Potential Mechanisms of Sodium-Glucose Co-Transporter 2 Inhibitors: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Gassan Moady.

Sodium-glucose co-transporter 2 (SGLT2) inhibitors, originally used for diabetes mellitus, are gaining more popularity for other indications, owing to their positive cardiovascular and renal effects. SGLT2 inhibitors reduce heart failure (HF) hospitalization and improve cardiovascular outcomes in patients with type 2 diabetes. SGLT2 inhibitors exhibit pleotropic effects on the cardiovascular system. It is clear that these beneficial effects are not related to the anti-glycemic properties. Cardiovascular benefits of SGLT2 inhibitors are mediated by several pathways. Enhanced diuresis and vascular dilation result in afterload reduction and coronary blood flow augmentation. Ketogenesis mediated by adipose tissue and hepatocytes promote ketogenesis, which serves as an energy source for the failing heart. In addition, SGLT2 inhibitors contribute to decreasing inflammation and improving systolic and diastolic functions by reducing fibrosis and inhibiting remodeling pathways.

  • heart failure
  • sodium-glucose co-transporter
  • diabetes

1. General Effects

Early afterload reduction and enhanced diuresis with the concomitant decrease in filling pressures probably play a role in the rapid reduction in HF hospitalizations that have been observed in the major clinical trials. In addition, SGLT2 inhibitors also cause a modest weigh reduction that may help in reducing blood pressure and improving glycemic control. The reduction in blood pressure is suggested to be secondary to improving endothelial function and decreasing arterial stiffness and sympathetic activity (discussed below) and unrelated to volume depletion or natriuresis [42,43][1][2]. Since osmotic diuresis associated with SGLT2 inhibitors is dependent on glucose level, this effect does not explain the benefits in non-diabetic patients with HF [42][1]. NT-proBNP is an important prognostic marker in HF, which reflects the volume status and the overall wall stress. However, in the DEFINE-HF trial, there was no decrease in NT-proBNP concentration despite the improvement in HF status [44][3]. This finding raises questions about the role of natriuresis associated with SGLT2 inhibition in improving HF outcomes [42][1]. Likewise, weight reduction is observed mainly in diabetic patients, and therefore, does not provide an explanation for cardiovascular benefits in non-diabetic patients.

2. Effects on Sympathetic Pathways

HF is associated with the activation of several neurohormonal pathways, including the sympathetic system, renin–angiotensin–aldosterone system, and the natriuretic peptides pathways. Despite a modest decline in blood pressure and intravascular volume depletion associated with SGLT2 inhibitors, there is no concomitant increase in heart rate. This finding may indicate a possible effect on the sympathetic system [45,46][4][5]. In experimental rat models with metabolic syndrome, luseogliflozin (a selective SGLT2 inhibitor) improved the circadian rhythm of the sympathetic nervous system [47][6]. In another model of hypertensive mice, chemical denervation caused a reduction in blood pressure, blood glucose, and renal SGLT2 protein expression [48][7]. Furthermore, the inhibition of SGLT2 in these models reduced the level of tyrosine hydroxylase and norepinephrine. Evidence for improving cardiac nerve activity with empagliflozin was demonstrated in diabetic patients with AMI in the EMBODY trial [49][8]. These findings may give insight to the role of SGLT2 inhibitors in preventing lethal arrhythmia in the acute phase of AMI.

3. Cardio-Renal Pathways

SGLT2 inhibitors exhibit natriuretic effects by acting on SGLT and Sodium/Hydrogen exchanger 3 (NHE3) receptors in the proximal tubule, resulting in an increase in the fractional excretion of sodium [50][9]. Afferent arteriolar vasoconstriction caused by SGLT2 inhibitors leads to a decrease in renal blood and an increase in erythropoietin (EPO) synthesis, which in turn promotes adenosine triphosphate (ATP) production and utilization in the cardiac tissue, and may help reduce inflammation [51][10]. In one study, empagliflozin administration induced erythropoiesis and iron utilization mediated by increased EPO secretion [52][11]. Nevertheless, it should be noted that the administration of darbepoetin alfa in patients with HF leads to an increase in hematocrit but without improvement in outcomes [53][12]. Another potential mediator in the cardiovascular–renal arena is serum uric acid. Uric acid is elevated in patients with diabetes and considered a part of the metabolic syndrome. Hyperuricemia in diabetic patients is associated with increased risk of hypertension, cardiovascular events, and progression of diabetic kidney disease. SGLT2 inhibitors reduce uric acid concentration by enhancing its urinary excretion facilitated by glucose transporter 9 [54][13].

4. Modulation of Energy Sources and Inflammatory Process

Glycosuria induced by SGLT2 inhibitors combined with the decrease in insulin and the increase in glucagon levels mimic starvation and cause lipolysis and fatty acid oxygenation, leading, finally, to ketogenesis [55][14]. Ketone bodies levels increase in conditions with oxidative stress, such as exercise, starvation, sepsis, and HF [56][15]. Under physiological conditions, the metabolism of healthy hearts depends on free fatty acids and glucose, whereas in the failing heart, a shift towards increased ketone utilization occurs [57,58][16][17]. In the failing heart, ketone bodies serve as an important source of energy as they require less oxygen per molecule of the ATP generated, and may also provide protective anti-inflammatory and antioxidant properties by inhibiting the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing 3 (NLRP3) inflammasome [59,60,61][18][19][20]. The modulation of mitochondrial autophagy is another pathway that may explain SGLT2 inhibitors’ positive cardiac effects. Autophagy is a catabolic process aimed at cleaning intracellular debris through lysosomes, and it contributes to cell hemostasis; however, this pathway is impaired in diabetic patients [62][21]. SGLT2 inhibitors, by mimicking nutrition deprivation, can stimulate autophagy mediated by Siritin-1 and adenosine 5′ monophosphate-activated protein kinas, thus adding another cardioprotective effect [63][22].

5. Microvascular Function

In an insulin resistance mouse model, empagliflozin increased the coronary flow reserve, indicating a possible positive effect on vascular function [64][23]. In diabetic patients, treatment with empagliflozin was associated with weight loss and reduction in several cardiac biomarkers; however, there was no improvement in coronary flow velocity reserve assesses by echo Doppler [65][24]. In another small trial, dapagliflozin administration improved myocardial blood flow and myocardial flow reserve measured by 13N-ammonia PET/CT in diabetic patients with stable coronary artery disease [66][25]. Differences in the results may reflect different methods in assessing coronary flow or the absence of class effect.

6. Effect on Lipid Profile

By mimicking starvation conditions, the inhibition of SGLT2 induces lipolysis in the adipose tissue and causes an increase in non-esterified fatty acids, which in turn promotes cholesterol biosynthesis in the liver and finally causes the downregulation of low-density lipoprotein cholesterol (LDL-C) [67,68][26][27]. This pathway may explain the tendency towards increase in circulating LDL-C, following SGLT2 inhibitors treatment [69][28]. Among the different SGLT2 inhibitor drugs, canagliflozin was associated with the worst effect on the lipid profile [69][28]. Along with the increase in LDL-C levels, a modest increase in high-density lipoprotein (HDL-C) has been reported with the use of SGLT2 inhibitors [67,68,69][26][27][28]. It should be noted, however, that the cardiovascular benefits of SGLT2 inhibitors are maintained regardless of the LDL-C or HDL-C levels [70][29].

7. Effect on Remodeling and Fibrosis

Several echocardiographic and animal-model studies have demonstrated the anti-remodeling effects of SGLT2 inhibitors. In non-diabetic rats, empagliflozin results in reduced collagen deposition and the inhibition of a fibrosis pathway when administered early after the induction of AMI [15][30]. In mouse models of pressure overload (induced by transverse aortic restriction), dapagliflozin reduced fibrosis and improved cardiac systolic function [71][31]. The anti-fibrotic effects of SGLT2 inhibitors may be mediated by inhibiting angiotensin II pathways, which finally leads to changes in the size, shape, geometry, and function of cardiac muscles [72,73,74,75][32][33][34][35]. In the EMPA-HEART trial, empagliflozin resulted in reduced LV mass index in diabetic patients with preserved LV function and coronary artery disease [76][36]. Likewise, in the SUGAR-DM-HF trial, empagliflozin led to reduction in LV end-systolic and end-diastolic volumes in diabetic and prediabetic patients with HFREF [77][37]. In a recent meta-analysis of randomized controlled trials of cardiac remodeling, as measured by cardiac magnetic resonance imaging, SGLT2 inhibitors’ treatment showed a decrease in LV mass [78][38]. Global longitudinal strain measured by speckle tracking was also shown to be positively affected by SGLT2 inhibition among patients with diabetes [79][39]. A significant improvement in diastolic parameters was also observed in patients with HFPEF [80][40]. This finding may in part explain the benefit observed in the clinical trials of SGLT2 inhibitors in HFPEF [10,11][41][42].

References

  1. Joshi, S.S.; Singh, T.; Newby, D.E.; Singh, J. Sodium-glucose co-transporter 2 inhibitor therapy: Mechanisms of action in heart failure. Heart 2021, 107, 1032–1038.
  2. Cherney, D.Z.; Perkins, B.A.; Soleymanlou, N.; Har, R.; Fagan, N.; Johansen, O.E.; Woerle, H.J.; von Eynatten, M.; Broedl, U.C. The effect of empagliflozin on arterial stiffness and heart rate variability in subjects with uncomplicated type 1 diabetes mellitus. Cardiovasc. Diabetol. 2014, 13, 28.
  3. Nassif, M.E.; Windsor, S.L.; Tang, F.; Khariton, Y.; Husain, M.; Inzucchi, S.E.; McGuire, D.K.; Pitt, B.; Scirica, B.M.; Austin, B.; et al. Dapagliflozin Effects on Biomarkers, Symptoms, and Functional Status in Patients with Heart Failure with Reduced Ejection Fraction: The DEFINE-HF Trial. Circulation 2019, 140, 1463–1476.
  4. Scheen, A.J. Effect of SGLT2 Inhibitors on the Sympathetic Nervous System and Blood Pressure. Curr. Cardiol. Rep. 2019, 21, 70.
  5. Sano, M. A new class of drugs for heart failure: SGLT2 inhibitors reduce sympathetic overactivity. J. Cardiol. 2018, 71, 471–476.
  6. Rahman, A.; Fujisawa, Y.; Nakano, D.; Hitomi, H.; Nishiyama, A. Effect of a selective SGLT2 inhibitor, luseogliflozin, on circadian rhythm of sympathetic nervous function and locomotor activities in metabolic syndrome rats. Clin. Exp. Pharmacol. Physiol. 2017, 44, 522–525.
  7. Herat, L.Y.; Magno, A.L.; Rudnicka, C.; Hricova, J.; Carnagarin, R.; Ward, N.C.; Arcambal, A.; Kiuchi, M.G.; Head, G.A.; Schlaich, M.P.; et al. SGLT2 Inhibitor-Induced Sympathoinhibition: A Novel Mechanism for Cardiorenal Protection. JACC Basic Transl. Sci. 2020, 5, 169–179.
  8. Shimizu, W.; Kubota, Y.; Hoshika, Y.; Mozawa, K.; Tara, S.; Tokita, Y.; Yodogawa, K.; Iwasaki, Y.K.; Yamamoto, T.; Takano, H.; et al. Effects of empagliflozin versus placebo on cardiac sympathetic activity in acute myocardial infarction patients with type 2 diabetes mellitus: The EMBODY trial. Cardiovasc. Diabetol. 2020, 19, 148.
  9. Onishi, A.; Fu, Y.; Patel, R.; Darshi, M.; Crespo-Masip, M.; Huang, W.; Song, P.; Freeman, B.; Kim, Y.C.; Soleimani, M.; et al. A role for tubular Na+/H+ exchanger NHE3 in the natriuretic effect of the SGLT2 inhibitor empagliflozin. Am. J. Physiol. Ren. Physiol. 2020, 319, F712–F728.
  10. Sano, M.; Goto, S. Possible Mechanism of Hematocrit Elevation by Sodium Glucose Cotransporter 2 Inhibitors and Associated Beneficial Renal and Cardiovascular Effects. Circulation 2019, 139, 1985–1987.
  11. Mazer, C.D.; Hare, G.M.T.; Connelly, P.W.; Gilbert, R.E.; Shehata, N.; Quan, A.; Teoh, H.; Leiter, L.A.; Zinman, B.; Jüni, P.; et al. Effect of Empagliflozin on Erythropoietin Levels, Iron Stores, and Red Blood Cell Morphology in Patients with Type 2 Diabetes Mellitus and Coronary Artery Disease. Circulation 2020, 141, 704–707.
  12. Swedberg, K.; Young, J.B.; Anand, I.S.; Cheng, S.; Desai, A.S.; Diaz, R.; Maggioni, A.P.; McMurray, J.J.; O’Connor, C.; Pfeffer, M.A.; et al. Treatment of anemia with darbepoetin alfa in systolic heart failure. N. Engl. J. Med. 2013, 368, 1210–1219.
  13. Cowie, M.R.; Fisher, M. SGLT2 inhibitors: Mechanisms of cardiovascular benefit beyond glycaemic control. Nat. Rev. Cardiol. 2020, 17, 761–772.
  14. Ferrannini, E.; Baldi, S.; Frascerra, S.; Astiarraga, B.; Heise, T.; Bizzotto, R.; Mari, A.; Pieber, T.R.; Muscelli, E. Shift to Fatty Substrate Utilization in Response to Sodium-Glucose Cotransporter 2 Inhibition in Subjects without Diabetes and Patients with Type 2 Diabetes. Diabetes 2016, 65, 1190–1195.
  15. Matsuura, T.R.; Puchalska, P.; Crawford, P.A.; Kelly, D.P. Ketones and the Heart: Metabolic Principles and Therapeutic Implications. Circ. Res. 2023, 132, 882–898.
  16. Lopaschuk, G.D.; Karwi, Q.G.; Tian, R.; Wende, A.R.; Abel, E.D. Cardiac Energy Metabolism in Heart Failure. Circ. Res. 2021, 128, 1487–1513.
  17. Murashige, D.; Jang, C.; Neinast, M.; Edwards, J.J.; Cowan, A.; Hyman, M.C.; Rabinowitz, J.D.; Frankel, D.S.; Arany, Z. Comprehensive quantification of fuel use by the failing and nonfailing human heart. Science 2020, 370, 364–368.
  18. Verma, S.; Rawat, S.; Ho, K.L.; Wagg, C.S.; Zhang, L.; Teoh, H.; Dyck, J.E.; Uddin, G.M.; Oudit, G.Y.; Mayoux, E.; et al. Empagliflozin Increases Cardiac Energy Production in Diabetes: Novel Translational Insights Into the Heart Failure Benefits of SGLT2 Inhibitors. JACC Basic Transl. Sci. 2018, 3, 575–587.
  19. Lopaschuk, G.D.; Verma, S. Mechanisms of Cardiovascular Benefits of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors: A State-of-the-Art Review. JACC Basic Transl. Sci. 2020, 5, 632–644.
  20. Youm, Y.H.; Nguyen, K.Y.; Grant, R.W.; Goldberg, E.L.; Bodogai, M.; Kim, D.; D’Agostino, D.; Planavsky, N.; Lupfer, C.; Kanneganti, T.D.; et al. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med. 2015, 21, 263–269.
  21. Dewanjee, S.; Vallamkondu, J.; Kalra, R.S.; John, A.; Reddy, P.H.; Kandimalla, R. Autophagy in the diabetic heart: A potential pharmacotherapeutic target in diabetic cardiomyopathy. Ageing Res. Rev. 2021, 68, 101338.
  22. Packer, M. Autophagy stimulation and intracellular sodium reduction as mediators of the cardioprotective effect of sodium-glucose cotransporter 2 inhibitors. Eur. J. Heart Fail. 2020, 22, 618–628.
  23. Adingupu, D.D.; Göpel, S.O.; Grönros, J.; Behrendt, M.; Sotak, M.; Miliotis, T.; Dahlqvist, U.; Gan, L.M.; Jönsson-Rylander, A.C. SGLT2 inhibition with empagliflozin improves coronary microvascular function and cardiac contractility in prediabetic ob/ob−/− mice. Cardiovasc. Diabetol. 2019, 18, 16.
  24. Suhrs, H.E.; Nilsson, M.; Bové, K.B.; Zander, M.; Prescott, E. Effect of empagliflozin on coronary microvascular function in patients with type 2 diabetes mellitus-A randomized, placebo-controlled cross-over study. PLoS ONE 2022, 17, e0263481.
  25. Leccisotti, L.; Cinti, F.; Sorice, G.P.; D’Amario, D.; Lorusso, M.; Guzzardi, M.A.; Mezza, T.; Gugliandolo, S.; Cocchi, C.; Capece, U.; et al. Dapagliflozin improves myocardial flow reserve in patients with type 2 diabetes: The DAPAHEART Trial: A preliminary report. Cardiovasc. Diabetol. 2022, 21, 173.
  26. Osto, E.; Bonacina, F.; Pirillo, A.; Norata, G.D. Neutral effect of SGLT2 inhibitors on lipoprotein metabolism: From clinical evidence to molecular mechanisms. Pharmacol. Res. 2023, 188, 106667.
  27. Sánchez-García, A.; Simental-Mendía, M.; Millán-Alanís, J.M.; Simental-Mendía, L.E. Effect of sodium-glucose co-transporter 2 inhibitors on lipid profile: A systematic review and meta-analysis of 48 randomized controlled trials. Pharmacol. Res. 2020, 160, 105068.
  28. Storgaard, H.; Gluud, L.L.; Bennett, C.; Grøndahl, M.F.; Christensen, M.B.; Knop, F.K.; Vilsbøll, T. Benefits and Harms of Sodium-Glucose Co-Transporter 2 Inhibitors in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0166125.
  29. Langslet, G.; Zinman, B.; Wanner, C.; Hantel, S.; Espadero, R.M.; Fitchett, D.; Johansen, O.E. Cardiovascular outcomes and LDL-cholesterol levels in EMPA-REG OUTCOME®. Diab. Vasc. Dis. Res. 2020, 17, 1479164120975256.
  30. Daud, E.; Ertracht, O.; Bandel, N.; Moady, G.; Shehadeh, M.; Reuveni, T.; Atar, S. The impact of empagliflozin on cardiac physiology and fibrosis early after myocardial infarction in non-diabetic rats. Cardiovasc. Diabetol. 2021, 20, 132.
  31. Shi, L.; Zhu, D.; Wang, S.; Jiang, A.; Li, F. Dapagliflozin Attenuates Cardiac Remodeling in Mice Model of Cardiac Pressure Overload. Am. J. Hypertens. 2019, 32, 452–459.
  32. Zhang, Y.; Lin, X.; Chu, Y.; Chen, X.; Du, H.; Zhang, H.; Xu, C.; Xie, H.; Ruan, Q.; Lin, J.; et al. Dapagliflozin: A sodium-glucose cotransporter 2 inhibitor, attenuates angiotensin II-induced cardiac fibrotic remodeling by regulating TGFβ1/Smad signaling. Cardiovasc. Diabetol. 2021, 20, 121.
  33. Salah, H.M.; Verma, S.; Santos-Gallego, C.G.; Bhatt, A.S.; Vaduganathan, M.; Khan, M.S.; Lopes, R.D.; Al’Aref, S.J.; McGuire, D.K.; Fudim, M. Sodium-Glucose Cotransporter 2 Inhibitors and Cardiac Remodeling. J. Cardiovasc. Transl. Res. 2022, 15, 944–956.
  34. Tian, J.; Zhang, M.; Suo, M.; Liu, D.; Wang, X.; Liu, M.; Pan, J.; Jin, T.; An, F. Dapagliflozin alleviates cardiac fibrosis through suppressing EndMT and fibroblast activation via AMPKα/TGF-β/Smad signalling in type 2 diabetic rats. J. Cell. Mol. Med. 2021, 25, 7642–7659.
  35. Pabel, S.; Hamdani, N.; Luedde, M.; Sossalla, S. SGLT2 Inhibitors and Their Mode of Action in Heart Failure-Has the Mystery Been Unravelled? Curr. Heart Fail. Rep. 2021, 18, 315–328.
  36. Verma, S.; Mazer, C.D.; Yan, A.T.; Mason, T.; Garg, V.; Teoh, H.; Zuo, F.; Quan, A.; Farkouh, M.E.; Fitchett, D.H.; et al. Effect of Empagliflozin on Left Ventricular Mass in Patients With Type 2 Diabetes Mellitus and Coronary Artery Disease: The EMPA-HEART CardioLink-6 Randomized Clinical Trial. Circulation 2019, 140, 1693–1702.
  37. Lee, M.M.Y.; Brooksbank, K.J.M.; Wetherall, K.; Mangion, K.; Roditi, G.; Campbell, R.T.; Berry, C.; Chong, V.; Coyle, L.; Docherty, K.F.; et al. Effect of Empagliflozin on Left Ventricular Volumes in Patients with Type 2 Diabetes, or Prediabetes, and Heart Failure with Reduced Ejection Fraction (SUGAR-DM-HF). Circulation 2021, 143, 516–525.
  38. Dhingra, N.K.; Mistry, N.; Puar, P.; Verma, R.; Anker, S.; Mazer, C.D.; Verma, S. SGLT2 inhibitors and cardiac remodelling: A systematic review and meta-analysis of randomized cardiac magnetic resonance imaging trials. ESC Heart Fail. 2021, 8, 4693–4700.
  39. Gamaza-Chulián, S.; Díaz-Retamino, E.; González-Testón, F.; Gaitero, J.C.; Castillo, M.J.; Alfaro, R.; Rodríguez, E.; González-Caballero, E.; Martín-Santana, A. Effect of sodium-glucose cotransporter 2 (SGLT2) inhibitors on left ventricular remodelling and longitudinal strain: A prospective observational study. BMC Cardiovasc. Disord. 2021, 21, 456.
  40. Tanaka, H.; Soga, F.; Tatsumi, K.; Mochizuki, Y.; Sano, H.; Toki, H.; Matsumoto, K.; Shite, J.; Takaoka, H.; Doi, T.; et al. Positive effect of dapagliflozin on left ventricular longitudinal function for type 2 diabetic mellitus patients with chronic heart failure. Cardiovasc. Diabetol. 2020, 19, 6.
  41. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Brunner-La Rocca, H.P.; Choi, D.J.; Chopra, V.; Chuquiure-Valenzuela, E.; et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021, 385, 1451–1461.
  42. Solomon, S.D.; McMurray, J.J.V.; Claggett, B.; de Boer, R.A.; De Mets, D.; Hernandez, A.F.; Inzucchi, S.E.; Kosiborod, M.N.; Lam, C.S.P.; Martinez, F.; et al. Dapagliflozin in Heart Failure with Mildly Reduced or Preserved Ejection Fraction. N. Engl. J. Med. 2022, 387, 1089–1098.
More
Video Production Service