1. Academic Validation
  2. TFE3, a potential therapeutic target for Spinal Cord Injury via augmenting autophagy flux and alleviating ER stress

TFE3, a potential therapeutic target for Spinal Cord Injury via augmenting autophagy flux and alleviating ER stress

  • Theranostics. 2020 Jul 23;10(20):9280-9302. doi: 10.7150/thno.46566.
Kailiang Zhou 1 2 Zhilong Zheng 2 Yao Li 1 Wen Han 2 Jing Zhang 2 Yuqin Mao 2 Huanwen Chen 3 Wanying Zhang 2 Mi Liu 2 Ling Xie 2 Hongyu Zhang 2 Huazi Xu 1 Jian Xiao 1 2
Affiliations

Affiliations

  • 1 Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
  • 2 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China.
  • 3 University of Maryland School of Medicine, Baltimore, MD 21201, USA.
Abstract

Background and Aim: Increasing evidence suggests that spinal cord injury (SCI)-induced defects in autophagic flux may contribute to an impaired ability for neurological repair following injury. Transcription factor E3 (TFE3) plays a crucial role in oxidative metabolism, lysosomal homeostasis, and Autophagy induction. Here, we investigated the role of TFE3 in modulating Autophagy following SCI and explored its impact on neurological recovery. Methods: Histological analysis via HE, Nissl and Mason staining, survival rate analysis, and behavioral testing via BMS and footprint analysis were used to determine functional recovery after SCI. Quantitative real-time polymerase chain reaction, Western blotting, immunofluorescence, TUNEL staining, enzyme-linked immunosorbent assays, and immunoprecipitation were applied to examine levels of Autophagy flux, ER-stress-induced Apoptosis, oxidative stress, and AMPK related signaling pathways. In vitro studies using PC12 cells were performed to discern the relationship between ROS accumulation and Autophagy flux blockade. Results: Our results showed that in SCI, defects in Autophagy flux contributes to ER stress, leading to neuronal death. Furthermore, SCI enhances the production of Reactive Oxygen Species (ROS) that induce lysosomal dysfunction to impair Autophagy flux. We also showed that TFE3 levels are inversely correlated with ROS levels, and increased TFE3 levels can lead to improved outcomes. Finally, we showed that activation of TFE3 after SCI is partly regulated by AMPK-mTOR and AMPK-SKP2-CARM1 signaling pathways. Conclusions: TFE3 is an important regulator in ROS-mediated Autophagy dysfunction following SCI, and TFE3 may serve as a promising target for developing treatments for SCI.

Keywords

AMPK signaling pathways; Autophagy; ER stress-induced apoptosis; Spinal cord injury; TFE3.

Figures
Products