Acute dapagliflozin administration exerts cardioprotective effects in rats with cardiac ischemia/reperfusion injury

Sarayut Lahnwong, Siripong Palee, Nattayaporn Apaijai, Sirawit Sriwichaiin, Sasiwan Kerdphoo, Thidarat Jaiwongkam, Siriporn C Chattipakorn, Nipon Chattipakorn, Sarayut Lahnwong, Siripong Palee, Nattayaporn Apaijai, Sirawit Sriwichaiin, Sasiwan Kerdphoo, Thidarat Jaiwongkam, Siriporn C Chattipakorn, Nipon Chattipakorn

Abstract

Background: A sodium-glucose co-transporter 2 (SGLT-2) inhibitor had favorable impact on the attenuation of hyperglycemia together with the severity of heart failure. However, the effects of acute dapagliflozin administration at the time of cardiac ischemia/reperfusion (I/R) injury are not established.

Methods: The effects of dapagliflozin on cardiac function were investigated by treating cardiac I/R injury at different time points. Cardiac I/R was instigated in forty-eight Wistar rats. These rats were then split into 4 interventional groups: control, dapagliflozin (SGLT2 inhibitor, 1 mg/kg) given pre-ischemia, at the time of ischemia and at the beginning of reperfusion. Left ventricular (LV) function and arrhythmia score were evaluated. The hearts were used to evaluate size of myocardial infarction, cardiomyocyte apoptosis, cardiac mitochondrial dynamics and function.

Results: Dapagliflozin given pre-ischemia conferred the maximum level of cardioprotection quantified through the decrease in arrhythmia, attenuated infarct size, decreased cardiac apoptosis and improved cardiac mitochondrial function, biogenesis and dynamics, leading to LV function improvement during cardiac I/R injury. Dapagliflozin given during ischemia also showed cardioprotection, but at a lower level of efficacy.

Conclusions: Acute dapagliflozin administration during cardiac I/R injury exerted cardioprotective effects by attenuating cardiac infarct size, increasing LV function and reducing arrhythmias. These benefits indicate its potential clinical usefulness.

Keywords: Dapagliflozin; Heart; Ischemia–reperfusion injury; Mitochondria; Sodium-glucose co-transporter 2 (SGLT-2) inhibitors.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
The protocol used to show the dapagliflozin effects on myocardial and mitochondrial function given at pre-ischemic, during ischemic and onset of reperfusion time points during acute I/R injury in rats, n = 12 per group. TTC triphenyltetrazolium chloride
Fig. 2
Fig. 2
The effects of dapagliflozin on myocardial infarction in rats with cardiac I/R injury. a Area at risk, n = 6 per group; and b Myocardial infarct size, n = 6 per group. *p < 0.05 vs control and †p < 0.05 vs pretreatment group. AAR area at risk
Fig. 3
Fig. 3
The effects of dapagliflozin on cardiac arrhythmia and gap junction-related proteins in rats with I/R injury. a Time to 1st VT/VF, n = 6 per group; b Arrhythmia score, n = 6 per group; c p-Cx43 Ser368/Cx43 protein expression in the ischemic area normalized with that in the remote area, n = 6 per group; and d Representative Western blot bands of p-Cx43 Ser368 and Cx43 proteins expression in the ischemic area and the remote area. *p < 0.05 vs control and †p < 0.05 vs pretreatment group. Cx43 connexin, I ischemic area, p-Cx43 Ser638 phosphorylation of Cx43 at serine638, R remote area, VF ventricular fibrillation, VT ventricular tachycardia
Fig. 4
Fig. 4
The effects of dapagliflozin on left ventricular function in rats with I/R injury. a Heart rate; b Stroke volume; c Left ventricular end systolic pressure; d Left ventricular end diastolic pressure; edP/dtmax; fdP/dtmin; and g Left ventricular ejection fraction, n = 6 per group. *p < 0.05 vs baseline of its group; †p < 0.05 vs control group at that period. dP/dt ventricular contractility assessment, EDP end diastolic pressure, ESP end systolic pressure, HR heart rate, LVEF left ventricular ejection fraction, SV stroke volume
Fig. 5
Fig. 5
The effects of dapagliflozin on cardiac cell apoptosis in rats with cardiac I/R injury. a Bax; b Bcl-2; c Caspase-3; and d Cleaved caspase-3 expression in the ischemic area normalized with that in the remote area, n = 6 per group. *p < 0.05 vs control. I ischemic area, R remote area
Fig. 6
Fig. 6
The effect of dapagliflozin on TUNEL-positive cells in the heart. a Representative images of TUNEL-positive cells; b Apoptotic index, n = 4 per group. *p < 0.05 vs control; †p < 0.05 vs pretreatment group; ‡p < 0.05 vs during ischemia
Fig. 7
Fig. 7
The effects of dapagliflozin on cardiac mitochondrial function in rats with cardiac I/R injury. a Mitochondrial ROS production; b Mitochondrial membrane potential changes; and c Mitochondrial swelling, n = 6 per group. *p < 0.05 vs control. MMP mitochondrial membrane potential, ROS reactive oxygen species
Fig. 8
Fig. 8
The effects of dapagliflozin on proteins associated with myocardial metabolism and oxidative phosphorylation in rats with I/R injury. a Myocardial PGC1-α; b Myocardial CPT-1; and c Cardiac mitochondrial complex I–V expression in the ischemic area normalized with that in the remote area, n = 6 per group. *p < 0.05 vs control. CPT1 carnitine palmitoyltransferase I, GAPDH glyceraldehyde 3-phosphate dehydrogenase, I ischemic area, PGC1-α peroxisome proliferator-activated receptor gamma coactivator 1-alpha, R remote area, VDAC voltage-dependent anion channel
Fig. 9
Fig. 9
The effects of dapagliflozin on the expression of cardiac mitochondrial fission and fusion protein in rats with I/R injury. a Cardiac mitochondrial DRP1; b MFN2; c OPA1 expression in the ischemic area normalized with that in the remote area, n = 6 per group; and d Representative western blot bands of a–c. *p < 0.05 vs control. DRP1 dynamin-related protein 1, I ischemic area, MFN2 mitofusin 2, OPA1 optic atrophy 1, R remote area, VDAC voltage-dependent anion channel

References

    1. Wong ND. Epidemiological studies of CHD and the evolution of preventive cardiology. Nat Rev Cardiol. 2014;11:276–289.
    1. Cassar A, Holmes DR, Rihal CS, Gersh BJ. Chronic coronary artery disease: diagnosis and management. Mayo Clin Proc. 2009;84:1130–1146.
    1. Iwasaki K, Matsumoto T, Aono H, Furukawa H, Nagamachi K, Samukawa M. Distribution of coronary atherosclerosis in patients with coronary artery disease. Heart Vessels. 2010;25:14–18.
    1. Danchin N. Systems of care for ST-segment elevation myocardial infarction: impact of different models on clinical outcomes. JACC Cardiovasc Interv. 2009;2:901–908.
    1. Reddy K, Khaliq A, Henning RJ. Recent advances in the diagnosis and treatment of acute myocardial infarction. World J Cardiol. 2015;7:243–276.
    1. Lip GYH, Chin BSP, Prasad N. ABC of antithrombotic therapy: antithrombotic therapy in myocardial infarction and stable angina. BMJ. 2002;325:1287–1289.
    1. Hausenloy DJ, Yellon DM. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J Clin Invest. 2013;123:92–100.
    1. Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N Engl J Med. 2007;357:1121–1135.
    1. Verbrugge FH. Role of SGLT2 inhibitors in patients with diabetes mellitus and heart failure. Curr Heart Fail Rep. 2017;14:275–283.
    1. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128.
    1. Tanajak P, Sa-Nguanmoo P, Sivasinprasasn S, Thummasorn S, Siri-Angkul N, Chattipakorn SC, et al. Cardioprotection of dapagliflozin and vildagliptin in rats with cardiac ischemia-reperfusion injury. J Endocrinol. 2018;236:69–84.
    1. Lee T-M, Chang N-C, Lin S-Z. Dapagliflozin, a selective SGLT2 Inhibitor, attenuated cardiac fibrosis by regulating the macrophage polarization via STAT3 signaling in infarcted rat hearts. Free Radic Biol Med. 2017;104:298–310.
    1. Chinda K, Sanit J, Chattipakorn S, Chattipakorn N. Dipeptidyl peptidase-4 inhibitor reduces infarct size and preserves cardiac function via mitochondrial protection in ischaemia-reperfusion rat heart. Diab Vasc Dis Res. 2014;11:75–83.
    1. Thummasorn S, Apaijai N, Kerdphoo S, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Humanin exerts cardioprotection against cardiac ischemia/reperfusion injury through attenuation of mitochondrial dysfunction. Cardiovasc Ther. 2016;34:404–414.
    1. Surinkaew S, Kumphune S, Chattipakorn S, Chattipakorn N. Inhibition of p38 MAPK during ischemia, but not reperfusion, effectively attenuates fatal arrhythmia in ischemia/reperfusion heart. J Cardiovasc Pharmacol. 2013;61:133–141.
    1. Curtis MJ, Hancox JC, Farkas A, Wainwright CL, Stables CL, Saint DA, et al. The lambeth conventions (II): guidelines for the study of animal and human ventricular and supraventricular arrhythmias. Pharmacol Ther. 2013;139:213–248.
    1. Pongkan W, Chattipakorn SC, Chattipakorn N. Chronic testosterone replacement exerts cardioprotection against cardiac ischemia-reperfusion injury by attenuating mitochondrial dysfunction in testosterone-deprived rats. PLoS ONE. 2015;10:e0122503.
    1. Pongkan W, Pintana H, Jaiwongkam T, Kredphoo S, Sivasinprasasn S, Chattipakorn SC, et al. Vildagliptin reduces cardiac ischemic-reperfusion injury in obese orchiectomized rats. J Endocrinol. 2016;231:81–95.
    1. Thummasorn S, Kumfu S, Chattipakorn S, Chattipakorn N. Granulocyte-colony stimulating factor attenuates mitochondrial dysfunction induced by oxidative stress in cardiac mitochondria. Mitochondrion. 2011;11:457–466.
    1. Tanajak P, Pintana H, Siri-Angkul N, Khamseekaew J, Apaijai N, Chattipakorn SC, et al. Vildagliptin and caloric restriction for cardioprotection in pre-diabetic rats. J Endocrinol. 2017;232:189–204.
    1. Nuntaphum W, Pongkan W, Wongjaikam S, Thummasorn S, Tanajak P, Khamseekaew J, et al. Vagus nerve stimulation exerts cardioprotection against myocardial ischemia/reperfusion injury predominantly through its efferent vagal fibers. Basic Res Cardiol. 2018;113:22.
    1. Tanaka A, Node K. Emerging roles of sodium-glucose cotransporter 2 inhibitors in cardiology. J Cardiol. 2017;69:501–507.
    1. Rajasekeran H, Lytvyn Y, Cherney DZI. Sodium-glucose cotransporter 2 inhibition and cardiovascular risk reduction in patients with type 2 diabetes: the emerging role of natriuresis. Kidney Int. 2016;89:524–526.
    1. Abdul-Ghani M, Del Prato S, Chilton R, DeFronzo RA. SGLT2 inhibitors and cardiovascular risk: lessons learned from the EMPA-REG OUTCOME study. Diabetes Care. 2016;39:717–725.
    1. Heerspink HJL, Perkins BA, Fitchett DH, Husain M, Cherney DZI. Sodium glucose cotransporter 2 inhibitors in the treatment of diabetes mellitus: cardiovascular and kidney effects, potential mechanisms, and clinical applications. Circulation. 2016;134:752–772.
    1. Marx N, McGuire DK. Sodium-glucose cotransporter-2 inhibition for the reduction of cardiovascular events in high-risk patients with diabetes mellitus. Eur Heart J. 2016;37:3192–3200.
    1. Lahnwong S, Chattipakorn SC, Chattipakorn N. Potential mechanisms responsible for cardioprotective effects of sodium-glucose co-transporter 2 inhibitors. Cardiovasc Diabetol. 2018;17:101.
    1. Soga F, Tanaka H, Tatsumi K, Mochizuki Y, Sano H, Toki H, et al. Impact of dapagliflozin on left ventricular diastolic function of patients with type 2 diabetic mellitus with chronic heart failure. Cardiovasc Diabetol. 2018;17:132.
    1. Matsutani D, Sakamoto M, Kayama Y, Takeda N, Horiuchi R, Utsunomiya K. Effect of canagliflozin on left ventricular diastolic function in patients with type 2 diabetes. Cardiovasc Diabetol. 2018;17:73.
    1. Kim Y-G, Han SJ, Kim DJ, Lee K-W, Kim HJ. Association between sodium glucose co-transporter 2 inhibitors and a reduced risk of heart failure in patients with type 2 diabetes mellitus: a real-world nationwide population-based cohort study. Cardiovasc Diabetol. 2018;17:91.
    1. Patorno E, Goldfine AB, Schneeweiss S, Everett BM, Glynn RJ, Liu J, et al. Cardiovascular outcomes associated with canagliflozin versus other non-gliflozin antidiabetic drugs: population based cohort study. BMJ. 2018;360:k119.
    1. Gallwitz B. The cardiovascular benefits associated with the use of sodium-glucose cotransporter 2 inhibitors—real-world data. Eur Endocrinol. 2018;14:17–23.
    1. Di Franco A, Cantini G, Tani A, Coppini R, Zecchi-Orlandini S, Raimondi L, et al. Sodium-dependent glucose transporters (SGLT) in human ischemic heart: a new potential pharmacological target. Int J Cardiol. 2017;243:86–90.
    1. Li C, Zhang J, Xue M, Li X, Han F, Liu X, et al. SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovasc Diabetol. 2019;18:15.
    1. Mori K, Tsuchiya K, Nakamura S, Miyachi Y, Shiba K, Ogawa Y, et al. Ipragliflozin-induced adipose expansion inhibits cuff-induced vascular remodeling in mice. Cardiovasc Diabetol. 2019;18:83.
    1. Lampe PD, TenBroek EM, Burt JM, Kurata WE, Johnson RG, Lau AF. Phosphorylation of connexin43 on serine368 by protein kinase C regulates gap junctional communication. J Cell Biol. 2000;149:1503–1512.
    1. Venditti P, Masullo P, Di Meo S. Effects of myocardial ischemia and reperfusion on mitochondrial function and susceptibility to oxidative stress. Cell Mol Life Sci. 2001;58:1528–1537.
    1. Zhou H, Zhu P, Wang J, Zhu H, Ren J, Chen Y. Pathogenesis of cardiac ischemia reperfusion injury is associated with CK2α-disturbed mitochondrial homeostasis via suppression of FUNDC1-related mitophagy. Cell Death Differ. 2018;25:1080–1093.
    1. Lucas E, Vila-Bedmar R, Arcones AC, Cruces-Sande M, Cachofeiro V, Mayor F, et al. Obesity-induced cardiac lipid accumulation in adult mice is modulated by G protein-coupled receptor kinase 2 levels. Cardiovasc Diabetol. 2016;15:155.
    1. Duncan JG. Peroxisome proliferator activated receptor-alpha (PPARα) and PPAR gamma coactivator-1alpha (PGC-1α) regulation of cardiac metabolism in diabetes. Pediatr Cardiol. 2011;32:323–328.
    1. Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol. 2010;11:872–884.
    1. Youle RJ, van der Bliek AM. Mitochondrial fission, fusion, and stress. Science. 2012;337:1062–1065.
    1. Galloway CA, Yoon Y. Mitochondrial dynamics in diabetic cardiomyopathy. Antioxid Redox Signal. 2015;22:1545–1562.
    1. Ong S-B, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation. 2010;121:2012–2022.
    1. Sharp WW, Beiser DG, Fang YH, Han M, Piao L, Varughese J, et al. Inhibition of the mitochondrial fission protein dynamin-related protein 1 improves survival in a murine cardiac arrest model. Crit Care Med. 2015;43:e38–e47.

Source: PubMed

3
S'abonner