1. Academic Validation
  2. Circular RNA circNFKB1 promotes osteoarthritis progression through interacting with ENO1 and sustaining NF-κB signaling

Circular RNA circNFKB1 promotes osteoarthritis progression through interacting with ENO1 and sustaining NF-κB signaling

  • Cell Death Dis. 2022 Aug 9;13(8):695. doi: 10.1038/s41419-022-05148-2.
Su'an Tang  # 1 2 Xiaoyu Nie  # 3 Jianzhao Ruan 3 Yumei Cao 3 4 Jingliang Kang 3 Changhai Ding 5 6
Affiliations

Affiliations

  • 1 Clinical Research Centre, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, Guangdong, China. [email protected].
  • 2 Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, Guangdong, China. [email protected].
  • 3 Clinical Research Centre, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, Guangdong, China.
  • 4 Department of Rheumatology and Immunology, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, Guangdong, China.
  • 5 Clinical Research Centre, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, Guangdong, China. [email protected].
  • 6 Menzies Institute for Medical Research, University of Tasmania, 7000, Hobart, Tasmania, Australia. [email protected].
  • # Contributed equally.
Abstract

Inflammatory cytokines-induced activation of the nuclear factor κB (NF-κB) pathway plays a critical role in the pathogenesis of osteoarthritis (OA). Circular RNA (circRNA) has been identified as important epigenetic factor in numerous diseases. However, the biological roles of inflammation-related circRNAs in regulating OA pathogenesis remain elusive. Here, we revealed circRNA expression profiles in human primary chondrocytes with interleukin-1β (IL-1β) stimulation by circRNA sequencing. We identified a highly upregulated circRNA, termed as circNFKB1 in inflamed chondrocytes and osteoarthritic cartilage. As a circRNA derived from exon 2-5 of NFKB1, circNFKB1 is located in both cytoplasm and nucleus of chondrocytes. Furthermore, knockdown of circNFKB1 inhibited extracellular matrix (ECM) catabolism and rescued IL-1β impaired ECM anabolism whereas ectopic expression of circNFKB1 significantly promoted chondrocytes degradation in vitro. Moreover, intraarticular injection of adenovirus-circNFKB1 in mouse joints triggered spontaneous cartilage loss and OA development. Mechanistically, circNFKB1 interacted with α-enolase (ENO1), regulated the expression of its parental gene NFKB1 and sustained the activation of NF-κB signaling pathway in chondrocytes. Therefore, this study highlights a novel ENO1-interacting circNFKB1 in OA pathogenesis, and provides valuable insights into understanding the regulatory mechanism of NF-κB signaling in chondrocytes and a promising therapeutic target for the treatment of OA.

Figures
Products