Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters

Publication year range
1.
Immunity ; 42(3): 499-511, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25769610

ABSTRACT

CD4(+) T cells differentiate into phenotypically distinct T helper cells upon antigenic stimulation. Regulation of plasticity between these CD4(+) T-cell lineages is critical for immune homeostasis and prevention of autoimmune disease. However, the factors that regulate lineage stability are largely unknown. Here we investigate a role for retinoic acid (RA) in the regulation of lineage stability using T helper 1 (Th1) cells, traditionally considered the most phenotypically stable Th subset. We found that RA, through its receptor RARα, sustains stable expression of Th1 lineage specifying genes, as well as repressing genes that instruct Th17-cell fate. RA signaling is essential for limiting Th1-cell conversion into Th17 effectors and for preventing pathogenic Th17 responses in vivo. Our study identifies RA-RARα as a key component of the regulatory network governing maintenance and plasticity of Th1-cell fate and defines an additional pathway for the development of Th17 cells.


Subject(s)
Cell Lineage/drug effects , Receptors, Retinoic Acid/genetics , T-Lymphocytes, Helper-Inducer/drug effects , Th1 Cells/drug effects , Th17 Cells/drug effects , Tretinoin/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Expression Regulation , Gene Regulatory Networks , Homeostasis/drug effects , Homeostasis/immunology , Integrases/genetics , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Retinoic Acid/immunology , Retinoic Acid Receptor alpha , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Tretinoin/immunology
2.
PLoS Biol ; 19(4): e3001199, 2021 04.
Article in English | MEDLINE | ID: mdl-33901179

ABSTRACT

Programmed cell death protein 1 (PD-1) is expressed on T cells upon T cell receptor (TCR) stimulation. PD-1 ligand 1 (PD-L1) is expressed in most tumor environments, and its binding to PD-1 on T cells drives them to apoptosis or into a regulatory phenotype. The fact that PD-L1 itself is also expressed on T cells upon activation has been largely neglected. Here, we demonstrate that PD-L1 ligation on human CD25-depleted CD4+ T cells, combined with CD3/TCR stimulation, induces their conversion into highly suppressive T cells. Furthermore, this effect was most prominent in memory (CD45RA-CD45RO+) T cells. PD-L1 engagement on T cells resulted in reduced ERK phosphorylation and decreased AKT/mTOR/S6 signaling. Importantly, T cells from rheumatoid arthritis patients exhibited high basal levels of phosphorylated ERK and following PD-L1 cross-linking both ERK signaling and the AKT/mTOR/S6 pathway failed to be down modulated, making them refractory to the acquisition of a regulatory phenotype. Altogether, our results suggest that PD-L1 signaling on memory T cells could play an important role in resolving inflammatory responses; maintaining a tolerogenic environment and its failure could contribute to ongoing autoimmunity.


Subject(s)
B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/physiology , T-Lymphocytes, Regulatory/physiology , B7-H1 Antigen/physiology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Transdifferentiation/genetics , Cell Transdifferentiation/immunology , Cohort Studies , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunologic Memory/physiology , Leukocyte Common Antigens/metabolism , Phenotype , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/physiology , Signal Transduction/physiology , T-Lymphocytes, Regulatory/metabolism
3.
Blood ; 137(8): 1090-1103, 2021 02 25.
Article in English | MEDLINE | ID: mdl-32976550

ABSTRACT

The nuclear receptor (NR) subclass, retinoid X receptors (RXRs), exert immunomodulatory functions that control inflammation and metabolism via homodimers and heterodimers, with several other NRs, including retinoic acid receptors. IRX4204 is a novel, highly specific RXR agonist in clinical trials that potently and selectively activates RXR homodimers, but not heterodimers. In this study, in vivo IRX4204 compared favorably with FK506 in abrogating acute graft-versus-host disease (GVHD), which was associated with inhibiting allogeneic donor T-cell proliferation, reducing T-helper 1 differentiation, and promoting regulatory T-cell (Treg) generation. Recipient IRX4204 treatment reduced intestinal injury and decreased IFN-γ and TNF-α serum levels. Transcriptional analysis of donor T cells isolated from intestines of GVHD mice treated with IRX4204 revealed significant decreases in transcripts regulating proinflammatory pathways. In vitro, inducible Treg differentiation from naive CD4+ T cells was enhanced by IRX4204. In vivo, IRX4204 increased the conversion of donor Foxp3- T cells into peripheral Foxp3+ Tregs in GVHD mice. Using Foxp3 lineage-tracer mice in which both the origin and current FoxP3 expression of Tregs can be tracked, we demonstrated that IRX4204 supports Treg stability. Despite favoring Tregs and reducing Th1 differentiation, IRX4204-treated recipients maintained graft-versus-leukemia responses against both leukemia and lymphoma cells. Notably, IRX4204 reduced in vitro human T-cell proliferation and enhanced Treg generation in mixed lymphocyte reaction cultures. Collectively, these beneficial effects indicate that targeting RXRs with IRX4204 could be a novel approach to preventing acute GVHD in the clinic.


Subject(s)
Bone Marrow Transplantation , Cyclopropanes/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Leukemia Effect/drug effects , Retinoid X Receptors/agonists , Animals , Bone Marrow Transplantation/adverse effects , Drug Repositioning , Female , Graft vs Host Disease/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology
4.
Semin Immunol ; 42: 101308, 2019 04.
Article in English | MEDLINE | ID: mdl-31604531

ABSTRACT

V-domain Ig suppressor of T cell activation (VISTA) is a novel checkpoint regulator with limited homology to other B7 family members. The constitutive expression of VISTA on both the myeloid and T lymphocyte lineages coupled to its important role in regulating innate and adaptive immune responses, qualifies VISTA to be a promising target for immunotherapeutic intervention. Studies have shown differential impact of agonistic and antagonistic targeting of VISTA, providing a unique landscape for influencing the outcome of cancer and inflammatory diseases.


Subject(s)
B7 Antigens/immunology , Neoplasms/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Immunotherapy , Neoplasms/therapy
5.
Int J Mol Sci ; 24(18)2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37762046

ABSTRACT

Radiation therapy (RT) has recently demonstrated promise at stimulating an enhanced immune response. The recent success of immunotherapies, such as checkpoint inhibitors, CART cells, and other immune modulators, affords new opportunities for combination with radiation. The aim of this study is to evaluate whether and to what extent blockade of VISTA, an immune checkpoint, can potentiate the tumor control ability of radiation therapy. Our study is novel in that it is the first comparison of two VISTA-blocking methods (antibody inhibition and genetic knockout) in combination with RT. VISTA was blocked either through genetic knockout (KO) or an inhibitory antibody and combined with RT in two syngeneic murine flank tumor models (B16 and MC38). Selected mRNA, immune cell infiltration, and tumor growth delay were used to assess the biological effects. When combined with a single 15Gy radiation dose, VISTA blockade via genetic knockout in the B16 model and via anti-VISTA antibodies in the MC38 model significantly improved survival compared to RT alone by an average of 5.5 days and 6.3 days, respectively (p < 0.05). The gene expression data suggest that the mechanism behind the enhanced tumor control is primarily a result of increased apoptosis and immune-mediated cytotoxicity. VISTA blockade significantly enhances the anti-tumor effect of a single dose of 15Gy radiation through increased expression and stimulation of cell-mediated apoptosis pathways. These results suggest that VISTA is a biologically relevant immune promoter that has the potential to enhance the efficacy of a large single radiation dose in a synergic manner.


Subject(s)
Adenocarcinoma , Melanoma , Animals , Mice , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Antibodies , Disease Models, Animal , Melanoma/drug therapy , Melanoma/radiotherapy , T-Lymphocytes , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
6.
Physiol Rev ; 95(1): 125-48, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25540140

ABSTRACT

Although vitamin A was recognized as an "anti-infective vitamin" over 90 years ago, the mechanism of how vitamin A regulates immunity is only beginning to be understood. Early studies which focused on the immune responses in vitamin A-deficient (VAD) animals clearly demonstrated compromised immunity and consequently increased susceptibility to infectious disease. The active form of vitamin A, retinoic acid (RA), has been shown to have a profound impact on the homing and differentiation of leukocytes. Both pharmacological and genetic approaches have been applied to the understanding of how RA regulates the development and differentiation of various immune cell subsets, and how RA influences the development of immunity versus tolerance. These studies clearly show that RA profoundly impacts on cell- and humoral-mediated immunity. In this review, the early findings on the complex relationship between VAD and immunity are discussed as well as vitamin A metabolism and signaling within hematopoietic cells. Particular attention is focused on how RA impacts on T-cell lineage commitment and plasticity in various diseases.


Subject(s)
Leukocytes/physiology , Tretinoin/metabolism , Animals , Humans , Immunity, Cellular , Immunity, Humoral , Leukocytes/drug effects , Leukocytes/immunology , Signal Transduction , Tretinoin/pharmacology
7.
J Immunol ; 202(9): 2795-2805, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30885956

ABSTRACT

Recent studies have underscored the critical role of retinoic acid (RA) in the development of lineage-committed CD4 and CD8 T cells in vivo. We have shown that under acute graft-versus-host disease (GVHD) inflammatory conditions, RA is upregulated in the intestine and is proinflammatory, as GVHD lethality was attenuated when donor allogeneic T cells selectively expressed a dominant negative RA receptor α that blunted RA signaling. RA can function in an autocrine and paracrine fashion, and as such, the host cell lineage responsible for the production of RA metabolism and the specific RA-metabolizing enzymes that potentiate GVHD severity are unknown. In this study, we demonstrate that enhancing RA degradation in the host and to a lesser extent donor hematopoietic cells by overexpressing the RA-catabolizing enzyme CYP26A1 reduced GVHD. RA production is facilitated by retinaldehyde isoform-2 (RALDH2) preferentially expressed in dendritic cells (DCs). Conditionally deleted RA-synthesizing enzyme RALDH2 in host or to a lesser extent donor DCs reduced GVHD lethality. Improved survival in recipients with RALDH2-deleted DCs was associated with increased T cell death, impaired T effector function, increased regulatory T cell frequency, and augmented coinhibitory molecule expression on donor CD4+ T cells. In contrast, retinaldehydrogenase isoform-1 (RALDH1) is dominantly expressed in intestinal epithelial cells. Unexpectedly, conditional host intestinal epithelial cells RALDH1 deletion failed to reduce GVHD. These data demonstrate the critical role of both donor and especially host RALDH2+ DCs in driving murine GVHD and suggest RALDH2 inhibition or CYP26A1 induction as novel therapeutic strategies to prevent GVHD.


Subject(s)
Aldehyde Oxidoreductases/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Expression Regulation, Enzymologic/immunology , Graft vs Host Disease/immunology , Aldehyde Oxidoreductases/genetics , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/immunology , Tretinoin/immunology
8.
Immunol Rev ; 276(1): 66-79, 2017 03.
Article in English | MEDLINE | ID: mdl-28258694

ABSTRACT

Utilization of negative checkpoint regulators (NCRs) for cancer immunotherapy has garnered significant interest with the completion of clinical trials demonstrating efficacy. While the results of monotherapy treatments are compelling, there is increasing emphasis on combination treatments in an effort to increase response rates to treatment. One of the most recently discovered NCRs is VISTA (V-domain Ig-containing Suppressor of T cell Activation). In this review, we describe the functions of this molecule in the context of cancer immunotherapy. We also discuss factors that may influence the use of anti-VISTA antibody in combination therapy and how genomic analysis may assist in providing indications for treatment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7 Antigens/metabolism , Immunotherapy/methods , Neoplasms/therapy , Animals , B7 Antigens/genetics , B7 Antigens/immunology , Combined Modality Therapy , Genome , Humans , Lymphocyte Activation , Neoplasms/immunology
9.
Immunity ; 35(4): 550-61, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22035846

ABSTRACT

Peripheral tolerance orchestrated by regulatory T cells, dendritic cells (DCs), and mast cells (MCs) has been studied in several models including skin allograft tolerance. We now define a role for MCs in controlling DC behavior ("conditioning") to facilitate tolerance. Under tolerant conditions, we show that MCs mediated a marked increase in tumor necrosis factor (TNFα)-dependent accumulation of graft-derived DCs in the dLN compared to nontolerant conditions. This increase of DCs in the dLN is due to the local production of granulocyte macrophage colony-stimulating factor (GM-CSF) by MCs that induces a survival advantage of graft-derived DCs. DCs that migrated to the dLN from the tolerant allograft were tolerogenic; i.e., they dominantly suppress T cell responses and control regional immunity. This study underscores the importance of MCs in conditioning DCs to mediate peripheral tolerance and shows a functional impact of peripherally produced TNFα and GM-CSF on the migration and function of tolerogenic DCs.


Subject(s)
Dendritic Cells/immunology , Mast Cells/immunology , Skin/immunology , Transplantation Tolerance , Animals , Cell Movement , Cells, Cultured , Dendritic Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Phenotype , Skin Transplantation , Tumor Necrosis Factor-alpha/immunology
10.
J Immunol ; 199(1): 186-203, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28539429

ABSTRACT

HSV type 1 (HSV-1) is a prevalent human pathogen that infects >3.72 billion individuals worldwide and can cause potentially blinding recurrent corneal herpetic disease. HSV-1 establishes latency within sensory neurons of trigeminal ganglia (TG), and TG-resident CD8+ T cells play a critical role in preventing its reactivation. The repertoire, phenotype, and function of protective CD8+ T cells are unknown. Bolstering the apparent feeble numbers of CD8+ T cells in TG remains a challenge for immunotherapeutic strategies. In this study, a comprehensive panel of 467 HLA-A*0201-restricted CD8+ T cell epitopes was predicted from the entire HSV-1 genome. CD8+ T cell responses to these genome-wide epitopes were compared in HSV-1-seropositive symptomatic individuals (with a history of numerous episodes of recurrent herpetic disease) and asymptomatic (ASYMP) individuals (who are infected but never experienced any recurrent herpetic disease). Frequent polyfunctional HSV-specific IFN-γ+CD107a/b+CD44highCD62LlowCD8+ effector memory T cells were detected in ASYMP individuals and were primarily directed against three "ASYMP" epitopes. In contrast, symptomatic individuals have more monofunctional CD44highCD62LhighCD8+ central memory T cells. Furthermore, therapeutic immunization with an innovative prime/pull vaccine, based on priming with multiple ASYMP epitopes (prime) and neurotropic TG delivery of the T cell-attracting chemokine CXCL10 (pull), boosted the number and function of CD44highCD62LlowCD8+ effector memory T cells and CD103highCD8+ tissue-resident T cells in TG of latently infected HLA-A*0201-transgenic mice and reduced recurrent ocular herpes following UV-B-induced reactivation. These findings have profound implications in the development of T cell-based immunotherapeutic strategies to treat blinding recurrent herpes infection and disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesvirus 1, Human/immunology , Immunologic Memory , Keratitis, Herpetic/immunology , Trigeminal Ganglion/immunology , Trigeminal Ganglion/virology , Virus Latency , Adult , Aged , Animals , CD8-Positive T-Lymphocytes/physiology , Chemokine CXCL10/immunology , Epitopes/chemistry , Epitopes/immunology , Epitopes/isolation & purification , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunization , Keratitis, Herpetic/therapy , Keratitis, Herpetic/virology , Male , Mice , Mice, Transgenic , Middle Aged , Recurrence , Trigeminal Ganglion/cytology , Young Adult
11.
Glia ; 66(12): 2645-2658, 2018 12.
Article in English | MEDLINE | ID: mdl-30306644

ABSTRACT

V-type immunoglobulin domain-containing suppressor of T-cell activation (VISTA) is a negative checkpoint regulator (NCR) involved in inhibition of T cell-mediated immunity. Expression changes of other NCRs (PD-1, PD-L1/L2, CTLA-4) during inflammation of the central nervous system (CNS) were previously demonstrated, but VISTA expression in the CNS has not yet been explored. Here, we report that in the human and mouse CNS, VISTA is most abundantly expressed by microglia, and to lower levels by endothelial cells. Upon TLR stimulation, VISTA expression was reduced in primary neonatal mouse and adult rhesus macaque microglia in vitro. In mice, microglial VISTA expression was reduced after lipopolysaccharide (LPS) injection, during experimental autoimmune encephalomyelitis (EAE), and in the accelerated aging Ercc1 Δ/- mouse model. After LPS injection, decreased VISTA expression in mouse microglia was accompanied by decreased acetylation of lysine residue 27 in histone 3 in both its promoter and enhancer region. ATAC-sequencing indicated a potential regulation of VISTA expression by Pu.1 and Mafb, two transcription factors crucial for microglia function. Finally, our data suggested that VISTA expression was decreased in microglia in multiple sclerosis lesion tissue, whereas it was increased in Alzheimer's disease patients. This study is the first to demonstrate that in the CNS, VISTA is expressed by microglia, and that VISTA is differentially expressed in CNS pathologies.


Subject(s)
Central Nervous System Diseases/complications , Inflammation/etiology , Inflammation/pathology , Membrane Proteins/metabolism , Microglia/metabolism , Microglia/pathology , Animals , Animals, Newborn , Brain/pathology , Calcium-Binding Proteins , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Endonucleases/genetics , Endonucleases/metabolism , Female , Freund's Adjuvant/toxicity , Gene Expression/physiology , Humans , Lipopolysaccharides/pharmacology , Macaca mulatta , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Microglia/drug effects , Myelin-Oligodendrocyte Glycoprotein/toxicity , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peptide Fragments/toxicity
12.
Cancer Immunol Immunother ; 67(7): 1113-1121, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29737375

ABSTRACT

Adaptive immune responses contribute to the pathogenesis of melanoma by facilitating immune evasion. V-domain Ig suppressor of T-cell activation (VISTA) is a potent negative regulator of T-cell function and is expressed at high levels on monocytes, granulocytes, and macrophages, and at lower densities on T-cell populations within the tumor microenvironment. In this study, 85 primary melanoma specimens were selected from pathology tissue archives and immunohistochemically stained for CD3, PD-1, PD-L1, and VISTA. Pearson's correlation coefficients identified associations in expression between VISTA and myeloid infiltrate (r = 0.28, p = 0.009) and the density of PD-1+ inflammatory cells (r = 0.31, p = 0.005). The presence of VISTA was associated with a significantly worse disease-specific survival in univariate analysis (hazard ratio = 3.57, p = 0.005) and multivariate analysis (hazard ratio = 3.02, p = 0.02). Our findings show that VISTA expression is an independent negative prognostic factor in primary cutaneous melanoma and suggests its potential as an adjuvant immunotherapeutic intervention in the future.


Subject(s)
B7 Antigens/metabolism , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/mortality , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/mortality , B7 Antigens/immunology , B7-H1 Antigen/immunology , Female , Follow-Up Studies , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Survival Rate , Tumor Microenvironment , Melanoma, Cutaneous Malignant
13.
Crit Rev Immunol ; 37(2-6): 371-420, 2017.
Article in English | MEDLINE | ID: mdl-29773027

ABSTRACT

Initially, a role for the interaction between CD40, expressed on B cells, and gp39 (CD40L), expressed on activated T cells, has been defined in humoral immunity. CD40-CD40L interaction is an essential signal for B cell proliferation, expression of activation markers, immunoglobulin production, and isotype switching. CD40-CD40L interaction is also required for formation of B memory cells and germinal centers, and signaling through CD40 prevents apoptosis of germinal center B cells. Defective expression of CD40L in humans leads to an inability to produce isotypes other than IgM (hyper IgM syndrome), and to an absence of germinal centers. More recent evidence indicates an expansion of the role of the CD40-CD40L axis in cellular interactions beyond antibody formation. Induced expression of CD40 on monocytes can lead to CD40L-activated monocyte effector mechanisms. In addition, CD40-CD40L interactions are crucially involved in development of autoimmune disease in a number of animal models. CD40-CD40L interactions also impact on growth regulation of certain carcinomas. Manipulation of CD40L has also been used to develop novel strategies for long-term antigen-specific tolerization of peripheral T cells. Finally, the CD40-CD40L axis is involved in thymic selection. Following is a comprehensive overview of CD40L-CD40 interactions in physiological and pathogenic cellular responses and a discussion of the therapeutic ramifications of these interactions.


Subject(s)
CD40 Antigens/immunology , CD40 Ligand/immunology , Cell Communication/immunology , Lymphocyte Activation/immunology , Animals , Apoptosis/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation/immunology , Germinal Center/immunology , Germinal Center/metabolism , Humans , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Monocytes/immunology , Monocytes/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
J Immunol ; 197(10): 3970-3981, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27742829

ABSTRACT

Plasmalemma vesicle-associated protein (Plvap) is an endothelial protein with roles in endothelial diaphragm formation and maintenance of basal vascular permeability. At the same time, Plvap has roles in immunity by facilitating leukocyte diapedesis at inflammatory sites and controlling peripheral lymph node morphogenesis and the entry of soluble Ags into lymph node conduits. Based on its postulated role in diapedesis, we have investigated the role of Plvap in hematopoiesis and show that deletion of Plvap results in a dramatic decrease of IgM+IgDlo B cells in both the spleen and the peritoneal cavity. Tissue-specific deletion of Plvap demonstrates that the defect is B cell extrinsic, because B cell and pan-hematopoietic Plvap deletion has no effect on IgM+IgDlo B cell numbers. Endothelial-specific deletion of Plvap in the embryo or at adult stage recapitulates the full Plvap knockout phenotype, whereas endothelial-specific reconstitution of Plvap under the Chd5 promoter rescues the IgM+IgDlo B cell phenotype. Taken together, these results show that Plvap expression in endothelial cells is important in the maintenance of IgM+ B cells in the spleen and peritoneal cavity.


Subject(s)
B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Homeostasis , Membrane Proteins/metabolism , Precursor Cells, B-Lymphoid/metabolism , Spleen/cytology , Animals , B-Lymphocytes/pathology , Capillary Permeability , Carrier Proteins/genetics , DNA Helicases/genetics , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Gene Expression Regulation , Immunoglobulin D/genetics , Immunoglobulin D/metabolism , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Peritoneal Cavity/cytology , Phenotype , Spleen/immunology , Transendothelial and Transepithelial Migration/immunology
15.
Arterioscler Thromb Vasc Biol ; 36(3): 482-90, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26821950

ABSTRACT

OBJECTIVE: Beyond their eminent role in hemostasis and thrombosis, platelets are recognized as mediators of inflammation. Platelet cluster of differentiation 40 (CD40) ligand (CD40L and CD154) plays a key role in mediating platelet-induced inflammation in atherosclerosis. CD40, the receptor for CD40L, is present on platelets; however, the role of CD40 on this cell type is until now undefined. APPROACH AND RESULTS: We found that in both mice and humans, platelet CD40 mediates the formation of platelet-leukocyte aggregates and the release of chemokine (C-X-C motif) ligand 4. Leukocytes were also less prone to adhere to CD40-deficient thrombi. However, platelet CD40 was not involved in platelet aggregation. Activated platelets isolated from Cd40(-/-)Apoe(-/-) mice adhered less to the endothelium upon injection into Apoe(-/-) mice when compared with CD40-sufficient platelets. Furthermore, lack of CD40 on injected platelets led to reduced leukocyte recruitment to the carotid artery as assayed by intravital microscopy. This was accompanied by a decrease in endothelial vascular cell adhesion molecule-1, platelet endothelial cell adhesion molecule, VE-cadherin, and P-selectin expression. To investigate the effect of platelet CD40 in atherosclerosis, Apoe(-/-) mice received thrombin-activated Apoe(-/-) or Cd40(-/-)Apoe(-/-) platelets every 5 days for 12 weeks, starting at the age of 17 weeks, when atherosclerotic plaques had already formed. When compared with mice that received Apoe(-/-) platelets, those receiving Cd40(-/-)Apoe(-/-) platelets exhibited a >2-fold reduction in atherosclerosis. Plaques of mice receiving CD40-deficient platelets were less advanced, contained less macrophages, neutrophils, and collagen, and displayed smaller lipid cores. CONCLUSIONS: Platelet CD40 plays a crucial role in inflammation by stimulating leukocyte activation and recruitment and activation of endothelial cells, thereby promoting atherosclerosis.


Subject(s)
Atherosclerosis/blood , Blood Platelets/metabolism , CD40 Antigens/blood , Endothelial Cells/metabolism , Inflammation/blood , Leukocytes/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Ligand/blood , Cells, Cultured , Chemotaxis , Coculture Techniques , Diet, High-Fat , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/blood , P-Selectin/genetics , Plaque, Atherosclerotic , Platelet Adhesiveness , Platelet Aggregation , Platelet Transfusion , Signal Transduction , Time Factors
16.
J Immunol ; 195(4): 1368-71, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26163586

ABSTRACT

Retinoic acid (RA) is a critical regulator of the intestinal adaptive immune response. However, the intrinsic impact of RA on B cell differentiation in the regulation of gut humoral immunity in vivo has never been directly shown. To address this issue, we have been able to generate a mouse model where B cells specifically express a dominant-negative receptor α for RA. In this study, we show that the silencing of RA signaling in B cells reduces the numbers of IgA(+) Ab-secreting cells both in vitro and in vivo, suggesting that RA has a direct effect on IgA plasma cell differentiation. Moreover, the lack of RA signaling in B cells abrogates Ag-specific IgA responses after oral immunization and affects the microbiota composition. In conclusion, these results suggest that RA signaling in B cells through the RA receptor α is important to generate an effective gut humoral response and to maintain a normal microbiota composition.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Immunization , Signal Transduction , Tretinoin/metabolism , Animals , B-Lymphocytes/cytology , Cell Differentiation/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Gene Expression , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Mice , Mice, Transgenic , Microbiota/immunology , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism
17.
Proc Natl Acad Sci U S A ; 111(41): 14846-51, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25267631

ABSTRACT

V domain-containing Ig suppressor of T-cell activation (VISTA) is a negative checkpoint regulator that suppresses T cell-mediated immune responses. Previous studies using a VISTA-neutralizing monoclonal antibody show that VISTA blockade enhances T-cell activation. The current study describes a comprehensive characterization of mice in which the gene for VISTA has been deleted. Despite the apparent normal hematopoietic development in young mice, VISTA genetic deficiency leads to a gradual accumulation of spontaneously activated T cells, accompanied by the production of a spectrum of inflammatory cytokines and chemokines. Enhanced T-cell responsiveness was also observed upon immunization with neoantigen. Despite the presence of multiorgan chronic inflammation, aged VISTA-deficient mice did not develop systemic or organ-specific autoimmune disease. Interbreeding of the VISTA-deficient mice with 2D2 T-cell receptor transgenic mice, which are predisposed to the development of experimental autoimmune encephalomyelitis, drastically enhanced disease incidence and intensity. Disease development is correlated with the increase in the activation of encephalitogenic T cells in the periphery and enhanced infiltration into the CNS. Taken together, our data suggest that VISTA is a negative checkpoint regulator whose loss of function lowers the threshold for T-cell activation, allowing for an enhanced proinflammatory phenotype and an increase in the frequency and intensity of autoimmunity under susceptible conditions.


Subject(s)
Autoimmunity/genetics , Autoimmunity/immunology , B7 Antigens/genetics , Genetic Predisposition to Disease , Inflammation/pathology , Aging/pathology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , B7 Antigens/deficiency , B7 Antigens/metabolism , Chemokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Hematopoiesis , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Th1 Cells/immunology , Th17 Cells/immunology
18.
Eur J Immunol ; 45(5): 1287-95, 2015 May.
Article in English | MEDLINE | ID: mdl-25808452

ABSTRACT

The importance of vitamin A for host defense is undeniable and the study of its mechanisms is paramount. Of the estimated 250 million preschool children who are vitamin A-deficient (VAD), 10% will die from their increased susceptibility to infectious disease. Vitamin A supplementation was established in the 1980s as one of the most successful interventions in the developing world. Understanding how vitamin A controls immunity will help curb the mortality and morbidity associated with vitamin A deficiency and exploit the immune-enhancing capacity of vitamin A to heighten host resistance to infectious disease. The discoveries that retinoic acid (RA) imprints the homing of leukocytes to the gut and enhances the induction of regulatory T cells, highlighted a potential role for RA in mucosal tolerance. However, more recently emerging data tell of a more profound systemic impact of RA on leukocyte function and commitment. In animal models using genetic manipulation of RA signaling, we learned when and how RA controls T cell fate. Here, we review the role for RA as a critical checkpoint regulator in the differentiation of CD4(+) T cells within the immune system.


Subject(s)
Vitamin A/immunology , Animals , Cell Differentiation , Forkhead Transcription Factors/metabolism , Humans , Immunity, Mucosal , Immunosuppressive Agents/therapeutic use , Immunotherapy , Mice , Models, Immunological , Retinoids/therapeutic use , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Tretinoin/immunology , Tretinoin/metabolism , Vitamin A Deficiency/immunology
19.
Eur J Immunol ; 45(7): 2017-27, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25944401

ABSTRACT

Although donor-specific transfusion (DST) plus CD154 blockade represents a robust protocol for inducing transplantation tolerance, the underlying mechanisms are incompletely understood. In a murine T-cell adoptive transfer model, we have visualized alloantigen-specific, TCR-transgenic for H2-A(b) /H2-K(d) 54-68 epitope (TCR75) CD4(+) T cells with indirect allospecificity during the course of tolerance induction. Three main observations were made. First, although the majority of TCR75 CD4(+) T cells were deleted following DST plus CD154 blockade, the surviving TCR75 CD4(+) T cells were capable of making IL-2, upregulating CD44, and undergoing cell division, suggesting that they were functionally active. Indeed, residual TCR75 CD4(+) T cells reisolated from the primary recipients given DST plus CD154 blockade were fully capable of rejecting allografts upon secondary transfer. Second, in tolerant mice, TCR75 CD4(+) T cells were not induced to express Foxp3 in the graft-draining lymph node. TCR75 CD4(+) T cells were also absent in accepted graft tissues in which endogenous Treg cells were enriched. Finally, DST plus CD154 blockade resulted in an abortive expansion of TCR75 CD4(+) T cells, a process that required the presence of endogenous Treg cells. Collectively, surviving TCR75 CD4(+) T cells are immunocompetent but kept in check by an endogenous immunosuppressive network induced by DST plus CD154 blockade.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Adoptive Transfer , Allografts , Animals , Graft Survival/immunology , Mice , Mice, Knockout , Mice, Transgenic
20.
J Virol ; 89(18): 9693-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26157131

ABSTRACT

Inhibition of T-cell responses in tumor microenvironments by myeloid-derived suppressor cells (MDSCs) is widely accepted. We demonstrated augmentation of monocytic MDSCs whose suppression of not only T-cell, but also B-cell, responsiveness paralleled the immunodeficiency during LP-BM5 retrovirus infection. MDSCs inhibited T cells by inducible nitric oxide synthase (iNOS)/nitric oxide (NO), but uniquely, inhibition of B cells was ~50% dependent each on iNOS/NO and the MDSC-expressed negative-checkpoint regulator VISTA. Blockade with a combination of iNOS/NO and VISTA caused additive or synergistic abrogation of MDSC-mediated suppression of B-cell responsiveness.


Subject(s)
B-Lymphocytes/immunology , Immunologic Deficiency Syndromes/immunology , Membrane Proteins/immunology , Monocytes/immunology , Retroviridae Infections/immunology , Animals , B-Lymphocytes/pathology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Membrane Proteins/genetics , Mice , Monocytes/pathology , Nitric Oxide/genetics , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Retroviridae Infections/genetics , Retroviridae Infections/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL