Abstract

The pathogenesis and progression of knee inflammatory pathologies is modulated partly by residing macrophages in the infrapatellar fat pad (IFP), thus, macrophage polarization towards pro-inflammatory (M1) or anti-inflammatory (M2) phenotypes is important in joint disease pathologies. Alteration of M1/M2 balance contributes to the initiation and progression of joint inflammation and can be potentially altered with mesenchymal stem cell (MSC) therapy. In an acute synovial/IFP inflammation rat model a single intra-articular injection of IFP-MSC was performed, having as controls (1) diseased rats not receiving IFP-MSC and (2) non-diseased rats. After 4 days, cell specific transcriptional profiling via single-cell RNA-sequencing was performed on isolated IFP tissue from each group. Eight transcriptomically distinct cell populations were identified within the IFP across all three treatment groups with a noted difference in the proportion of myeloid cells across the groups. Largely myeloid cells consisted of macrophages (>90%); one M1 sub-cluster highly expressing pro-inflammatory markers and two M2 sub-clusters with one of them expressing higher levels of canonical M2 markers. Notably, the diseased samples (11.9%) had the lowest proportion of cells expressing M2 markers relative to healthy (14.8%) and MSC treated (19.4%) samples. These results suggest a phenotypic polarization of IFP macrophages towards the pro-inflammatory M1 phenotype in an acute model of inflammation, which are alleviated by IFP-MSC therapy inducing a switch towards an alternate M2 status. Understanding the IFP cellular heterogeneity and associated transcriptional programs may offer insights into novel therapeutic strategies for disabling joint disease pathologies.

Highlights

  • We dissected by single-cell RNA-sequencing the infrapatellar fat pad (IFP) cellular heterogeneity and associated transcriptional programs to help elucidate the dynamic cellular programs involved in homeostasis and potentially synovitis/IFP fibrosis

  • We clearly demonstrated that these structural transitional changes between diseased and treated animals were reflected in the cell population proportions, with IFP-mesenchymal stem cell (MSC) infused fat pads increasing the proportion of macrophages making up the IFP

  • We propose that synoviumbut IFP-derived resident macrophages are present in homeostasis, and together with infiltrating macrophages are strongly involved in both the progression and resolution of knee joint inflammation through phenotypic polarization towards pro- and anti-inflammatory states

Read more

Summary

Introduction

The IFP and synovium undergo cycles of inflammation inducing progressive fibrotic changes, supporting synovitis and IFP fibrosis as key elements during the onset and progression of joint disease [2,3,4,5,6,7,8,9,10], in those conditions possessing an inflammatory phenotype [19,20] On this basis, the IFP is a potential therapeutic target to mitigate synovitis/IFP fibrosis incidence and progression [9,21,22]. An IFPbased therapeutic strategy aimed at restoring joint homeostasis could potentially mitigate progression of synovitis/IFP fibrosis-related disabling pain and joint degeneration

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call