Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 203(4): 844-852, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31324724

ABSTRACT

Programmed death-1 (PD-1) inhibits T and B cell function upon ligand binding. PD-1 blockade revolutionized cancer treatment, and although numerous patients respond, some develop autoimmune-like symptoms or overt autoimmunity characterized by autoantibody production. PD-1 inhibition accelerates autoimmunity in mice, but its role in regulating germinal centers (GC) is controversial. To address the role of PD-1 in the GC reaction in type 1 diabetes, we used tetramers to phenotype insulin-specific CD4+ T and B cells in NOD mice. PD-1 or PD-L1 deficiency, and PD-1 but not PD-L2 blockade, unleashed insulin-specific T follicular helper CD4+ T cells and enhanced their survival. This was concomitant with an increase in GC B cells and augmented insulin autoantibody production. The effect of PD-1 blockade on the GC was reduced when mice were treated with a mAb targeting the insulin peptide:MHC class II complex. This work provides an explanation for autoimmune side effects following PD-1 pathway inhibition and suggests that targeting the self-peptide:MHC class II complex might limit autoimmunity arising from checkpoint blockade.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , B7-H1 Antigen/immunology , Diabetes Mellitus, Experimental/immunology , Female , Germinal Center/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD
2.
Sci Rep ; 8(1): 8295, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844327

ABSTRACT

Type 1 diabetes is caused by autoreactive T cell-mediated ß cell destruction. Even though co-inhibitory receptor programmed death-1 (PD-1) restrains autoimmunity, the expression and regulation of its cognate ligands on ß cell remains unknown. Here, we interrogated ß cell-intrinsic programmed death ligand-1 (PD-L1) expression in mouse and human islets. We measured a significant increase in the level of PD-L1 surface expression and the frequency of PD-L1+ ß cells as non-obese diabetic (NOD) mice aged and developed diabetes. Increased ß cell PD-L1 expression was dependent on T cell infiltration, as ß cells from Rag1-deficient mice lacked PD-L1. Using Rag1-deficient NOD mouse islets, we determined that IFN-γ promotes ß cell PD-L1 expression. We performed analogous experiments using human samples, and found a significant increase in ß cell PD-L1 expression in type 1 diabetic samples compared to type 2 diabetic, autoantibody positive, and non-diabetic samples. Among type 1 diabetic samples, ß cell PD-L1 expression correlated with insulitis. In vitro experiments with human islets from non-diabetic individuals showed that IFN-γ promoted ß cell PD-L1 expression. These results suggest that insulin-producing ß cells respond to pancreatic inflammation and IFN-γ production by upregulating PD-L1 expression to limit self-reactive T cells.


Subject(s)
B7-H1 Antigen/metabolism , Diabetes Mellitus, Type 1/immunology , Interferon-gamma/metabolism , Islets of Langerhans/metabolism , T-Lymphocytes/immunology , Animals , Female , Humans , Interferon-gamma/biosynthesis , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD
3.
Diabetes ; 66(12): 3051-3060, 2017 12.
Article in English | MEDLINE | ID: mdl-28842400

ABSTRACT

Type 1 diabetes (T1D) results from T cell-mediated destruction of insulin-producing ß-cells. Insulin represents a key self-antigen in disease pathogenesis, as recent studies identified proinsulin-responding T cells from inflamed pancreatic islets of organ donors with recent-onset T1D. These cells respond to an insulin B-chain (InsB) epitope presented by the HLA-DQ8 molecule associated with high T1D risk. Understanding insulin-specific T-cell frequency and phenotype in peripheral blood is now critical. We constructed fluorescent InsB10-23:DQ8 tetramers, stained peripheral blood lymphocytes directly ex vivo, and show DQ8+ patients with T1D have increased tetramer+ CD4+ T cells compared with HLA-matched control subjects without diabetes. Patients with a shorter disease duration had higher frequencies of insulin-reactive CD4+ T cells, with most of these cells being antigen experienced. We also demonstrate that the number of insulin tetramer+ effector memory cells is directly correlated with insulin antibody titers, suggesting insulin-specific T- and B-cell interactions. Notably, one of four control subjects with tetramer+ cells was a first-degree relative who had insulin-specific cells with an effector memory phenotype, potentially representing an early marker of T-cell autoimmunity. Our results suggest that studying InsB10-23:DQ8 reactive T-cell frequency and phenotype may provide a biomarker of disease activity in patients with T1D and those at risk.


Subject(s)
Autoantibodies/blood , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immunologic Memory , Insulin Antibodies/blood , Insulin/immunology , Adult , Animals , Female , Histocompatibility Testing , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
4.
J Immunol ; 194(8): 3551-3555, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25769925

ABSTRACT

Programmed death-1 (PD-1) promotes T cell tolerance. Despite therapeutically targeting this pathway for chronic infections and tumors, little is known about how different T cell subsets are affected during blockade. We examined PD-1/PD ligand 1 (PD-L1) regulation of self-antigen-specific CD4 and CD8 T cells in autoimmune-susceptible models. PD-L1 blockade increased insulin-specific effector CD4 T cells in type 1 diabetes. However, anergic islet-specific CD4 T cells were resistant to PD-L1 blockade. Additionally, PD-L1 was critical for induction, but not maintenance, of CD8 T cell intestinal tolerance. PD-L1 blockade enhanced functionality of effector T cells, whereas established tolerant or anergic T cells were not dependent on PD-1/PD-L1 signaling to remain unresponsive. This highlights the existence of Ag-experienced T cell subsets that do not rely on PD-1/PD-L1 regulation. These findings illustrate how positive treatment outcomes and autoimmunity development during PD-1/PD-L1 inhibition are linked to the differentiation state of a T cell.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Anergy , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Disease Models, Animal , Disease Susceptibility/immunology , Disease Susceptibility/pathology , Female , Immune Tolerance/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Signal Transduction/genetics
5.
J Immunol ; 191(10): 4913-7, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24123682

ABSTRACT

Insulin-specific CD4(+) T cells are required for type 1 diabetes. How these cells are regulated and how tolerance breaks down are poorly understood because of a lack of reagents. Therefore, we used an enrichment method and tetramer reagents to track insulin-specific CD4(+) T cells in diabetes-susceptible NOD and resistant B6 mice expressing I-A(g7). Insulin-specific cells were detected in both strains, but they only became activated, produced IFN-γ, and infiltrated the pancreas in NOD mice. Unexpectedly, the majority of Ag-experienced cells in NOD mice displayed an anergic phenotype, but this population decreased with age as tolerance was lost. B6 mice expressing I-A(g7) were protected because insulin-specific cells did not become effector or anergic T cells but remained naive. These data suggest that NOD mice promote tolerance through anergy induction, but a small proportion of autoreactive T cells escape anergy to provoke type 1 diabetes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Animals , Insulin/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred NOD , Pancreas/cytology , Pancreas/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...