Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Transplant ; 20(9): 2551-2558, 2020 09.
Article in English | MEDLINE | ID: mdl-32185859

ABSTRACT

Alloreactive memory T cells play a key role in transplantation by accelerating allograft rejection and preventing tolerance induction. Some studies using µMT mice, which are constitutionally devoid of B cells, showed that B cells were required for the generation of memory T cells after allotransplantation. However, whether B cell depletion in normal adult mice has the same effect on memory responses by CD4+ and CD8+ T cells activated after transplantation has not been thoroughly investigated. In this study, we tested the effect of anti-CD20 antibody-mediated B cell depletion on CD4+ and CD8+ memory T cell alloresponses after skin transplantation in wild-type mice. We found that B cell depletion prevented the development of memory alloresponses by CD4+ T cells but enhanced that of CD8+ memory T cells. Next, we tested the influence of B cell depletion on hematopoietic chimerism. In OT-II CD4+ anti-OVA TCR transgenic mice sensitized to ovalbumin antigen, B cell depletion also impaired allospecific memory T cell responses and thereby enhanced donor hematopoietic chimerism and T cell deletion after bone marrow transplantation. This study underscores the complexity of the relationships between B and T cells in the generation and reactivation of different memory T cell subsets after transplantation.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Animals , B-Lymphocytes , CD4-Positive T-Lymphocytes , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin Transplantation
2.
Am J Transplant ; 18(8): 1843-1856, 2018 08.
Article in English | MEDLINE | ID: mdl-29939471

ABSTRACT

Achieving host immune tolerance of allogeneic transplants represents the ultimate challenge in clinical transplantation. It has become clear that different cells and mechanisms participate in acquisition versus maintenance of allograft tolerance. Indeed, manipulations which prevent tolerance induction often fail to abrogate tolerance once it has been established. Hence, elucidation of the immunological mechanisms underlying maintenance of T cell tolerance to alloantigens is essential for the development of novel interventions that preserve a robust and long lasting state of allograft tolerance that relies on T cell deletion in addition to intra-graft suppression of inflammatory immune responses. In this review, we discuss some essential elements of the mechanisms involved in the maintenance of naturally occurring or experimentally induced allograft tolerance, including the newly described role of antigen cross-dressing mediated by extracellular vesicles.


Subject(s)
Graft Survival/immunology , Isoantigens/immunology , T-Lymphocytes/immunology , Transplantation Tolerance/immunology , Animals
3.
Methods Mol Biol ; 1585: 111-125, 2017.
Article in English | MEDLINE | ID: mdl-28477191

ABSTRACT

Over the past decade, multiple effector T cell subsets have been identified with varying differentiation conditions in the milieu as well as a broad diversity of cytokine expression. Interleukin-9 (IL-9) secreting T helper 9 (Th9) cells are the newest member of this family. T helper cell differentiation including Th9 cells appears to be an epigenetic phenomenon requiring the coordination of a large variety of transcription factors to reshape the chromatin landscape and generate various T helper phenotypes. This chapter details methods for both predicting and validating potential transcription factor binding sites as well as their downstream epigenetic effect using a variety of in silico and in vitro methods in both primary Th9 cells and IL-9-producing T cell lines.


Subject(s)
Interleukin-9/metabolism , Transcription Factors/metabolism , Chromatin Immunoprecipitation , Humans , Protein Binding , Signal Transduction/physiology , T-Lymphocytes, Helper-Inducer/metabolism
4.
Nat Commun ; 7: 13048, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27725632

ABSTRACT

RORγt is a master transcription factor of Th17 cells and considered as a promising drug target for the treatment of autoimmune diseases. Here, we show the guanine nucleotide exchange factor, Tiam1, and its cognate Rho-family G protein, Rac1, regulate interleukin (IL)17A transcription and autoimmunity. Whereas Tiam1 genetic deficiency weakens IL-17A expression partially and inhibits the development of experimental autoimmune encephalomyelitis (EAE), deletion of Rac1 in T cells exhibits more robust effects on Th17 cells and EAE. We demonstrate Tiam1 and Rac1 form a complex with RORγt in the nuclear compartment of Th17 cells, and together bind and activate the Il17 promoter. The clinical relevance of these findings is emphasized by pharmacological targeting of Rac1 that suppresses both murine and human Th17 cells as well as EAE. Thus, our findings highlight a regulatory pathway of Tiam1/Rac1 in Th17 cells and suggest that it may be a therapeutic target in multiple sclerosis.


Subject(s)
Autoimmunity , Interleukin-17/metabolism , T-Lymphoma Invasion and Metastasis-inducing Protein 1/metabolism , Transcription, Genetic , rac1 GTP-Binding Protein/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Interleukin-17/genetics , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Promoter Regions, Genetic , Protein Binding , T-Lymphoma Invasion and Metastasis-inducing Protein 1/deficiency , Th17 Cells/metabolism
5.
Hum Mol Genet ; 25(2): 404-17, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26604133

ABSTRACT

Notch signaling has recently emerged as an important regulator of immune responses in autoimmune diseases. The recombination signal-binding protein for immunoglobulin kappa J region (RBPJ) is a transcriptional repressor, but converts into a transcriptional activator upon activation of the canonical Notch pathway. Genome-wide association studies of rheumatoid arthritis (RA) identified a susceptibility locus, rs874040(CC), which implicated the RBPJ gene. Here, chromatin state mapping generated using the chromHMM algorithm reveals strong enhancer regions containing DNase I hypersensitive sites overlapping the rs874040 linkage disequilibrium block in human memory, but not in naïve CD4(+) T cells. The rs874040 overlapping this chromatin state was associated with increased RBPJ expression in stimulated memory CD4(+) T cells from healthy subjects homozygous for the risk allele (CC) compared with memory CD4(+) T cells bearing the protective allele (GG). Transcriptomic analysis of rs874040(CC) memory T cells showed a repression of canonical Notch target genes IL (interleukin)-9, IL-17 and interferon (IFN)γ in the basal state. Interestingly, activation of the Notch pathway using soluble Notch ligand, Jagged2-Fc, induced IL-9 and IL-17A while delta-like 4Fc, another Notch ligand, induced higher IFNγ expression in the rs874040(CC) memory CD4(+) T cells compared with their rs874040(GG) counterparts. In RA, RBPJ expression is elevated in memory T cells from RA patients compared with control subjects, and this was associated with induced inflammatory cytokines IL-9, IL-17A and IFNγ in response to Notch ligation in vitro. These findings demonstrate that the rs874040(CC) allele skews memory T cells toward a pro-inflammatory phenotype involving Notch signaling, thus increasing the susceptibility to develop RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/immunology , Genetic Predisposition to Disease , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Polymorphism, Single Nucleotide , Adult , Arthritis, Rheumatoid/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cytokines , Female , Gene Expression , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/immunology , Immunologic Memory , Male , Receptors, Notch , Signal Transduction , Young Adult
6.
J Immunol ; 193(1): 198-207, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24879792

ABSTRACT

The transcriptional repressor B cell lymphoma 6 (BCL6) is required for the development of Th follicular cells, and it has been shown to suppress Th2 cell differentiation. We demonstrate that BCL6 is a key regulator of Th9 cell development. BCL6 expression is transiently downregulated in polarized Th9 cells, and forced expression of BCL6 in Th9 cells impairs Th9 cell differentiation. In contrast, BCL6 knockdown upregulated IL-9 production in Th9 cells. The function of BCL6 in Th9 cells is under the control of IL-2/JAK3/STAT5 signaling pathway. Using chromatin immunoprecipitation, we show that, in Th9 cells, BCL6 and STAT5 bind to adjacent motifs in the Il9 promoter. Furthermore, we found that STAT5 binding was associated with the abundance of a permissive histone mark at the Il9 promoter, whereas under conditions in which BCL6 binding was predominant, a repressive histone mark was prevalent. The effects of STAT5 and BCL6 on IL-9 transcription were further demonstrated using an IL-9 luciferase reporter assay in which BCL6 repressed STAT5-mediated Il9 transactivation. In experimental autoimmune encephalomyelitis, forced expression of BCL6 in myelin oligodendrocyte glycoprotein35-55-specific Th9 cells resulted in decreased IL-9 production and induction of IFN-γ, causing an exacerbation of the clinical disease. Our findings demonstrate a novel role of BCL6 in the regulation of Th9 cell development and their encephalitogenicity.


Subject(s)
DNA-Binding Proteins/immunology , Interleukin-9/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transcription, Genetic/immunology , Transcriptional Activation/immunology , Animals , DNA-Binding Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-9/genetics , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6 , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction/genetics , T-Lymphocytes, Helper-Inducer/pathology , Transcription, Genetic/genetics , Transcriptional Activation/genetics
7.
Clin Dev Immunol ; 2013: 570731, 2013.
Article in English | MEDLINE | ID: mdl-24324509

ABSTRACT

The Notch signaling pathway preservation across species hints to the indispensable role it plays during evolution. Over the last decade the science community has extensively studied the Notch signaling pathway, with Notch emerging as a key player in embryogenesis, tissue homeostasis, angiogenesis, and immunoregulation. Multiple sclerosis (MS) is an incurable yet treatable autoimmune chronic inflammatory disease of the central nervous system. The aim of this review is to provide a brief description of the Notch signaling pathway, and summarize the current literature implicating Notch in the pathogenesis of MS.


Subject(s)
Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Receptors, Notch/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Oligodendroglia/immunology , Oligodendroglia/metabolism , Serrate-Jagged Proteins , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/cytology
8.
J Immunol ; 191(6): 3139-51, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23960232

ABSTRACT

Although activated inflammatory monocytes (IMCs) and inflammatory dendritic cells (IDCs) are potent T cell suppressors, nonactivated IMCs and IDCs promote T cell activation and Th1/Th17 cell differentiation. In this study, we investigated how to reduce the proinflammatory properties of IMCs and IDCs and further convert them into immune regulatory dendritic cells (DCs). We found that IL-4 and retinoic acid (RA) cotreatment of GM-CSF-differentiated IDCs synergistically induced the expression of aldehyde dehydrogenase family 1, subfamily A2, a rate-limiting enzyme for RA synthesis in DCs. IL-4 plus RA-treated IDCs upregulated CD103 expression and markedly reduced the production of proinflammatory cytokines upon activation. IL-4 plus RA-treated IDCs strongly induced CD4⁺Foxp3⁺ regulatory T cell differentiation and suppressed Th1 and Th17 differentiation. Mechanistically, the transcription factors Stat6 and RA receptor ß play important roles in aldehyde dehydrogenase family 1, subfamily A2, induction. In addition, IL-4 and RA signaling pathways interact closely to enhance the regulatory function of treated DCs. Adoptive transfer of IL-4 plus RA-treated DCs significantly increased regulatory T cell frequency in vivo. Direct treatment with IL-4 and RA also markedly suppressed actively induced experimental autoimmune encephalomyelitis. Our data demonstrate the synergistic effect of IL-4 and RA in inducing a regulatory phenotype in IDCs, providing a potential treatment strategy for autoimmune diseases.


Subject(s)
Aldehyde Dehydrogenase/biosynthesis , Cell Differentiation/immunology , Dendritic Cells/immunology , Interleukin-4/metabolism , Tretinoin/metabolism , Aldehyde Dehydrogenase/immunology , Aldehyde Dehydrogenase 1 Family , Animals , Cell Differentiation/drug effects , Chromatin Immunoprecipitation , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Flow Cytometry , Immunoblotting , Interleukin-4/immunology , Interleukin-4/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Real-Time Polymerase Chain Reaction , Retinal Dehydrogenase , Tretinoin/immunology , Tretinoin/pharmacology
9.
Immunity ; 36(4): 623-34, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22503540

ABSTRACT

Interleukin 9 (IL-9) is a pleiotropic cytokine that can regulate autoimmune responses by enhancing regulatory CD4(+)FoxP3(+) T regulatory (Treg) cell survival and T helper 17 (Th17) cell proliferation. Here, we analyzed the costimulatory requirements for the induction of Th9 cells, and demonstrated that Notch pathway cooperated with TGF-ß signaling to induce IL-9. Conditional ablation of Notch1 and Notch2 receptors inhibited the development of Th9 cells. Notch1 intracellular domain (NICD1) recruited Smad3, downstream of TGF-ß cytokine signaling, and together with recombining binding protein (RBP)-Jκ bound the Il9 promoter and induced its transactivation. In experimental autoimmune encephalomyelitis (EAE), Jagged2 ligation regulated clinical disease in an IL-9-dependent fashion. Signaling through Jagged2 expanded Treg cells and suppressed EAE when administered before antigen immunization, but worsened EAE when administered concurrently with immunization by favoring Th17 cell expansion. We propose that Notch and Smad3 cooperate to induce IL-9 and participate in regulating the immune response.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-9/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Smad3 Protein/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Communication , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Interleukin-9/biosynthesis , Jagged-2 Protein , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Receptors, Interleukin-9/genetics , Signal Transduction , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL
...