Endocrinol Metab.  2022 Dec;37(6):901-917. 10.3803/EnM.2022.1565.

Inhibition of miR-146a-5p and miR-8114 in Insulin-Secreting Cells Contributes to the Protection of Melatonin against Stearic Acid-Induced Cellular Senescence by Targeting Mafa

Affiliations
  • 1Department of Nutrition and Food Hygiene (National Key Discipline), Public Health College, Harbin Medical University, Harbin, China
  • 2Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
  • 3Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland

Abstract

Background
Chronic exposure to elevated levels of saturated fatty acids results in pancreatic β-cell senescence. However, targets and effective agents for preventing stearic acid-induced β-cell senescence are still lacking. Although melatonin administration can protect β-cells against lipotoxicity through anti-senescence processes, the precise underlying mechanisms still need to be explored. Therefore, we investigated the anti-senescence effect of melatonin on stearic acid-treated mouse β-cells and elucidated the possible role of microRNAs in this process.
Methods
β-Cell senescence was identified by measuring the expression of senescence-related genes and senescence-associated β-galactosidase staining. Gain- and loss-of-function approaches were used to investigate the involvement of microRNAs in stearic acid-evoked β-cell senescence and dysfunction. Bioinformatics analyses and luciferase reporter activity assays were applied to predict the direct targets of microRNAs.
Results
Long-term exposure to a high concentration of stearic acid-induced senescence and upregulated miR-146a-5p and miR- 8114 expression in both mouse islets and β-TC6 cell lines. Melatonin effectively suppressed this process and reduced the levels of these two miRNAs. A remarkable reversibility of stearic acid-induced β-cell senescence and dysfunction was observed after silencing miR-146a-5p and miR-8114. Moreover, V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (Mafa) was verified as a direct target of miR-146a-5p and miR-8114. Melatonin also significantly ameliorated senescence and dysfunction in miR-146a-5pand miR-8114-transfected β-cells.
Conclusion
These data demonstrate that melatonin protects against stearic acid-induced β-cell senescence by inhibiting miR-146a- 5p and miR-8114 and upregulating Mafa expression. This not only provides novel targets for preventing stearic acid-induced β-cell dysfunction, but also points to melatonin as a promising drug to combat type 2 diabetes progression.

Keyword

Stearic acid; Melatonin; MicroRNAs; Cellular senescence; Mafa

Figure

  • Fig. 1. Long-term high-stearic-acid diet (HSD) feeding induces senescence in mouse islets. Changes in the expressions of senescence-related genes in HSD-fed mice islets (n=5 mice per group). aP<0.05, bP<0.01, compared with the control (Ctrl) group.

  • Fig. 2. Melatonin (Mel) protects mouse β-TC6 cells against stearic acid-induced senescence and dysfunction. (A) Cytotoxic effect on β-TC6 cells of treatment with different concentrations of melatonin for 24 hours, measured by cell counting kit 8 (CCK-8) assays. (B) Effect of Mel (0.1 mmol/L) on cell viability at different times. (C, D) Protective effect of Mel on stearic acid-decreased glucose-stimulated insulin secretion assay (GSIS) and cell viability. (E) Changes in mRNA levels of senescence-related genes after incubation with Mel in the presence and absence of stearic acid. Each experiment was independently repeated three times. (F) Effect of Mel on stearic acid-induced β-cell senescence measured by X-Gal staining (scale bar, 100 μm). Glu, glucose. aP<0.01, compared with 0 mmol/L; bP<0.01, compared with 0 hour; cP<0.05, dP<0.01, eP<0.001, compared with the control (Ctrl) group; fP<0.05, gP<0.01, hP<0.001, compared with the stearic acid (SA) group; iP<0.05; jP<0.01.

  • Fig. 3. Melatonin (Mel) significantly reversed the stearic acid (SA)-induced increase in the expression of miR-146a-5p and miR-8114 in β-TC6 cells and mouse islet cells. (A, B) Changes in levels of miR-146a-5p and miR-8114 in mouse islets fed with high-stearic-acid diet (HSD) (n=5 mice per group). (C, D) Expression of miR-146a-5p and miR-8114 in SA-treated β-TC6 cells in the presence or absence of Mel. Each experiment was repeated independently three (C) or four (D) times. (E, F) The miR-146a-5p and miR-8114 level in islet cells exposed to SA followed by Mel treatment. Each experiment was repeated independently five times.aP<0.05, bP<0.001, compared with the control (Ctrl) group; cP<0.05, dP<0.01, compared with the SA group; eP<0.01, compared with the Ctrl group.

  • Fig. 4. Inhibition of miR-146a-5p alleviated stearic acid-induced cell senescence and glucose-stimulated insulin secretion assay (GSIS) in β-TC6 cells. (A) Change in miR-146a-5p expression after transfection of stearic acid-treated β-TC6 cells with anti-miRNA oligonucleotides (AMO)-146a-5p. (B, C) Effect of AMO-146a-5p on cell viability and GSIS in the presence and absence of stearic acid, respectively. (D) Alterations in senescence-related gene expression in stearic acid-treated β-TC6 cells transfected with AMO-146a-5p. (E) Inhibition of miR- 146a-5p reversed stearic acid-reduced β-gal activity. Each experiment was repeated independently three times (scale bar, 100 μm). Glu, glucose. aP<0.05, bP<0.01, compared with control (Ctrl)+AMO-negative control (NC) group; cP<0.05, dP<0.01, eP<0.001, compared with the stearic acid (SA)+AMO-NC group; fP<0.05; gP<0.01.

  • Fig. 5. Silencing miR-8114 improved stearic acid (SA)-induced cell senescence and glucose-stimulated insulin secretion assay (GSIS) in β-TC6 cells. (A) Change in miR-8114 level after knockdown of miR-8114 in the presence and absence of SA. (B, C) Silencing miR-8114 improved SA-impaired cell viability and GSIS, respectively. (D) Effect of anti-miRNA oligonucleotides (AMO)-8114 on senescence-related gene expression in SA-treated β-TC6 cells. Each experiment was repeated independently three times. (E) Transfection of SA-treated β-TC6 cells with AMO-8114 reversed the decrease in β-gal activity (scale bar, 100 μm). aP<0.05, bP<0.01, cP<0.001, compared with control (Ctrl)+AMO-negative control (NC) group; dP<0.05, eP<0.01, fP<0.001, compared with SA+AMO-NC group; gP<0.05.

  • Fig. 6. V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (Mafa) is a direct target of miR-146a-5p and miR-8114. (A) Effect of melatonin on Mafa protein levels in stearic acid (SA)-treated β-TC6 cells. (B, C) Predicted binding sites between miR-146a/miR-8114 and Mafa, respectively. (D, E) Changes in levels of Mafa mRNA and Mafa protein after transfection of anti-miRNA oligonucleotides (AMO)- 146a-5p in SA-treated β-TC6 cells, respectively. (F, G) Mafa mRNA and Mafa protein levels after inhibition of miR-8114 in the presence of SA, respectively. (H, I) Overexpression of miR-146a-5p decreased Mafa mRNA and Mafa protein levels in the absence of SA, respectively. (J, K) Transfection of miR-8114 mimic alone inhibited the expression of Mafa mRNA and Mafa protein, respectively. (L) Luciferase activity between Mafa and miR-146a-5p or miR-8114, respectively. Each experiment was repeated independently three (A-K) or four (L) times. UTR, untranslated region; WT, wild-type; MUT, mutant; NS, no significance. aP<0.05, bP<0.01, compared with control (Ctrl)/Ctrl+AMO-negative control (NC)/mimic-NC group; cP<0.05, dP<0.01, compared with SA/SA+AMO-NC/miR-146a-5p mimic (mimic-146a-5p)/8114 group.

  • Fig. 7. Melatonin (Mel) rescued miR-146a-5p mimic-induced senescence and dysfunction in β-TC6 cells. (A) Change in miR-146a-5p expression after exposure to Mel with a miR-146a-5p mimic. (B, C, D, E, F) Effect of Mel on cell viability, glucose-stimulated insulin secretion assay (GSIS), senescence gene expression, and V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (Mafa) protein levels after overexpression of miR-146a-5p (D: scale bar, 100 μm). Each experiment was repeated independently three times. Glu, glucose. aP<0.05, bP<0.01, cP<0.001, compared with the control (Ctrl)+mimic-negative control (NC) group; dP<0.05, eP<0.01, fP<0.001, compared with the miR-146a-5p mimic (mimic-146a-5p) group; gP<0.05.

  • Fig. 8. Melatonin (Mel) blocked miR-8114 overexpression-induced β-cell senescence and dysfunction in β-TC6 cells. (A) Change in miR- 8114 expression after incubation with Mel after miR-8114 mimic transfection. (B, C, D, E, F) Effects of Mel on cell viability, glucose-stimulated insulin secretion assay (GSIS), senescence gene expression and V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (Mafa) protein levels after overexpression of miR-8114 (D: scale bar, 100 μm). Each experiment was repeated independently three times. aP<0.05, bP<0.01, cP<0.001, compared with the control (Ctrl)+mimic-negative control (NC) group; dP<0.05, eP<0.01, fP<0.001, compared with the miR-8114 mimic (mimic-8114) group; gP<0.05.

  • Fig. 9. Schematic diagram of the molecular mechanisms underlying the protective effect of melatonin (Mel) on stearic acid (SA)-induced β-cell senescence through miR-146a-5p and miR-8114. ORF, Open Reading Frame; Mafa, V-maf musculoaponeurotic fibrosarcoma oncogene homolog A.


Reference

1. Arvan P, Bernal-Mizrachi E, Liu M, Pietropaolo M, Satin L, Schnell S, et al. Molecular aspects of pancreatic beta cell failure and diabetes. Mol Aspects Med. 2015; 42:1–2.
Article
2. Forouhi NG, Koulman A, Sharp SJ, Imamura F, Kroger J, Schulze MB, et al. Differences in the prospective association between individual plasma phospholipid saturated fatty acids and incident type 2 diabetes: the EPIC-InterAct case-cohort study. Lancet Diabetes Endocrinol. 2014; 2:810–8.
Article
3. Lu H, Hao L, Li S, Lin S, Lv L, Chen Y, et al. Elevated circulating stearic acid leads to a major lipotoxic effect on mouse pancreatic beta cells in hyperlipidaemia via a miR-34a-5p-mediated PERK/p53-dependent pathway. Diabetologia. 2016; 59:1247–57.
Article
4. Chu X, Liu L, Na L, Lu H, Li S, Li Y, et al. Sterol regulatory element-binding protein-1c mediates increase of postprandial stearic acid, a potential target for improving insulin resistance, in hyperlipidemia. Diabetes. 2013; 62:561–71.
Article
5. Liu L, Li Y, Guan C, Li K, Wang C, Feng R, et al. Free fatty acid metabolic profile and biomarkers of isolated post-challenge diabetes and type 2 diabetes mellitus based on GCMS and multivariate statistical analysis. J Chromatogr B Analyt Technol Biomed Life Sci. 2010; 878:2817–25.
Article
6. Remedi MS, Emfinger C. Pancreatic β-cell identity in diabetes. Diabetes Obes Metab. 2016; 18(Suppl 1):110–6.
Article
7. Brereton MF, Rohm M, Ashcroft FM. β-Cell dysfunction in diabetes: a crisis of identity? Diabetes Obes Metab. 2016; 18(Suppl 1):102–9.
8. Weir GC, Bonner-Weir S. Five stages of evolving beta-cell dysfunction during progression to diabetes. Diabetes. 2004; 53 Suppl 3:S16–21.
Article
9. Marchetti P, Bugliani M, Lupi R, Marselli L, Masini M, Boggi U, et al. The endoplasmic reticulum in pancreatic beta cells of type 2 diabetes patients. Diabetologia. 2007; 50:2486–94.
Article
10. Kitamura T. The role of FOXO1 in β-cell failure and type 2 diabetes mellitus. Nat Rev Endocrinol. 2013; 9:615–23.
Article
11. Eguchi K, Manabe I, Oishi-Tanaka Y, Ohsugi M, Kono N, Ogata F, et al. Saturated fatty acid and TLR signaling link β cell dysfunction and islet inflammation. Cell Metab. 2012; 15:518–33.
Article
12. Aguayo-Mazzucato C. Functional changes in beta cells during ageing and senescence. Diabetologia. 2020; 63:2022–9.
Article
13. Aguayo-Mazzucato C, Andle J, Lee TB Jr, Midha A, Talemal L, Chipashvili V, et al. Acceleration of β cell aging determines diabetes and senolysis improves disease outcomes. Cell Metab. 2019; 30:129–42.
Article
14. Zisapel N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharmacol. 2018; 175:3190–9.
Article
15. Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M, Qin L. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016; 61:253–78.
Article
16. Tamura H, Kawamoto M, Sato S, Tamura I, Maekawa R, Taketani T, et al. Long-term melatonin treatment delays ovarian aging. J Pineal Res. 2017; 62:e12381.
Article
17. Carbajo-Pescador S, Ordonez R, Benet M, Jover R, GarciaPalomo A, Mauriz JL, et al. Inhibition of VEGF expression through blockade of Hif1α and STAT3 signalling mediates the anti-angiogenic effect of melatonin in HepG2 liver cancer cells. Br J Cancer. 2013; 109:83–91.
Article
18. Xia Y, Chen S, Zeng S, Zhao Y, Zhu C, Deng B, et al. Melatonin in macrophage biology: current understanding and future perspectives. J Pineal Res. 2019; 66:e12547.
Article
19. Garaulet M, Qian J, Florez JC, Arendt J, Saxena R, Scheer FA. Melatonin effects on glucose metabolism: time to unlock the controversy. Trends Endocrinol Metab. 2020; 31:192–204.
Article
20. Li X, Zhang M, Tang W. Effects of melatonin on streptozotocin-induced retina neuronal apoptosis in high blood glucose rat. Neurochem Res. 2013; 38:669–76.
Article
21. Costes S, Boss M, Thomas AP, Matveyenko AV. Activation of melatonin signaling promotes β-cell survival and function. Mol Endocrinol. 2015; 29:682–92.
Article
22. Lee YH, Jung HS, Kwon MJ, Jang JE, Kim TN, Lee SH, et al. Melatonin protects INS-1 pancreatic β-cells from apoptosis and senescence induced by glucotoxicity and glucolipotoxicity. Islets. 2020; 12:87–98.
Article
23. He H, Dong W, Huang F. Anti-amyloidogenic and antiapoptotic role of melatonin in Alzheimer disease. Curr Neuropharmacol. 2010; 8:211–7.
Article
24. Park JH, Shim HM, Na AY, Bae KC, Bae JH, Im SS, et al. Melatonin prevents pancreatic β-cell loss due to glucotoxicity: the relationship between oxidative stress and endoplasmic reticulum stress. J Pineal Res. 2014; 56:143–53.
25. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011; 12:861–74.
Article
26. Flynt AS, Lai EC. Biological principles of microRNA-mediated regulation: shared themes amid diversity. Nat Rev Genet. 2008; 9:831–42.
Article
27. Martinez-Sanchez A, Rutter GA, Latreille M. MiRNAs in β-cell development, identity, and disease. Front Genet. 2017; 7:226.
Article
28. Yu Y, Guo R, Zhang Y, Shi H, Sun H, Chu X, et al. miRNAmRNA profile and regulatory network in stearic acid-treated β-cell dysfunction. J Endocrinol. 2020; 246:13–27.
Article
29. Goldstein JL, Basu SK, Brown MS. Receptor-mediated endocytosis of low-density lipoprotein in cultured cells. Methods Enzymol. 1983; 98:241–60.
30. Kim HS, Han TY, Yoo YM. Melatonin-mediated intracellular insulin during 2-deoxy-d-glucose treatment is reduced through autophagy and EDC3 protein in insulinoma INS-1E cells. Oxid Med Cell Longev. 2016; 2016:2594703.
Article
31. Guo F, Huang C, Liao X, Wang Y, He Y, Feng R, et al. Beneficial effects of mangiferin on hyperlipidemia in high-fatfed hamsters. Mol Nutr Food Res. 2011; 55:1809–18.
Article
32. Guo R, Yu Y, Zhang Y, Li Y, Chu X, Lu H, et al. Overexpression of miR-297b-5p protects against stearic acid-induced pancreatic β-cell apoptosis by targeting LATS2. Am J Physiol Endocrinol Metab. 2020; 318:E430–9.
Article
33. Zhu MJ, Liu BY, Shi L, Wang X, Wang Y. mTOR-autophagy promotes pulmonary senescence through IMP1 in chronic toxicity of methamphetamine. J Cell Mol Med. 2020; 24:12082–93.
Article
34. Lee BY, Han JA, Im JS, Morrone A, Johung K, Goodwin EC, et al. Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell. 2006; 5:187–95.
35. Shi H, Bressan R. RNA extraction. Methods Mol Biol. 2006; 323:345–8.
Article
36. Fleischer N, Chen C, Surana M, Leiser M, Rossetti L, Pralong W, et al. Functional analysis of a conditionally transformed pancreatic beta-cell line. Diabetes. 1998; 47:1419–25.
Article
37. Knaack D, Fiore DM, Surana M, Leiser M, Laurance M, Fusco-DeMane D, et al. Clonal insulinoma cell line that stably maintains correct glucose responsiveness. Diabetes. 1994; 43:1413–7.
Article
38. Fan X, Gu S, Lei J, Gu S, Yang L. Controlled release of insulin based on temperature and glucose dual responsive biomicrocapsules. Molecules. 2022; 27:1686.
Article
39. Mziaut H, Henniger G, Ganss K, Hempel S, Wolk S, McChord J, et al. MiR-132 controls pancreatic beta cell proliferation and survival through Pten/Akt/Foxo3 signaling. Mol Metab. 2020; 31:150–62.
Article
40. Giuliani A, Prattichizzo F, Micolucci L, Ceriello A, Procopio AD, Rippo MR. Mitochondrial (Dys) function in inflammaging: do MitomiRs influence the energetic, oxidative, and inflammatory status of senescent cells? Mediators Inflamm. 2017; 2017:2309034.
Article
41. Iacona JR, Lutz CS. miR-146a-5p: expression, regulation, and functions in cancer. Wiley Interdiscip Rev RNA. 2019; 10:e1533.
Article
42. Olivieri F, Prattichizzo F, Giuliani A, Matacchione G, Rippo MR, Sabbatinelli J, et al. miR-21 and miR-146a: the microRNAs of inflammaging and age-related diseases. Ageing Res Rev. 2021; 70:101374.
Article
43. Lo WY, Wang SJ, Wang HJ. Non-canonical interaction between O-linked N-acetylglucosamine transferase and miR146a-5p aggravates high glucose-induced endothelial inflammation. Front Physiol. 2020; 11:1091.
Article
44. Lovis P, Roggli E, Laybutt DR, Gattesco S, Yang JY, Widmann C, et al. Alterations in microRNA expression contribute to fatty acid-induced pancreatic beta-cell dysfunction. Diabetes. 2008; 57:2728–36.
45. Roggli E, Britan A, Gattesco S, Lin-Marq N, Abderrahmani A, Meda P, et al. Involvement of microRNAs in the cytotoxic effects exerted by proinflammatory cytokines on pancreatic beta-cells. Diabetes. 2010; 59:978–86.
46. Ebrahimi AG, Hollister-Lock J, Sullivan BA, Tsuchida R, Bonner-Weir S, Weir GC. Beta cell identity changes with mild hyperglycemia: implications for function, growth, and vulnerability. Mol Metab. 2020; 35:100959.
Article
47. Hang Y, Stein R. MafA and MafB activity in pancreatic β cells. Trends Endocrinol Metab. 2011; 22:364–73.
Article
48. Palmer AK, Tchkonia T, LeBrasseur NK, Chini EN, Xu M, Kirkland JL. Cellular senescence in type 2 diabetes: a therapeutic opportunity. Diabetes. 2015; 64:2289–98.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr