Int J Biol Sci 2024; 20(5):1905-1926. doi:10.7150/ijbs.91480 This issue Cite

Research Paper

Impaired TFEB-mediated autophagy-lysosome fusion promotes tubular cell cycle G2/M arrest and renal fibrosis by suppressing ATP6V0C expression and interacting with SNAREs

Xiang Ren1,2, Jing Wang1,2, Huizhi Wei3,4, Xing Li1,2, Yiqun Tian1,2, Zhixian Wang5, Yisheng Yin1,2, Zihao Guo1,2, Zhenliang Qin1,2, Minglong Wu6✉, Xiaoyong Zeng1,2✉

1. Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
2. Hubei Provincial Institute of Urology, Wuhan, China.
3. School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, China.
4. Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Taiyuan, China.
5. Department of Urology, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
6. Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.

Citation:
Ren X, Wang J, Wei H, Li X, Tian Y, Wang Z, Yin Y, Guo Z, Qin Z, Wu M, Zeng X. Impaired TFEB-mediated autophagy-lysosome fusion promotes tubular cell cycle G2/M arrest and renal fibrosis by suppressing ATP6V0C expression and interacting with SNAREs. Int J Biol Sci 2024; 20(5):1905-1926. doi:10.7150/ijbs.91480. https://www.ijbs.com/v20p1905.htm
Other styles

File import instruction

Abstract

Graphic abstract

Increasing evidence suggests that autophagy plays a major role during renal fibrosis. Transcription factor EB (TFEB) is a critical regulator of autophagy- and lysosome-related gene transcription. However, the pathophysiological roles of TFEB in renal fibrosis and fine-tuned mechanisms by which TFEB regulates fibrosis remain largely unknown. Here, we found that TFEB was downregulated in unilateral ureteral obstruction (UUO)-induced human and mouse fibrotic kidneys, and kidney-specific TFEB overexpression using recombinant AAV serotype 9 (rAAV9)-TFEB in UUO mice alleviated renal fibrosis pathogenesis. Mechanically, we found that TFEB's prevention of extracellular matrix (ECM) deposition depended on autophagic flux integrity and its subsequent blockade of G2/M arrest in tubular cells, rather than the autophagosome synthesis. In addition, we together RNA-seq with CUT&Tag analysis to determine the TFEB targeted gene ATP6V0C, and revealed that TFEB was directly bound to the ATP6V0C promoter only at specific site to promote its expression through CUT&Run-qPCR and luciferase reporter assay. Interestingly, TFEB induced autophagic flux integrity, mainly dependent on scaffold protein ATP6V0C-mediated autophagosome-lysosome fusion by bridging with STX17 and VAMP8 (major SNARE complex) by co-immunoprecipitation analysis, rather than its mediated lysosomal acidification and degradation function. Moreover, we further investigated the underlying mechanism behind the low expression of TEFB in UUO-induced renal fibrosis, and clearly revealed that TFEB suppression in fibrotic kidney was due to DNMT3a-associated TFEB promoter hypermethylation by utilizing methylation specific PCR (MSP) and bisulfite-sequencing PCR (BSP), which could be effectively recovered by 5-Aza-2'-deoxycytidine (5A-za) to alleviate renal fibrosis pathogenesis. These findings reveal for the first time that impaired TFEB-mediated autophagosome-lysosome fusion disorder, tubular cell G2/M arrest and renal fibrosis appear to be sequentially linked in UUO-induced renal fibrosis and suggest that DNMT3a/TFEB/ATP6V0C may serve as potential therapeutic targets to prevent renal fibrosis.

Keywords: Renal fibrosis, transcription factor EB, autophagy, cell cycle, V-ATPase, methylation


Citation styles

APA
Ren, X., Wang, J., Wei, H., Li, X., Tian, Y., Wang, Z., Yin, Y., Guo, Z., Qin, Z., Wu, M., Zeng, X. (2024). Impaired TFEB-mediated autophagy-lysosome fusion promotes tubular cell cycle G2/M arrest and renal fibrosis by suppressing ATP6V0C expression and interacting with SNAREs. International Journal of Biological Sciences, 20(5), 1905-1926. https://doi.org/10.7150/ijbs.91480.

ACS
Ren, X.; Wang, J.; Wei, H.; Li, X.; Tian, Y.; Wang, Z.; Yin, Y.; Guo, Z.; Qin, Z.; Wu, M.; Zeng, X. Impaired TFEB-mediated autophagy-lysosome fusion promotes tubular cell cycle G2/M arrest and renal fibrosis by suppressing ATP6V0C expression and interacting with SNAREs. Int. J. Biol. Sci. 2024, 20 (5), 1905-1926. DOI: 10.7150/ijbs.91480.

NLM
Ren X, Wang J, Wei H, Li X, Tian Y, Wang Z, Yin Y, Guo Z, Qin Z, Wu M, Zeng X. Impaired TFEB-mediated autophagy-lysosome fusion promotes tubular cell cycle G2/M arrest and renal fibrosis by suppressing ATP6V0C expression and interacting with SNAREs. Int J Biol Sci 2024; 20(5):1905-1926. doi:10.7150/ijbs.91480. https://www.ijbs.com/v20p1905.htm

CSE
Ren X, Wang J, Wei H, Li X, Tian Y, Wang Z, Yin Y, Guo Z, Qin Z, Wu M, Zeng X. 2024. Impaired TFEB-mediated autophagy-lysosome fusion promotes tubular cell cycle G2/M arrest and renal fibrosis by suppressing ATP6V0C expression and interacting with SNAREs. Int J Biol Sci. 20(5):1905-1926.

This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
Popup Image