Abstract

Spinal cord injury (SCI) is a devastating disease and causes tissue loss and neurologic dysfunction, contributing to high morbidity and disability among human. However, the underlying molecular mechanisms still remain unclear. Tumor necrosis factor-α-induced protein 8 (TNFAIP8) is a member of the TNFAIP8/TIPE family, and has been implicated in different diseases associated with inflammation, infection, and immunity. Nevertheless, its effects on SCI have not been well investigated. In our study, we found time course of TNFAIP8 following SCI in mice, along with time-dependent increases of pro-inflammatory cytokines. The in vitro results confirmed the up-regulation of TNFAIP8 induced by lipopolysaccharide (LPS). Subsequently, we found that reducing TNFAIP8 by transfection with its specific siRNA (siTNFAIP8) markedly alleviated cell viability and inflammatory response caused by LPS in mouse microglial BV2 cells. Importantly, LPS-enhanced activation of inhibitor of κBα/nuclear factor-κB (IκBα/NF-κB) and phosphoinositide 3-kinase/serine-threonine kinase (PI3K/AKT) signaling pathways was considerably blunted by siTNFAIP8. Intriguingly, our results further showed that siTNFAIP8-restrained inflammation and IκBα/NF-κB in LPS-stimulated BV2 cells were almost abolished by the pre-treatment of AKT activator SC-79, demonstrating that TNFAIP8-regulated inflammatory response was largely dependent on AKT activation. Then, the in vivo studies were performed using the wild type (WT) and TNFAIP8-knockout (KO) mice with or without SCI operation. Results showed that TNFAIP8-KO mice exhibited improved neuron injury and locomotor function along with decreased microglial activity. Furthermore, compared with the WT/SCI mice, the expression of pro-inflammatory cytokines in spinal cords was markedly down-regulated by TNFAIP8-deficiency through blocking IκBα/NF-κB and PI3K/AKT signaling pathways. Taken together, these findings elucidated the novel role of TNFAIP8 in regulating SCI via the AKT signaling, and thus TNFAIP8 may be served as a promising therapeutic target for SCI treatment.

Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.