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.
Eur J Immunol ; 53(2): e2250059, 2023 02.
Article in English | MEDLINE | ID: mdl-36458588

ABSTRACT

Toll-like receptors (TLR) control the activation of dendritic cells that prime CD4+ T cells in draining lymph nodes, where these T cells then undergo massive clonal expansion. The mechanisms controlling this clonal T cell expansion are poorly defined. Using the CD4+ T cell-mediated disease experimental autoimmune encephalomyelitis (EAE), we show here that this process is markedly suppressed when TLR9 signaling is increased, without noticeably affecting the transcriptome of primed T cells, indicating a purely quantitative effect on CD4+ T cell expansion. Addressing the underpinning mechanisms revealed that CD4+ T cell expansion was preceded and depended on the accumulation of neutrophils in lymph nodes a few days after immunization. Underlying the importance of this immune regulation pathway, blocking neutrophil accumulation in lymph nodes by treating mice with a TLR9 agonist inhibited EAE progression in mice with defects in regulatory T cells or regulatory B cells, which otherwise developed a severe chronic disease. Collectively, this study demonstrates the key role of neutrophils in the quantitative regulation of antigen-specific CD4+ T cell expansion in lymph nodes, and the counter-regulatory role of TLR signaling in this process.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , Neutrophils/pathology , Toll-Like Receptor 9/metabolism , CD4-Positive T-Lymphocytes , Lymph Nodes , Toll-Like Receptors/metabolism , Mice, Inbred C57BL
2.
Br J Pharmacol ; 165(7): 2044-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22035209

ABSTRACT

Development of novel cell migration modulators for anti-inflammatory and cardiovascular therapy is a complex task since any modulator will necessarily interfere with a balanced system of physiological regulators directing proper positioning of diverse immune cell types within the body. Whereas this shall serve efficient pathogen elimination, lack of proper control over these processes may result in counterproductive chronic inflammation and progressive tissue injury instead of healing. Prediction of the therapeutic potential or side effects of any migration modulator is not possible based on theoretical considerations alone but needs to be experimentally evaluated in preclinical disease models and by clinical studies. Here, we briefly summarize basic mechanism of cell migration, and groups of synthetic drugs currently in use for migration modulation. We then discuss one fundamental problem encountered with single-target approaches that arises from the complexity of any inflammation, with multiple interacting and often redundant factors being involved. This issue is likely to arise for any class of therapeutic agent (small molecules, peptides, antibodies, regulatory RNAs) addressing a single gene or protein. Against this background of studies on synthetic migration modulators addressing single targets, we then discuss the potential of endogenous proteins as therapeutic migration modulators, or as parent compounds for the development of mimetic drugs. Regulatory proteins of this type commonly address multiple receptors and signalling pathways and act upon the immune response in a phase-specific manner. Based on recent evidence, we suggest investigation of such endogenous migration modulators as novel starting points for anti-inflammatory and cardiovascular drug development.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cardiovascular Agents/pharmacology , Cell Movement/drug effects , Cell Movement/physiology , Animals , Drug Delivery Systems , Drug Discovery , Heart Diseases/drug therapy , Humans , Immune System/cytology , Immune System/drug effects , MicroRNAs/therapeutic use , Models, Biological
3.
Eur J Immunol ; 41(8): 2323-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21538348

ABSTRACT

Adiponectin (APN), a cytokine constitutively produced in fat tissue, has been shown to exert anti-inflammatory effects in various disease models. While the influence of APN on monocytic cells has been extensively studied in vitro, little is known about its role in T cells. In this study, we show that while <10% of human peripheral blood T cells express adiponectin receptors (AdipoRs) on their surface, most T cells store AdipoRs in intracellular compartments. AdipoRs colocalized with immune regulatory molecules CTLA-4 and TIRC7 within clathrin-coated vesicles. After stimulation, the expression of adiponectin receptor 1 (AdipoR1) and AdipoR2 was upregulated on the surface of antigen-specific T cells, as determined by tetramer or CD137 staining, and AdipoR1 and AdipoR2 coexpressed with CTLA-4. Addition of APN resulted in a significant diminution of antigen-specific T-cell expansion. Mechanistically, APN enhanced apoptosis and inhibited proliferation of antigen-specific T-cell lines. Further, APN directly inhibited cytokine production in response to antigen stimulation. In line with the in vitro data, APN-deficient (knockout, KO) mice had higher frequencies of CD137(+) T cells upon Coxsackie B virus infection. Altogether, our data suggest that APN is a novel negative T-cell regulator. In contrast to the CTLA-4 ligand B7 only expressed on APCs, APN is abundant in human plasma.


Subject(s)
Adiponectin/immunology , Antigens/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adiponectin/genetics , Adiponectin/pharmacology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Proliferation/drug effects , Cells, Cultured , Clathrin-Coated Vesicles/immunology , Clathrin-Coated Vesicles/metabolism , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Flow Cytometry , Gene Expression , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Jurkat Cells , K562 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Receptors, Adiponectin/genetics , Receptors, Adiponectin/immunology , Receptors, Adiponectin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Vacuolar Proton-Translocating ATPases/immunology , Vacuolar Proton-Translocating ATPases/metabolism
4.
Circulation ; 122(25): 2688-98, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21135363

ABSTRACT

BACKGROUND: CCN1 is an evolutionary ancient matricellular protein that modulates biological processes associated with tissue repair. Induction at sites of injury was observed in conditions ranging from skin wounds to cardiac diseases, including ischemic and inflammatory cardiomyopathy. Here, we provide evidence of a novel function of CCN1 as a modulator of immune cell migration. METHODS AND RESULTS: to understand the role of CCN1 in cardiomyopathies and to evaluate its therapeutic potential, we overexpressed CCN1 using an adenoviral hepatotropic vector in murine experimental autoimmune myocarditis, a model of human inflammatory cardiomyopathy. CCN1 gene transfer significantly reduced cardiac disease score and immune cell infiltration. In vivo tracking of hemagglutinin epitope-tagged CCN1 revealed binding to spleen macrophages but not to cardiomyocytes. Unexpectedly, CCN1 therapy left cardiac chemokine and cytokine expression unchanged but instead strongly inhibited the migration of spleen macrophages and lymphocytes, as evidenced by ex vivo transwell assays. In accordance with the ex vivo data, in vitro preincubation with CCN1 diminished transwell migration of human monocytes and abrogated their chemotactic response to monocyte chemoattractant protein-1, macrophage inflammatory protein-1α, and stromal cell-derived factor-1α. Further mechanistic studies showed that CCN1-driven modulation of immune cell migration is mimicked in part by cyclic RGD peptides currently in clinical evaluation for cancer therapy. CONCLUSIONS: our proof-of-concept study suggests investigation of CCN1 as a novel, endogenous "parent compound" for chemotaxis modulation and of cyclic RGD peptides as a class of partially CCN1-mimetic drugs with immediate potential for clinical evaluation in cardiac diseases associated with chronic pathogenic inflammation.


Subject(s)
Autoimmune Diseases/metabolism , Autoimmune Diseases/prevention & control , Cell Movement/physiology , Cysteine-Rich Protein 61/metabolism , Myocarditis/metabolism , Myocarditis/prevention & control , Adult , Animals , Autoimmune Diseases/pathology , Biomimetics , Cell Movement/drug effects , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/pharmacology , Disease Models, Animal , Female , Gene Transfer Techniques , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Liver/cytology , Liver/drug effects , Male , Mice , Mice, Inbred BALB C , Middle Aged , Monocytes/cytology , Monocytes/drug effects , Myocarditis/pathology , Peptides, Cyclic/pharmacology , Recombinant Proteins/pharmacology , Spleen/cytology , Spleen/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...