Article

The lncRNA Malat1 functions as a ceRNA to contribute to berberine-mediated inhibition of HMGB1 by sponging miR-181c-5p in poststroke inflammation

Ding-wen Cao1,2, Man-man Liu1,2, Rui Duan2, Yi-fu Tao1, Jun-shan Zhou3, Wei-rong Fang2, Jun-rong Zhu1,2, Li Niu4, Jian-guo Sun5
1 Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
2 School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
3 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
4 Jinling Hospital Department of Medicine, Nanjing University, School of Medicine, Nanjing 210002, China
5 Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
Correspondence to: Jun-rong Zhu: junrong_zhu@aliyun.com, Li Niu: jsnjnl@sina.com, Jian-guo Sun: jgsun@cpu.edu.cn,
DOI: 10.1038/s41401-019-0284-y
Received: 11 April 2019
Accepted: 30 June 2019
Advance online: 20 August 2019

Abstract

Long non-coding RNAs (lncRNAs) have been identified as essential mediators in neurological dysfunction. Our previous study shows that berberine (BBR) hampers the nuclear-to-cytosolic translocation of high-mobility group box 1 (HMGB1) in the process of poststroke inflammation. In this study, we explored the role of lncRNA metastasis‐associated lung adenocarcinoma transcript 1 (Malat1) in the process of BBR-induced inhibition of HMGB1 in ischemic brain. Before the 60-min MCAO surgery, the mice were pretreated with BBR (50 mg· kg−1 per day, ig) for 14 days or ICV injected with specific lentiviral vector or shRNA. We showed that MCAO caused marked increase in the expression Malat1 and HMGB1 in the ipsilateral cortex, which was significantly attenuated by pretreatment with BBR. Knockdown of Malat1 attenuated the inflammatory injury after brain ischemia, whereas overexpression of Malat1 exacerbated ischemic brain inflammation. Overexpression of Malat1 also reversed BBR-induced reduction of HMGB1 and proinflammatory cytokines. The above results suggested a potential correlation between Malat1 and stroke inflammation. Based on informatics analysis we predicted that HMGB1 was a direct downstream target of miR-181c-5p, whereas Malat1 acted as a competitive endogenous RNA (ceRNA) for miR-181c-5p targeted the 3′-UTR of HMGB1 to promote inflammation after ischemic stroke. Knockdown of Malat1 significantly decreased HMGB1 level, which could be abrogated by transfection with miR-181c-5p inhibitors. Taken together, our results demonstrate for the first time that Malat1/miR-181c-5p/HMGB1 axis may be a key pathway of BBR-induced antiinflammation effects in stroke, and they may provide a novel avenue for targeted therapy.
Keywords: Malat1; miR-181c-5p; HMGB1; stroke; inflammation; berberine

Article Options

Download Citation

Cited times in Scopus