Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Neuroinflammation ; 19(1): 158, 2022 Jun 19.
Article in English | MEDLINE | ID: mdl-35718775

ABSTRACT

BACKGROUND: Immune activation, neuroinflammation, and cell death are the hallmarks of multiple sclerosis (MS), which is an autoimmune demyelinating disease of the central nervous system (CNS). It is well-documented that the cellular inhibitor of apoptosis 2 (cIAP2) is induced by inflammatory stimuli and regulates adaptive and innate immune responses, cell death, and the production of inflammatory mediators. However, the impact of cIAP2 on neuroinflammation associated with MS and disease severity remains unknown. METHODS: We used experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of MS, to assess the effect of cIAP2 deletion on disease outcomes. We performed a detailed analysis on the histological, cellular, and molecular levels. We generated and examined bone-marrow chimeras to identify the cIAP2-deficient cells that are critical to the disease outcomes. RESULTS: cIAP2-/- mice exhibited increased EAE severity, increased CD4+ T cell infiltration, enhanced proinflammatory cytokine/chemokine expression, and augmented demyelination. This phenotype was driven by cIAP2-deficient non-hematopoietic cells. cIAP2 protected oligodendrocytes from cell death during EAE by limiting proliferation and activation of brain microglia. This protective role was likely exerted by cIAP2-mediated inhibition of the non-canonical NLRP3/caspase-8-dependent myeloid cell activation during EAE. CONCLUSIONS: Our findings suggest that cIAP2 is needed to modulate neuroinflammation, cell death, and survival during EAE. Significantly, our data demonstrate the critical role of cIAP2 in limiting the activation of microglia during EAE, which could be explored for developing MS therapeutics in the future.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Microglia/metabolism , Multiple Sclerosis/pathology , Neuroinflammatory Diseases
2.
FASEB J ; 29(12): 4853-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26246404

ABSTRACT

The neuroinflammation associated with multiple sclerosis involves activation of astrocytes that secrete and respond to inflammatory mediators such as IL-1. IL-1 stimulates expression of many chemokines, including C-C motif ligand (CCL) 5, that recruit immune cells, but it also stimulates sphingosine kinase-1, an enzyme that generates sphingosine-1-phosphate (S1P), a bioactive lipid mediator essential for inflammation. We found that whereas S1P promotes IL-1-induced expression of IL-6, it inhibits IL-1-induced CCL5 expression in astrocytes. This inhibition is mediated by the S1P receptor (S1PR)-2 via an inhibitory G-dependent mechanism. Consistent with this surprising finding, infiltration of macrophages into sites of inflammation increased significantly in S1PR2(-/-) animals. However, activation of NF-κB, IFN regulatory factor-1, and MAPKs, all of which regulate CCL5 expression in response to IL-1, was not diminished by the S1P in astrocytes. Instead, S1PR2 stimulated inositol 1,4,5-trisphosphate-dependent Ca(++) release and Elk-1 phosphorylation and enhanced c-Fos expression. In our study, IL-1 induced the IFNß production that supports CCL5 expression. An intriguing finding was that S1P induced c-Fos-inhibited CCL5 directly and also indirectly through inhibition of the IFN-ß amplification loop. We propose that in addition to S1PR1, which promotes inflammation, S1PR2 mediates opposing inhibitory functions that limit CCL5 expression and diminish the recruitment of immune cells.


Subject(s)
Chemokine CCL5/antagonists & inhibitors , Interferon-beta/metabolism , Interleukin-1/antagonists & inhibitors , Lysophospholipids/physiology , Proto-Oncogene Proteins c-fos/metabolism , Sphingosine/analogs & derivatives , Animals , Cells, Cultured , Humans , Interferon Regulatory Factor-1/biosynthesis , Interferon-beta/biosynthesis , Ligands , Mice , Mice, Knockout , Phosphorylation , Protein Kinases/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Sphingosine/physiology
3.
J Immunol ; 194(6): 2862-70, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25681350

ABSTRACT

The secreted protein, YKL-40, has been proposed as a biomarker of a variety of human diseases characterized by ongoing inflammation, including chronic neurologic pathologies such as multiple sclerosis and Alzheimer's disease. However, inflammatory mediators and the molecular mechanism responsible for enhanced expression of YKL-40 remained elusive. Using several mouse models of inflammation, we now show that YKL-40 expression correlated with increased expression of both IL-1 and IL-6. Furthermore, IL-1 together with IL-6 or the IL-6 family cytokine, oncostatin M, synergistically upregulated YKL-40 expression in both primary human and mouse astrocytes in vitro. The robust cytokine-driven expression of YKL-40 in astrocytes required both STAT3 and NF-κB binding elements of the YKL-40 promoter. In addition, YKL-40 expression was enhanced by constitutively active STAT3 and inhibited by dominant-negative IκBα. Surprisingly, cytokine-driven expression of YKL-40 in astrocytes was independent of the p65 subunit of NF-κB and instead required subunits RelB and p50. Mechanistically, we show that IL-1-induced RelB/p50 complex formation was further promoted by oncostatin M and that these complexes directly bound to the YKL-40 promoter. Moreover, we found that expression of RelB was strongly upregulated during inflammation in vivo and by IL-1 in astrocytes in vitro. We propose that IL-1 and the IL-6 family of cytokines regulate YKL-40 expression during sterile inflammation via both STAT3 and RelB/p50 complexes. These results suggest that IL-1 may regulate the expression of specific anti-inflammatory genes in nonlymphoid tissues via the canonical activation of the RelB/p50 complexes.


Subject(s)
Adipokines/genetics , Cytokines/pharmacology , Gene Expression/drug effects , Glycoproteins/genetics , Lectins/genetics , NF-kappa B p50 Subunit/metabolism , Transcription Factor RelB/metabolism , Adipokines/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Chitinase-3-Like Protein 1 , Cytokines/genetics , Female , Glycoproteins/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-1/genetics , Interleukin-1/pharmacology , Interleukin-6/genetics , Interleukin-6/pharmacology , Lectins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/metabolism , NF-kappa B p50 Subunit/genetics , Oncostatin M/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Transcription Factor RelB/genetics
4.
Nat Immunol ; 15(3): 231-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24464131

ABSTRACT

Although interleukin 1 (IL-1) induces expression of the transcription factor IRF1 (interferon-regulatory factor 1), the roles of IRF1 in immune and inflammatory responses and mechanisms of its activation remain elusive. Here we found that IRF1 was essential for IL-1-induced expression of the chemokines CXCL10 and CCL5, which recruit mononuclear cells into sites of sterile inflammation. Newly synthesized IRF1 acquired Lys63 (K63)-linked polyubiquitination mediated by the apoptosis inhibitor cIAP2 that was enhanced by the bioactive lipid S1P. In response to IL-1, cIAP2 and the sphingosine kinase SphK1 (the enzyme that generates S1P) formed a complex with IRF1, which led to its activation. Thus, IL-1 triggered a hitherto unknown signaling cascade that controlled the induction of IRF1-dependent genes that encode molecules important for sterile inflammation.


Subject(s)
Chemokine CCL5/biosynthesis , Chemokine CXCL10/biosynthesis , Interferon Regulatory Factor-1/metabolism , Interleukin-1/metabolism , Signal Transduction/immunology , Animals , Chemokine CCL5/immunology , Chemokine CXCL10/immunology , Chemotaxis, Leukocyte/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunoprecipitation , Inflammation/immunology , Inflammation/metabolism , Interferon Regulatory Factor-1/immunology , Interleukin-1/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lysine , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Ubiquitination
5.
J Biol Chem ; 286(46): 39893-903, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21953450

ABSTRACT

Nuclear factor I-X3 (NFI-X3) is a newly identified splice variant of NFI-X that regulates expression of several astrocyte-specific markers, such as glial fibrillary acidic protein. Here, we identified a set of genes regulated by NFI-X3 that includes a gene encoding a secreted glycoprotein YKL-40. Although YKL-40 expression is up-regulated in glioblastoma multiforme, its regulation and functions in nontransformed cells of the central nervous system are widely unexplored. We find that expression of YKL-40 is activated during brain development and also differentiation of neural progenitors into astrocytes in vitro. Furthermore, YKL-40 is a migration factor for primary astrocytes, and its expression is controlled by both NFI-X3 and STAT3, which are known regulators of gliogenesis. Knockdown of NFI-X3 and STAT3 significantly reduced YKL-40 expression in astrocytes, whereas overexpression of NFI-X3 dramatically enhanced YKL-40 expression in glioma cells. Activation of STAT3 by oncostatin M induced YKL-40 expression in astrocytes, whereas expression of a dominant-negative STAT3 had a suppressive effect. Mechanistically, NFI-X3 and STAT3 form a complex that binds to weak regulatory elements in the YKL-40 promoter and activates transcription. We propose that NFI-X3 and STAT3 control the migration of differentiating astrocytes as well as migration and invasion of glioma cells via regulating YKL-40 expression.


Subject(s)
Adipokines/biosynthesis , Astrocytes/metabolism , Cell Movement , Glioma/metabolism , Lectins/biosynthesis , Multiprotein Complexes/metabolism , NFI Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , STAT3 Transcription Factor/metabolism , Adipokines/genetics , Animals , Brain/embryology , Brain/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Chitinase-3-Like Protein 1 , Coculture Techniques , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Humans , Lectins/genetics , Mice , Multiprotein Complexes/genetics , NFI Transcription Factors/genetics , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Oncostatin M/metabolism , Oncostatin M/pharmacology , Response Elements/genetics , STAT3 Transcription Factor/genetics , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...