Abstract

Posttraumatic activation of the catabolic cascade plays a major role in degradation of cartilage. Interleukin-1β (IL-1β), a primary instigator in the catabolic axis, is upregulated in chondrocytes following injury. IL-1β activates key degradative enzymes, including MMPs and aggrecanases, and other proinflammatory mediators such as PGE2 which contribute to ECM breakdown. Posttranscriptional silencing of IL-1β by RNA interference (RNAi) may drive a reduction in IL-1β. We hypothesized that transduction of chondrocytes using rAAV2 expressing a short hairpin RNAi motif targeting IL-1β (shIL-1β) would significantly decrease IL-1β expression and, in turn, decrease expression of other catabolic enzymes. Chondrocyte cultures were transduced with rAAV2-tdT-shIL-1β in serum-free media. The fluorescent protein, tdTomato, was used to determine transduction efficiency via flow cytometry and fluorescent microscopy. Cells were stimulated with lipopolysaccharide (LPS) 48 hours following transduction. After 24-hour stimulation, supernatants were collected for cytokine analysis, and cells lysed for gene expression analysis. IL-1β knockdown led to significantly decreased expression of IL-1β, TNF-α, and ADAMTS5. PGE2 synthesis was also significantly downregulated. Overall, effective silencing of IL-1β using rAAV2 vector expressing a short hairpin IL-1β knockdown sequence was shown. Additionally, significant downstream effects were evident, including decreased expression of TNF-α and ADAMTS5. Targeted silencing of catabolic cytokines may provide a promising treatment avenue for osteoarthritic (OA) joints.

Highlights

  • Osteoarthritis (OA) is a prevalent, debilitating disease that currently lacks any effective treatment and often culminates in total joint replacement surgery

  • The objective of this study was to evaluate the effects of rAAV2 mediated knockdown of IL-1β on gene expression and protein synthesis in chondrocytes cultured in an OA model

  • Posttranscriptional silencing of IL-1β with an rAAV2 vector expressing a short hairpin sequence targeting equine IL-1β led to effective reduction in IL-1β in chondrocytes stimulated with LPS

Read more

Summary

Introduction

Osteoarthritis (OA) is a prevalent, debilitating disease that currently lacks any effective treatment and often culminates in total joint replacement surgery. It is characterized by progressive degradation of the extracellular matrix (ECM) leading to joint swelling, reduced mobility, and pain. IL-1β decreases expression of collagen type II and inhibits aggrecan synthesis [7, 8]. Inflammatory mediators, such as prostaglandin E2 (PGE2) [9] and nitric oxide (NO) [10], are increased by IL-1β and likely play a significant role in the pain response associated with OA

Objectives
Methods
Findings
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