Empagliflozin reduces the senescence of cardiac stromal cells and improves cardiac function in a murine model of diabetes

Rosalinda Madonna, Vanessa Doria, Ilaria Minnucci, Angela Pucci, Donato Sante Pierdomenico, Raffaele De Caterina, Rosalinda Madonna, Vanessa Doria, Ilaria Minnucci, Angela Pucci, Donato Sante Pierdomenico, Raffaele De Caterina

Abstract

The sodium-glucose cotransporter 2 (SGLT2) inhibitor empagliflozin reduces heart failure in diabetes, but underlying mechanisms remain elusive. We hypothesized that empagliflozin could counteract the senescence of cardiac stromal cells (CSC), the action of which limits cardiac damage and cardiac fibrosis in diabetic-like conditions in vitro and in vivo. CSC were isolated from murine heart biopsies (n = 5) through cardiosphere (CSp) formation and incubated for 3 or 48 hours with 5.5 mmol/L normal glucose (NG), high glucose (12-5 and 30.5 mmol/L, HG) or a hyperosmolar control of mannitol (HM) in the presence or absence of empagliflozin 100 nmol/L. The senescent CSC status was verified by β-gal staining and expression of the pro-survival marker Akt (pAkt) and the pro-inflammatory marker p38 (p-P38). The cardiac effects of empagliflozin were also studied in vivo by echocardiography and by histology in a murine model of streptozotocin (STZ)-induced diabetes. Compared to NG, incubations with HG and HM significantly reduced the number of CSps, increased the β-gal-positive CSC and P-p38, while decreasing pAkt, all reversed by empagliflozin (P < .01). Empagliflozin also reversed cardiac dysfunction, cardiac fibrosis and cell senescence in mice with (STZ)-induced diabetes (P < .01). Empagliflozin counteracts the pro-senescence effect of HG and of hyperosmolar stress on CSC, and improves cardiac function via decreasing cardiac fibrosis and senescence in diabetic mice, possibly through SGLT2 off-target effects. These effects may explain empagliflozin unexpected benefits on cardiac function in diabetic patients.

Keywords: cardiac function; cardiac stromal cells; diabetes; empagliflozin; glucose; senescence.

Conflict of interest statement

The authors declare no conflict of interest.

© 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Effects of high glucose and high mannitol with or without empagliflozin on cardiosphere number and senescence of murine cardiac stromal cells. Panels A and B, Quantitation of murine cardiospheres in multiple microscope fields, treated with normal glucose (5.5 mmol/L) or high glucose (12.5 and 30 mmol/L) or high mannitol (7.0 and 25 mmol/L) for 3 h (panel A) and 48 h (Panel B), in the presence or absence of 100 nmol/L empagliflozin. The graph bars showing the total number of cardiospheres over 5 microscope fields per sample represent means ± standard deviations (n = 3 independent experiments). Panel C, Quantitation of senescent cells in multiple microscope fields from murine cardiac stromal cells (CSC) treated with low (5.5 mmol/L) or high glucose (30 mmol/L) or high mannitol (25 mmol/L) for 48 h, in the presence or absence of 100 nmol/L empagliflozin. The graph bars, showing the ratio between the number of senescence‐associated β‐galactosidase (SA‐βGal) positive cells and the total number of cells over 5 microscope fields per sample, represent means ± standard deviations (n = 3 independent experiments). Panel D, Representative SA‐βGal staining images of CSC treated with normal glucose (5.5 mmol/L) or high glucose (30 mmol/L) or high mannitol (25 mmol/L) for 48 h, in the presence or absence of 100 nmol/L empagliflozin, as indicated by the blue staining (arrows). All images are shown at 10× magnification. *P < .05 and **P < .01 vs. normal glucose; °P < .05 and °°P < .01 vs without empagliflozin. E, empagliflozin; Glu, glucose; Man, mannitol; SA‐βGal, senescence‐associated β‐galactosidase
Figure 2
Figure 2
Effects of high glucose and high mannitol with or without empagliflozin on the expression of pAkt, P‐P38, IR‐1α and IRS‐1 in murine cardiac stromal cells. Western analysis of the pro‐survival marker pAkt and AKT (A), pro‐inflammatory marker P‐P38 (B), IR‐1α (C), IRS‐1 and pIRS‐1 (D) in murine cardiac stromal cells treated with normal glucose (5.5 mmol/L) or high glucose (12.5 and 30 mmol/L) or high mannitol (7.0 and 25 mmol/L) for 48 h, in the presence or absence of 100 nmol/L empagliflozin, with β‐actin serving as a loading control. The graph bars represent means ± standard deviations (n = 3 independent experiments). **P < .01 vs normal glucose; °°P < .01 vs without empagliflozin. Glu, glucose; IR, insulin receptor; IRS, Insulin receptor substrate; Man, mannitol; pAkt, phosphorilated Akt; P‐p38, phosphorylated p38
Figure 3
Figure 3
Effects of streptozotocin and empagliflozin on cardiac function in mice. Panels A, Ejection Fraction (EF) and B, Fractional Shortening (FS) measured by echocardiography in the different treatment groups such as control (vehicle), streptozotocin‐treated mice and streptozotocin + empagliflozin‐treated mice. Panels C, Representative M‐mode and B‐mode images recorded by echocardiography in the different treatment groups. Data are expressed as means ± standard deviations (n = 5 mice per treatment group). **P < .01 vs vehicle‐treated mice; °°P < .01 vs without empagliflozin. E, empagliflozin; STZ, streptozotocin
Figure 4
Figure 4
Effects of streptozotocin and empagliflozin on cardiac fibrosis in mice. Panels A‐C, Picro‐sirius red staining showing the collagen content in left ventricle sections from control (vehicle), streptozotocin‐treated mice and streptozotocin + empagliflozin‐treated mice. All images are shown at 20× original magnification. Panel D, Collagen content quantified on picro‐sirius red staining using the ImageJ software. Panel E, Quantification of collagen type III expression by western analysis. Panel F, Representative western blot showing type III collagen expression in each treatment group. β‐actin levels were assessed as a loading control. Data are expressed as means ± standard deviation (n = 5 mice per treatment group). **P < .01 vs vehicle‐treated mice; °°P < .01 vs without empagliflozin. E, empagliflozin; STZ, streptozotocin
Figure 5
Figure 5
Effects of streptozotocin and empagliflozin on cardiac senescence‐associated β‐galactosidase activity in mice. Panels A‐C, Senescence β‐galactosidase (SA β‐Gal) staining (magnification 5× and 40×) and quantitative evaluations (Panel D) in cardiac tissue. Data are expressed as means ± standard deviations (n = 5 mice per treatment group). **P < .01 vs vehicle‐treated mice; °°P < .01 vs without empagliflozin. E, empagliflozin; STZ, streptozotocin
Figure 6
Figure 6
A scheme of the putative signalling pathway through which empagliflozin target cardiac stromal cell senescence induced by hyperglycaemia and hyperosmotic, based on findings from the in vitro part of the present study. In the plasma membrane, hyperglycaemia induces hyperosmotic stress, and together, these metabolic and biophysical stressors impair the ability of cardiac stromal cells to respond to insulin. This causes the perturbation of the pro‐survival PI3K/Akt and pro‐inflammatory p38 pathways with insulin preserving its signalling capacity through the mitogenic and pro‐inflammatory arm, leading to the development and progression of a pro‐senescent phenotype. Empagliflozin is able to revert CSC senescence by targeting the insulin signalling pathway

References

    1. Timmis A, Townsend N, Gale C, et al. European society of cardiology: cardiovascular disease statistics 2017. Eur Heart J. 2018;39:508‐579.
    1. Xin M, Olson EN, Bassel‐Duby R. Mending broken hearts: cardiac development as a basis for adult heart regeneration and repair. Nat Rev Mol Cell Biol. 2013;14:529‐541.
    1. Olson EN. A decade of discoveries in cardiac biology. Nat Med. 2004;10:467‐474.
    1. Mayourian J, Ceholski DK, Gonzalez DM, et al. Physiologic, pathologic, and therapeutic paracrine modulation of cardiac excitation‐contraction coupling. Circ Res. 2018;122:167‐183.
    1. Shen D, Shen M, Liang H, et al. Therapeutic benefits of CD90‐negative cardiac stromal cells in rats with a 30‐day chronic infarct. J Cell Mol Med. 2018;22:1984‐1991.
    1. Forbes SJ, Rosenthal N. Preparing the ground for tissue regeneration: from mechanism to therapy. Nat Med. 2014;20:857‐869.
    1. Cencioni C, Atlante S, Savoia M, et al. The double life of cardiac mesenchymal cells: epimetabolic sensors and therapeutic assets for heart regeneration. Pharmacol Ther. 2017;171:43‐55.
    1. Perkisas S, Vandewoude M. Where frailty meets diabetes. Diabetes Metab Res Rev. 2016;32(Suppl 1):261‐267.
    1. Morley JE. Diabetes and aging: epidemiologic overview. Clin Geriatr Med. 2008;24:395‐405.
    1. Moore‐Morris T, Guimarães‐Camboa N, Yutzey K, Pucéat M, Evans S. Cardiac fibroblasts: from development to heart failure. J Mol Med. 2015;93:823‐830.
    1. Persson F, Nystrom T, Jorgensen ME, et al. Dapagliflozin is associated with lower risk of cardiovascular events and all‐cause mortality in people with type 2 diabetes (CVD‐REAL Nordic) when compared with dipeptidyl peptidase‐4 inhibitor therapy: a multinational observational study. Diabetes Obes Metab. 2018;20:344‐351.
    1. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117‐2128.
    1. Wu KK, Huan Y. Streptozotocin‐induced diabetic models in mice and rats. Curr Protoc Pharmacol. 2008;Chapter 5:Unit 5 47.
    1. Madonna R, Pieragostino D, Cufaro MC, et al. Ponatinib induces vascular toxicity through the notch‐1 signaling pathway. J Clin Med. 2020;9(3):820.
    1. Madonna R, Di Napoli P, Massaro M, et al. Simvastatin attenuates expression of cytokine‐inducible nitric‐oxide synthase in embryonic cardiac myoblasts. J Biol Chem. 2005;280:13503‐13511.
    1. Nystrom F, Quon M. Insulin signalling: metabolic pathways and mechanisms for specificity. Cell Signal. 1999;11:563‐574.
    1. Lee C‐H, Shieh Y‐S, Hsiao F‐C, et al. High glucose induces human endothelial dysfunction through an Axl‐dependent mechanism. Cardiovasc Diabetol. 2014;13:53.
    1. Madonna R, Pieragostino D, Rossi C, et al. Simulated hyperglycemia impairs insulin signaling in endothelial cells through a hyperosmolar mechanism. Vascul Pharmacol. 2020;130:106678.
    1. Liao Y, Takashima S, Zhao H, et al. Control of plasma glucose with alpha‐glucosidase inhibitor attenuates oxidative stress and slows the progression of heart failure in mice. Cardiovasc Res. 2006;70:107‐116.
    1. Guo Y, Zhuang X, Huang Z, et al. Klotho protects the heart from hyperglycemia‐induced injury by inactivating ROS and NF‐κB‐mediated inflammation both in vitro and in vivo. Biochim Biophys Acta Mol Basis Dis. 2018;1864:238‐251.
    1. Andreadou I, Efentakis P, Balafas E, et al. Empagliflozin limits myocardial infarction in vivo and cell death in vitro: role of STAT3, mitochondria, and redox aspects. Front Physiol. 2017;8:1077.
    1. Penna C, Andreadou I, Aragno M, et al. Effect of hyperglycaemia and diabetes on acute myocardial ischaemia‐reperfusion injury and cardioprotection by ischaemic conditioning protocols. Br J Pharmacol. 2020.
    1. Penna C, Perrelli M‐G, Tullio F, et al. Diazoxide postconditioning induces mitochondrial protein S‐nitrosylation and a redox‐sensitive mitochondrial phosphorylation/translocation of RISK elements: no role for SAFE. Basic Res Cardiol. 2013;108:371.
    1. Nikolaou PE, Efentakis P, Qourah FA, et al. Chronic Empaglifozin treatment reduces myocardial infarct size in non‐diabetic mice through STAT‐3 mediated protection on microvascular endothelial cells and reduction of oxidative stress. Antioxid Redox Signal. 2020.
    1. Madonna R, Novo G, Balistreri C. Cellular and molecular basis of the imbalance between vascular damage and repair in ageing and age‐related diseases: as biomarkers and targets for new treatments. Mech Ageing Dev. 2016;159:22‐30.
    1. Datta S, Brunet A, Greenberg M. Cellular survival: a play in three Akts. Genes Dev. 1999;13:2905‐2927.
    1. Medema R, Kops G, Bos J, Burgering B. AFX‐like Forkhead transcription factors mediate cell‐cycle regulation by Ras and PKB through p27kip1. Nature. 2000;404:782‐787.
    1. Shin I, Yakes FM, Rojo F, et al. PKB/Akt mediates cell‐cycle progression by phosphorylation of p27(Kip1) at threonine 157 and modulation of its cellular localization. Nat Med. 2002;8:1145‐1152.
    1. Viglietto G, Motti ML, Bruni P, et al. Cytoplasmic relocalization and inhibition of the cyclin‐dependent kinase inhibitor p27(Kip1) by PKB/Akt‐mediated phosphorylation in breast cancer. Nat Med. 2002;8:1136‐1144.
    1. Diehl J, Cheng M, Roussel M, Sherr C. Glycogen synthase kinase‐3beta regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 1998;12:3499‐3511.
    1. Datta SR, Dudek H, Tao XU, et al. Akt phosphorylation of BAD couples survival signals to the cell‐intrinsic death machinery. Cell. 1997;91:231‐241.
    1. Cardone M, Roy N, Stennicke H, et al. Regulation of cell death protease caspase‐9 by phosphorylation. Science. 1998;282:1318‐1321.
    1. Kusaka H, Koibuchi N, Hasegawa Y, Ogawa H, Kim‐Mitsuyama S. Empagliflozin lessened cardiac injury and reduced visceral adipocyte hypertrophy in prediabetic rats with metabolic syndrome. Cardiovasc Diabetol. 2016;15:1‐14.
    1. Lin B, Koibuchi N, Hasegawa YU, et al. Glycemic control with empagliflozin, a novel selective SGLT2 inhibitor, ameliorates cardiovascular injury and cognitive dysfunction in obese and type 2 diabetic mice. Cardiovasc Diabetol. 2014;13:1‐15.
    1. Habibi J, Aroor AR, Sowers JR, et al. Sodium glucose transporter 2 (SGLT2) inhibition with empagliflozin improves cardiac diastolic function in a female rodent model of diabetes. Cardiovasc Diabetol. 2017;16:9.
    1. Vrhovac I, Balen Eror D, Klessen D, et al. Localizations of Na(+)‐D‐glucose cotransporters SGLT1 and SGLT2 in human kidney and of SGLT1 in human small intestine, liver, lung, and heart. Pflugers Arch. 2015;467:1881‐1898.
    1. Maly K, Strese K, Kampfer S. Critical role of protein kinase C a and calcium in growth factor induced activation of the Na�/H� exchanger NHE1. FEBS Lett. 2002;521:205‐210.
    1. Orlowski J, Grinstein S. Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pflugers Arch. 2004;447:549‐565.
    1. Uthman L, Baartscheer A, Bleijlevens B, et al. Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: inhibition of Na+/H+ exchanger, lowering of cytosolic Na+ and vasodilation. Diabetologia. 2018;61:722‐726.
    1. Madonna R, Montebello E, Lazzerini G, Zurro M, De Caterina R. NA+/H+ exchanger 1‐ and aquaporin‐1‐dependent hyperosmolarity changes decrease nitric oxide production and induce VCAM‐1 expression in endothelial cells exposed to high glucose. Int J Immunopathol Pharmacol. 2010;23:755‐765.
    1. Madonna R, De Caterina R. Sodium‐hydrogen exchangers (NHE) in human cardiovascular diseases: interfering strategies and their therapeutic applications. Vascul Pharmacol. 2013;59:127‐130.
    1. Madonna R, De Caterina R. Aquaporin‐1 and sodium‐hydrogen exchangers as pharmacological targets in diabetic atherosclerosis. Curr Drug Targets. 2015;16:361‐365.
    1. Darmellah A, Baetz D, Prunier F, Tamareille S, Rücker‐Martin C, Feuvray D. Enhanced activity of the myocardial Na+/H+ exchanger contributes to left ventricular hypertrophy in the Goto‐Kakizaki rat model of type 2 diabetes: critical role of Akt. Diabetologia. 2007;50:1335‐1344.
    1. Engelhardt S, Hein L, Keller U, Klambt K, Lohse M. Inhibition of Na+/H+ exchange prevents hypertrophy, fibrosis, and heart failure in β1‐adrenergic receptor transgenic mice. Circ Res. 2002;90:814‐819.
    1. Linz W, Busch A. NHE‐1 inhibition: from protection during acute ischaemia/reperfusion to prevention/reversal of myocardial remodeling. Naunyn Schmiedebergs Arch Pharmacol. 2003;368:239‐246.

Source: PubMed

3
Tilaa