Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 606
Filter
1.
Cell Biochem Funct ; 42(1): e3895, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38050849

ABSTRACT

Autoimmune diseases are diseases in which the regulatory mechanisms of the immune response are disturbed. As a result, the body loses self-tolerance. Since one of the main regulatory mechanisms of the immune response is the CTLA4-CD80/86 axis, this hypothesis suggests that autoimmune diseases potentially share a similar molecular basis of pathogenesis. Hence, investigating the CTLA4-CD80/86 axis may be helpful in finding an appropriate treatment strategy. Therefore, this study aims to investigate the molecular basis of the CTLA4-CD80/86 axis in the regulation of the immune response, and then its role in developing some autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. As well, the main therapeutic strategies affecting the CTLA4-CD80/86 axis have been summarized to highlight the importance of this axis in management of autoimmune diseases.


Subject(s)
Autoimmune Diseases , Immunoconjugates , Humans , CTLA-4 Antigen , Antigens, CD , B7-2 Antigen , B7-1 Antigen/physiology , Autoimmune Diseases/therapy , Cell Adhesion Molecules
2.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34301886

ABSTRACT

Foxp3-expressing CD4+CD25+ regulatory T cells (Tregs) constitutively and highly express the immune checkpoint receptor cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), whose Treg-specific deficiency causes severe systemic autoimmunity. As a key mechanism of Treg-mediated suppression, Treg-expressed CTLA-4 down-regulates the expression of CD80/CD86 costimulatory molecules on antigen-presenting cells (APCs). Here, we show that Treg-expressed CTLA-4 facilitated Treg-APC conjugation and immune synapse formation. The immune synapses thus formed provided a stable platform whereby Tregs were able to deplete CD80/CD86 molecules on APCs by extracting them via CTLA-4-dependent trogocytosis. The depletion occurred even with Tregs solely expressing a mutant CTLA-4 form lacking the cytoplasmic portion required for its endocytosis. The CTLA-4-dependent trogocytosis of CD80/CD86 also accelerated in vitro and in vivo passive transfer of other membrane proteins and lipid molecules from APCs to Tregs without their significant reduction on the APC surface. Furthermore, CD80 down-regulation or blockade by Treg-expressed membrane CTLA-4 or soluble CTLA-4-immunoglobulin (CTLA-4-Ig), respectively, disrupted cis-CD80/programmed death ligand-1 (PD-L1) heterodimers and increased free PD-L1 on dendritic cells (DCs), expanding a phenotypically distinct population of CD80lo free PD-L1hi DCs. Thus, Tregs are able to inhibit the T cell stimulatory activity of APCs by reducing their CD80/CD86 expression via CTLA-4-dependent trogocytosis. This CD80/CD86 reduction on APCs is able to exert dual suppressive effects on T cell immune responses by limiting CD80/CD86 costimulation to naïve T cells and by increasing free PD-L1 available for the inhibition of programmed death-1 (PD-1)-expressing effector T cells. Blockade of CTLA-4 and PD-1/PD-L1 in combination may therefore synergistically hinder Treg-mediated immune suppression, thereby effectively enhancing immune responses, including tumor immunity.


Subject(s)
Antigen-Presenting Cells/immunology , B7-1 Antigen/physiology , B7-2 Antigen/physiology , B7-H1 Antigen/metabolism , CTLA-4 Antigen/physiology , T-Lymphocytes, Regulatory/immunology , Trogocytosis , Animals , B7-H1 Antigen/genetics , Dendritic Cells/immunology , Female , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
3.
Cancer Res ; 76(20): 6017-6029, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27569212

ABSTRACT

Certain cytotoxic chemotherapeutic drugs are immunogenic, stimulating tumor immunity through mechanisms that are not completely understood. Here we show how the DNA-damaging drug cisplatin modulates tumor immunity. At the maximum tolerated dose (MTD), cisplatin cured 50% of mice with established murine TC-1 or C3 tumors, which are preclinical models of human papillomavirus (HPV)-associated cancer. Notably, the curative benefit of cisplatin relied entirely upon induction of tumor-specific CD8+ T cells. Mechanistic investigations showed that cisplatin stimulated tumor infiltration of inflammatory antigen-presenting cells (APC) expressing relatively higher levels of the T-cell costimulatory ligands CD70, CD80, and CD86. Cell death triggered by cisplatin was associated with the release of at least 19 proteins in the tumor environment that could act as damage-associated molecular patterns and upregulate costimulatory molecules, either alone or in concert, but the responsible proteins remain unknown. Essentially, the curative effect of cisplatin was abrogated in mice lacking expression of CD80 and CD86 on APCs. Furthermore, cisplatin treatment was improved by CTLA-4 blockade, which increases the availability of CD80/86 to bind to CD28. In contrast, there was no effect of CD27 stimulation, which replaces CD70 interaction. At the cisplatin MTD, cure rates could also be increased by vaccination with synthetic long peptides, whereas cures could also be achieved at similar rates at 80% of the MTD with reduced side effects. Our findings reveal an essential basis for the immunogenic properties of cisplatin, which are mediated by the induction of costimulatory signals for CD8+ T-cell-dependent tumor destruction. Cancer Res; 76(20); 6017-29. ©2016 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , B7-1 Antigen/physiology , B7-2 Antigen/physiology , CD8-Positive T-Lymphocytes/immunology , Cisplatin/therapeutic use , Neoplasms, Experimental/drug therapy , Animals , Antigen-Presenting Cells/physiology , CD27 Ligand/physiology , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/physiology , Neoplasms, Experimental/immunology , Vaccination
4.
Mediators Inflamm ; 2015: 593075, 2015.
Article in English | MEDLINE | ID: mdl-26146464

ABSTRACT

Type 2 diabetes mellitus (T2D) is a metabolic disease that is strongly tied to obesity and often preceded by insulin resistance (IR). It has been established that chronic inflammation of hypertrophic adipose tissue depots in obese individuals leads to obesity-associated IR and is mediated by cells of the innate immune system, particularly macrophages. More recently, cells of the adaptive immune system, B and T lymphocytes, have also emerged as important regulators of glucose homeostasis, raising the intriguing possibility that antigen-driven immune responses play a role in disease. In this review, we critically evaluate the roles that various B and T cell subsets play in IR, and then we examine the data suggesting that antigen-driven mechanisms, such as antigen presentation and costimulation, may drive the activity of these lymphocytes.


Subject(s)
Adaptive Immunity , Insulin Resistance , Obesity/metabolism , Animals , Antigen Presentation , B-Lymphocytes/immunology , B7-1 Antigen/physiology , B7-2 Antigen/physiology , Humans , Immunologic Memory , T-Lymphocytes/immunology
5.
Nat Immunol ; 15(7): 631-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880458

ABSTRACT

Memory B cells (MBCs) are long-lived sources of rapid, isotype-switched secondary antibody-forming cell (AFC) responses. Whether MBCs homogeneously retain the ability to self-renew and terminally differentiate or if these functions are compartmentalized into MBC subsets has remained unclear. It has been suggested that antibody isotype controls MBC differentiation upon restimulation. Here we demonstrate that subcategorizing MBCs on the basis of their expression of CD80 and PD-L2, independently of isotype, identified MBC subsets with distinct functions upon rechallenge. CD80(+)PD-L2(+) MBCs differentiated rapidly into AFCs but did not generate germinal centers (GCs); conversely, CD80(-)PD-L2(-) MBCs generated few early AFCs but robustly seeded GCs. The gene-expression patterns of the subsets supported both the identity and function of these distinct MBC types. Hence, the differentiation and regeneration of MBCs are compartmentalized.


Subject(s)
B-Lymphocyte Subsets/immunology , B7-1 Antigen/physiology , Immunoglobulin Isotypes/physiology , Immunologic Memory , Programmed Cell Death 1 Ligand 2 Protein/physiology , Amino Acid Sequence , Animals , Antibody-Producing Cells/physiology , Germinal Center/immunology , Immunization , Mice , Mice, Inbred BALB C , Molecular Sequence Data , T-Lymphocytes/physiology
6.
Drug Des Devel Ther ; 8: 447-57, 2014.
Article in English | MEDLINE | ID: mdl-24872677

ABSTRACT

BACKGROUND: Atherosclerosis is based on a chronic inflammatory process including the innate and adaptive immune response. Costimulatory molecules and their receptors provide decisive signals for antigen-specific cell activation. The contribution of B7-related pathways to atherosclerosis has hardly been explored. METHODS: In the present study, we investigated the contribution of B7-1 to inflammation and tissue injury in the human plaque microenvironment in order to identify possible target structures of future therapeutic agents ex vivo and in vitro. RESULTS: Carotid artery plaque stimulation with lipopolysaccharides (LPS) could be significantly inhibited by RhuDex(®), a specific inhibitor of the costimulatory molecule B7-1 ex vivo (P<0.001). Coculture of antigen-presenting cells with T-cells demonstrated that the inhibitory effects of RhuDex(®) derived from reduced T-cell activation. In addition, incubation of monocytes/macrophages with LPS and RhuDex(®) resulted in an inhibitory negative feedback on antigen-presenting cells. Signaling pathways affected by RhuDex(®) seem to be nuclear transcription factor kappa B, activator protein-1, and extracellular signal-regulated kinase 1/2. CONCLUSION: The present data support B7-1 alone as an important costimulatory molecule in the context of LPS-mediated inflammation in atherosclerotic lesions. Due to its marked inhibitory effects, RhuDex(®) may be a useful therapy to modulate the inflammatory milieu in atherosclerosis.


Subject(s)
Atherosclerosis/pathology , B7-1 Antigen/antagonists & inhibitors , Inflammation/prevention & control , Atherosclerosis/immunology , B7-1 Antigen/physiology , Chemokines/biosynthesis , Cytokines/biosynthesis , Humans , Inflammation/etiology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , MAP Kinase Signaling System/drug effects , T-Lymphocytes/immunology
9.
J Am Soc Nephrol ; 25(7): 1415-29, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24676639

ABSTRACT

Podocyte injury and resulting albuminuria are hallmarks of diabetic nephropathy, but targeted therapies to halt or prevent these complications are currently not available. Here, we show that the immune-related molecule B7-1/CD80 is a critical mediator of podocyte injury in type 2 diabetic nephropathy. We report the induction of podocyte B7-1 in kidney biopsy specimens from patients with type 2 diabetes. Genetic and epidemiologic studies revealed the association of two single nucleotide polymorphisms at the B7-1 gene with diabetic nephropathy. Furthermore, increased levels of the soluble isoform of the B7-1 ligand CD28 correlated with the progression to ESRD in individuals with type 2 diabetes. In vitro, high glucose conditions prompted the phosphatidylinositol 3 kinase-dependent upregulation of B7-1 in podocytes, and the ectopic expression of B7-1 in podocytes increased apoptosis and induced disruption of the cytoskeleton that were reversed by the B7-1 inhibitor CTLA4-Ig. Podocyte expression of B7-1 was also induced in vivo in two murine models of diabetic nephropathy, and treatment with CTLA4-Ig prevented increased urinary albumin excretion and improved kidney pathology in these animals. Taken together, these results identify B7-1 inhibition as a potential therapeutic strategy for the prevention or treatment of diabetic nephropathy.


Subject(s)
B7-1 Antigen/physiology , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/etiology , Podocytes , Adult , Aged , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Up-Regulation
10.
Diabetes ; 63(4): 1289-302, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24222350

ABSTRACT

A key pathophysiologic role for activated T-cells in mediating adipose inflammation and insulin resistance (IR) has been recently postulated. However, mechanisms underlying their activation are poorly understood. In this study, we demonstrated a previously unrecognized homeostatic role for the costimulatory B7 molecules (CD80 and CD86) in preventing adipose inflammation. Instead of promoting inflammation, which was found in many other disease conditions, B7 costimulation reduced adipose inflammation by maintaining regulatory T-cell (Treg) numbers in adipose tissue. In both humans and mice, expression of CD80 and CD86 was negatively correlated with the degree of IR and adipose tissue macrophage infiltration. Decreased B7 expression in obesity appeared to directly impair Treg proliferation and function that lead to excessive proinflammatory macrophages and the development of IR. CD80/CD86 double knockout (B7 KO) mice had enhanced adipose macrophage inflammation and IR under both high-fat and normal diet conditions, accompanied by reduced Treg development and proliferation. Adoptive transfer of Tregs reversed IR and adipose inflammation in B7 KO mice. Our results suggest an essential role for B7 in maintaining Tregs and adipose homeostasis and may have important implications for therapies that target costimulation in type 2 diabetes.


Subject(s)
Adipose Tissue/pathology , B7-1 Antigen/physiology , B7-2 Antigen/physiology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/physiology , Adipose Tissue/immunology , Adoptive Transfer , Animals , Cell Proliferation , Homeostasis/physiology , Humans , Inflammation/immunology , Insulin Resistance/immunology , Macrophages/immunology , Mice , Mice, Knockout
11.
Chin Med J (Engl) ; 126(11): 2139-44, 2013.
Article in English | MEDLINE | ID: mdl-23769573

ABSTRACT

BACKGROUND: Despite extensive research, the mechanism of immature dendritic cells (DCs) induced immune hyporesponsiveness remains incompletely understood. METHODS: Recipient DCs from C3H mouse bone marrow cells were incubated with donor antigen from splenic lymphocytes of C57BL/6 mouse; these DCs were transfected with CD80/86 specific siRNA using lentiviral vectors. Flow cytometry was used to evaluate expression of CD80/86 on the antigen-pulsed recipient DCs. Immune regulatory activity was examined by mixed lymphocyte reaction, in which irradiated DCs were cultured with C3H spleen T cells. After the reaction, interleukin (IL)-2, IL-4, IL-10, and interferon (INF)-γ levels of mixed lymphocyte reaction culture supernatant were measured by enzyme-linked immunosorbent assay. The apoptotic T lymphocytes were identified by Annexin V and CD3 staining. RESULTS: There was a significant inhibition of CD80/86 expression in DCs transfected with CD80/86 lentiviral vectors compared with the control groups (P < 0.05), indicating the specificity of RNA interference. Enzyme-linked immunosorbent assay results showed a significant reduction of INF-γ, IL-2 and IL-10 in the CD80/86 lentivirus transfected group compared to the control groups (P < 0.05). There was no significant difference in IL-4 levels between the groups (P > 0.05). We also showed that CD80/86 low DCs loaded with alloantigen (1) stimulated low T cell proliferative responses via the indirect recognition pathway and (2) enhanced apoptotic activity (P < 0.05) in co-cultured T cells. CONCLUSIONS: Lentiviral vector transfection can effectively and specifically knock down target genes in DCs. The CD80/86 low DCs may show tolerogenic activity via induction of T-cell apoptosis, thereby modulating the activity of recipient-derived DCs. The use of this approach may potentially be clinically applicable.


Subject(s)
Apoptosis , B7-1 Antigen/physiology , B7-2 Antigen/physiology , Dendritic Cells/immunology , RNA Interference , T-Lymphocytes/immunology , Animals , B7-1 Antigen/genetics , B7-2 Antigen/genetics , Lentivirus/genetics , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , T-Lymphocytes/cytology
12.
J Leukoc Biol ; 94(2): 367-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23744647

ABSTRACT

Although T cell activation has been classically described to require distinct, positive stimulation signals that include B7-1 (CD80) and B7-2 (CD86) costimulation, overriding suppression signals that avert immune-mediated host injury are equally important. How these opposing stimulation and suppression signals work together remains incompletely defined. Our recent studies demonstrate that CD8 Teff activation in response to cognate peptide stimulation is actively suppressed by the Foxp3(+) subset of CD4 cells, called Tregs. Here, we show that the elimination of Treg suppression does not bypass the requirement for positive B7-1/B7-2 costimulation. The expansion, IFN-γ cytokine production, cytolytic, and protective features of antigen-specific CD8 T cells stimulated with purified cognate peptide in Treg-ablated mice were each neutralized effectively by CTLA-4-Ig that blocks B7-1/B7-2. In turn, given the efficiency whereby CTLA-4-Ig overrides the effects of Treg ablation, the role of Foxp3(+) cell-intrinsic CTLA-4 in mitigating CD8 Teff activation was also investigated. With the use of mixed chimera mice that contain CTLA-4-deficient Tregs exclusively after the ablation of WT Foxp3(+) cells, a critical role for Treg CTLA-4 in suppressing the expansion, cytokine production, cytotoxicity, and protective features of peptide-stimulated CD8 T cells is revealed. Thus, the activation of protective CD8 T cells requires positive B7-1/B7-2 costimulation even when suppression by Tregs and in particular, Treg-intrinsic CTLA-4 is circumvented.


Subject(s)
B7-1 Antigen/antagonists & inhibitors , B7-2 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Abatacept , Adoptive Transfer , Animals , B7-1 Antigen/deficiency , B7-1 Antigen/physiology , B7-2 Antigen/deficiency , B7-2 Antigen/physiology , Cytotoxicity, Immunologic , Forkhead Transcription Factors/analysis , Immunoconjugates/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Peptide Fragments/immunology , Radiation Chimera
13.
J Immunol ; 189(9): 4221-5, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018463

ABSTRACT

The mechanisms that link bacterial infection to solid organ rejection remain unclear. In this study, we show that following the establishment of lung allograft acceptance in mice, Pseudomonas aeruginosa airway infection induces a G-CSF-dependent neutrophilia that stimulates acute rejection. Graft-infiltrating neutrophils sharply upregulate the B7 molecules CD80 and CD86, but they do not express CD40 or MHC class II in response to P. aeruginosa infection. Neutrophil B7 promotes naive CD4(+) T cell activation and intragraft IL-2(+), IFN-γ(+), and IL-17(+) T lymphocyte accumulation. Intravital two-photon microscopy reveals direct interactions between neutrophils and CD4(+) T cells within pulmonary allografts. Importantly, lung rejection in P. aeruginosa-infected recipients is triggered by CD80/86 on neutrophils and can be prevented by B7 blockade without affecting clearance of this pathogen. These data show that neutrophils enhance T cell activation through B7 trans-costimulation and suggest that inhibiting neutrophil-mediated alloimmunity can be accomplished without compromising bacterial immune surveillance.


Subject(s)
B7-1 Antigen/physiology , Graft Rejection/etiology , Lung Transplantation/adverse effects , Neutrophils/immunology , Neutrophils/microbiology , Pseudomonas Infections/immunology , Transplantation Tolerance/immunology , Up-Regulation/immunology , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , Graft Rejection/immunology , Graft Rejection/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology
14.
Eur J Immunol ; 42(6): 1375-84, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22678893

ABSTRACT

Elevated levels of intracellular cyclic adenosine monophosphate (cAMP) in naturally occurring T regulatory (nTreg) cells play a key role in nTreg-cell-mediated suppression. Upon contact with nTreg cells, cAMP is transferred from nTreg cells into activated target CD4(+) T cells and/or antigen-presenting cells (APCs) via gap junctions to suppress CD4(+) T-cell function. cAMP facilitates the expression and nuclear function of a potent transcriptional inhibitor, inducible cAMP early repressor (ICER), resulting in ICER-mediated suppression of interleukin-2 (IL-2). Furthermore, ICER inhibits transcription of nuclear factor of activated T cell c1/α (NFATc1/α) and forms inhibitory complexes with preexisting NFATc1/c2, thereby inhibiting NFAT-driven transcription, including that of IL-2. In addition to its suppressive effects mediated via ICER, cAMP can also modulate the levels of surface-expressed cytotoxic T lymphocyte antigen-4 (CTLA-4) and its cognate B7 ligands on conventional CD4(+) T cells and/or APCs, fine-tuning suppression. These cAMP-driven nTreg-cell suppression mechanisms are the focus of this review.


Subject(s)
Cyclic AMP/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Antigen-Presenting Cells/physiology , B7-1 Antigen/physiology , CD28 Antigens/physiology , CTLA-4 Antigen/physiology , Cyclic AMP Response Element Modulator/physiology , Forkhead Transcription Factors/physiology , Homeostasis , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , NFATC Transcription Factors/metabolism , Signal Transduction
16.
Semin Nephrol ; 31(4): 320-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21839364

ABSTRACT

Minimal change disease is the most common nephrotic syndrome in children. Although the etiology of minimal change disease remains to be elucidated, it has been postulated that it is the result of a circulating T-cell factor that causes podocyte cytoskeleton disorganization leading to increased glomerular capillary permeability and/or changes in glomerular basement membrane heparan sulfate glycosaminoglycans resulting in proteinuria. Minimal change disease has been associated with allergies and Hodgkin disease. Consistent with these associations, a role for interleukin-13 with minimal change disease has been proposed. Furthermore, studies evaluating podocytes also have evolved. Recently, increased expression of CD80 (also termed B7-1) on podocytes was identified as a mechanism for proteinuria. CD80 is inhibited by binding to CTLA-4, which is expressed on regulatory T cells. Recently, we showed that urinary CD80 is increased in minimal change disease patients and limited studies have suggested that it is not commonly present in the urine of patients with other glomerular diseases. Interleukin-13 or microbial products via Toll-like receptors could be factors that induce CD80 expression on podocytes. CTLA-4 appears to regulate CD80 expression in podocytes, and to be altered in minimal change disease patients. These findings lead us to suggest that proteinuria in minimal change disease is caused by persistent CD80 expression in podocytes, possibly initiated by stimulation of these cells by antigens or cytokines.


Subject(s)
B7-1 Antigen/physiology , Nephrosis, Lipoid/etiology , Podocytes/physiology , Animals , CTLA-4 Antigen/physiology , Glomerulosclerosis, Focal Segmental/etiology , Graft vs Host Reaction , Humans , Interleukin-13/physiology , Nephrosis, Lipoid/immunology , Nephrotic Syndrome/etiology , Nephrotic Syndrome/immunology , T-Lymphocytes/immunology
17.
J Immunol ; 187(3): 1097-105, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21697456

ABSTRACT

Programmed death-1 ligand 1 (PD-L1) is a coinhibitory molecule that negatively regulates multiple tolerance checkpoints. In the NOD mouse model, PD-L1 regulates the development of diabetes. PD-L1 has two binding partners, programmed death-1 and B7-1, but the significance of the PD-L1:B7-1 interaction in regulating self-reactive T cell responses is not yet clear. To investigate this issue in NOD mice, we have compared the effects of two anti-PD-L1 Abs that have different blocking activities. Anti-PD-L1 mAb 10F.2H11 sterically and functionally blocks only PD-L1:B7-1 interactions, whereas anti-PD-L1 mAb 10F.9G2 blocks both PD-L1:B7-1 and PD-L1:programmed death-1 interactions. Both Abs had potent, yet distinct effects in accelerating diabetes in NOD mice: the single-blocker 10F.2H11 mAb was more effective at precipitating diabetes in older (13-wk-old) than in younger (6- to 7-wk-old) mice, whereas the dual-blocker 10F.9G2 mAb rapidly induced diabetes in NOD mice of both ages. Similarly, 10F.2H11 accelerated diabetes in recipients of T cells from diabetic, but not prediabetic mice, whereas 10F.9G2 was effective in both settings. Both anti-PD-L1 mAbs precipitated diabetes in adoptive transfer models of CD4(+) and CD8(+) T cell-driven diabetes. Taken together, these data demonstrate that the PD-L1:B7-1 pathway inhibits potentially pathogenic self-reactive effector CD4(+) and CD8(+) T cell responses in vivo, and suggest that the immunoinhibitory functions of this pathway may be particularly important during the later phases of diabetogenesis.


Subject(s)
B7-1 Antigen/physiology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Growth Inhibitors/physiology , Membrane Glycoproteins/physiology , Peptides/physiology , Signal Transduction/immunology , Adoptive Transfer , Animals , Antibodies, Blocking/administration & dosage , Antigens, CD/physiology , Apoptosis Regulatory Proteins/physiology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-H1 Antigen , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Diabetes Mellitus, Type 1/genetics , Female , Ligands , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Peptides/deficiency , Peptides/immunology , Programmed Cell Death 1 Receptor , Protein Binding/immunology , Signal Transduction/genetics
18.
J Immunol ; 187(3): 1113-9, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21697455

ABSTRACT

The programmed death ligand 1 (PDL1)/programmed death 1 (PD1) costimulatory pathway plays an important role in the inhibition of alloimmune responses as well as in the induction and maintenance of peripheral tolerance. It has been demonstrated recently that PDL1 also can bind B7.1 to inhibit T cell responses in vitro. Using the bm12 into B6 heart transplant model, we investigated the functional significance of this interaction in alloimmune responses in vivo. PD1 blockade unlike PDL1 blockade failed to accelerate bm12 allograft rejection, suggesting a role for an additional binding partner for PDL1 other than PD1 in transplant rejection. PDL1 blockade was able to accelerate allograft rejection in B7.2-deficient recipients but not B7.1-deficient recipients, indicating that PDL1 interaction with B7.1 was important in inhibiting rejection. Administration of the novel 2H11 anti-PDL1 mAb, which only blocks the PDL1-B7.1 interaction, aggravated chronic injury of bm12 allografts in B6 recipients. Aggravated chronic injury was associated with an increased frequency of alloreactive IFN-γ-, IL-4-, and IL-6-producing splenocytes and a decreased percentage of regulatory T cells in the recipients. Using an in vitro cell culture assay, blockade of the interaction of PDL1 on dendritic cells with B7.1 on T cells increased IFN-γ production from alloreactive CD4(+) T cells, whereas blockade of dendritic cell B7.1 interaction with T cell PDL1 did not. These data indicate that PDL1 interaction with B7.1 plays an important role in the inhibition of alloimmune responses in vivo and suggests a dominant direction for PDL1 and B7.1 interaction.


Subject(s)
Antigens, CD/physiology , Apoptosis Regulatory Proteins/physiology , B7-1 Antigen/physiology , Lymphocyte Activation/immunology , Self Tolerance/immunology , Signal Transduction/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Apoptosis Regulatory Proteins/deficiency , B7-1 Antigen/genetics , Cells, Cultured , Graft Survival/genetics , Graft Survival/immunology , Heart Transplantation/immunology , Ligands , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor , Self Tolerance/genetics , Signal Transduction/genetics , Skin Transplantation/immunology
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 27(4): 357-9, 363, 2011 Apr.
Article in Chinese | MEDLINE | ID: mdl-21481304

ABSTRACT

AIM: To establish a ubiquitously expressed PD-L1 transgenic mouse model and evaluate its recovery of motor function after spinal cord injury. METHODS: Clone and sequence the complete mouse PD-L1 cDNA and construct the pCAG-PD-L1 transgenic vector by inserting the PD-L1 cDNA into the pCAGGS vector. The PD-L1 transgenic (TgPD-L1) mice were established by pronuclear micro-injection with fertilized eggs from C57BL/6 mice and the genotypes were confirmed by PCR with tail genomic DNA. The expression of PD-L1 on T and B lymphocytes from mouse spleen were detected by flow cytometry. The expression of PD-L1 in peripheral tissues was displayed by immunohistochemistry. The expression level of PD-L1 in spinal tissue was evaluated by RT-PCR. The recovery of motor function was analyzed by Basso-Beattie-Bresnahan(BBB) locomotion testing system at 3, 7, 14, 21, 28 and 35 day after spinal severe crush with forceps in mice. RESULTS: Three lines of TgPD-L1 mice in C57BL/6 background were generated and the exogenous PD-L1 gene can be heritable steadily to offsprings. PD-L1 was highly exppressed in spinal tissue, peripheral tissues, T and B lymphocytes using RT-PCR, immunohistochemistry and flow cytometry respectively in TgPD-L1 mice. The BBB scores were obviously higher at 21 day post-injury in TgPD-L1 than those of in WT mice (P<0.05). CONCLUSION: The TgPD-L1 mice whose background are C57BL/6 were established successfully and high expression level of PD-L1 in tissues promotes locomotion recovery after spinal cord injury in TgPD-L1 mice.


Subject(s)
B7-1 Antigen/genetics , Locomotion , Membrane Glycoproteins/genetics , Peptides/genetics , Spinal Cord Injuries/physiopathology , Animals , B7-1 Antigen/analysis , B7-1 Antigen/physiology , B7-H1 Antigen , Disease Models, Animal , Female , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Peptides/analysis , Peptides/physiology
20.
Am J Transplant ; 11(4): 832-40, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21401869

ABSTRACT

The PD1:PDL1 pathway is an essential negative costimulatory pathway that plays a key role in regulating the alloimune response. PDL1 is expressed not only on antigen-presenting cells (APCs) but also cardiac endothelium. In this study, we investigated the importance of PDL1 expression on donor cardiac allograft in acquired transplantation tolerance in a fully MHC-mismatched model. We generated PDL1 chimeric mice on B6 background that expressed PDL1 on either hematopoietic cells or nonhematopoietic cells of the heart. Sham animals were used as controls. These hearts were then transplanted into BALB/c recipients and treated with CTLA4-Ig to induce tolerance. Cardiac endothelium showed significant expression of PDL1, which was upregulated upon transplantation. While the absence of PDL1 on hematopoietic cells of the heart resulted in delayed rejection and prevented long-term tolerance in most but not all recipients, we observed an accelerated and early graft rejection of all donor allografts that lacked PDL1 on the endothelium. Moreover, PDL1-deficient endothelium hearts had significant higher frequency of IFN-γ-producing alloreactive cells as well as higher frequency of CD8(+) effector T cells. These findings demonstrate that PDL1 expression mainly on donor endothelium is functionally important in a fully allogeneic mismatched model for the induction of cardiac allograft tolerance.


Subject(s)
B7-1 Antigen/physiology , Bone Marrow/metabolism , Endothelium, Vascular/metabolism , Heart Transplantation , Membrane Glycoproteins/physiology , Peptides/physiology , Transplantation Tolerance , Animals , B7-H1 Antigen , Flow Cytometry , Fluorescent Antibody Technique , Graft Rejection , Hematopoietic Stem Cells/metabolism , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...