TFEB, a potential therapeutic target for osteoarthritis via autophagy regulation

The blockage of autophagic flux in chondrocytes has been considered as a major reason for the excessive cellular apoptosis and senescence in osteoarthritis (OA) development; however, the molecular mechanism and therapeutic strategy for interrupted autophagic flux is still not clear. Most recently, t...

Full description

Saved in:
Bibliographic Details
Published inCell death & disease Vol. 9; no. 9; pp. 858 - 15
Main Authors Zheng, Gang, Zhan, Yu, Li, Xiaobin, Pan, Zongyou, Zheng, Fanghong, Zhang, Zengjie, Zhou, Yifei, Wu, Yaosen, Wang, Xiangyang, Gao, Weiyang, Xu, Huazi, Tian, Naifeng, Zhang, Xiaolei
Format Journal Article
LanguageEnglish
Published London Nature Publishing Group UK 28.08.2018
Springer Nature B.V
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:The blockage of autophagic flux in chondrocytes has been considered as a major reason for the excessive cellular apoptosis and senescence in osteoarthritis (OA) development; however, the molecular mechanism and therapeutic strategy for interrupted autophagic flux is still not clear. Most recently, the transcription factor EB (TFEB) is identified as a master regulator for autophagic flux via initiating the expression of multiple autophagy-related genes and lysosomal biogenesis. This research was performed to confirm whether TFEB expression and activity are impacted in OA development and to confirm the effect of genetic up-regulation of TFEB on autophagic flux and cellular protection in the in vitro and in vivo models of OA. We demonstrated that the expression and nuclear localization of TFEB is decreased in human and mouse OA cartilage as well as in tert-Butyl hydroperoxide (TBHP)-treated chondrocytes. Applying lentivirus to transfect chondrocytes, we found that TFEB overexpression rescues the TBHP-induced the autophagic flux damage, lysosome dysfunction and protects chondrocyte against TBHP induced apoptosis and senescence; these protections of TFEB are diminished by chloroquine-medicated autophagy inhibition. Our destabilized medial meniscus (DMM) mouse OA model shows that TFEB overexpression ameliorates the surgery-induced cartilage degradation, restrains the apoptosis and senescence of chondrocyte, and enhances the autophagic flux. In summary, our study indicates that the activity of TFEB in chondrocyte is involved in OA development, also TFEB overexpression may be a promising strategy for OA treatment.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2041-4889
2041-4889
DOI:10.1038/s41419-018-0909-y