Increased Galectin-9 Levels Correlate with Disease Activity in Patients with DMARD-Naïve Rheumatoid Arthritis and Modulate the Secretion of MCP-1 and IL-6 from Synovial Fibroblasts

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Dokumenter

  • Fulltext

    Forlagets udgivne version, 1,76 MB, PDF-dokument

  • Morten A. Nielsen
  • Ditte Køster
  • Akul Y. Mehta
  • Kristian Stengaard-Pedersen
  • Pierre Busson
  • Peter Junker
  • Kim Hørslev-Petersen
  • Hetland, Merete Lund
  • Østergaard, Mikkel
  • Malene Hvid
  • Hakon Leffler
  • Tue W. Kragstrup
  • Richard D. Cummings
  • Bent Deleuran
Background: Fibroblast-like synoviocytes (FLSs) are essential mediators in the expansive growth and invasiveness of rheumatoid synovitis, and patients with a fibroblastic-rich pauci-immune pathotype respond poorly to currently approved antirheumatic drugs. Galectin-9 (Gal-9) has been reported to directly modulate rheumatoid arthritis (RA) FLSs and to hold both pro- and anti-inflammatory properties. The objective of this study was to evaluate clinical and pathogenic aspects of Gal-9 in RA, combining national patient cohorts and cellular models. Methods: Soluble Gal-9 was measured in plasma from patients with newly diagnosed, treatment-naïve RA (n = 98). The disease activity score 28-joint count C-reactive protein (DAS28CRP) and total Sharp score were used to evaluate the disease course serially over a two-year period. Plasma and synovial fluid samples were examined for soluble Gal-9 in patients with established RA (n = 18). A protein array was established to identify Gal-9 binding partners in the extracellular matrix (ECM). Synovial fluid mononuclear cells (SFMCs), harvested from RA patients, were used to obtain synovial-fluid derived FLSs (SF-FLSs) (n = 7). FLSs from patients suffering from knee Osteoarthritis (OA) were collected from patients when undergoing joint replacement surgery (n = 5). Monocultures of SF-FLSs (n = 6) and autologous co-cultures of SF-FLSs and peripheral blood mononuclear cells (PBMCs) were cultured with and without a neutralizing anti-Gal-9 antibody (n = 7). The mono- and co-cultures were subsequently analyzed by flow cytometry, MTT assay, and ELISA. Results: Patients with early and established RA had persistently increased plasma levels of Gal-9 compared with healthy controls (HC). The plasma levels of Gal-9 were associated with disease activity and remained unaffected when adding a TNF-inhibitor to their standard treatment. Gal-9 levels were elevated in the synovial fluid of established RA patients with advanced disease, compared with corresponding plasma samples. Gal-9 adhered to fibronectin, laminin and thrombospondin, while not to interstitial collagens in the ECM protein array. In vitro, a neutralizing Gal-9 antibody decreased MCP-1 and IL-6 production from both RA FLSs and OA FLSs. In co-cultures of autologous RA FLSs and PBMCs, the neutralization of Gal-9 also decreased MCP-1 and IL-6 production, without affecting the proportion of inflammatory FLSs. Conclusions: In RA, pretreatment plasma Gal-9 levels in early RA were increased and correlated with clinical disease activity. Gal-9 levels remained increased despite a significant reduction in the disease activity score in patients with early RA. The in vitro neutralization of Gal-9 decreased both MCP-1 and IL-6 production in an inflammatory subset of RA FLSs. Collectively these findings indicate that the persistent overexpression of Gal-9 in RA may modulate synovial FLS activities and could be involved in the maintenance of subclinical disease activity in RA.
OriginalsprogEngelsk
Artikelnummer327
TidsskriftCells
Vol/bind12
Udgave nummer2
Antal sider13
ISSN2073-4409
DOI
StatusUdgivet - 2023

Bibliografisk note

Funding Information:
We owe a debt of gratitude to Karin Skovgård Sørensen (Dept. of Biomedicine, Aarhus University) for technical assistance concerning the ELISA data and the FACS Core Facility (Aarhus University, Denmark) for technical assistant regarding Flow cytometry. We greatly acknowledge the assistance of medical doctors and nurses at the Department of Rheumatology, Aarhus University Hospital for helping to collect the patient samples. We also acknowledge the generous grants from Aarhus University Research Foundation and the Danish Rheumatism Association. Graphical abstract: Created with BioRender.com , License was granted to permit sublicensed for use in journal publications, agreement nr: WX24VRNF8E.

Funding Information:
This research was funded by the National Institute for General Medical Sciences NIH grants R24GM137763 and P41GM103694 to RDC and by a grant from Aarhus University Research Foundation (NOVA, AUFF-E-2016-9-27) and the Danish Rheumatoid Association (R188-A6589).

Publisher Copyright:
© 2023 by the authors.

ID: 362892691