Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

LncRNA SNHG16 induces proliferation and fibrogenesis via modulating miR-141-3p and CCND1 in diabetic nephropathy

Abstract

LncRNAs are reported to participate in the progression of various diseases including diabetic nephropathy. Currently, we reported that SNHG16 was obviously upregulated in db/db mice and high glucose-treated mice mesangial cells. Then, functional experiments showed that SNHG16 silencing significantly inhibited proliferation of mice mesangial cells, which induced the apoptosis and triggered cell cycle arrest. Meanwhile, proliferation-related biomarkers PCNA and Cyclin D1 (CCND1) were greatly repressed. Furthermore, western blot analysis was conducted to test fibrogenesis-associated genes Fibronectin and α-SMA. Meanwhile, the increased protein expression levels of Fibronectin and α-SMA under high glucose conditions were reversed by loss of SNHG16. miR-141-3p has been reported to be involved in various diseases. Then, RNA immunoprecipitation assay revealed the relation between SNHG16 and miR-141-3p. Downregulation of SNHG16 was able to induce expression of miR-141-3p, which was obviously reduced in db/db diabetic nephropathy mice. In addition, CCND1 is a crucial cell cycle master in human diseases. CCND1 was speculated as the target of miR-141-3p and miR-141-3p inhibited CCND1 expression significantly. Meanwhile, we observed that loss of CCND1 greatly repressed mice mesangial cell proliferation and induced cell apoptosis. Taken these together, we revealed for the first time that SNHG16 induced proliferation and fibrogenesis via modulating miR-141-3p and CCND1 in diabetic nephropathy. SNHG16/miR-141-3p/CCND1 axis can suggest a pathological mechanism of progression of diabetic nephropathy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: SNHG16 was increased in db/db mice.
Fig. 2: SNHG16 was elevated in mesangial cell indicated with HG.
Fig. 3: Effect of SNHG16 on mesangial cell proliferation.
Fig. 4: Effect of SNHG16 on mesangial cells apoptosis and cell cycle.
Fig. 5: Effect of SNHG16 on mesangial cell fibrogenesis.
Fig. 6: miR-141-3p was a direct target of SNHG16.
Fig. 7: CCND1 was a direct target of miR-141-3p.

Similar content being viewed by others

Data availability

All relevant data are within the paper.

References

  1. Tsai YC, Lee CS, Chiu YW, Lee JJ, Lee SC, Hsu YL, et al. Angiopoietin-2, renal deterioration, major adverse cardiovascular events and all-cause mortality in patients with diabetic nephropathy. Kidney Blood Press Res. 2018;43:545–54.

    Article  CAS  Google Scholar 

  2. Oates PJ. Aldose reductase inhibitors and diabetic kidney disease. Curr Opin Investig Drugs. 2010;11:402–17.

    CAS  PubMed  Google Scholar 

  3. Zhu L, Han J, Yuan R, Xue L, Pang W. Berberine ameliorates diabetic nephropathy by inhibiting TLR4/NF-kappaB pathway. Biol Res. 2018;51:9.

    Article  Google Scholar 

  4. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136:629–41.

    Article  CAS  Google Scholar 

  5. Neumann P, Jae N, Knau A, Glaser SF, Fouani Y, Rossbach O, et al. The lncRNA GATA6-AS epigenetically regulates endothelial gene expression via interaction with LOXL2. Nat Commun. 2018;9:237.

    Article  Google Scholar 

  6. Yan X, Zhang D, Wu W, Wu S, Qian J, Hao Y, et al. Mesenchymal stem cells promote hepatocarcinogenesis via lncRNA-MUF interaction with ANXA2 and miR-34a. Cancer Res. 2017;77:6704–16.

    Article  CAS  Google Scholar 

  7. Zhang L, Yang Z, Trottier J, Barbier O, Wang L. Long noncoding RNA MEG3 induces cholestatic liver injury by interaction with PTBP1 to facilitate shp mRNA decay. Hepatology. 2017;65:604–15.

    Article  CAS  Google Scholar 

  8. Roy S, Awasthi A. Emerging roles of noncoding RNAs in T cell differentiation and functions in autoimmune diseases. Int Rev Immunol. 2019;38:232–45.

    Article  CAS  Google Scholar 

  9. Lopez-Urrutia E, Bustamante Montes LP, Ladron de Guevara Cervantes D, Perez-Plasencia C, Campos-Parra AD. Crosstalk between long non-coding RNAs, micro-RNAs and mRNAs: deciphering molecular mechanisms of master regulators in cancer. Front Oncol. 2019;9:669.

    Article  Google Scholar 

  10. Zhao J, Li L, Han ZY, Wang ZX, Qin LX. Long noncoding RNAs, emerging and versatile regulators of tumor-induced angiogenesis. Am J Cancer Res. 2019;9:1367–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ignarski M, Islam R, Muller RU. Long non-coding RNAs in kidney disease. Int J Mol Sci. 2019;20:3276.

    Article  CAS  Google Scholar 

  12. Li Y, Xu K, Xu K, Chen S, Cao Y, Zhan H. Roles of identified long noncoding RNA in diabetic nephropathy. J Diabetes Res. 2019;2019:5383010.

    PubMed  PubMed Central  Google Scholar 

  13. Zhang J, Jiang T, Liang X, Shu S, Xiang X, Zhang W, et al. lncRNA MALAT1 mediated high glucose-induced HK-2 cell epithelial-to-mesenchymal transition and injury. J Physiol Biochem. 2019;75:443–52.

    Article  CAS  Google Scholar 

  14. Liu DW, Zhang JH, Liu FX, Wang XT, Pan SK, Jiang DK, et al. Silencing of long noncoding RNA PVT1 inhibits podocyte damage and apoptosis in diabetic nephropathy by upregulating FOXA1. Exp Mol Med. 2019;51:88.

    Article  Google Scholar 

  15. Zhang Y, Chang B, Zhang J, Wu X. LncRNA SOX2OT alleviates the high glucose-induced podocytes injury through autophagy induction by the miR-9/SIRT1 axis. Exp Mol Pathol. 2019;110:104283.

    Article  CAS  Google Scholar 

  16. Liu H, Sun HL. LncRNA TCF7 triggered endoplasmic reticulum stress through a sponge action with miR-200c in patients with diabetic nephropathy. Eur Rev Med pharmacol Sci. 2019;23:5912–22.

    CAS  PubMed  Google Scholar 

  17. Xie X, Xu X, Sun C, Yu Z. Long intergenic noncoding RNA SNHG16 interacts with miR-195 to promote proliferation, invasion and tumorigenesis in hepatocellular carcinoma. Exp Cell Res. 2019;383:111501.

    Article  CAS  Google Scholar 

  18. Xu C, Hu C, Wang Y, Liu S. Long noncoding RNA SNHG16 promotes human retinoblastoma progression via sponging miR-140-5p. Biomed Pharmacother. 2019;117:109153.

    Article  CAS  Google Scholar 

  19. Zhou XY, Liu H, Ding ZB, Xi HP, Wang GW. lncRNA SNHG16 promotes glioma tumorigenicity through miR-373/EGFR axis by activating PI3K/AKT pathway. Genomics. 2020;112:1021–9.

    Article  CAS  Google Scholar 

  20. Leti F, Morrison E, DiStefano JK. Long noncoding RNAs in the pathogenesis of diabetic kidney disease: implications for novel therapeutic strategies. Personal Med. 2017;14:271–8.

    Article  CAS  Google Scholar 

  21. Alvarez ML, Distefano JK. The role of non-coding RNAs in diabetic nephropathy: potential applications as biomarkers for disease development and progression. Diabetes Res Clin Pract. 2013;99:1–11.

    Article  CAS  Google Scholar 

  22. Christensen LL, True K, Hamilton MP, Nielsen MM, Damas ND, Damgaard CK, et al. SNHG16 is regulated by the Wnt pathway in colorectal cancer and affects genes involved in lipid metabolism. Mol Oncol. 2016;10:1266–82.

    Article  CAS  Google Scholar 

  23. Cai C, Huo Q, Wang X, Chen B, Yang Q. SNHG16 contributes to breast cancer cell migration by competitively binding miR-98 with E2F5. Biochem Biophys Res Commun. 2017;485:272–8.

    Article  CAS  Google Scholar 

  24. Yu Y, Chen F, Yang Y, Jin Y, Shi J, Han S, et al. lncRNA SNHG16 is associated with proliferation and poor prognosis of pediatric neuroblastoma. Int J Oncol. 2019;55:93–102.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Guo G, Kang Q, Zhu X, Chen Q, Wang X, Chen Y, et al. A long noncoding RNA critically regulates Bcr-Abl-mediated cellular transformation by acting as a competitive endogenous RNA. Oncogene. 2015;34:1768–79.

    Article  CAS  Google Scholar 

  26. Liang Z, Li X, Liu S, Li C, Wang X, Xing J. MiR-141-3p inhibits cell proliferation, migration and invasion by targeting TRAF5 in colorectal cancer. Biochem Biophys Res Commun. 2019;514:699–705.

    Article  CAS  Google Scholar 

  27. Li JH, Zhang Z, Du MZ, Guan YC, Yao JN, Yu HY, et al. microRNA-141-3p fosters the growth, invasion, and tumorigenesis of cervical cancer cells by targeting FOXA2. Arch Biochem Biophys. 2018;657:23–30.

    Article  CAS  Google Scholar 

  28. Zhang MN, Tang QY, Li RM, Song MG. MicroRNA-141-3p/200a-3p target and may be involved in post-transcriptional repression of RNA decapping enzyme Dcp2 during renal development. Biosci Biotechnol Biochem. 2018;82:1724–32.

    Article  CAS  Google Scholar 

  29. Wang W, Medeiros LJ. Utility of cyclin D1 in the diagnostic workup of hematopoietic neoplasms: what can cyclin D1 do for us? Ad Anatomic Pathol. 2019;26:281–91.

    Article  CAS  Google Scholar 

  30. Ramos-Garcia P, Gil-Montoya JA, Scully C, Ayen A, Gonzalez-Ruiz L, Navarro-Trivino FJ, et al. An update on the implications of cyclin D1 in oral carcinogenesis. Oral Dis. 2017;23:897–912.

    Article  CAS  Google Scholar 

  31. Neumeister P, Pixley FJ, Xiong Y, Xie H, Wu K, Ashton A, et al. Cyclin D1 governs adhesion and motility of macrophages. Mol Biol Cell. 2003;14:2005–15.

    Article  CAS  Google Scholar 

  32. Zhong Z, Yeow WS, Zou C, Wassell R, Wang C, Pestell RG, et al. Cyclin D1/cyclin-dependent kinase 4 interacts with filamin A and affects the migration and invasion potential of breast cancer cells. Cancer Res. 2010;70:2105–14.

    Article  CAS  Google Scholar 

  33. Zhou Q, Zhang W, Wang Z, Liu S. Long non-coding RNA PTTG3P functions as an oncogene by sponging miR-383 and up-regulating CCND1 and PARP2 in hepatocellular carcinoma. BMC Cancer. 2019;19:731.

    Article  Google Scholar 

  34. Wang M, Yu W, Gao J, Ma W, Frentsch M, Thiel A, et al. MicroRNA-487a-3p functions as a new tumor suppressor in prostate cancer by targeting CCND1. J Cell Physiol. 2020;235:1588–600.

    Article  CAS  Google Scholar 

Download references

Funding

This work was supported by the Key Program of Science and Technology Committee of Changning District, Shanghai, China, [Grant No. WKW2016 Z03].

Author information

Authors and Affiliations

Authors

Contributions

SH, XJ, PL, and QR designed the study and the experiments. XJ, PL, and QR performed the experiments. XG and KS collected the data. LX, BX, and QW did the analysis. XJ drafted the manuscript. SH revised the manuscript. All of the authors finally approved the proof for submission.

Corresponding author

Correspondence to Shan Huang.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jiang, X., Ru, Q., Li, P. et al. LncRNA SNHG16 induces proliferation and fibrogenesis via modulating miR-141-3p and CCND1 in diabetic nephropathy. Gene Ther 27, 557–566 (2020). https://doi.org/10.1038/s41434-020-0160-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41434-020-0160-x

This article is cited by

Search

Quick links