Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol Res ; 2022: 9926305, 2022.
Article in English | MEDLINE | ID: mdl-35252461

ABSTRACT

C-Type lectin receptor 5A (CLEC5A) is a spleen tyrosine kinase- (Syk-) coupled pattern recognition receptor expressed on myeloid cells and involved in the innate immune response to viral and bacterial infections. Activation of the CLEC5A receptor with pathogen-derived antigens leads to a secretion of proinflammatory mediators such as TNF-α and IL-6 that may provoke a systemic cytokine storm, and CLEC5A gene polymorphisms are associated with the severity of DV infection. In addition, the CLEC5A receptor was mentioned in the context of noninfectious disorders like chronic obstructive pulmonary disease (COPD) or arthritis. Altogether, CLEC5A may be considered as an innate immune checkpoint capable to amplify proinflammatory signals, and this way contributes to infection or to aseptic inflammation. In this study, we determined CLEC5A receptor expression on different macrophage subsets (in vitro and ex vivo) and the functional consequences of its activation in aseptic conditions. The CLEC5A surface expression appeared the highest on proinflammatory M1 macrophages while intermediate on tumor-associated phenotypes (M2c or TAM). In contrast, the CLEC5A expression on ex vivo-derived alveolar macrophages from healthy donors or macrophages from ovarian cancer patients was hardly detectable. Targeting CLEC5A on noninflammatory macrophages with an agonistic α-CLEC5A antibody triggered a release of proinflammatory cytokines, resembling a response to dengue virus, and led to phenotypic changes in myeloid cells that may suggest their reprogramming towards a proinflammatory phenotype, e.g., upregulation of CD80 and downregulation of CD163. Interestingly, the CLEC5A agonist upregulated immune-regulatory molecules like CD206, PD-L1, and cytokines like IL-10, macrophage-derived chemokine (MDC/CCL22), and thymus and activation chemokine (TARC/CCL17) which are associated with an anti-inflammatory or a protumorigenic macrophage phenotype. In the absence of concomitant pathogenic or endogenous danger signals, the CLEC5A receptor activation did not amplify an autologous T cell response, which may represent a protective innate mechanism to avoid an undesirable autoimmune adaptive response.


Subject(s)
Lectins, C-Type , T-Lymphocytes , Cytokines/metabolism , Humans , Immunity, Innate , Lectins, C-Type/metabolism , Macrophages , Receptors, Cell Surface , T-Lymphocytes/metabolism
2.
Nat Commun ; 7: 10888, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26964669

ABSTRACT

The ability of CD4+ T cells to change their phenotype and to specialize into different functional subsets may enhance the risk of autoimmune diseases. Here we investigate how a pleiotropic cytokine interleukin (IL)-15 may modify the functional commitment of CD4+ T cells expressing the lineage-associated transcription factors: forkhead box P3 (Foxp3; Treg) and RORγt (Th17) in the context of inflammatory bowel disease (IBD). We demonstrate in mice that impaired delivery of IL-15 to CD4+ T cells in the colon downmodulates Foxp3 expression (diminishing STAT5 phosphorylation) and enhances RORγt expression (by upregulating the expression of Runx1). In consequence, CD4+ T cells deprived of IL-15 rapidly trigger IBD characterized by enhanced production of pro-inflammatory cytokines (interferon-γ, IL-6) and accumulation of Th1/Th17 cells. Overall, our findings indicate a potentially beneficial role of IL-15 in IBD by fine-tuning the balance between Treg and Th17 cells and controlling intestinal inflammation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Colon/immunology , Cytokines/immunology , Forkhead Transcription Factors/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-15/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Adoptive Transfer , Animals , Colon/metabolism , Colon/pathology , Down-Regulation , Flow Cytometry , Forkhead Transcription Factors/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interleukin-15/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology
3.
Am J Respir Cell Mol Biol ; 47(6): 869-78, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22984087

ABSTRACT

Although the contribution of CD8(+) T cells to the pathogenesis of noncommunicable lung diseases has become increasingly appreciated, our knowledge about the mechanisms controlling self-reactive CD8(+) T cells in the respiratory tract remains largely elusive. The outcome of the encounter between pulmonary self-antigen and naive CD8(+) T cells, in the presence or absence of inflammation, was traced after adoptive transfer of fluorescence-labeled CD8(+) T cells specific for the neo-self-antigen influenza A hemagglutinin into transgenic mice expressing hemagglutinin specifically in alveolar type II epithelial cells in order: to study the outcome of alveolar antigen encounter in the steady state and under inflammatory conditions; to define the phenotype and fate of CD8(+) T cells primed in the respiratory tract; and, finally, to correlate these findings with the onset of autoimmunity in the lung. We found that CD8(+) T cells remain ignorant in the steady state, whereas transient proliferation of self-reactive CD8(+) T cells is induced by forced maturation or licensing of dendritic cells, increases in the antigenic threshold, and targeted release of alveolar self-antigen by epithelial injury. However, these cells fail to acquire effector functions, lack the expression of the high-affinity IL-2 receptor CD25, and do not precipitate autoimmunity in the lung. We conclude that inadvertent activation of CD8(+) T cells in the lung is prevented in the absence of "danger signals," whereas tissue damage after infection or noninfectious inflammation creates an environment that allows the priming of previously ignorant T cells. Failure in effector cell differentiation after abortive priming, however, precludes the establishment of self-perpetuating autoimmunity in the lung.


Subject(s)
Autoantigens/immunology , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Pulmonary Alveoli/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation , Cell Proliferation , Cytotoxicity, Immunologic , Inflammation Mediators/metabolism , Interleukin-2/physiology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Signal Transduction
4.
J Immunol ; 186(11): 6106-18, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21518973

ABSTRACT

Every person harbors a population of potentially self-reactive lymphocytes controlled by tightly balanced tolerance mechanisms. Failures in this balance evoke immune activation and autoimmunity. In this study, we investigated the contribution of self-reactive CD8(+) T lymphocytes to chronic pulmonary inflammation and a possible role for naturally occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells (nTregs) in counterbalancing this process. Using a transgenic murine model for autoimmune-mediated lung disease, we demonstrated that despite pulmonary inflammation, lung-specific CD8(+) T cells can reside quiescently in close proximity to self-antigen. Whereas self-reactive CD8(+) T cells in the inflamed lung and lung-draining lymph nodes downregulated the expression of effector molecules, those located in the spleen appeared to be partly Ag-experienced and displayed a memory-like phenotype. Because ex vivo-reisolated self-reactive CD8(+) T cells were very well capable of responding to the Ag in vitro, we investigated a possible contribution of nTregs to the immune control over autoaggressive CD8(+) T cells in the lung. Notably, CD8(+) T cell tolerance established in the lung depends only partially on the function of nTregs, because self-reactive CD8(+) T cells underwent only biased activation and did not acquire effector function after nTreg depletion. However, although transient ablation of nTregs did not expand the population of self-reactive CD8(+) T cells or exacerbate the disease, it provoked rapid accumulation of activated CD103(+)CD62L(lo) Tregs in bronchial lymph nodes, a finding suggesting an adaptive phenotypic switch in the nTreg population that acts in concert with other yet-undefined mechanisms to prevent the detrimental activation of self-reactive CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Pneumonia/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Immune Tolerance/immunology , Immunologic Memory/immunology , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , L-Selectin/genetics , L-Selectin/immunology , L-Selectin/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Pneumonia/genetics , Pneumonia/metabolism , Respiratory Function Tests , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...