SGLT2 promotes cardiac fibrosis following myocardial infarction and is regulated by miR-141

Gang Li, Congchun Zhao, Shanhua Fang, Gang Li, Congchun Zhao, Shanhua Fang

Abstract

Cardiac fibrosis is a primary event during myocardial infarction (MI) progression, which impairs cardiac function. The present study aimed to investigate the effect of SGLT2 on cardiac fibrosis following MI. To validate the role of SGLT2 in the regulation of cardiac fibrosis in vivo, an MI rat model was established. Echocardiography was performed to determine cardiac function at 4 weeks post-MI. MI model rats were transfected with short hairpin RNA (sh)-SGLT2 or sh-negative control lentiviruses to investigate the effect of SGLT2 on rat heart function post-MI. Subsequently, the effects of SGLT2 on the cardiac fibrosis of infarcted hearts were assessed by performing Masson's trichrome staining. To further clarify the effect of SGLT2 on cardiac fibroblast proliferation, TGFβ was used to stimulate primary cardiac fibroblasts in vitro. The results demonstrated that SGLT2 served a key role in cardiac fibrosis. SGLT2 expression levels in infarct tissues were significantly increased at week 1 post-MI compared with the sham group. Compared with the control group, SGLT2 knockdown attenuated cardiac fibrosis by inhibiting the expression of collagen I and collagen III in cardiac fibroblasts in vitro and in vivo. Furthermore, the results indicated that SGLT2 expression was modulated by miR-141 in cardiac fibroblasts. In summary, the present study indicated that upregulated SGLT2 expression in cardiac fibrosis following MI was regulated by miR-141 and SGLT2 that knockdown reduced cardiac fibrosis and improved cardiac function after MI.

Keywords: cardiac fibrosis; microRNA-141; myocardial infarction; sodium-glucose linked transporter 1.

Conflict of interest statement

The authors declare that they have no competing interests.

Copyright: © Li et al.

Figures

Figure 1
Figure 1
SGLT2 expression was analyzed in the infarcted myocardial tissues of MI model rats. SGLT2 (A) mRNA and (B) protein expression levels in infarcted areas of MI model rats at indicated times (n=5). SGLT2 mRNA expression levels in the (C) border and (D) far zones (n=5). **P<0.01, ***P<0.001 vs. sham. SGLT2, sodium-glucose linked transporter 2; MI, myocardial infarction; W, week; NS, not significant.
Figure 2
Figure 2
SGLT2 knockdown improves rat heart function following MI in vivo. SGLT2 (A) mRNA and (B) protein expression levels in infarcted areas treated with sh-SGLT2 or sh-NC (n=5). (C) Echocardiography results of rats in the different groups (n=5). At 4 weeks post-MI, ventricular parameters were measured and analyzed by echocardiography, including (D) LVEF, (E) LVEDV, (F) LVESV, (G) (-) LVdp/dtmax, (H) (+) LVdp/dtmax, (I) HR and (J) E/A ratio (n=5). *P<0.05, **P<0.01, ***P<0.001. SGLT2, sodium-glucose linked transporter 2; MI, myocardial infarction; sh, short hairpin RNA; NC, negative control; LVEF, left ventricular ejection fractions; LVEDV, left ventricular end diastolic volume; LVESV, left ventricular end systolic volume; (-)/(+) LVdp/dtmax, the maximum left ventricular change in pressure/time; HR, heart rate; E/A ratio, ratio of the peak early transmitral flow velocity to peak late transmitral flow velocity.
Figure 3
Figure 3
SGLT2 knockdown inhibits cardiac fibrosis following MI in vivo. (A) Representative photomicrographs of Masson trichrome staining of hearts of MI model rats treated with PBS, sh-NC or sh-SGLT2 (n=5, original magnification x12.5). (B) Fibrotic area was determined as the ratio of the average circumference of fibrotic scars (blue)/average inner circumference of the left ventricle (n=5). Collagen I and collagen III (C) mRNA and (D) protein expression levels in cardiac tissues (n=5). *P<0.05, **P<0.01, ***P<0.001. SGLT2, sodium-glucose linked transporter 2; MI, myocardial infarction; sh, short hairpin RNA; NC, negative control.
Figure 4
Figure 4
SGLT2 knockdown inhibits TGFβ-induced proliferation and collagen synthesis in cardiac fibroblasts. Primary cardiac fibroblasts were transfected with sh-NC or sh-SGLT2 and then treated with 20 ng/ml recombinant TGFβ for 72 h. (A) SGLT2 expression levels. (B) The MTT assay was performed to examine cell proliferation. (C) Protein and (D) mRNA expression levels and of collagen I and collagen III in primary cardiac fibroblasts. *P<0.05, **P<0.01, ***P<0.001. SGLT2, sodium-glucose linked transporter 2; TGFβ, transforming growth factor β; sh, short hairpin RNA; NC, negative control.
Figure 5
Figure 5
SGLT2 is targeted by miR-141. (A) The binding site between the 3'-UTR of SGLT2 mRNA and miR-141. SGLT2 3'-UTR-mutants were generated in which 6 complementary binding site nucleotides were altered. (B) miR-141 expression levels in the infarcted areas of MI model rats at indicated times. (C) Relative luciferase activities were determined by conducting a luciferase reporter assay. (D) Transfection efficiency of miR-141 mimics. SGLT2 (E) protein and (F) mRNA expression levels in primary cardiac fibroblasts transfected with miR-141 mimics. *P<0.05, **P<0.01, ***P<0.001. SGLT2, sodium-glucose linked transporter 2; miR, microRNA; UTR, untranslated region; miR, microRNA; W, week; WT, wild-type; MUT, mutant; NC, negative control.
Figure 6
Figure 6
TGFβ-induced proliferation and collagen synthesis in cardiac fibrosis is regulated by the miR-141/SGLT2 axis. Primary cardiac fibroblasts were transfected with miR-141 NC, miR-141 mimics or miR-141 mimics + pcDNA3.1-SGLT2 and treated with 20 ng/ml recombinant TGFβ for 72 h. (A) Transfection efficiency of pcDNA3.1-SGLT2. (B) The MTT assay was performed to assess cell proliferation. Collagen I and collagen III (C) mRNA and protein expression levels in primary cardiac fibroblasts. *P<0.05. TGFβ, transforming growth factor β; miR, microRNA; SGLT2, sodium-glucose linked transporter 2; NC, negative control; NS, not significant.

References

    1. Mcmanus DD, Gore J, Yarzebski J, Spencer F, Lessard D, Goldberg RJ. Recent trends in the incidence, treatment, and outcomes of patients with STEMI and NSTEMI. Am J Med. 2011;124:40–47. doi: 10.1016/j.amjmed.2010.07.023.
    1. Pontecorboli G, Ventura RMFI, Carlosena A, Benito EM, Pratgonzales S, Padeletti L, Mont L. Use of delayed-enhancement magnetic resonance imaging for fibrosis detection in the atria: A review. Europace. 2017;19:180–189. doi: 10.1093/europace/euw053.
    1. Nagalingam RS, Safi HA, Czubryt MP. Gaining myocytes or losing fibroblasts: Challenges in cardiac fibroblast reprogramming for infarct repair. J Mol Cell Cardiol. 2016;93:108–114. doi: 10.1016/j.yjmcc.2015.11.029.
    1. Tahrani AA, Barnett AH, Bailey CJ. SGLT inhibitors in management of diabetes. Lancet Diabetes Endocrinol. 2013;1:140–151. doi: 10.1016/S2213-8587(13)70050-0.
    1. Wells RG, Mohandas TK, Hediger MA. Localization of the Na+/glucose cotransporter gene SGLT2 to human chromosome 16 close to the centromere. Genomics. 1993;17:787–789. doi: 10.1006/geno.1993.1411.
    1. Kashiwagi Y, Nagoshi T, Yoshino T, Tanaka TD, Ito K, Harada T, Takahashi H, Ikegami M, Anzawa R, Yoshimura M. Expression of SGLT1 in human hearts and impairment of cardiac glucose uptake by phlorizin during ischemia-reperfusion injury in mice. PLoS One. 2015;10(e0130605) doi: 10.1371/journal.pone.0130605.
    1. Koepsell H. The Na(+)-D-glucose cotransporters SGLT1 and SGLT2 are targets for the treatment of diabetes and cancer. Pharmacol Ther. 2017;170:148–165. doi: 10.1016/j.pharmthera.2016.10.017.
    1. Furtado RHM, Bonaca MP, Raz I, Zelniker TA, Mosenzon O, Cahn A, Kuder J, Murphy SA, Bhatt DL, Leiter LA, et al. Dapagliflozin and cardiovascular outcomes in patients with type 2 diabetes mellitus and previous myocardial infarction. Circulation. 2019;139:2516–2527. doi: 10.1161/CIRCULATIONAHA.119.039996.
    1. Reid J, Rana K, Niman S, Sheikh-Ali M, Lewis T, Choksi RR, Goldfaden RF. Sodium-glucose cotransporter-2 (SGLT-2) inhibitors for cardiovascular disease prevention. Am J Cardiovasc Drugs. 2020;20:419–429. doi: 10.1007/s40256-020-00397-y.
    1. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128. doi: 10.1056/NEJMoa1504720.
    1. Habibi J, Aroor AR, Sowers JR, Jia G, Hayden MR, Garro M, Barron BJ, Mayoux E, Rector RS, Whaley-Connell A, DeMarco VG. Sodium glucose transporter 2 (SGLT2) inhibition with empagliflozin improves cardiac diastolic function in a female rodent model of diabetes. Cardiovasc Diabetol. 2017;16(9) doi: 10.1186/s12933-016-0489-z.
    1. Ye Y, Bajaj M, Yang HC, Perez-Polo JR, Birnbaum Y. SGLT-2 inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with type 2 diabetes. Further augmentation of the effects with saxagliptin, a DPP4 inhibitor. Cardiovasc Drugs Ther. 2017;31:119–132. doi: 10.1007/s10557-017-6725-2.
    1. Piccoli MT, Bar C, Thum T. Non-coding RNAs as modulators of the cardiac fibroblast phenotype. J Mol Cell Cardiol. 2016;92:75–81. doi: 10.1016/j.yjmcc.2015.12.023.
    1. Wang H, Zhang X, Li Y, Ma Y, Zhang Y, Liu Z, Zhou J, Lin Q, Wang Y, Duan C, Wang C. Improved myocardial performance in infarcted rat heart by co-injection of basic fibroblast growth factor with temperature-responsive chitosan hydrogel. J Heart Lung Transplant. 2010;29:881–887. doi: 10.1016/j.healun.2010.03.016.
    1. Yuan J, Chen H, Ge D, Xu Y, Xu H, Yang Y, Gu M, Zhou Y, Zhu J, Ge T, et al. Mir-21 promotes cardiac fibrosis after myocardial infarction via targeting Smad7. Cell Physiol Biochem. 2017;42:2207–2219. doi: 10.1159/000479995.
    1. Hao K, Lei W, Wu H, Wu J, Yang Z, Yan S, Lu XA, Li J, Xia X, Han X, et al. LncRNA-Safe contributes to cardiac fibrosis through Safe-Sfrp2-HuR complex in mouse myocardial infarction. Theranostics. 2019;9:7282–7297. doi: 10.7150/thno.33920.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Wang H, Qi C, Wan D. MicroRNA-377-3p targeting MMP-16 inhibits ovarian cancer cell growth, invasion, and interstitial transition. Ann Transl Med. 2021;9(124) doi: 10.21037/atm-20-8027.
    1. Guo J, Gan Q, Gan C, Zhang X, Ma X, Dong M. LncRNA MIR205HG regulates melanomagenesis via the miR-299-3p/VEGFA axis. Aging (Albany NY) 2021;13:5297–5311. doi: 10.18632/aging.202450.
    1. Wu H, Zhou X, Wang X, Cheng W, Hu X, Wang Y, Luo B, Huang W, Gu J. miR-34a in extracellular vesicles from bone marrow mesenchymal stem cells reduces rheumatoid arthritis inflammation via the cyclin I/ATM/ATR/p53 axis. J Cell Mol Med. 2021;25:1896–1910. doi: 10.1111/jcmm.15857.
    1. Qu X, Du Y, Shu Y, Gao M, Sun F, Luo S, Yang T, Zhan L, Yuan Y, Chu W, et al. MIAT is a pro-fibrotic long non-coding RNA governing cardiac fibrosis in post-infarct myocardium. Sci Rep. 2017;7(42657) doi: 10.1038/srep42657.
    1. Zhou B, Yu JW. A novel identified circular RNA, circRNA_010567, promotes myocardial fibrosis via suppressing miR-141 by targeting TGF-β1. Biochem Biophys Res Commun. 2017;487:769–775. doi: 10.1016/j.bbrc.2017.04.044.
    1. Travers JG, Kamal FA, Robbins J, Yutzey KE, Blaxall BC. Cardiac fibrosis: The fibroblast awakens. Circ Res. 2016;118:1021–1040. doi: 10.1161/CIRCRESAHA.115.306565.
    1. Moore-Morris T, Guimaraes-Camboa N, Yutzey KE, Puceat M, Evans SM. Cardiac fibroblasts: From development to heart failure. J Mol Med (Berl) 2015;93:823–830. doi: 10.1007/s00109-015-1314-y.
    1. Leask A. Getting to the heart of the matter new insights into cardiac fibrosis. Circ Res. 2015;116:1269–1276. doi: 10.1161/CIRCRESAHA.116.305381.
    1. Schelbert EB, Fonarow GC, Bonow RO, Butler J, Gheorghiade M. Therapeutic targets in heart failure: Refocusing on the myocardial interstitium. J Am Coll Cardiol. 2014;63:2188–2198. doi: 10.1016/j.jacc.2014.01.068.
    1. Okada J, Yamada E, Saito T, Yokoo H, Osaki A, Shimoda Y, Ozawa A, Nakajima Y, Pessin JE, Okada S, Yamada M. Dapagliflozin inhibits cell adhesion to collagen I and IV and increases ectodomain proteolytic cleavage of DDR1 by increasing ADAM10 activity. Molecules. 2020;25(495) doi: 10.3390/molecules25030495.
    1. Grimaldi V, De Pascale MR, Zullo A, Soricelli A, Infante T, Mancini FP, Napoli C. Evidence of epigenetic tags in cardiac fibrosis. J Cardiol. 2017;69:401–408. doi: 10.1016/j.jjcc.2016.10.004.
    1. Cheng R, Dang R, Zhou Y, Ding M, Hua H. MicroRNA-98 inhibits TGF-β1-induced differentiation and collagen production of cardiac fibroblasts by targeting TGFBR1. Human Cell. 2017;30:192–200. doi: 10.1007/s13577-017-0163-0.
    1. Van Rooij E, Sutherland LB, Thatcher JE, Dimaio JM, Naseem RH, Marshall WS, Hill JA, Olson EN. Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci USA. 2008;105:13027–13032. doi: 10.1073/pnas.0805038105.
    1. Li L, Bounds KR, Chatterjee P, Gupta S. MicroRNA-130a, a potential antifibrotic target in cardiac fibrosis. J Am Heart Assoc. 2017;6(e006763) doi: 10.1161/JAHA.117.006763.

Source: PubMed

3
Subscribe