Nature Communications (Feb 2024)

A single cell atlas of frozen shoulder capsule identifies features associated with inflammatory fibrosis resolution

  • Michael T. H. Ng,
  • Rowie Borst,
  • Hamez Gacaferi,
  • Sarah Davidson,
  • Jessica E. Ackerman,
  • Peter A. Johnson,
  • Caio C. Machado,
  • Ian Reekie,
  • Moustafa Attar,
  • Dylan Windell,
  • Mariola Kurowska-Stolarska,
  • Lucy MacDonald,
  • Stefano Alivernini,
  • Micon Garvilles,
  • Kathrin Jansen,
  • Ananya Bhalla,
  • Angela Lee,
  • James Charlesworth,
  • Rajat Chowdhury,
  • Paul Klenerman,
  • Kate Powell,
  • Carl-Philip Hackstein,
  • ICECAP Consortium,
  • Dominic Furniss,
  • Jonathan Rees,
  • Derek Gilroy,
  • Mark Coles,
  • Andrew J. Carr,
  • Stephen N. Sansom,
  • Christopher D. Buckley,
  • Stephanie G. Dakin

DOI
https://doi.org/10.1038/s41467-024-45341-9
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 21

Abstract

Read online

Abstract Frozen shoulder is a spontaneously self-resolving chronic inflammatory fibrotic human disease, which distinguishes the condition from most fibrotic diseases that are progressive and irreversible. Using single-cell analysis, we identify pro-inflammatory MERTKlowCD48+ macrophages and MERTK + LYVE1 + MRC1+ macrophages enriched for negative regulators of inflammation which co-exist in frozen shoulder capsule tissues. Micro-cultures of patient-derived cells identify integrin-mediated cell-matrix interactions between MERTK+ macrophages and pro-resolving DKK3+ and POSTN+ fibroblasts, suggesting that matrix remodelling plays a role in frozen shoulder resolution. Cross-tissue analysis reveals a shared gene expression cassette between shoulder capsule MERTK+ macrophages and a respective population enriched in synovial tissues of rheumatoid arthritis patients in disease remission, supporting the concept that MERTK+ macrophages mediate resolution of inflammation and fibrosis. Single-cell transcriptomic profiling and spatial analysis of human foetal shoulder tissues identify MERTK + LYVE1 + MRC1+ macrophages and DKK3+ and POSTN+ fibroblast populations analogous to those in frozen shoulder, suggesting that the template to resolve fibrosis is established during shoulder development. Crosstalk between MerTK+ macrophages and pro-resolving DKK3+ and POSTN+ fibroblasts could facilitate resolution of frozen shoulder, providing a basis for potential therapeutic resolution of persistent fibrotic diseases.