193 research outputs found

    High-mobility group box protein 1 (HMGB1): an alarmin mediating the pathogenesis of rheumatic disease

    Get PDF
    High-mobility group box protein 1 (HMGB1) is a non-histone nuclear protein that has a dual function. Inside the cell, HMGB1 binds DNA, regulating transcription and determining chromosomal architecture. Outside the cell, HMGB1 can serve as an alarmin to activate the innate system and mediate a wide range of physiological and pathological responses. To function as an alarmin, HMGB1 translocates from the nucleus of the cell to the extra-cellular milieu, a process that can take place with cell activation as well as cell death. HMGB1 can interact with receptors that include RAGE (receptor for advanced glycation endproducts) as well as Toll-like receptor-2 (TLR-2) and TLR-4 and function in a synergistic fashion with other proinflammatory mediators to induce responses. As shown in studies on patients as well as animal models, HMGB1 can play an important role in the pathogenesis of rheumatic disease, including rheumatoid arthritis, systemic lupus erythematosus, and polymyositis among others. New approaches to therapy for these diseases may involve strategies to inhibit HMGB1 release from cells, its interaction with receptors, and downstream signaling

    Citrullinated proteins in arthritis: presence in joints and effects on immunogenicity

    Get PDF
    Contains fulltext : 60098.pdf (publisher's version ) (Open Access

    Transcriptomic Profiling Reveals That HMGB1 Induces Macrophage Polarization Different from Classical M1

    Get PDF
    Macrophages are key inflammatory immune cells that display dynamic phenotypes and functions in response to their local microenvironment. In different conditions, macrophage polarization can be induced by high-mobility group box 1 (HMGB1), a nuclear DNA-binding protein that activates innate immunity via the Toll-like receptor (TLR) 4, the receptor for advanced glycation end products (RAGE), and C-X-C chemokine receptor (CXCR) 4. This study investigated the phenotypes of murine bone-marrow-derived macrophages (BMDMs) stimulated with different HMGB1 redox isoforms using bulk RNA sequencing (RNA-Seq). Disulfide HMGB1 (dsHMGB1)-stimulated BMDMs showed a similar but distinct transcriptomic profile to LPS/IFNγ- and LPS-stimulated BMDMs. Fully reduced HMGB1 (frHMGB1) did not induce any significant transcriptomic change. Interestingly, compared to LPS/IFNγ- and LPS-, dsHMGB1-stimulated BMDMs showed lipid metabolism and foam cell differentiation gene set enrichment, and oil red O staining revealed that both dsHMGB1 and frHMGB1 alleviated oxidized low-density lipoprotein (oxLDL)-induced foam cells formation. Overall, this work, for the first time, used transcriptomic analysis by RNA-Seq to investigate the impact of HMGB1 stimulation on BMDM polarization. Our results demonstrated that dsHMGB1 and frHMGB1 induced distinct BMDM polarization phenotypes compared to LPS/IFNγ- and LPS- induced phenotypes.publishedVersio

    Morphological characterization of intra-articular HMGB1 expression during the course of collagen-induced arthritis

    Get PDF
    High-mobility group chromosomal box protein 1 (HMGB1) is a structural nuclear protein that promotes inflammation when present extracellularly. Aberrant, extracellular HMGB1 expression has been demonstrated in human and experimental synovitis. The aim of the present study was to elucidate the temporal and spatial expression of HMGB1 compared to that of the central mediators tumor necrosis factor (TNF) and interleukin-1-beta (IL-1β) during the course of collagen-induced arthritis. Thus, Dark Agouti rats were immunized with homologous type II collagen and synovial tissue specimens were obtained at various time points prior to and during the course of clinical arthritis. Local cytokine responses were assessed by immunohistochemistry and by in situ hybridization. We demonstrate a distinct nuclear expression of HMGB1 at early disease-preceding time points. Preceding clinical onset by a few days, cytoplasmic HMGB1 expression was evident in synoviocytes within the non-proliferative lining layer. Pronounced cytoplasmic and additional extracellular HMGB1 expression coincided with the progression of clinical disease. In advanced arthritis, the number of cells with cytoplasmic HMGB1 expression was quantitatively comparable to that of cells expressing TNF and IL-1β. Interestingly, although HMGB1 was abundantly expressed throughout the inflamed synovium at a protein level, upregulation of HMGB1 mRNA was restricted mainly to areas of cartilage and bone destruction. In conclusion, these new findings implicate a role for HMGB1 in both inducing and perpetuating inflammatory events of significant importance in the destructive processes in chronic arthritis

    High mobility group box protein 1 in complex with lipopolysaccharide or IL-1 promotes an increased inflammatory phenotype in synovial fibroblasts

    Get PDF
    Inflammation can be infectious and/or sterile depending on the initiating event. The proinflammatory mediator High mobility group box protein 1 (HMGB1) is a nuclear protein released from cells during both sterile and infectious inflammation and once extracellular, initates and potentiates inflammation by inducing cytokine production and by recruiting inflammatory cells. Autoimmune diseases are characterised by chronic sterile inflammation leading to tissue destruction. HMGB1 has been implicated in the pathogenesis of several autoimmune diseases including rheumatoid arthritis (RA), systemic lupus erythematosis, multiple sclerosis and myositis. The involvement of HMGB1 in arthritis has been shown by overexpression of HMGB1 in RA synovial tissue and synovial fluid, by beneficial outcome of therapeutic HMGB1-blockade in several experimental arthritis models and by the induction of arthritis by intra-articular injection of recombinant HMGB1 into mice. In this thesis work I set out to investigate the potential role of HMGB1 in juvenile idiopathic arthritis (JIA), to further delineate mechanisms by which HMGB1 can contribute to arthritis pathogenesis and to study the means by which HMGB1 activity can be suppressed. I could report for the first time that HMGB1 levels were increased in synovial fluid as compared to plasma during JIA. HMGB1 levels in synovial fluid did not correlate to disease duration. In contrast, the recorded levels of IL-8 and S100 proteins were higher in synovial fluid during early phases of disease. This indicates a change in the inflammatory phenotype during the progression of JIA. High HMGB1 levels in synovial fluid correlated with early JIA onset, suggesting differences in immunopathogenesis between patient groups. I have also demonstrated that HMGB1 may form complexes with the exogenous TLR ligand LPS or the endogenous inflammatory mediators IL-1α and IL-1β, respectively. Compared to each mediator alone such complexes stimulated synovial fibroblasts from arthritis patients to enhanced production of cytokines and tissue degrading enzymes. This enhancement is mediated via the reciprocal receptor for each HMGB1-partner molecule. Since all the studied mediators are present in arthritic joint during inflammation, this is a potential mechanism through which HMGB1 enhances ongoing inflammation and destruction during rheumatic diseases. Finally, I have demonstrated that the proinflammatory activity of HMGB1 can be therapeutically targeted, either by inhibiting its active release by clinically approved anti-rheumatic drugs or by neutralization with a HMGB1-specific monoclonal antibody. Extracellular secretion of HMGB1 from LPS+IFN-γ stimulated human primary monocytes was inhibited by dexamethasone, chloroquine and gold sodium thiomalate in vitro as recorded using an ELISPOT assay. Therapeutic administration of an HMGB1-specific HMGB1 monoclonal antibody ameliorated arthritis in two separate experimental models. In conclusion, my thesis work has added to the growing evidence that HMGB1 is involved in the pathogenesis of arthritis, has revealed a potential mechanism for its proinflammatory function and has demonstrated a means by which HMGB1-mediated activities can be counteracted

    Citrullinated proteins have increased immunogenicity and arthritogenicity and their presence in arthritic joints correlates with disease severity

    Get PDF
    Autoantibodies directed against citrulline-containing proteins have an impressive specificity of nearly 100% in patients with rheumatoid arthritis and have been suggested to be involved in the disease pathogenesis. The targeted epitopes are generated by a post-translational modification catalysed by the calcium-dependent enzyme peptidyl arginine deiminase (PAD), which converts positively charged arginine to polar but uncharged citrulline. The aim of this study was to explore the effects of citrullination on the immunogenicity of autoantigens as well as on potential arthritogenicity. Thus, immune responses to citrullinated rat serum albumin (Cit-RSA) and to unmodified rat serum albumin (RSA) were examined as well as arthritis development induced by immunisation with citrullinated rat collagen type II (Cit-CII) or unmodified CII. In addition, to correlate the presence of citrullinated proteins and the enzyme PAD4 with different stages of arthritis, synovial tissues obtained at different time points from rats with collagen-induced arthritis were examined immunohistochemically. Our results demonstrate that citrullination of the endogenous antigen RSA broke immunological tolerance, as was evident by the generation of antibodies directed against the modified protein and cross-reacting with the native protein. Furthermore we could demonstrate that Cit-CII induced arthritis with higher incidence and earlier onset than did the native counterpart. Finally, this study reveals that clinical signs of arthritis precede the presence of citrullinated proteins and the enzyme PAD4. As disease progressed into a more severe and chronic state, products of citrullination appeared specifically in the joints. Citrullinated proteins were detected mainly in extracellular deposits but could also be found in infiltrating cells and on the cartilage surface. PAD4 was detected in the cytoplasm of infiltrating mononuclear cells, from day 21 after immunisation and onwards. In conclusion, our data reveal the potency of citrullination to break tolerance against the self antigen RSA and to increase the arthritogenic properties of the cartilage antigen CII. We also show that citrullinated proteins and the enzyme PAD4 are not detectable in healthy joints, and that the appearance and amounts in arthritic joints of experimental animals are correlated with the severity of inflammation

    Expression of Concern: Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1) (Molecular Medicine (2012) 18 (250-259) DOI: 10.2119/molmed.2011.00389)

    Get PDF
    © The Author(s). 2020. The Editors-in-Chief would like to alert readers that this article (Yang et al. 2012) is part of an investigation being conducted by the journal following the conclusions of an institutional enquiry at the University of Liverpool with respect to the quantitative mass spectrometrygenerated results regarding acetylated and redox-modified HMGB1. Appropriate editorial action will be taken once the investigation is concluded. Huan Yang, Peter Lundbäck, Lars Ottosson, Helena Erlandsson-Harris, Emilie Venereau, Marco E. Bianchi, Yousef Al-Abed, Ulf Andersson, and Kevin J. Tracey agree to this editorial expression of concern. Daniel J. Antoine has not responded to any correspondence from the editor/publisher about this editorial expression of concern
    • …
    corecore