Abstract

Evidence links aryl hydrocarbon receptor (AHR) activation to rheumatoid arthritis (RA) pathogenesis, although results are inconsistent. AHR agonists inhibit pro-inflammatory cytokine expression in macrophages, pivotal cells in RA aetiopathogenesis, which hints at specific circuits that regulate the AHR pathway in RA macrophages. We compared microRNA (miR) expression in CD14+ cells from patients with active RA or with osteoarthritis (OA). Seven miR were downregulated and one (miR-223) upregulated in RA compared to OA cells. miR-223 upregulation correlated with reduced Notch3 and Notch effector expression in RA patients. Overexpression of the Notch-induced repressor HEY-1 and co-culture of healthy donor monocytes with Notch ligand-expressing cells showed direct Notch-mediated downregulation of miR-223. Bioinformatics predicted the AHR regulator ARNT (AHR nuclear translocator) as a miR-223 target. Pre-miR-223 overexpression silenced ARNT 3’UTR-driven reporter expression, reduced ARNT (but not AHR) protein levels and prevented AHR/ARNT-mediated inhibition of pro-inflammatory cytokine expression. miR-223 counteracted AHR/ARNT-induced Notch3 upregulation in monocytes. Levels of ARNT and of CYP1B1, an AHR/ARNT signalling effector, were reduced in RA compared to OA synovial tissue, which correlated with miR-223 levels. Our results associate Notch signalling to miR-223 downregulation in RA macrophages, and identify miR-223 as a negative regulator of the AHR/ARNT pathway through ARNT targeting.

Highlights

  • Targets; this causes their degradation or translation repression depending on the degree of base-pair complementarity between the miR and its target mRNA5

  • We found no differences in aryl hydrocarbon receptor (AHR) mRNA levels between OA and RA patients in our cohort (Supplementary Fig. S7b), which suggests that the functional impairment of the AHR/ ARNT pathway is a result of reduced ARNT protein levels

  • We combined gene expression and advanced bioinformatics analyses to demonstrate that (i) miR-223 is selectively upregulated in CD14+ cells from synovial fluid (SF) of RA patients with active disease; (ii) Notch activation represses miR-223 expression in myeloid cells; (iii) miR-223 targets ARNT, a necessary co-receptor for AHR-dependent transcriptional activity; (iv) miR-223 prevents AHR-mediated inhibition of pro-inflammatory cytokines; and (v) AHR target genes and ARNT protein itself are downmodulated in RA compared to OA synovial tissues

Read more

Summary

Introduction

Targets; this causes their degradation or translation repression depending on the degree of base-pair complementarity between the miR and its target mRNA5. MiR participate in immune cell differentiation, maturation and function[6], and their aberrant expression is associated with pathogenesis in several osteoarticular diseases. The miR profile in RA patients differs from that in patients with OA9, a non-autoimmune disease characterised by low inflammation levels. The relevance of these miR in RA aetiopathogenesis is poorly understood. AHR stimulation inhibits TNF-α , IL-6 and IL-1β expression in lipopolysaccharide (LPS)-stimulated macrophages[21,22,23]. These disparate results hint at cell type-specific circuits that regulate the AHR/ARNT pathway in specific contexts. We analysed the role of miR-223 as a part of these regulatory circuits in RA-derived macrophages

Methods
Results
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