Suppression of NF-κB activity via nanoparticle-based siRNA delivery alters early cartilage responses to injury

H Yan, X Duan, H Pan, N Holguin… - Proceedings of the …, 2016 - National Acad Sciences
H Yan, X Duan, H Pan, N Holguin, MF Rai, A Akk, LE Springer, SA Wickline, LJ Sandell
Proceedings of the National Academy of Sciences, 2016National Acad Sciences
Osteoarthritis (OA) is a major cause of disability and morbidity in the aging population. Joint
injury leads to cartilage damage, a known determinant for subsequent development of
posttraumatic OA, which accounts for 12% of all OA. Understanding the early molecular and
cellular responses postinjury may provide targets for therapeutic interventions that limit
articular degeneration. Using a murine model of controlled knee joint impact injury that
allows the examination of cartilage responses to injury at specific time points, we show that …
Osteoarthritis (OA) is a major cause of disability and morbidity in the aging population. Joint injury leads to cartilage damage, a known determinant for subsequent development of posttraumatic OA, which accounts for 12% of all OA. Understanding the early molecular and cellular responses postinjury may provide targets for therapeutic interventions that limit articular degeneration. Using a murine model of controlled knee joint impact injury that allows the examination of cartilage responses to injury at specific time points, we show that intraarticular delivery of a peptidic nanoparticle complexed to NF-κB siRNA significantly reduces early chondrocyte apoptosis and reactive synovitis. Our data suggest that NF-κB siRNA nanotherapy maintains cartilage homeostasis by enhancing AMPK signaling while suppressing mTORC1 and Wnt/β-catenin activity. These findings delineate an extensive crosstalk between NF-κB and signaling pathways that govern cartilage responses postinjury and suggest that delivery of NF-κB siRNA nanotherapy to attenuate early inflammation may limit the chronic consequences of joint injury. Therapeutic benefits of siRNA nanotherapy may also apply to primary OA in which NF-κB activation mediates chondrocyte catabolic responses. Additionally, a critical barrier to the successful development of OA treatment includes ineffective delivery of therapeutic agents to the resident chondrocytes in the avascular cartilage. Here, we show that the peptide–siRNA nanocomplexes are nonimmunogenic, are freely and deeply penetrant to human OA cartilage, and persist in chondrocyte lacunae for at least 2 wk. The peptide–siRNA platform thus provides a clinically relevant and promising approach to overcoming the obstacles of drug delivery to the highly inaccessible chondrocytes.
National Acad Sciences