTLA-4 expression returned to baseline. This suggested that Ag exposure while CTLA-4 expression is enhanced is sufficient to induce long-term engraftment. To substantiate this, on day 0, anti-CD45RB-treated mice received BALB/c vs unrelated alloantigen, followed by transplantation of BALB/c islets 10 days later. Whereas recipients exposed to unrelated Ag experienced acute rejection, recipients exposed to donor Ag achieved long-term engraftment. Anti-CD45RB-treated mice exposed to alloantigen exhibited anergic CD4(+)CD25(-) effector cells and regulatory CD4(+)CD25(+) cells. Moreover, CD25 depletion in the peritransplant period prevented anti-CD45RB-mediated engraftment. Thus, exposure of CD4 cells expressing CTLA-4 to donor Ag is necessary and sufficient to induce long-term engraftment which appears to be mediated by both regulation and anergy.


Subject(s)
Antigens, Differentiation/metabolism , Antigens/immunology , Islets of Langerhans Transplantation/immunology , Transplantation Tolerance/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, CD , CTLA-4 Antigen , Leukocyte Common Antigens/immunology , Male , Mice , Mice, Inbred BALB C , Thymectomy , Time Factors , Transplantation, Homologous
18.
J Immunol ; 174(10): 6161-8, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879112

ABSTRACT

Immune responses are suppressed in immunologically privileged sites, which may provide a unique opportunity to prolong allograft survival. However, it is unknown whether testicular immune privilege promotes transplantation tolerance. Mechanisms underlying immune privilege are also not well understood. Here we found that islet transplantation in the testis, an immunologically privileged site, generates much less memory CD8(+) T cells but induces more Ag-specific CD4(+)CD25(+) regulatory T cells than in a conventional site. These CD4(+)CD25(+) cells exhibited the suppression of alloimmune responses in vivo and in vitro. Despite the immune regulation, intratesticular islet allografts all were rejected within 42 days after transplantation although they survived longer than renal subcapsular islet allografts. However, blocking CD40/CD40L costimulation induced the tolerance of intratesticular, but not renal subcapsular, islet allografts. Tolerance to intratesticular islet allografts spread to skin allografts in the non-privileged sites. Either transfer of memory CD8(+) T cells or deletion of CD25(+) T cells in vivo broke islet allograft tolerance. Thus, transplantation tolerance requires both costimulatory blockade, which suppresses acute allograft rejection, and a favorable balance between memory and regulatory T cells that could favorably prevent late allograft failure. These findings reveal novel mechanisms of immune privilege and provide direct evidence that testicular immune privilege fosters the induction of transplantation tolerance to allografts in both immunologically privileged and non-privileged sites.


Subject(s)
Immunologic Memory , Islets of Langerhans Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Testis/immunology , Transplantation Tolerance/immunology , Animals , Antibodies, Blocking/pharmacology , Apoptosis/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Graft Rejection/immunology , Graft Rejection/pathology , Growth Inhibitors/pharmacology , Islets of Langerhans Transplantation/pathology , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-2/biosynthesis , T-Lymphocytes, Regulatory/metabolism , Transplantation, Heterotopic/immunology , Transplantation, Heterotopic/pathology
19.
J Immunol ; 174(3): 1165-70, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661869

ABSTRACT

Foreign Ags that enter immunologically privileged sites such as the eye, brain, and testis persist for an extended period of time, whereas the same Ags are rapidly eliminated at conventional sites. Immune privilege, therefore, provides unwanted refuge for pathogens and tumor cells but is beneficial for the survival of allogeneic grafts. In this study, we asked whether memory T cells can eliminate foreign Ags deposited at an immunologically privileged site by studying CD8 memory T cell-mediated rejection of pancreatic islet allografts placed either in the testis (a privileged organ) or under the kidney capsule (a nonprivileged site) of diabetic mice. We found that CD8 memory T cells reject intratesticular grafts at a significantly slower rate than the rejection of intrarenal grafts. Delayed graft rejection in the testis was not due to reduced homing or proliferation of memory T cells but due to their increased apoptosis at that site. Apoptosis was mediated by the combined actions of two TNFR family members that are up-regulated on activated memory T cells, Fas, and CD30. Therefore, memory T cells survey immunologically privileged tissues but are subject to the immunosuppressive mechanisms present at these sites.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Testis/cytology , Testis/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Movement/genetics , Cell Movement/immunology , Epitopes, T-Lymphocyte/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/methods , Islets of Langerhans Transplantation/pathology , Ki-1 Antigen/physiology , Kidney/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Testis/pathology , Transplantation, Heterotopic/immunology , Transplantation, Heterotopic/methods , Transplantation, Heterotopic/pathology , fas Receptor/physiology
20.
J Immunol ; 171(11): 5673-7, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14634073

ABSTRACT

Knockout and blocking studies have shown a critical role for CTLA-4 in peripheral tolerance, however, it is unknown whether augmenting CTLA-4 expression actually promotes tolerance. Here we demonstrate a specific and requisite role for CTLA-4 and its up-regulation in tolerance through anti-CD45RB. First, long-term murine islet allograft survival induced by anti-CD45RB is prevented by CTLA4-Ig, which interferes with B7:CTLA-4 interactions. Second, anti-CD45RB is ineffective in recipients lacking CTLA-4, B7-1, and B7-2. In contrast, CTLA4-Ig, which targets B7 on allogeneic cells, promotes long-term engraftment in these mice. Moreover, anti-CD45RB was effective in B7-deficient controls expressing CTLA-4. Finally, in wild-type mice, CTLA-4 expression returned to baseline 17 days after receiving anti-CD45RB, and was refractory to further increase. Transplantation and anti-CD45RB therapy at this time could neither augment CTLA-4 nor prolong engraftment. These data demonstrate a specific role for CTLA-4 in anti-CD45RB-mediated tolerance and indicate that CTLA-4 up-regulation can directly promote allograft survival.


Subject(s)
Adjuvants, Immunologic/biosynthesis , Antigens, Differentiation/biosynthesis , Graft Enhancement, Immunologic , Transplantation Tolerance/immunology , Abatacept , Adjuvants, Immunologic/deficiency , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD , Antigens, Differentiation/genetics , Antigens, Differentiation/physiology , CTLA-4 Antigen , Graft Enhancement, Immunologic/methods , Graft Survival/genetics , Graft Survival/immunology , Immunoconjugates/administration & dosage , Injections, Intraperitoneal , Injections, Intravenous , Islets of Langerhans Transplantation/immunology , Leukocyte Common Antigens/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Transplantation Tolerance/genetics , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...