0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Antibodies to a Citrullinated Porphyromonas gingivalis Epitope Are Increased in Early Rheumatoid Arthritis, and Can Be Produced by Gingival Tissue B Cells: Implications for a Bacterial Origin in RA Etiology

      research-article
      1 , 1 , 1 , 1 , 2 , 3 , 1 , 4 , 1 , 5 , 6 , 1 , 1 , 7 , 8 , 9 , 1 , 10 , 11 , 12 , 11 , 13 , 14 , 15 , 1 , 10 , 6 , 1 , 11 , 13 , 3 , 16 , 1 , 14 , 1 , 1 , 1 ,
      Frontiers in Immunology
      Frontiers Media S.A.
      rheumatoid arthritis (RA), anti-citrullinated protein antibodies (ACPA), Porphyromonas gingivalis (Pg), periodontitis (PD), monoclonal antibodies (mAbs), B cells

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Based on the epidemiological link between periodontitis and rheumatoid arthritis (RA), and the unique feature of the periodontal bacterium Porphyromonas gingivalis to citrullinate proteins, it has been suggested that production of anti-citrullinated protein antibodies (ACPA), which are present in a majority of RA patients, may be triggered in the gum mucosa. To address this hypothesis, we investigated the antibody response to a citrullinated P. gingivalis peptide in relation to the autoimmune ACPA response in early RA, and examined citrulline-reactivity in monoclonal antibodies derived from human gingival B cells. Antibodies to a citrullinated peptide derived from P. gingivalis (denoted CPP3) and human citrullinated peptides were analyzed by multiplex array in 2,807 RA patients and 372 controls; associations with RA risk factors and clinical features were examined. B cells from inflamed gingival tissue were single-cell sorted, and immunoglobulin (Ig) genes were amplified, sequenced, cloned and expressed (n=63) as recombinant monoclonal antibodies, and assayed for citrulline-reactivities by enzyme-linked immunosorbent assay. Additionally, affinity-purified polyclonal anti-cyclic-citrullinated peptide (CCP2) IgG, and monoclonal antibodies derived from RA blood and synovial fluid B cells (n=175), were screened for CPP3-reactivity. Elevated anti-CPP3 antibody levels were detected in RA (11%), mainly CCP2+ RA, compared to controls (2%), p<0.0001, with a significant association to HLA-DRB1 shared epitope alleles, smoking and baseline pain, but with low correlation to autoimmune ACPA fine-specificities. Monoclonal antibodies derived from gingival B cells showed cross-reactivity between P. gingivalis CPP3 and human citrullinated peptides, and a CPP3+/CCP2+ clone, derived from an RA blood memory B cell, was identified. Our data support the possibility that immunity to P. gingivalis derived citrullinated antigens, triggered in the inflamed gum mucosa, may contribute to the presence of ACPA in RA patients, through mechanisms of molecular mimicry.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Porphyromonas gingivalis: An Overview of Periodontopathic Pathogen below the Gum Line

          Periodontal disease represents a group of oral inflammatory infections initiated by oral pathogens which exist as a complex biofilms on the tooth surface and cause destruction to tooth supporting tissues. The severity of this disease ranges from mild and reversible inflammation of the gingiva (gingivitis) to chronic destruction of connective tissues, the formation of periodontal pocket and ultimately result in loss of teeth. While human subgingival plaque harbors more than 500 bacterial species, considerable research has shown that Porphyromonas gingivalis, a Gram-negative anaerobic bacterium, is the major etiologic agent which contributes to chronic periodontitis. This black-pigmented bacterium produces a myriad of virulence factors that cause destruction to periodontal tissues either directly or indirectly by modulating the host inflammatory response. Here, this review provides an overview of P. gingivalis and how its virulence factors contribute to the pathogenesis with other microbiome consortium in oral cavity.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Long-Lived Plasma Cells Are Contained within the CD19(-)CD38(hi)CD138(+) Subset in Human Bone Marrow.

            Antibody responses to viral infections are sustained for decades by long-lived plasma cells (LLPCs). However, LLPCs have yet to be characterized in humans. Here we used CD19, CD38, and CD138 to identify four PC subsets in human bone marrow (BM). We found that the CD19(-)CD38(hi)CD138(+) subset was morphologically distinct, differentially expressed PC-associated genes, and exclusively contained PCs specific for viral antigens to which the subjects had not been exposed for more than 40 years. Protein sequences of measles- and mumps-specific circulating antibodies were encoded for by CD19(-)CD38(hi)CD138(+) PCs in the BM. Finally, we found that CD19(-)CD38(hi)CD138(+) PCs had a distinct RNA transcriptome signature and human immunoglobulin heavy chain (VH) repertoire that was relatively uncoupled from other BM PC subsets and probably represents the B cell response's "historical record" of antigenic exposure. Thus, our studies define human LLPCs and provide a mechanism for the life-long maintenance of anti-viral antibodies in the serum.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cardiolipin polyspecific autoreactivity in two broadly neutralizing HIV-1 antibodies.

              The design of a human immunodeficiency virus-1 (HIV-1) immunogen that can induce broadly reactive neutralizing antibodies is a major goal of HIV-1 vaccine development. Although rare human monoclonal antibodies (mAbs) exist that broadly neutralize HIV-1, HIV-1 envelope immunogens do not induce these antibody specificities. Here we demonstrate that the two most broadly reactive HIV-1 envelope gp41 human mAbs, 2F5 and 4E10, are polyspecific autoantibodies reactive with the phospholipid cardiolipin. Thus, current HIV-1 vaccines may not induce these types of antibodies because of autoantigen mimicry of the conserved membrane-proximal epitopes of the virus. These results may have important implications for generating effective neutralizing antibody responses by using HIV-1 vaccines.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                20 April 2022
                2022
                : 13
                : 804822
                Affiliations
                [1] 1 Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital , Stockholm, Sweden
                [2] 2 Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital , Stockholm, Sweden
                [3] 3 Institute of Environmental Medicine, Karolinska Institutet , Stockholm, Sweden
                [4] 4 Wallenberg Centre for Molecular Medicine, Umeå University , Umeå, Sweden
                [5] 5 Thermo Fisher Scientific, ImmunoDiagnositic Division , Uppsala, Sweden
                [6] 6 Department of Immunology, Genetics and Pathology, Uppsala University , Uppsala, Sweden
                [7] 7 Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet , Stockholm, Sweden
                [8] 8 Faculty of Medicine, School of Health Sciences, University of Iceland , Reykjavik, Iceland
                [9] 9 Section for Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
                [10] 10 Center for Rheumatology, Academic Specialist Center, Stockholm Health Region , Stockholm, Sweden
                [11] 11 Division of Periodontology, Department of Dental Medicine, Karolinska Institutet , Stockholm, Sweden
                [12] 12 Danakliniken Specialisttandvård, Praktikertjänst AB , Danderyd, Sweden
                [13] 13 Division of Orthodontics and Pediatric Dentistry, Department of Dental Medicine, Karolinska Institutet , Stockholm, Sweden
                [14] 14 Institute for Research in Biomedicine, Universita dell a Svizzera Italiana , Bellinzona, Switzerland
                [15] 15 Institute of Microbiology, ETH Zurich , Zurich, Switzerland
                [16] 16 Centre of Occupational and Environmental Medicine, Region Stockholm , Stockholm, Sweden
                Author notes

                Edited by: Carmelo Carmona-Rivera, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIH), United States

                Reviewed by: Piotr Mydel, University of Bergen, Norway; Shauna Culshaw, University of Glasgow, United Kingdom

                *Correspondence: Karin Lundberg, Karin.Lundberg@ 123456ki.se

                †Professor Anca I. Catrina passed away in February 2021 during the preparation of the manuscript

                This article was submitted to Autoimmune and Autoinflammatory Disorders, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2022.804822
                9066602
                35514991
                9e591a4e-2405-4d85-80ce-4e954b956f1b
                Copyright © 2022 Sherina, de Vries, Kharlamova, Sippl, Jiang, Brynedal, Kindstedt, Hansson, Mathsson-Alm, Israelsson, Stålesen, Saevarsdottir, Holmdahl, Hensvold, Johannsen, Eriksson, Sallusto, Catrina, Rönnelid, Grönwall, Yucel-Lindberg, Alfredsson, Klareskog, Piccoli, Malmström, Amara and Lundberg

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 29 October 2021
                : 17 March 2022
                Page count
                Figures: 5, Tables: 2, Equations: 0, References: 60, Pages: 13, Words: 6334
                Funding
                Funded by: Vetenskapsrådet , doi 10.13039/501100004359;
                Funded by: Stiftelsen Konung Gustaf V:s 80-årsfond , doi 10.13039/501100007857;
                Funded by: Reumatikerförbundet , doi 10.13039/501100007949;
                Funded by: Stiftelsen Professor Nanna Svartz Fond , doi 10.13039/501100009800;
                Funded by: Innovative Medicines Initiative , doi 10.13039/501100010767;
                Funded by: Helmut Horten Stiftung , doi 10.13039/501100013850;
                Categories
                Immunology
                Original Research

                Immunology
                rheumatoid arthritis (ra),anti-citrullinated protein antibodies (acpa),porphyromonas gingivalis (pg),periodontitis (pd),monoclonal antibodies (mabs),b cells

                Comments

                Comment on this article