Article Text

Download PDFPDF

Anticitrullinated protein antibodies facilitate migration of synovial tissue-derived fibroblasts
  1. Meng Sun1,
  2. Bence Rethi1,
  3. Akilan Krishnamurthy1,
  4. Vijay Joshua1,
  5. Alexandra Circiumaru1,
  6. Aase Haj Hensvold1,
  7. Elena Ossipova1,
  8. Caroline Grönwall1,
  9. Yanying Liu2,
  10. Marianne Engstrom1,
  11. Sergiu Bogdan Catrina3,
  12. Johanna Steen1,
  13. Vivianne Malmstrom1,
  14. Lars Klareskog1,
  15. Camilla Svensson4,
  16. Caroline Ospelt5,
  17. Heidi Wähämaa1,
  18. Anca Irinel Catrina1
  1. 1 Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
  2. 2 Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
  3. 3 Molecular Medicine and Surgery, Karolinska University Hospital and Institutet, Stockholm, Sweden
  4. 4 Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
  5. 5 Center of Experimental Rheumatology, Zurich, Switzerland
  1. Correspondence to Dr Anca Irinel Catrina, Rheumatology Unit, Stockholm SE-17176, Sweden; anca.catrina{at}ki.se

Abstract

Objectives Rheumatoid arthritis (RA)-specific anti-citrullinated protein/peptide antibodies (ACPAs) might contribute to bone loss and arthralgia before the onset of joint inflammation. We aimed to dissect additional mechanisms by which ACPAs might contribute to development of joint pathology.

Methods Fibroblast-like synoviocytes (FLS) were isolated from the synovial membrane of patients with RA. The FLS cultures were stimulated with polyclonal ACPAs (anti-CCP-2 antibodies) purified from the peripheral blood of patients with RA or with monoclonal ACPAs derived from single synovial fluid B cells. We analysed how ACPAs modulate FLS by measuring cell adhesion and mobility as well as cytokine production. Expression of protein arginine deiminase (PAD) enzymes and protein citrullination were analysed by immunofluorescence, and signal transduction was studied using immunoblotting.

Results Challenge of FLS by starvation-induced stress or by exposure to the chemokine interleukin-8 was essential to sensitise the cells to ACPAs. These challenges led to an increased PAD expression and protein citrullination and an ACPA-mediated induction of FLS migration through a mechanism involving phosphoinositide 3-kinase activation. Inhibition of the PAD enzymes or competition with soluble citrullinated proteins or peptides completely abolished the ACPA-induced FLS migration. Different monoclonal ACPAs triggered distinct cellular effects in either fibroblasts or osteoclasts, suggesting unique roles for individual ACPA clones in disease pathogenesis.

Conclusion We propose that transient synovial insults in the presence of a certain pre-existing ACPA repertoire might result in an ACPA-mediated increase of FLS migration.

  • Anti-CCP
  • Rheumatoid Arthritis
  • Fibroblasts
  • Autoantibodies
  • Autoimmune Diseases

This is an open access article distributed in accordance with the Creative Commons Attribution 4.0 Unported (CC BY 4.0) license, which permits others to copy, redistribute, remix, transform and build upon this work for any purpose, provided the original work is properly cited, a link to the licence is given, and indication of whether changes were made. See: https://creativecommons.org/licenses/by/4.0/.

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Footnotes

  • Handling editor Josef S Smolen

  • Contributors MS, BR and AIC designed the experiments, analysed the data and wrote the manuscript along with input from VM, HW, CS, AHH, SBC and LK. MS designed and performed all fibroblast experiments with help from BR. MS and CO designed and performed adhesion assay. VJ, YL and HW measured chemokines and cytokines in supernatants. MS and ME performed all immunohistochemistry experiments with help from AC. EO perform and analysed mass spectrometry data. AK conducted osteoclast assays. AHH recruited patients and characterised all clinical data. HW purified polyclonal ACPAs. JS, CG and VM produced and validated monoclonal ACPAs. AIC, MS, BR, AK, VJ, HW, AC, VM and LK discussed and developed the concept. All authors critically reviewed and approved the final form of the manuscript.

  • Funding This project has received funding from FOREUM, Foundation for Research in Rheumatology, from the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (grant agreement CoG 2017—7722209_PREVENT RA and grant agreement 777357_RTCure), from the Swedish Research Council and Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse.

  • Competing interests None declared.

  • Patient consent for publication Not required.

  • Ethics approval This study involves human participants with ethical permit listed in blow: (1) Kartläggning av prediktiva biomarkörer vid kronisk artrit ID: 2009-358-31-3; (2) Kartläggning av inflammatoriska mediatorers betydelse för sjukdomsförlopp vid kroniskaledsjukdomar ID:2009-1262-31-3.

  • Provenance and peer review Not commissioned; externally peer reviewed.

  • Data availability statement Data are available on reasonable request.