Article

Satellite cell-derived exosome-mediated delivery of microRNA-23a/27a/26a cluster ameliorates the renal tubulointerstitial fibrosis in mouse diabetic nephropathy

Jia-ling Ji1, Hui-min Shi1, Zuo-lin Li2, Ran Jin1, Gao-ting Qu1, Hui Zheng1, E. Wang1, Yun-yang Qiao1, Xing-yue Li1, Ling Ding3, Da-fa Ding4, Liu-cheng Ding5, Wei-hua Gan1, Bin Wang2, Ai-qing Zhang3
1 Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China
2 Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
3 Department of Pediatrics, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China
4 Department of Endocrinology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China
5 Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China
Correspondence to: Bin Wang: wangbinhewei@126.com, Ai-qing Zhang: njaiqing@njmu.edu.cn,
DOI: 10.1038/s41401-023-01140-4
Received: 26 March 2023
Accepted: 12 July 2023
Advance online: 18 August 2023

Abstract

Renal tubulointerstitial fibrosis (TIF) is considered as the final convergent pathway of diabetic nephropathy (DN) without effective therapies currently. MiRNAs play a key role in fibrotic diseases and become promising therapeutic targets for kidney diseases, while miRNA clusters, formed by the cluster arrangement of miRNAs on chromosomes, can regulate diverse biological functions alone or synergistically. In this study, we developed clustered miR-23a/27a/26a-loaded skeletal muscle satellite cells–derived exosomes (Exos) engineered with RVG peptide, and investigated their therapeutic efficacy in a murine model of DN. Firstly, we showed that miR-23a-3p, miR-26a-5p and miR-27a-3p were markedly decreased in serum samples of DN patients using miRNA sequencing. Meanwhile, we confirmed that miR-23a-3p, miR-26a-5p and miR-27a-3p were primarily located in proximal renal tubules and highly negatively correlated with TIF in db/db mice at 20 weeks of age. We then engineered RVG-miR-23a/27a/26a cluster loaded Exos derived from muscle satellite cells, which not only enhanced the stability of miR-23a/27a/26a cluster, but also efficiently delivered more miR-23a/27a/26a cluster homing to the injured kidney. More importantly, administration of RVG-miR-23a/27a/26a-Exos (100 μg, i.v., once a week for 8 weeks) significantly ameliorated tubular injury and TIF in db/db mice at 20 weeks of age. We revealed that miR-23a/27a/26a-Exos enhanced antifibrotic effects by repressing miRNA cluster-targeting Lpp simultaneously, as well as miR-27a-3p-targeting Zbtb20 and miR-26a-5p-targeting Klhl42, respectively. Knockdown of Lpp by injection of AAV-Lpp-RNAi effectively ameliorated the progression of TIF in DN mice. Taken together, we established a novel kidney-targeting Exo-based delivery system by manipulating the miRNA-23a/27a/26a cluster to ameliorate TIF in DN, thus providing a promising therapeutic strategy for DN.

Keywords: diabetic nephropathy; tubulointerstitial fibrosis; skeletal muscle satellite cell; exosomes; miR-23a/27a/26a cluster; Lpp; Zbtb20; Klhl42

Article Options

Download Citation

Cited times in Scopus