Next Article in Journal
Cytokine Dynamics in Autism: Analysis of BMAC Therapy Outcomes
Previous Article in Journal
Rhodamine-Based Cyclic Hydroxamate as Fluorescent pH Probe for Imaging of Lysosomes
Previous Article in Special Issue
Improvement of Vascular Insulin Sensitivity by Ranolazine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

mTORC1 and SGLT2 Inhibitors—A Therapeutic Perspective for Diabetic Cardiomyopathy

1
Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
2
Division of Cardiology, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(20), 15078; https://doi.org/10.3390/ijms242015078
Submission received: 1 September 2023 / Revised: 27 September 2023 / Accepted: 4 October 2023 / Published: 11 October 2023
(This article belongs to the Special Issue Molecular Pharmacology in Diabetes 2.0)

Abstract

:
Diabetic cardiomyopathy is a critical diabetes-mediated co-morbidity characterized by cardiac dysfunction and heart failure, without predisposing hypertensive or atherosclerotic conditions. Metabolic insulin resistance, promoting hyperglycemia and hyperlipidemia, is the primary cause of diabetes-related disorders, but ambiguous tissue-specific insulin sensitivity has shed light on the importance of identifying a unified target paradigm for both the glycemic and non-glycemic context of type 2 diabetes (T2D). Several studies have indicated hyperactivation of the mammalian target of rapamycin (mTOR), specifically complex 1 (mTORC1), as a critical mediator of T2D pathophysiology by promoting insulin resistance, hyperlipidemia, inflammation, vasoconstriction, and stress. Moreover, mTORC1 inhibitors like rapamycin and their analogs have shown significant benefits in diabetes and related cardiac dysfunction. Recently, FDA-approved anti-hyperglycemic sodium–glucose co-transporter 2 inhibitors (SGLT2is) have gained therapeutic popularity for T2D and diabetic cardiomyopathy, even acknowledging the absence of SGLT2 channels in the heart. Recent studies have proposed SGLT2-independent drug mechanisms to ascertain their cardioprotective benefits by regulating sodium homeostasis and mimicking energy deprivation. In this review, we systematically discuss the role of mTORC1 as a unified, eminent target to treat T2D-mediated cardiac dysfunction and scrutinize whether SGLT2is can target mTORC1 signaling to benefit patients with diabetic cardiomyopathy. Further studies are warranted to establish the underlying cardioprotective mechanisms of SGLT2is under diabetic conditions, with selective inhibition of cardiac mTORC1 but the concomitant activation of mTORC2 (mTOR complex 2) signaling.

1. Diabetic Cardiomyopathy

Diabetes mellitus (DM) is a chronic metabolic disorder with a current prevalence of 573 million individuals worldwide, estimated to reach 643 million by 2030 and 783 million by 2045 [1]. Globally, around 18 million more men than women have diabetes [2] and the number of aged patients (>65 years) with DM is projected to reach ~276 million by 2045 as it is more prevalent in middle-aged and older adults [3]. DM ranks among the top 10 global mortality causes and is riddled with several health complications like cardiovascular dysfunction, leading to progressive heart failure. Cardiovascular diseases are the main cause of morbidity and account for two out of three overall deaths in diabetic patients [4], arising from conditions like hypertension, obesity, and dyslipidemia [5]. The concept of diabetes-related cardiomyopathy was first suggested in 1954 by Lundbæk [6]; later, in 1972, Rubler et al. reported the post-mortem findings of four diabetic patients who showed advanced symptoms of heart failure without any direct relation to congenital, valvular, or atherosclerotic heart conditions [7]. In 2013, to set a standard clinical perspective of diabetic cardiomyopathy, the American Heart Association [8] and the European Society of Cardiology [9] defined it as a pathophysiological condition of ventricular dysfunction in diabetic patients, without predisposing atherosclerosis and coronary artery disorders, or hypertensive, congenital, or valvular heart diseases.
DM can be broadly classified into type 1 diabetes mellitus (T1D) and type 2 diabetes mellitus (T2D). T1D, associated with autoimmune insulin deficiency, accounts for 5–10%, while T2D, characterized by insulin resistance, accounts for 90–95% of all diabetes cases [10]. Although cardiovascular complications are associated with both types of diabetes mellitus, the incidence of cardiac dysfunction in T1D and T2D patients has been reported to be 14.5% and 35%, respectively [11], making T2D-related cardiomyopathy the more prevalent pathophysiology.
The most common metabolic dysregulations in T2D, arising from insulin resistance [12], involve hyperglycemia and hyperlipidemia, which directly or indirectly provoke cardiac dysfunction in patients [13,14]. The heart shows reduced mitochondrial glucose oxidation due to insulin resistance along with excess fatty acid uptake, impaired mitochondrial fatty acid β-oxidation, and increased reactive oxygen species (ROS), resulting in lipotoxicity and cellular stress. These insults typically affect ventricular compliances with increased systemic pressure and manifest as impaired diastolic function with cardiac inflammation, abnormal calcium transport, and cardiac remodeling, which are linked to a slow progression into systolic dysfunction and ultimately heart failure [15,16,17,18] (Figure 1).
The systemic hyperglycemia, hyperlipidemia, oxidative stress, and inflammation associated with diabetes contribute to diabetic cardiomyopathy through several molecular pathways that provide significant therapeutic targets against diabetes-associated cardiac dysfunction. Some preclinical studies concerning diabetic cardiomyopathy have reported improved endothelial function, reduced cardiac fibrosis/hypertrophy, and reduced inflammation with metformin (via 5′ adenosine monophosphate-activated protein kinase, AMPK-dependent and AMPK-independent mechanisms) [19], tadalafil (phosphodiesterase 5, PDE5 inhibitor) [20], and MCC950 (nucleotide-binding oligomerization domain- leucine rich repeat- and pyrin domain-containing protein 3, NLRP3 inflammasome inhibitor) [21], respectively, while other preclinical studies with sulforaphane (nuclear factor erythroid 2-related factor 2, NRF2 activator) [22] and saxagliptin (dipeptidyl peptidase-4, DPP4 inhibitor) [23] showed cardioprotective benefits by ameliorating cardiac oxidative stress and lipotoxicity. Owing to these therapeutic targets, several clinical trials were conducted in the last decade with a focus on T2D patients (Table 1). However, these studies lacked an overall benefit curve in terms of both the glycemic and non-glycemic context of diabetic cardiomyopathy, emphasizing the critical need for identifying new molecular targets for the development of novel and more effective therapeutics.

2. Differential Role of mTORC1 and mTORC2 in Diabetic Cardiomyopathy

Although the central theme of T2D is highlighted as insulin resistance and hyperglycemia, several disorders in diabetes patients tend to be partially insulin responsive and therefore the glycemic aspect in the case of diabetes is just one part of a deranged metabolic network [34]. Obesity, hypertension, and cancers in diabetic patients have earlier been reported to reflect insulin-responsive pathology [35,36,37], thereby suggesting that insulin may promote, rather than benefit, non-glycemic disorders, particularly cardiovascular diseases [38], which account for the majority of mortality in patients with T2D [39]. In this context, it is crucial to recognize a target that links the glycemic and non-glycemic aspects of diabetes to effectively treat diabetic cardiomyopathy.
The mammalian target of rapamycin (mTOR), a member of the phosphatidylinositol 3-kinase (PI3K)-related protein kinase superfamily, is crucial for insulin and insulin-like growth factor 1 (IGF-1) signaling and plays an important role in cell growth, proliferation, autophagy, apoptosis, inflammation, and metabolism. There are two distinct mTOR complexes, termed mTORC1 and mTORC2, with a common core catalytic subunit that harbors specific mTOR-interacting units that designate specific cellular functions to these complexes. mTORC1 has three core components: regulatory subunit Raptor (regulatory-associated protein of mTOR), catalytic subunit mTOR, and mLST8/GβL (mammalian lethal with SEC13 protein 8/G protein beta subunit-like). Raptor helps in substrate recruitment to the complex and ensures proper subcellular localization. mLST8 associates with the catalytic domain and stabilizes the kinase activation loop. Besides these components, mTORC1 has two inhibitory subunits PRAS40 (proline-rich Akt substrate of 40 kDa) and DEPTOR (disheveled EGL-10 and pleckstrin (DEP)-domain containing mTOR-interacting protein). mTORC2 consists of mLST8 and the catalytic mTOR subunit, but Raptor is replaced by an analogous subunit Rictor (rapamycin-insensitive companion of mTOR). mTORC2 also contains the inhibitory DEPTOR subunit along with other regulatory subunits mSin1 and Protor1/2. mTORC1 is extensively involved in protein synthesis, nucleotide synthesis, lipid synthesis, autophagy, and mitochondrial biogenesis, while mTORC2 is associated with cell survival, apoptosis, cytoskeletal organization, and glucose metabolism (Figure 2). The mTOR complexes and their upstream/downstream signaling processes have been extensively discussed in several reviews with respect to various pathophysiologies [40,41,42,43].
In the glycemic context, insulin resistance, driven by mTORC1 hyperactivation-mediated deregulation of the insulin receptor (IR)-PI3K/Akt substrate (IRS) signaling axis, leads to elevated blood glucose levels (hyperglycemia). Hyperactivation of mTORC1 and its downstream S6K1 kinase (Ribosomal S6 kinase) phosphorylates IRS-1 at Ser307 and Ser636/639 to initiate IRS-1 degradation [44] and PI3K/Akt signaling suppression, thereby causing insulin resistance via the mTORC1/S6K1 negative feedback loop. mTORC1 also regulates insulin signaling via Grb10 (growth factor receptor-bound protein 10), which inhibits threonine phosphorylation of insulin/IGF receptors and blocks PI3K/Akt signaling [45], thereby disrupting the IRS axis leading to insulin resistance and increased hyperglycemia [46].
In the non-glycemic context, closely associated with diabetic cardiomyopathy and endothelial disorders, mTORC1 has been prompted as a key player. The mTORC1 pathway has been linked to cardiac hypertrophy and hypertension. An upregulated mTORC1/S6K1 activity contributes to deregulated insulin-stimulated vasodilation by suppressing eNOS (endothelial nitric oxide synthase), resulting in vasoconstriction and hypertension. Several reports of diabetic cardiomyopathy and heart failure also demonstrate upregulated mTORC1 activity, whereas studies concerning mTORC1 inhibitors (rapamycin and PRAS40) [47,48] and induced cardiac autophagy (inhibited by mTORC1) show beneficial effects in diabetic cardiac dysfunction, implying a pathogenic role of mTORC1 hyperactivation [49]. In T2D patients, ischemia and cardiomyopathy go hand-in-hand, leading to heart failure. The condition results in a fibrotic phenotype of the heart which initially faces ejection fraction-preserved diastolic dysfunction and develops systolic dysfunction in the later stages [50]. mTORC1 regulates ischemic injury conditions by preserving energy homeostasis, and, as per literature, Rheb (Ras homolog enriched in brain) inhibition, which subsequently inhibits mTORC1 and protects cardiomyocytes by activating autophagy during energy deprivation and ischemia [51]. Studies have indicated that AMPK inhibition in glucose-deprived and ischemia conditions, resulting in mTORC1 hyperactivation, lead to worsening of cardiac dysfunction and cardiomyocyte death [52], while AMPK activation attenuates pressure overload or diabetes-related cardiac remodeling [53,54]. Moreover, AMPK activators like resveratrol, berberine, and metformin have also been associated with cardiovascular benefits in T2D. Resveratrol has been linked to upregulated adiponectin levels to prevent myocardial ischemia injury in diabetic mice [55], whereas berberine studies in diabetic cardiomyopathic rats have showed attenuated hypertrophy via activated AMPK and reduced GSK3β (glycogen synthase kinase 3 beta) [56]. A recent study by Yang F. et al., in 2019, further emphasized the role of AMPK activators in diabetic cardiomyopathy benefit and reported metformin-mediated NLRP3 inflammasome inhibition via AMPK/mTOR pathway [57]. These reports cumulatively suggest a critical role of mTORC1 downregulation in diabetic cardiomyopathy benefits. Figure 3 depicts mTOR signaling and its hyperactivation in cardiomyocytes that contributes to hyperlipidemia, inflammation, and stress.
Ionic imbalance, including a state of calcium overload as well as increased intracellular sodium, is a key player in the development of cardiac dysfunction and characteristics of diabetic cardiomyopathy [58,59,60]. mTOR signaling is involved in diverse biological pathways by regulating ionic homeostasis, specifically by regulating the activity and expression of various Ca2+ channels [61,62,63]. Intertwined links between sarcoplasmic reticulum calcium homeostasis and mTORC1 signaling are critical for physiological and pathological cardiac hypertrophy [64,65]. Inhibition of mTORC1 with rapamycin induces Ca2+ release from lysosomes through the activation of two-pore segment channel 2, TPC2 [66]. The downregulation of the inward rectifier potassium (IK1) channel with intracellular Ca2+ overload is a hallmark in cardiac hypertrophy, interstitial fibrosis, and electrical remodeling and failure [67]. Specifically, mTORC1 regulates the lysoNaATP, which determines the sensitivity of endolysosome’s resting membrane potential to Na+ and cytosolic ATP as well as controls lysosomal pH stability and whole-body amino acid homeostasis [68,69]. Selective IK1 agonist attenuates cardiac remodeling by promoting autophagy via negatively regulating calcium-activated CaMKII and mTOR signaling [69]. The activation of IK1 channel protects the heart against myocardial ischemia-induced cardiac dysfunction by inhibiting mTOR-p70S6 signaling pathway [68]. mTOR also acutely controls endosomal and lysosomal functions through the endolysosomal ATP-sensitive Na+ channel (lysoNaATP) in response to changes under different nutrition status and metabolic conditions [62]. mTORC1 regulates the endolysosomal ATP-sensitive Na+ channel (lysoNaATP), which determines the sensitivity of endolysosome’s resting membrane potential to Na+ and cytosolic ATP as well as controls lysosomal pH stability and whole-body amino acid homeostasis. Under nutrient deprivation, mTORC1 interacts with lysosomal TPC2 and regulates authophagy [62,70].
From a therapeutic perspective, direct and indirect mTORC1 inhibitors have been broadly used for treating T2D and co-existing diabetic cardiac conditions. Metformin is a widely used, FDA-approved anti-diabetes drug that regulates mTORC1 via mitochondrial complex I-mediated AMPK activation [71] or AMPK-independent Rag GTPase inhibition [72]. Recent preclinical reports on metformin have shown attenuated myocardial hypertrophy [73] and reduced inflammation [57] in diabetic animal models but earlier clinical trials with metformin, as an overall cardioprotective drug in diabetic patients, are inconspicuous [19]. A 2014 study by Mirko Volkers et al. on PRAS40, a direct mTORC1 inhibitor, reported diabetic cardiomyopathy prevention besides improved hepatic insulin sensitivity in a diabetic mouse model [74] but further PRAS40 studies in diabetic cardiac dysfunction are yet to emerge.
Rapamycin, another direct mTORC1 inhibitor, has been closely linked to improved T2D and diabetic cardiac dysfunction [75,76,77], but multiple studies have reported controversial effects of chronic treatment with rapamycin [78,79]. Studies with analogs of rapamycin or rapalogs, like everolimus, have shown beneficial effects and an improvement in glucose metabolism in diabetes by disrupting the mTORC1/S6K1 feedback loop [80], but similar to rapamycin, chronic treatment with rapalogs has shown detrimental effects in T2D patients [78]. These poor outcomes of chronic rapamycin treatment might involve the inhibition of mTORC2 activity [81]. However, chronic treatment with a sub-clinical dose (0.25 mg/kg/day) of rapamycin or nano-formulated micelles of rapamycin, rapatar, has ameliorated the metabolic status of diabetic mice, with an improvement in cardiac function by preferentially inhibiting mTORC1 [75,82]. Our studies on rapamycin also reported mTORC2 activation in diabetic mice and rabbits, which might be associated with improved cardiac function and reduced myocardial infarction following ischemia-reperfusion injury [83,84]. A 2014 study using an ischemia-reperfusion injury mouse model suggested that miR-144 improves cardioprotection via suppressing mTORC1 and simultaneously activating mTORC2 [85], while another study implicated the role of mTORC2 in preserving cardiac function in pressure-overload hypertrophy [86], therefore highlighting mTORC1 inhibition, alongside mTORC2 activation, as crucial mechanisms to consider in diabetic cardiomyopathy treatment.

3. Sodium and Glucose Co-Transporter Inhibitors (SGLT2is)—Do They Regulate mTORC1 in Diabetic Cardiomyopathy?

Despite the boom in preclinical and clinical studies on diabetic cardiomyopathy, its pathogenesis and unified paradigm remain unclear for devising specific therapeutic strategies. As a result, a plethora of research is still based on figuring out the most effective therapeutic approach to treating diabetes, diabetic cardiomyopathy, and related heart failure. To fill this critical gap in therapeutics, mTORC1 might be a key target for diabetic therapeutics, which also addresses other co-morbidities like cardiomyopathy in a glycemic and non-glycemic context. In T2D, metformin is usually the first-line standard treatment [87] and though a few studies have shown metformin to exert cardioprotective effects via mTORC1 regulation [57], these reports are very limited despite a long history of metformin use [88]. Recently, sodium–glucose co-transporter 2 inhibitors (SGLT2i), a new class of FDA-approved [89] anti-diabetic drugs, have shown a promising risk reduction of cardiomyopathy and other cardiovascular diseases in patients with T2D [90,91], owing to the overexpression of SGLTs in diabetes mellitus [92].
Glucose homeostasis, a crucial diabetes parameter associated with mTORC1 [93], is actively regulated by transporters like GLUTs and SGLTs that mediate D-glucose transport. While SLC2 genes encode for facilitated glucose diffusion transporters GLUT, sodium glucose co-transporters SGLT1-5 are encoded by SLC5. SGLT transports glucose into cells via Na+/K+-ATPase pump gradient and two major SGLT isoforms are SGLT1 and SGLT2. SGLT1 is expressed in the small intestine, kidneys, brain, and heart, while SGLT2 is expressed in kidney and pancreatic β cells. SGLT1 primarily acts as rate limiting intestinal glucose absorption whereas SGLT2 manages bulk glucose reabsorption in the kidneys [94]. In the kidneys, SGLT2 in the S1/S2 segment of the convoluted proximal tubule in kidney nephrons reabsorbs 90% of the glomerular filtrate glucose, aided by a positive sodium gradient [95], while SGLT1 reabsorbs the remaining 10% in the S3 segment of the proximal tubule [96]. The reabsorbed glucose in the tubular epithelial cells is flushed back into circulation through GLUT2 and this whole unidirectional transport of glucose is coupled to and regulated by the Na+K+ ATPase pump on the basolateral side of the cells [97]. SGLT2is primarily work as anti-hyperglycemic agents by blocking the SGLT2 channel and preventing glucose reabsorption, thus preventing hyperglycemia in diabetes mellitus. Preclinical studies with phlorizin, the first SGLT2 inhibitor, in the 19th century, improved insulin sensitivity in diabetic rat models but did not have any scope for oral bioavailability and showed adverse gastrointestinal concerns [98]. In 1990, T-1095, a phlorizin derivative, was developed as the first orally available SGLT2 inhibitor but was discontinued after phase II clinical trials owing to its non-selective nature and safety concerns [98]. In current therapeutic use, the common FDA-approved SGLT2is are empagliflozin, dapagliflozin, and canagliflozin, which have demonstrated cardiorenal benefits in diabetic patients [99,100].
Over the years, besides significant glucose-lowering efficacy, SGLT2is have also shown remarkable cardiovascular benefits in renal-impaired patients with lower glomerular filtration rates, indicating a major role of SGLT2is in promoting diabetic cardiac dysfunction benefits [101]. Table 2 provides a summary of SGLT2i clinical trials (phase III) in assessing cardiac dysfunction and heart failure with diabetes, while current trials are also focusing on SGLT2is in heart failure mediated by other metabolic insults like obesity [95,102]. A large multinational observational cohort study on T2D patients, CVD-REAL, associated the early initiation of SGLT2is with a lower risk of heart failure, myocardial infarction, and stroke, compared to other glucose-lowering drugs [100,103,104]. The EMPA-REG trial [100,105] in T2D patients with established cardiovascular risks showed a reduction in cardiovascular-related deaths with empagliflozin, while the DAPA-HF trial [106] in diabetic/non-diabetic patients with heart failure reported that dapagliflozin reduces the worsening of cardiovascular risk and heart failure by 26%, regardless of T2D status. These clinical trials suggested a moderate/no risk of genital infections with SGLT2i treatment but, interestingly, amputation risks were closely associated with SGLT2is like canagliflozin and ertugliflozin, which have a higher degree of non-selectivity towards SGLT2. Compared to other anti-diabetic drugs like DPP4i, safety analyses of SGLT2i treatment indicate a higher risk of genital infections, urinary tract infections, hypertension, and diabetic ketoacidosis, and a lower risk of acute kidney injury and decreased bone mineral density [107]. Other clinical trials concerning diabetic cardiomyopathy like the EMPEROR-Reduced trial [108] of SGLT2-selective empagliflozin also show reduced cardiovascular risks and heart failure in diabetic and non-diabetic patients with uncomplicated adverse genital infections, thus strongly suggesting that SGLT2is work in both glycemic and non-glycemic contexts. Therefore, deciphering the mechanism of SGLT2is and whether it concerns mTORC1 regulation in terms of cardiac dysfunction is crucial to understand its therapeutic role in diabetic cardiomyopathy.
Although SGLT2is have shown interesting benefits in terms of cardiac function with or without T2D, the expression of SGLT2 channels is negligible in the heart, further highlighting SGLT2-independent functions of these inhibitors in diabetic cardiomyopathy and cardiac dysfunction [134,135]. The presence of SGLT1 in the heart is well established and is reportedly overexpressed in T2DM patients [136]. Along with glucose transporters (GLUT), SGLT1 is involved in cardiomyocyte glucose uptake, and in ischemic conditions, SGLT1 is reported to supplement the ATP reserve by increasing glucose utilization, thus playing a role in myocardial energy metabolism [137,138]. Several clinical trials with SGLT2is have emphasized cardiac benefits, but most of these drugs show a variable affinity for SGLT1. To provide some evidence on specificity, Kondo et al. reported that non-selective SGLT2is, like canagliflozin, can mediate anti-inflammatory and anti-apoptotic effects, which are related to SGLT1-binding-mediated improved NOS coupling in cardiomyocytes, thus indicating SGLT1 inhibition by SGLT2is [138,139]. Studies on dapagliflozin acting as a SGLT1/2 dual inhibitor, also suggest the involvement of myocardial SGLT1 in mediating SGLT2i effects [140]. Some researchers therefore emphasize that SGLT1/2 dual inhibitors provide greater cardiac benefit and heart failure prevention compared to selective SGLT2is [141], but the role of SGLT1 inhibition in terms of cardiovascular benefits has a fair share of contradictory reports. While some studies reported reduced heart failure incidence [142] and enhanced AMPK [139] with SGLT1 inhibition, others showed that AMPK activation resulted in increased SGLT1 expression, which can lead to hypertrophy and ischemia [143]. To bypass these contradictions, trials to treat diabetic cardiomyopathy or cardiac dysfunction (without diabetes) use empagliflozin, which is more SGLT2-selective as compared to canagliflozin and dapagliflozin, thus limiting the chances of SGLT1 channel cross-targeting [144,145,146]. For such selective SGLT2i scenarios, off-target effects on GLUT receptors might be involved. While some researchers have hypothesized that the SGLT2i-GLUT binding inhibits glucose uptake in cardiac tissue in an SGLT1/2-independent manner [147] and empagliflozin has been proposed to dock on GLUT1 and GLUT4 [148], confirmed research is yet to be documented in terms of cardiac pathophysiology.
Amidst some theories to explain the role of SGLT2is in cardiac function that involves the renin–angiotensin–aldosterone system (RAAS) and diuretic pathways [149], a prominent hypothesis to support the role of SGLT2is in the heart concerns the involvement of sodium ion homeostasis [150]. Cardiac Na+ and Ca2+ homeostasis plays a major role in maintaining heart physiology, rhythm, and contraction [151]. Increased intracellular sodium (Nai+) due to hyperactive sodium hydrogen exchanger (NHE) is well known in cardiac dysfunction pathologies and leads to elevated Ca2+ efflux from the mitochondria, resulting in oxidative stress and deteriorated cellular function [152,153]. Besides variable degrees of cross-reactivity with SGLT1 [154], several reports on SGLT2is have demonstrated non-SGLT2 cardiac-based off-target effects in reducing ventricular myocyte systolic Ca2+ and lowering myocardial cytoplasmic Na+/Ca2+ via NHE regulation [155]. A reduction in myocardial Nai+ via the inhibition of Na+/H+ or Na+/Ca2+ exchangers has been reported to improve cardiac hypertrophy and heart failure [156,157]. Preclinical studies have shown that SGLT2is can directly bind and reduce NHE activity in cardiomyocytes, leading to decreased Nai+ and restored Ca2+ homeostasis, resulting in improved cardiac function and antioxidative capacity of cardiomyocytes [158,159], but the observations are not consistent [148,160]. Baartscheer et al. showed direct effects of empagliflozin on Na+ and Ca2+ alteration, independent of SGLT2 binding in isolated ventricular cardiomyocytes, thus indicating a similar role of empagliflozin to that of NHE inhibitors [158]. SGLT2is like empagliflozin and dapagliflozin have also been implicated in enhanced sarcoplasmic endoplasmic reticulum Ca2+-ATPase (SERCA2a) activity, which improves cardiac contractility via sarcoplasmic reticulum Ca2+ reuptake [161,162]. Under normal physiological conditions, the Na+/H+ exchanger pumps Na+ inside the cell and H+ outside the cell to maintain ionic homeostasis but in case of prolonged NHE activation during T2D, excess Nai+ increases sodium–calcium exchanger (NCX)-mediated intracellular Ca2i+, leading to oxidative stress and an acidic intracellular environment [163,164]. Therefore, by reducing NHE hyperactivity, SGLT2is can promote an alkaline intracellular environment. A recent bioRxiv preprint study by Kazyken and colleagues reported that alkaline intracellular pH can activate the AMPK/mTORC2 pathway and inhibit mTORC1 activity [165]. This links the role of SGLT2is to AMPK activation and subsequent mTORC2 activity via NHE-mediated pH homeostasis and Cai2+ modulation [64].
From another perspective, SGLT2 inhibitors have been shown in some studies to promote ketogenesis, lipid oxidation, and erythrocytosis, which can reflect a fasting-like transcriptional paradigm by mimicking nutrient deprivation and hypoxia [166] but can also be responsible for the moderate adverse effects in SGLT2i clinical trials (Table 2). Although systemic glucose lowering or ketogenesis by SGLT2is in vivo can modulate a starvation and nutrient deprivation environment, a possible GLUT inhibition by selective SGLT2is might be responsible for glucose deprivation in isolated cardiomyocytes. A depletion in the glucose environment caused by SGLT2is can reduce the ATP/ADP ratio, leading to increased AMP that stimulates the phosphorylation of AMPK and subsequently, phosphorylates GAPDH to activate SIRT1 [167]. AMPK (nutrient sensor) and SIRT1 (redox rheostat) activation help cardiomyocytes to adapt in response to SGLT2i-mediated nutrient-deprived conditions. AMPK and SIRT1 activation have been reported to negatively regulate mTORC1 in a Tsc1/2-dependent manner [49,168], thus indicating that SGLT2is indirectly inactivate mTORC1 by mimicking a nutrition-deprivation setting. Furthermore, a recent study of SGLT2is in obesity-related diabetic cardiomyopathy reported sestrin2-mediated AMPK activation/mTORC1 inactivation in cardiomyocytes upon empagliflozin treatment [169]. Some studies have reported energy deprivation as a cause for sestrin2 activation [170], which can be correlated with AMPK/mTORC2 activation besides inhibiting mTORC1 and promoting autophagy [171]. This adds another revelation to the role of SGLT2is in attenuating diabetic cardiomyopathy conditions by promoting an energy-deprived environment. A recent study by Zhang et al. in 2023 suggested that empagliflozin significantly reduced diabetic cardiomyopathy by promoting branched-chain amino acid catabolism, inhibiting mTORC1/p-ULK1, and reactivating autophagy [172], while another study by Feng et al. reported that dapagliflozin prevented cardiac dysfunction in diabetic rats by restoring autophagy by repressing mTORC1 and activating AMPK [173]. Figure 4 shows a summary of SGLT2 inhibitor mechanisms to target renal glucose absorption (canonical) and cardiac mTORC1 signaling (non-canonical) in diabetic cardiomyopathy.
SGLT2is have been broadly assertive in terms of cardiac benefits via potential mechanisms of anti-inflammation, oxidative stress reduction, and apoptosis prevention [174]. Although the cardioprotective effects of SGLT2is were previously regarded as glucose-lowering systemic effects, current research points out several direct cardiac effects of SGLT2is and mTORC1 might be the missing link. While Leet et al. reported that dapagliflozin reduced inflammatory cytokines IL-6/IL-1β and superoxide levels in a myocardial infarction model [175], Shi X et al. showed attenuation of pro-inflammatory cyclooxygenase-2 and IL-1β in a heart failure model [176]. Taking a step deeper into the mechanism, Ye Y et al. showed that dapagliflozin reduced diabetic-induced activation of cardiac nucleotide-binding oligomerization domain-like receptor 3 (NLRP3) inflammasome and the subsequent stimulation of pro-inflammatory cytokine production, which are associated with T2DM cardiac inflammation [177]. The researchers also found that dapagliflozin reduced apoptosis speck-like protein containing a caspase recruitment domain (ASC) and IL-1β in cardiofibroblasts in vitro, indicating that these effects are not SGLT2-related or glycemic. Besides inflammation, oxidative stress is a major player in diabetic cardiomyopathy and cardiac hypertrophy [178]. High doses of empagliflozin have been reported in some studies to reduce cardiac superoxide levels, advanced glycation end products (AGE), and AGE receptors (RAGE) in diabetic female rodent models [179], while dapaglifozin in myocardial infarction models has been implicated as an antioxidant modulator through direct reactive oxygen and nitrogen species (RONS)-dependent STAT3 signaling, independent of SGLT2-binding or glucose-lowering anti-diabetic effects [175]. Endoplasmic reticulum stress pathway (ERS)-mediated cardiac apoptosis via ROS is also a prominent pathological condition in diabetic cardiomyopathy and studies on empagliflozin have reported decreased ERS-associated caspase-12 [180]. However, some studies on different SGLT2i dosages show no anti-apoptotic benefit [162], making the evidence inconclusive. Although more studies are required to conclude the direct role of SGLT2is in NLRP3, ROS, and ERS-associated signaling, SGLT2i-mediated mTORC1 regulation can be hypothesized as the medium. Several reports indicate that mTORC1 activation induces NLRP3 inflammasome, while rapamycin and AMPK activation can inhibit mTOR/NLRP3 [57,181]. Moreover, literature reports also establish AMPK-mediated STAT3 inhibition via attenuation of JAK signaling and activation of redox-regulating NRF2 [182]. Therefore, aside from the systemic glucose-lowering advantage of SGLT2i cardioprotection, mTORC1 regulation by selective SGLT2is can also explain the direct mechanism of documented SGLT2i action in cardiac tissue to prevent cardiac dysfunction in diabetic cardiomyopathy patients.

4. Future Directions

With the advent of SGLT2is as potential anti-diabetes drugs with independent cardioprotective effects, several preclinical comparative studies with existing FDA-approved drugs, like metformin, sulfonylurea, DPP4i-inhibitors, and GLP-1 agonists, have come to the forefront. Sulfonylureas are the oldest form of anti-diabetic drug which stimulate insulin from pancreatic beta cells [183], while metformin is regarded as the first line of diabetic drugs, which reduces glucose production in the liver and enhances insulin sensitivity [184]. DPP4i and GLP-1 agonists both work by stimulating insulin secretion after an oral glucose load via incretin effect [185,186]. An observational multidatabase cohort study reported reduced myocardial infarction, stroke, and heart failure (MACEs—major adverse cardiovascular events) with SGLT2is as compared to DPP4is [187], whereas another database study concerning SGLT2is vs. metformin reported a trend of decreased heart failure hospitalizations and mortality events with SGLT2is in T2D patients [188]. Other cohort studies in terms of combination therapy showed SGLT2i–metformin to have a reduced all-cause mortality risk compared to SGLT2i monotherapy or sulfonylurea–metformin [189]. Recently, several observational comparative studies are focusing on SGLT2i and GLP-1 agonists as they are the first anti-diabetic drugs to demonstrate definite direct cardiac benefits with a reduction in glycated hemoglobin level in T2DM [190]. In a meta-analysis of several cardiovascular outcome trials regarding SGLT2i and GLP-1 agonists, Zelniker et al. found that MACE reduction was restricted to SGLT2i-administered patients with established atherosclerosis [191], while Wright et al. demonstrated that both SGLT2i monotherapy and SGLT2–GLP-1 agonist combination therapy may have a beneficial primary MACE risk reduction [192]. Emphasizing mTORC1 inhibition/mTORC2 activation as our proposed key to cardiovascular improvements in diabetic cardiomyopathy, several reports of GLP-1 agonists show mTORC2 activation [193] besides SGLT2i-mediated mTORC1 inhibition, making their combination therapy ideal, but there are limitations of increased hypoglycemia risk [194]. To date, there are no randomized controlled trials that compare SGLT2is with GLP-1 agonists head-to-head in diabetic cardiomyopathy and hence the current inconsistent meta-analysis-based interpretations have their limitations, thus demanding further preclinical and clinical studies in the future.
Besides diving into a deeper cardiovascular understanding of SGLT2is, there is a dire need to focus on the adverse effects of SGLT2i treatment in diabetic patients. The association of SGLT2i treatment with genital infections has been reported in several trials like EMPA-REG [109], DECLARE-TIMI [118], and EMPEROR-Reduced [130], which might be due to higher glucose concentrations in the urine, which promote bacterial growth [195,196], but the statistical data are inconclusive. Currently, a one-year observational study is recruiting female T2DM patients taking empagliflozin or dapagliflozin to correlate SGLT2is with urinary tract infections [197], but further studies are required to document statistical significance and mitigate any severe adverse effects of SGLT2i monotherapy or combination therapy.
With several completed and ongoing clinical trials, SGLT2 inhibitors are rapidly emerging as the miracle anti-diabetic drug. Along with beneficial renal outcomes and reduced kidney insults in T2DM patients, SGLT2is have also galvanized their position as a potential treatment candidate for diabetic cardiomyopathy and other cardiac disorders owing to their direct SGLT2-independent cardiac effects, thus encouraging further applications of SGLT2is in other metabolic co-morbidities like non-alcoholic liver steatohepatitis (NASH), with or without diabetes/obesity. A placebo-controlled interventional phase II study (LEGEND) by Inventiva Pharma is currently recruiting participants to compare the effects of lanifibranor (a pan-peroxisome proliferator-activated receptor agonist) monotherapy and lanifibronor–empagliflozin combination therapy in patients with NASH and T2DM [198], whereas another phase IV interventional clinical trial is underway to assess the effect of empagliflozin on fatty liver in non-diabetic patients [199]. With several other preclinical and clinical trials [200,201,202] lined up to assess the potential of SGLT2 inhibitors in various metabolic pathophysiologies, our review presents a crucial molecular perspective of SGLT2is’ mechanism of action mediated by mTOR complexes. A better understanding of how off-target SGLT2i effects can regulate mTOR complexes might be the stairway to repurposing these miracle drugs in the near future.

5. Conclusions

Cardiovascular diseases like diabetic cardiomyopathy are critical co-manifestations in patients with diabetes mellitus and several medical approaches are currently targeting diabetes with a significant consideration for treatments that also improve cardiac dysfunction and cardiomyopathy. A thorough analysis of T2D and its co-morbidities, in both glycemic and non-glycemic contexts, translates into mTORC1 being a unified therapeutic target. Besides the standard metformin treatment for diabetes, SGLT2 inhibitors have come up recently as potential anti-diabetic drugs that show very promising cardiovascular protection. Although the mechanisms of the cardiac effects of SGLT2is are still being explored, the existing hypotheses consistently point to mTORC1 regulation via ionic dyshomeostasis/stress and mimics of nutrient deprivation. While the role of SGLT2is in downregulating mTORC1 is very critical for cardioprotective effects in diabetic patients, owing to the evidence of improved cardiac function by mTORC1-inhibitor rapamycin, all roads do not lead to Rome; several studies have indicated that a fine balance between mTORC2 activation and mTORC1 inhibition is optimal for cardiac benefits in patients with/without diabetes. An in-depth understanding of off-target SGLT2i effects might open up novel treatment regimes with SGLT2is in several other cardiac, renal, pancreatic, cerebral, and hepatic pathologies as a single drug or combination therapy. With further clinical progress in exploring the mechanisms of how SGLT2is regulate cell signaling, drug modifications might also help in bypassing the adverse effects of genital infections, thus ameliorating the standard of patient care. Therefore, further research is pivotal to understand novel mechanisms of SGLT2is in the non-glycemic SGLT2-independent context besides shedding light on the role of SGLT2is in regulating the mTORC1/mTORC2 switch for a holistic approach towards diabetic cardiomyopathy and related metabolic disorders.

Author Contributions

S.S. and A.D. participated in original draft preparation of the article and A.D. revisited it critically for important intellectual content. A.D., C.D.G. and X.F. reviewed and edited the document. All authors have read and agreed to the published version of the manuscript.

Funding

The authors were supported in part by grants from the National Institutes of Health RO1HL134366 and RO1HL158951 (A.D.), R01CA266124 (C.D.G.), R01CA229812, Astar Biotech Research Award, and VA Merit Award 1I01BX005992-01A (X.F.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. IDF Diabetes Atlas 2021|IDF Diabetes Atlas. Available online: https://diabetesatlas.org/atlas/tenth-edition/ (accessed on 21 April 2023).
  2. Kautzky-Willer, A.; Leutner, M.; Harreiter, J. Sex Differences in Type 2 Diabetes. Diabetologia 2023, 66, 986–1002. [Google Scholar] [CrossRef] [PubMed]
  3. Francisco, P.M.S.B.; de Assumpção, D.; Bacurau, A.G.d.M.; da Silva, D.S.M.; Yassuda, M.S.; Borim, F.S.A. Diabetes Mellitus in Older Adults, Prevalence and Incidence: Results of the FIBRA Study. Rev. Bras. Geriatr. Gerontol. 2022, 25, e210203. [Google Scholar] [CrossRef]
  4. Tancredi, M.; Rosengren, A.; Svensson, A.-M.; Kosiborod, M.; Pivodic, A.; Gudbjörnsdottir, S.; Wedel, H.; Clements, M.; Dahlqvist, S.; Lind, M. Excess Mortality among Persons with Type 2 Diabetes. N. Engl. J. Med. 2015, 373, 1720–1732. [Google Scholar] [CrossRef] [PubMed]
  5. Kannel, W.B.; McGee, D.L. Diabetes and Cardiovascular Disease. The Framingham Study. JAMA 1979, 241, 2035–2038. [Google Scholar] [CrossRef]
  6. Lundbaek, K. Diabetic Angiopathy: A Specific Vascular Disease. Lancet 1954, 266, 377–379. [Google Scholar] [CrossRef]
  7. Rubler, S.; Dlugash, J.; Yuceoglu, Y.Z.; Kumral, T.; Branwood, A.W.; Grishman, A. New Type of Cardiomyopathy Associated with Diabetic Glomerulosclerosis. Am. J. Cardiol. 1972, 30, 595–602. [Google Scholar] [CrossRef]
  8. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E.; Drazner, M.H.; Fonarow, G.C.; Geraci, S.A.; Horwich, T.; Januzzi, J.L.; et al. 2013 ACCF/AHA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2013, 62, e147–e239. [Google Scholar] [CrossRef]
  9. Authors/Task Force Members; Rydén, L.; Grant, P.J.; Anker, S.D.; Berne, C.; Cosentino, F.; Danchin, N.; Deaton, C.; Escaned, J.; Hammes, H.-P.; et al. ESC Guidelines on Diabetes, Pre-Diabetes, and Cardiovascular Diseases Developed in Collaboration with the EASD: The Task Force on Diabetes, Pre-Diabetes, and Cardiovascular Diseases of the European Society of Cardiology (ESC) and Developed in Collaboration with the European Association for the Study of Diabetes (EASD). Eur. Heart J. 2013, 34, 3035–3087. [Google Scholar] [CrossRef]
  10. Goyal, R.; Jialal, I. Type 2 Diabetes. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  11. Tan, Y.; Zhang, Z.; Zheng, C.; Wintergerst, K.A.; Keller, B.B.; Cai, L. Mechanisms of Diabetic Cardiomyopathy and Potential Therapeutic Strategies: Preclinical and Clinical Evidence. Nat. Rev. Cardiol. 2020, 17, 585–607. [Google Scholar] [CrossRef]
  12. Taylor, R. Insulin Resistance and Type 2 Diabetes. Diabetes 2012, 61, 778–779. [Google Scholar] [CrossRef]
  13. Riehle, C.; Bauersachs, J. Of Mice and Men: Models and Mechanisms of Diabetic Cardiomyopathy. Basic Res. Cardiol. 2018, 114, 2. [Google Scholar] [CrossRef] [PubMed]
  14. Hölscher, M.E.; Bode, C.; Bugger, H. Diabetic Cardiomyopathy: Does the Type of Diabetes Matter? Int. J. Mol. Sci. 2016, 17, 2136. [Google Scholar] [CrossRef] [PubMed]
  15. Lebeche, D.; Davidoff, A.J.; Hajjar, R.J. Interplay between Impaired Calcium Regulation and Insulin Signaling Abnormalities in Diabetic Cardiomyopathy. Nat. Clin. Pract. Cardiovasc. Med. 2008, 5, 715–724. [Google Scholar] [CrossRef]
  16. Buchanan, J.; Mazumder, P.K.; Hu, P.; Chakrabarti, G.; Roberts, M.W.; Yun, U.J.; Cooksey, R.C.; Litwin, S.E.; Abel, E.D. Reduced Cardiac Efficiency and Altered Substrate Metabolism Precedes the Onset of Hyperglycemia and Contractile Dysfunction in Two Mouse Models of Insulin Resistance and Obesity. Endocrinology 2005, 146, 5341–5349. [Google Scholar] [CrossRef] [PubMed]
  17. Bonen, A.; Jain, S.S.; Snook, L.A.; Han, X.-X.; Yoshida, Y.; Buddo, K.H.; Lally, J.S.; Pask, E.D.; Paglialunga, S.; Beaudoin, M.-S.; et al. Extremely Rapid Increase in Fatty Acid Transport and Intramyocellular Lipid Accumulation but Markedly Delayed Insulin Resistance after High Fat Feeding in Rats. Diabetologia 2015, 58, 2381–2391. [Google Scholar] [CrossRef] [PubMed]
  18. Jia, G.; Whaley-Connell, A.; Sowers, J.R. Diabetic Cardiomyopathy: A Hyperglycaemia- and Insulin-Resistance-Induced Heart Disease. Diabetologia 2018, 61, 21–28. [Google Scholar] [CrossRef]
  19. Nesti, L.; Natali, A. Metformin Effects on the Heart and the Cardiovascular System: A Review of Experimental and Clinical Data. Nutr. Metab. Cardiovasc. Dis. 2017, 27, 657–669. [Google Scholar] [CrossRef]
  20. Koka, S.; Das, A.; Salloum, F.N.; Kukreja, R.C. Phosphodiesterase-5 Inhibitor Tadalafil Attenuates Oxidative Stress and Protects against Myocardial Ischemia/Reperfusion Injury in Type 2 Diabetic Mice. Free Radic. Biol. Med. 2013, 60, 80–88. [Google Scholar] [CrossRef]
  21. Pavillard, L.E.; Cañadas-Lozano, D.; Alcocer-Gómez, E.; Marín-Aguilar, F.; Pereira, S.; Robertson, A.A.B.; Muntané, J.; Ryffel, B.; Cooper, M.A.; Quiles, J.L.; et al. NLRP3-Inflammasome Inhibition Prevents High Fat and High Sugar Diets-Induced Heart Damage through Autophagy Induction. Oncotarget 2017, 8, 99740–99756. [Google Scholar] [CrossRef]
  22. Gu, J.; Cheng, Y.; Wu, H.; Kong, L.; Wang, S.; Xu, Z.; Zhang, Z.; Tan, Y.; Keller, B.B.; Zhou, H.; et al. Metallothionein Is Downstream of Nrf2 and Partially Mediates Sulforaphane Prevention of Diabetic Cardiomyopathy. Diabetes 2017, 66, 529–542. [Google Scholar] [CrossRef]
  23. Wu, L.; Wang, K.; Wang, W.; Wen, Z.; Wang, P.; Liu, L.; Wang, D.W. Glucagon-like Peptide-1 Ameliorates Cardiac Lipotoxicity in Diabetic Cardiomyopathy via the PPARα Pathway. Aging Cell 2018, 17, e12763. [Google Scholar] [CrossRef] [PubMed]
  24. National Heart, Lung, and Blood Institute (NHLBI). Action to Control Cardiovascular Risk in Diabetes (ACCORD); National Library of Medicine: Bethesda, MD, USA, 2016; clinicaltrials.gov.
  25. Margolis, K.L.; O’Connor, P.J.; Morgan, T.M.; Buse, J.B.; Cohen, R.M.; Cushman, W.C.; Cutler, J.A.; Evans, G.W.; Gerstein, H.C.; Grimm, R.H.; et al. Outcomes of Combined Cardiovascular Risk Factor Management Strategies in Type 2 Diabetes: The ACCORD Randomized Trial. Diabetes Care 2014, 37, 1721–1728. [Google Scholar] [CrossRef] [PubMed]
  26. Isidori, A.M. Cardiovascular Effects of Chronic Sildenafil (Viagra) Treatment in Diabetic Subjects with Endothelial Dysfunction; National Library of Medicine: Bethesda, MD, USA, 2013; clinicaltrials.gov.
  27. Giannetta, E.; Isidori, A.M.; Galea, N.; Carbone, I.; Mandosi, E.; Vizza, C.D.; Naro, F.; Morano, S.; Fedele, F.; Lenzi, A. Chronic Inhibition of cGMP Phosphodiesterase 5A Improves Diabetic Cardiomyopathy. Circulation 2012, 125, 2323–2333. [Google Scholar] [CrossRef] [PubMed]
  28. Novo Nordisk, A.S. A Long-Term, Multi-Centre, International, Randomised Double-Blind, Placebo-Controlled Trial to Determine Liraglutide Effects on Cardiovascular Events; National Library of Medicine: Bethesda, MD, USA, 2019; clinicaltrials.gov.
  29. Verma, S.; Poulter, N.R.; Bhatt, D.L.; Bain, S.C.; Buse, J.B.; Leiter, L.A.; Nauck, M.A.; Pratley, R.E.; Zinman, B.; Ørsted, D.D.; et al. Effects of Liraglutide on Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus With or Without History of Myocardial Infarction or Stroke. Circulation 2018, 138, 2884–2894. [Google Scholar] [CrossRef]
  30. Janssen Research & Development, LLC. A Randomized, Multicenter, Double-Blind, Parallel, Placebo-Controlled Study of the Effects of JNJ-28431754 on Cardiovascular Outcomes in Adult Subjects With Type 2 Diabetes Mellitus; National Library of Medicine: Bethesda, MD, USA, 2018; clinicaltrials.gov.
  31. Everett, B.M.; Donath, M.Y.; Pradhan, A.D.; Thuren, T.; Pais, P.; Nicolau, J.C.; Glynn, R.J.; Libby, P.; Ridker, P.M. Anti-Inflammatory Therapy With Canakinumab for the Prevention and Management of Diabetes. J. Am. Coll. Cardiol. 2018, 71, 2392–2401. [Google Scholar] [CrossRef]
  32. Rosengren, A. Randomized Clinical Trial with Broccoli Sprout Extract to Patients with Type 2 Diabetes; National Library of Medicine: Bethesda, MD, USA, 2022; clinicaltrials.gov.
  33. Axelsson, A.S.; Tubbs, E.; Mecham, B.; Chacko, S.; Nenonen, H.A.; Tang, Y.; Fahey, J.W.; Derry, J.M.J.; Wollheim, C.B.; Wierup, N.; et al. Sulforaphane Reduces Hepatic Glucose Production and Improves Glucose Control in Patients with Type 2 Diabetes. Sci. Transl. Med. 2017, 9, eaah4477. [Google Scholar] [CrossRef]
  34. Brown, M.S.; Goldstein, J.L. Selective versus Total Insulin Resistance: A Pathogenic Paradox. Cell Metab. 2008, 7, 95–96. [Google Scholar] [CrossRef]
  35. da Silva, A.A.; do Carmo, J.M.; Li, X.; Wang, Z.; Mouton, A.J.; Hall, J.E. Role of Hyperinsulinemia and Insulin Resistance in Hypertension: Metabolic Syndrome Revisited. Can. J. Cardiol. 2020, 36, 671–682. [Google Scholar] [CrossRef]
  36. Gallagher, E.J.; LeRoith, D. Hyperinsulinaemia in Cancer. Nat. Rev. Cancer 2020, 20, 629–644. [Google Scholar] [CrossRef]
  37. Kolb, H.; Stumvoll, M.; Kramer, W.; Kempf, K.; Martin, S. Insulin Translates Unfavourable Lifestyle into Obesity. BMC Med. 2018, 16, 232. [Google Scholar] [CrossRef]
  38. Herman, M.E.; O’Keefe, J.H.; Bell, D.S.H.; Schwartz, S.S. Insulin Therapy Increases Cardiovascular Risk in Type 2 Diabetes. Prog. Cardiovasc. Dis. 2017, 60, 422–434. [Google Scholar] [CrossRef] [PubMed]
  39. Einarson, T.R.; Acs, A.; Ludwig, C.; Panton, U.H. Prevalence of Cardiovascular Disease in Type 2 Diabetes: A Systematic Literature Review of Scientific Evidence from across the World in 2007–2017. Cardiovasc. Diabetol. 2018, 17, 83. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, G.Y.; Sabatini, D.M. mTOR at the Nexus of Nutrition, Growth, Ageing and Disease. Nat. Rev. Mol. Cell Biol. 2020, 21, 183–203. [Google Scholar] [CrossRef]
  41. Sabatini, D.M. Twenty-Five Years of mTOR: Uncovering the Link from Nutrients to Growth. Proc. Natl. Acad. Sci. USA 2017, 114, 11818–11825. [Google Scholar] [CrossRef] [PubMed]
  42. Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [PubMed]
  43. Samidurai, A.; Kukreja, R.C.; Das, A. Emerging Role of mTOR Signaling-Related miRNAs in Cardiovascular Diseases. Oxid. Med. Cell Longev. 2018, 2018, 6141902. [Google Scholar] [CrossRef] [PubMed]
  44. Yoneyama, Y.; Inamitsu, T.; Chida, K.; Iemura, S.-I.; Natsume, T.; Maeda, T.; Hakuno, F.; Takahashi, S.-I. Serine Phosphorylation by mTORC1 Promotes IRS-1 Degradation through SCFβ-TRCP E3 Ubiquitin Ligase. iScience 2018, 5, 1–18. [Google Scholar] [CrossRef]
  45. Hsu, P.P.; Kang, S.A.; Rameseder, J.; Zhang, Y.; Ottina, K.A.; Lim, D.; Peterson, T.R.; Choi, Y.; Gray, N.S.; Yaffe, M.B.; et al. The mTOR-Regulated Phosphoproteome Reveals a Mechanism of mTORC1-Mediated Inhibition of Growth Factor Signaling. Science 2011, 332, 1317–1322. [Google Scholar] [CrossRef]
  46. Um, S.H.; Frigerio, F.; Watanabe, M.; Picard, F.; Joaquin, M.; Sticker, M.; Fumagalli, S.; Allegrini, P.R.; Kozma, S.C.; Auwerx, J.; et al. Absence of S6K1 Protects against Age- and Diet-Induced Obesity While Enhancing Insulin Sensitivity. Nature 2004, 431, 200–205. [Google Scholar] [CrossRef]
  47. Malhowski, A.J.; Hira, H.; Bashiruddin, S.; Warburton, R.; Goto, J.; Robert, B.; Kwiatkowski, D.J.; Finlay, G.A. Smooth Muscle Protein-22-Mediated Deletion of Tsc1 Results in Cardiac Hypertrophy That Is mTORC1-Mediated and Reversed by Rapamycin. Hum. Mol. Genet. 2011, 20, 1290–1305. [Google Scholar] [CrossRef]
  48. Völkers, M.; Toko, H.; Doroudgar, S.; Din, S.; Quijada, P.; Joyo, A.Y.; Ornelas, L.; Joyo, E.; Thuerauf, D.J.; Konstandin, M.H.; et al. Pathological Hypertrophy Amelioration by PRAS40-Mediated Inhibition of mTORC1. Proc. Natl. Acad. Sci. USA 2013, 110, 12661–12666. [Google Scholar] [CrossRef] [PubMed]
  49. Sciarretta, S.; Forte, M.; Frati, G.; Sadoshima, J. New Insights Into the Role of mTOR Signaling in the Cardiovascular System. Circ. Res. 2018, 122, 489–505. [Google Scholar] [CrossRef] [PubMed]
  50. Bell, D.S.H. Heart Failure: The Frequent, Forgotten, and Often Fatal Complication of Diabetes. Diabetes Care 2003, 26, 2433–2441. [Google Scholar] [CrossRef] [PubMed]
  51. Sciarretta, S.; Zhai, P.; Shao, D.; Maejima, Y.; Robbins, J.; Volpe, M.; Condorelli, G.; Sadoshima, J. Rheb Is a Critical Regulator of Autophagy during Myocardial Ischemia: Pathophysiological Implications in Obesity and Metabolic Syndrome. Circulation 2012, 125, 1134–1146. [Google Scholar] [CrossRef]
  52. Matsui, Y.; Takagi, H.; Qu, X.; Abdellatif, M.; Sakoda, H.; Asano, T.; Levine, B.; Sadoshima, J. Distinct Roles of Autophagy in the Heart During Ischemia and Reperfusion. Circ. Res. 2007, 100, 914–922. [Google Scholar] [CrossRef]
  53. Ma, Z.-G.; Yuan, Y.-P.; Xu, S.-C.; Wei, W.-Y.; Xu, C.-R.; Zhang, X.; Wu, Q.-Q.; Liao, H.-H.; Ni, J.; Tang, Q.-Z. CTRP3 Attenuates Cardiac Dysfunction, Inflammation, Oxidative Stress and Cell Death in Diabetic Cardiomyopathy in Rats. Diabetologia 2017, 60, 1126–1137. [Google Scholar] [CrossRef]
  54. Ma, Z.-G.; Dai, J.; Zhang, W.-B.; Yuan, Y.; Liao, H.-H.; Zhang, N.; Bian, Z.-Y.; Tang, Q.-Z. Protection against Cardiac Hypertrophy by Geniposide Involves the GLP-1 Receptor / AMPKα Signalling Pathway. Br. J. Pharmacol. 2016, 173, 1502–1516. [Google Scholar] [CrossRef]
  55. Yang, Q.; Wang, H.-C.; Liu, Y.; Gao, C.; Sun, L.; Tao, L. Resveratrol Cardioprotection Against Myocardial Ischemia/Reperfusion Injury Involves Upregulation of Adiponectin Levels and Multimerization in Type 2 Diabetic Mice. J. Cardiovasc. Pharmacol. 2016, 68, 304–312. [Google Scholar] [CrossRef]
  56. Chang, W.; Zhang, M.; Meng, Z.; Yu, Y.; Yao, F.; Hatch, G.M.; Chen, L. Berberine Treatment Prevents Cardiac Dysfunction and Remodeling through Activation of 5′-Adenosine Monophosphate-Activated Protein Kinase in Type 2 Diabetic Rats and in Palmitate-Induced Hypertrophic H9c2 Cells. Eur. J. Pharmacol. 2015, 769, 55–63. [Google Scholar] [CrossRef]
  57. Yang, F.; Qin, Y.; Wang, Y.; Meng, S.; Xian, H.; Che, H.; Lv, J.; Li, Y.; Yu, Y.; Bai, Y.; et al. Metformin Inhibits the NLRP3 Inflammasome via AMPK/mTOR-Dependent Effects in Diabetic Cardiomyopathy. Int. J. Biol. Sci. 2019, 15, 1010–1019. [Google Scholar] [CrossRef]
  58. Despa, S.; Islam, M.A.; Weber, C.R.; Pogwizd, S.M.; Bers, D.M. Intracellular Na+ Concentration Is Elevated in Heart Failure but Na/K Pump Function Is Unchanged. Circulation 2002, 105, 2543–2548. [Google Scholar] [CrossRef] [PubMed]
  59. Jia, G.; Hill, M.A.; Sowers, J.R. Diabetic Cardiomyopathy: An Update of Mechanisms Contributing to This Clinical Entity. Circ. Res. 2018, 122, 624–638. [Google Scholar] [CrossRef] [PubMed]
  60. Luo, M.; Anderson, M.E. Mechanisms of Altered Ca2+ Handling in Heart Failure. Circ. Res. 2013, 113, 690–708. [Google Scholar] [CrossRef] [PubMed]
  61. Ogawa, A.; Firth, A.L.; Smith, K.A.; Maliakal, M.V.; Yuan, J.X.-J. PDGF Enhances Store-Operated Ca2+ Entry by Upregulating STIM1/Orai1 via Activation of Akt/mTOR in Human Pulmonary Arterial Smooth Muscle Cells. Am. J. Physiol. Cell Physiol. 2012, 302, C405–C411. [Google Scholar] [CrossRef] [PubMed]
  62. Cang, C.; Zhou, Y.; Navarro, B.; Seo, Y.-J.; Aranda, K.; Shi, L.; Battaglia-Hsu, S.; Nissim, I.; Clapham, D.E.; Ren, D. mTOR Regulates Lysosomal ATP-Sensitive Two-Pore Na+ Channels to Adapt to Metabolic State. Cell 2013, 152, 778–790. [Google Scholar] [CrossRef]
  63. Hisatsune, C.; Shimada, T.; Miyamoto, A.; Lee, A.; Yamagata, K. Tuberous Sclerosis Complex (TSC) Inactivation Increases Neuronal Network Activity by Enhancing Ca2+ Influx via L-Type Ca2+ Channels. J. Neurosci. 2021, 41, 8134–8149. [Google Scholar] [CrossRef]
  64. Amemiya, Y.; Maki, M.; Shibata, H.; Takahara, T. New Insights into the Regulation of mTOR Signaling via Ca2+-Binding Proteins. Int. J. Mol. Sci. 2023, 24, 3923. [Google Scholar] [CrossRef]
  65. Sanlialp, A.; Schumacher, D.; Kiper, L.; Varma, E.; Riechert, E.; Ho, T.C.; Hofmann, C.; Kmietczyk, V.; Zimmermann, F.; Dlugosz, S.; et al. Saraf-Dependent Activation of mTORC1 Regulates Cardiac Growth. J. Mol. Cell Cardiol. 2020, 141, 30–42. [Google Scholar] [CrossRef]
  66. Ogunbayo, O.A.; Duan, J.; Xiong, J.; Wang, Q.; Feng, X.; Ma, J.; Zhu, M.X.; Evans, A.M. mTORC1 Controls Lysosomal Ca2+ Release through the Two-Pore Channel TPC2. Sci. Signal 2018, 11, eaao5775. [Google Scholar] [CrossRef]
  67. Janse, M.J. Electrophysiological Changes in Heart Failure and Their Relationship to Arrhythmogenesis. Cardiovasc. Res. 2004, 61, 208–217. [Google Scholar] [CrossRef]
  68. Liu, C.; Liu, E.; Luo, T.; Zhang, W.; He, R. Opening of the Inward Rectifier Potassium Channel Alleviates Maladaptive Tissue Repair Following Myocardial Infarction. Acta Biochim. Biophys. Sin. 2016, 48, 687–695. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, Q.-H.; Zhang, L.-J.; Wang, J.; Wu, B.-W.; Cao, J.-M. Cardioprotection of an IK1 Channel Agonist on L-Thyroxine Induced Rat Ventricular Remodeling. Am. J. Transl. Res. 2021, 13, 8683–8696. [Google Scholar] [PubMed]
  70. Lin, P.-H.; Duann, P.; Komazaki, S.; Park, K.H.; Li, H.; Sun, M.; Sermersheim, M.; Gumpper, K.; Parrington, J.; Galione, A.; et al. Lysosomal Two-Pore Channel Subtype 2 (TPC2) Regulates Skeletal Muscle Autophagic Signaling. J. Biol. Chem. 2015, 290, 3377–3389. [Google Scholar] [CrossRef] [PubMed]
  71. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-Activated Protein Kinase in Mechanism of Metformin Action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
  72. Kalender, A.; Selvaraj, A.; Kim, S.Y.; Gulati, P.; Brûlé, S.; Viollet, B.; Kemp, B.E.; Bardeesy, N.; Dennis, P.; Schlager, J.J.; et al. Metformin, Independent of AMPK, Inhibits mTORC1 in a Rag GTPase-Dependent Manner. Cell Metab. 2010, 11, 390–401. [Google Scholar] [CrossRef] [PubMed]
  73. Lu, J.; Liu, J.; Zhang, L.; Wang, X.; Zhang, Y.; Tang, Q. Morphological and Functional Characterization of Diabetic Cardiomyopathy in Db/Db Mice Following Exercise, Metformin Alone, or Combination Treatments. Biochem. Biophys. Res. Commun. 2021, 584, 80–86. [Google Scholar] [CrossRef]
  74. Völkers, M.; Doroudgar, S.; Nguyen, N.; Konstandin, M.H.; Quijada, P.; Din, S.; Ornelas, L.; Thuerauf, D.J.; Gude, N.; Friedrich, K.; et al. PRAS40 Prevents Development of Diabetic Cardiomyopathy and Improves Hepatic Insulin Sensitivity in Obesity. EMBO Mol. Med. 2014, 6, 57–65. [Google Scholar] [CrossRef]
  75. Das, A.; Durrant, D.; Koka, S.; Salloum, F.N.; Xi, L.; Kukreja, R.C. Mammalian Target of Rapamycin (mTOR) Inhibition with Rapamycin Improves Cardiac Function in Type 2 Diabetic Mice. J. Biol. Chem. 2014, 289, 4145–4160. [Google Scholar] [CrossRef]
  76. Reifsnyder, P.C.; Flurkey, K.; Te, A.; Harrison, D.E. Rapamycin Treatment Benefits Glucose Metabolism in Mouse Models of Type 2 Diabetes. Aging 2016, 8, 3120–3130. [Google Scholar] [CrossRef]
  77. McMullen, J.R.; Sherwood, M.C.; Tarnavski, O.; Zhang, L.; Dorfman, A.L.; Shioi, T.; Izumo, S. Inhibition of mTOR Signaling With Rapamycin Regresses Established Cardiac Hypertrophy Induced by Pressure Overload. Circulation 2004, 109, 3050–3055. [Google Scholar] [CrossRef]
  78. Houde, V.P.; Brûlé, S.; Festuccia, W.T.; Blanchard, P.-G.; Bellmann, K.; Deshaies, Y.; Marette, A. Chronic Rapamycin Treatment Causes Glucose Intolerance and Hyperlipidemia by Upregulating Hepatic Gluconeogenesis and Impairing Lipid Deposition in Adipose Tissue. Diabetes 2010, 59, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  79. Salmon, A.B. About-Face on the Metabolic Side Effects of Rapamycin. Oncotarget 2015, 6, 2585–2586. [Google Scholar] [CrossRef] [PubMed]
  80. Krebs, M.; Brunmair, B.; Brehm, A.; Artwohl, M.; Szendroedi, J.; Nowotny, P.; Roth, E.; Fürnsinn, C.; Promintzer, M.; Anderwald, C.; et al. The Mammalian Target of Rapamycin Pathway Regulates Nutrient-Sensitive Glucose Uptake in Man. Diabetes 2007, 56, 1600–1607. [Google Scholar] [CrossRef]
  81. Lamming, D.W.; Ye, L.; Katajisto, P.; Goncalves, M.D.; Saitoh, M.; Stevens, D.M.; Davis, J.G.; Salmon, A.B.; Richardson, A.; Ahima, R.S.; et al. Rapamycin-Induced Insulin Resistance Is Mediated by mTORC2 Loss and Uncoupled from Longevity. Science 2012, 335, 1638–1643. [Google Scholar] [CrossRef] [PubMed]
  82. Samidurai, A.; Salloum, F.N.; Durrant, D.; Chernova, O.B.; Kukreja, R.C.; Das, A. Chronic Treatment with Novel Nanoformulated Micelles of Rapamycin, Rapatar, Protects Diabetic Heart against Ischaemia/Reperfusion Injury. Br. J. Pharmacol. 2017, 174, 4771–4784. [Google Scholar] [CrossRef]
  83. Samidurai, A.; Ockaili, R.; Cain, C.; Roh, S.K.; Filippone, S.M.; Kraskauskas, D.; Kukreja, R.C.; Das, A. Differential Regulation of mTOR Complexes with miR-302a Attenuates Myocardial Reperfusion Injury in Diabetes. iScience 2020, 23, 101863. [Google Scholar] [CrossRef] [PubMed]
  84. Samidurai, A.; Roh, S.K.; Prakash, M.; Durrant, D.; Salloum, F.N.; Kukreja, R.C.; Das, A. STAT3-miR-17/20 Signalling Axis Plays a Critical Role in Attenuating Myocardial Infarction Following Rapamycin Treatment in Diabetic Mice. Cardiovasc. Res. 2020, 116, 2103–2115. [Google Scholar] [CrossRef] [PubMed]
  85. Li, J.; Rohailla, S.; Gelber, N.; Rutka, J.; Sabah, N.; Gladstone, R.A.; Wei, C.; Hu, P.; Kharbanda, R.K.; Redington, A.N. MicroRNA-144 Is a Circulating Effector of Remote Ischemic Preconditioning. Basic Res. Cardiol. 2014, 109, 423. [Google Scholar] [CrossRef] [PubMed]
  86. Shende, P.; Xu, L.; Morandi, C.; Pentassuglia, L.; Heim, P.; Lebboukh, S.; Berthonneche, C.; Pedrazzini, T.; Kaufmann, B.A.; Hall, M.N.; et al. Cardiac mTOR Complex 2 Preserves Ventricular Function in Pressure-Overload Hypertrophy. Cardiovasc. Res. 2016, 109, 103–114. [Google Scholar] [CrossRef]
  87. American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes—2021. Diabetes Care 2020, 44, S111–S124. [Google Scholar] [CrossRef]
  88. Baker, C.; Retzik-Stahr, C.; Singh, V.; Plomondon, R.; Anderson, V.; Rasouli, N. Should Metformin Remain the First-Line Therapy for Treatment of Type 2 Diabetes? Ther. Adv. Endocrinol. Metab. 2021, 12, 2042018820980225. [Google Scholar] [CrossRef] [PubMed]
  89. Research, C. for D.E. and Sodium-Glucose Cotransporter-2 (SGLT2) Inhibitors. FDA 2018, 9, 1273–1281. [Google Scholar]
  90. Zheng, S.L.; Roddick, A.J.; Aghar-Jaffar, R.; Shun-Shin, M.J.; Francis, D.; Oliver, N.; Meeran, K. Association Between Use of Sodium-Glucose Cotransporter 2 Inhibitors, Glucagon-like Peptide 1 Agonists, and Dipeptidyl Peptidase 4 Inhibitors With All-Cause Mortality in Patients With Type 2 Diabetes: A Systematic Review and Meta-Analysis. JAMA 2018, 319, 1580–1591. [Google Scholar] [CrossRef] [PubMed]
  91. Kato, E.T.; Silverman, M.G.; Mosenzon, O.; Zelniker, T.A.; Cahn, A.; Furtado, R.H.M.; Kuder, J.; Murphy, S.A.; Bhatt, D.L.; Leiter, L.A.; et al. Effect of Dapagliflozin on Heart Failure and Mortality in Type 2 Diabetes Mellitus. Circulation 2019, 139, 2528–2536. [Google Scholar] [CrossRef]
  92. Cefalu, W.T.; Stenlöf, K.; Leiter, L.A.; Wilding, J.P.H.; Blonde, L.; Polidori, D.; Xie, J.; Sullivan, D.; Usiskin, K.; Canovatchel, W.; et al. Effects of Canagliflozin on Body Weight and Relationship to HbA1c and Blood Pressure Changes in Patients with Type 2 Diabetes. Diabetologia 2015, 58, 1183–1187. [Google Scholar] [CrossRef]
  93. Mao, Z.; Zhang, W. Role of mTOR in Glucose and Lipid Metabolism. Int. J. Mol. Sci. 2018, 19, 2043. [Google Scholar] [CrossRef]
  94. 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. [Google Scholar] [CrossRef]
  95. Keller, D.M.; Ahmed, N.; Tariq, H.; Walgamage, M.; Walgamage, T.; Mohammed, A.; Chou, J.T.-T.; Kałużna-Oleksy, M.; Lesiak, M.; Straburzyńska-Migaj, E. SGLT2 Inhibitors in Type 2 Diabetes Mellitus and Heart Failure—A Concise Review. J. Clin. Med. 2022, 11, 1470. [Google Scholar] [CrossRef]
  96. Bakris, G.L.; Fonseca, V.A.; Sharma, K.; Wright, E.M. Renal Sodium–Glucose Transport: Role in Diabetes Mellitus and Potential Clinical Implications. Kidney Int. 2009, 75, 1272–1277. [Google Scholar] [CrossRef]
  97. Wilcox, C.S. Antihypertensive and Renal Mechanisms of SGLT2 (Sodium-Glucose Linked Transporter 2) Inhibitors. Hypertension 2020, 75, 894–901. [Google Scholar] [CrossRef]
  98. Abdul-Ghani, M.A.; Norton, L.; DeFronzo, R.A. Role of Sodium-Glucose Cotransporter 2 (SGLT 2) Inhibitors in the Treatment of Type 2 Diabetes. Endocr. Rev. 2011, 32, 515–531. [Google Scholar] [CrossRef] [PubMed]
  99. Fonseca-Correa, J.I.; Correa-Rotter, R. Sodium-Glucose Cotransporter 2 Inhibitors Mechanisms of Action: A Review. Front. Med. (Lausanne) 2021, 8, 777861. [Google Scholar] [CrossRef] [PubMed]
  100. Kyriakos, G.; Quiles-Sanchez, L.V.; Garmpi, A.; Farmaki, P.; Kyre, K.; Savvanis, S.; Antoniou, V.K.; Memi, E. SGLT2 Inhibitors and Cardiovascular Outcomes: Do They Differ or There Is a Class Effect? New Insights from the EMPA-REG OUTCOME Trial and the CVD-REAL Study. Curr. Cardiol. Rev. 2020, 16, 258–265. [Google Scholar] [CrossRef] [PubMed]
  101. Kosiborod, M.; Lam, C.S.P.; Kohsaka, S.; Kim, D.J.; Karasik, A.; Shaw, J.; Tangri, N.; Goh, S.-Y.; Thuresson, M.; Chen, H.; et al. Cardiovascular Events Associated With SGLT-2 Inhibitors Versus Other Glucose-Lowering Drugs: The CVD-REAL 2 Study. J. Am. Coll. Cardiol. 2018, 71, 2628–2639. [Google Scholar] [CrossRef]
  102. The Metabolic Effects of Empagliflozin in Patients With High Risk of Heart Failure—Full Text View—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT05042973 (accessed on 29 April 2023).
  103. AstraZeneca. Characteristics and Cardiovascular and Mortality Outcomes in Patients with Type 2 Diabetes Mellitus Initiating Treatment with Sodium-Glucose Co-Transporter-2 Inhibitors (SGLT-2i) and Other Glucose Lowering Drugs; National Library of Medicine: Bethesda, MD, USA, 2021; clinicaltrials.gov.
  104. Khunti, K.; Kosiborod, M.; Kim, D.J.; Kohsaka, S.; Lam, C.S.P.; Goh, S.-Y.; Chiang, C.-E.; Shaw, J.E.; Cavender, M.A.; Tangri, N.; et al. Cardiovascular Outcomes with Sodium–Glucose Cotransporter-2 Inhibitors vs Other Glucose-Lowering Drugs in 13 Countries across Three Continents: Analysis of CVD-REAL Data. Cardiovasc. Diabetol. 2021, 20, 159. [Google Scholar] [CrossRef]
  105. Scheen, A.J. [EMPA-REG OUTCOME: Empagliflozin reduces mortality in patients with type 2 diabetes at high cardiovascular risk]. Rev. Med. Liege 2015, 70, 583–589. [Google Scholar]
  106. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef]
  107. D’Andrea, E.; Wexler, D.J.; Kim, S.C.; Paik, J.M.; Alt, E.; Patorno, E. Comparing Effectiveness and Safety of SGLT2 Inhibitors vs DPP-4 Inhibitors in Patients With Type 2 Diabetes and Varying Baseline HbA1c Levels. JAMA Intern. Med. 2023, 183, 242–254. [Google Scholar] [CrossRef]
  108. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  109. Ingelheim, B. A Phase III, Multicentre, International, Randomised, Parallel Group, Double Blind Cardiovascular Safety Study of BI 10773 (10 Mg and 25 Mg Administered Orally Once Daily) Compared to Usual Care in Type 2 Diabetes Mellitus Patients With Increased Cardiovascular Risk; National Library of Medicine: Bethesda, MD, USA, 2016; clinicaltrials.gov.
  110. Abdul-Ghani, M.; Del Prato, S.; Chilton, R.; DeFronzo, R.A. SGLT2 Inhibitors and Cardiovascular Risk: Lessons Learned From the EMPA-REG OUTCOME Study. Diabetes Care 2016, 39, 717–725. [Google Scholar] [CrossRef]
  111. Verma, S.; Leiter, L.A.; Zinman, B.; Sharma, A.; Mattheus, M.; Fitchett, D.; George, J.; Ofstad, A.P.; Kosiborod, M.N.; Wanner, C.; et al. Time to Cardiovascular Benefits of Empagliflozin: A Post Hoc Observation from the EMPA-REG OUTCOME Trial. ESC Heart Fail. 2021, 8, 2603–2607. [Google Scholar] [CrossRef] [PubMed]
  112. Ferreira, J.P.; Kraus, B.J.; Zwiener, I.; Lauer, S.; Zinman, B.; Fitchett, D.H.; Koitka-Weber, A.; George, J.T.; Ofstad, A.P.; Wanner, C.; et al. Cardio/Kidney Composite End Points: A Post Hoc Analysis of the EMPA-REG OUTCOME Trial. J. Am. Heart Assoc. 2021, 10, e020053. [Google Scholar] [CrossRef] [PubMed]
  113. Fitchett, D.; Inzucchi, S.E.; Cannon, C.P.; McGuire, D.K.; Scirica, B.M.; Johansen, O.E.; Sambevski, S.; Kaspers, S.; Pfarr, E.; George, J.T.; et al. Empagliflozin Reduced Mortality and Hospitalization for Heart Failure Across the Spectrum of Cardiovascular Risk in the EMPA-REG OUTCOME Trial. Circulation 2019, 139, 1384–1395. [Google Scholar] [CrossRef] [PubMed]
  114. Alzaid, A. Empa’s New Clothes: The Untold Story of the Empa-Reg Outcome Trial. Diabetes Technol. Ther. 2017, 19, 324–327. [Google Scholar] [CrossRef]
  115. Neal, B.; Perkovic, V.; Mahaffey, K.W.; de Zeeuw, D.; Fulcher, G.; Erondu, N.; Shaw, W.; Law, G.; Desai, M.; Matthews, D.R. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 644–657. [Google Scholar] [CrossRef] [PubMed]
  116. Yu, J.; Li, J.; Leaver, P.J.; Arnott, C.; Huffman, M.D.; Udell, J.A.; Perkovic, V.; Mahaffey, K.W.; de Zeeuw, D.; Fulcher, G.; et al. Effects of Canagliflozin on Myocardial Infarction: A Post Hoc Analysis of the CANVAS Programme and CREDENCE Trial. Cardiovasc. Res. 2022, 118, 1103–1114. [Google Scholar] [CrossRef]
  117. Yu, J.; Arnott, C.; Neuen, B.L.; Heersprink, H.L.; Mahaffey, K.W.; Cannon, C.P.; Khan, S.S.; Baldridge, A.S.; Shah, S.J.; Huang, Y.; et al. Cardiovascular and Renal Outcomes with Canagliflozin According to Baseline Diuretic Use: A Post Hoc Analysis from the CANVAS Program. ESC Heart Fail. 2021, 8, 1482–1493. [Google Scholar] [CrossRef]
  118. AstraZeneca. Dapagliflozin Effect on Cardiovascular Events A Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial to Evaluate the Effect of Dapagliflozin 10 Mg Once Daily on the Incidence of Cardiovascular Death, Myocardial Infarction or Ischemic Stroke in Patients With Type 2 Diabetes; National Library of Medicine: Bethesda, MD, USA, 2019; clinicaltrials.gov.
  119. Wiviott, S.D.; Raz, I.; Bonaca, M.P.; Mosenzon, O.; Kato, E.T.; Cahn, A.; Silverman, M.G.; Zelniker, T.A.; Kuder, J.F.; Murphy, S.A.; et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2019, 380, 347–357. [Google Scholar] [CrossRef]
  120. Schechter, M.; Wiviott, S.D.; Raz, I.; Goodrich, E.L.; Rozenberg, A.; Yanuv, I.; Murphy, S.A.; Zelniker, T.A.; Fredriksson, M.; Johansson, P.A.; et al. Effects of Dapagliflozin on Hospitalisations in People with Type 2 Diabetes: Post-Hoc Analyses of the DECLARE-TIMI 58 Trial. Lancet Diabetes Endocrinol. 2023, 11, 233–241. [Google Scholar] [CrossRef]
  121. Furtado, R.H.M.; Raz, I.; Goodrich, E.L.; Murphy, S.A.; Bhatt, D.L.; Leiter, L.A.; McGuire, D.K.; Wilding, J.P.H.; Aylward, P.; Dalby, A.J.; et al. Efficacy and Safety of Dapagliflozin in Type 2 Diabetes According to Baseline Blood Pressure: Observations From DECLARE-TIMI 58 Trial. Circulation 2022, 145, 1581–1591. [Google Scholar] [CrossRef]
  122. Mosenzon, O.; Wiviott, S.D.; Heerspink, H.J.L.; Dwyer, J.P.; Cahn, A.; Goodrich, E.L.; Rozenberg, A.; Schechter, M.; Yanuv, I.; Murphy, S.A.; et al. The Effect of Dapagliflozin on Albuminuria in DECLARE-TIMI 58. Diabetes Care 2021, 44, 1805–1815. [Google Scholar] [CrossRef] [PubMed]
  123. Merck Sharp & Dohme LLC. Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study to Assess Cardiovascular Outcomes Following Treatment with Ertugliflozin (MK-8835/PF-04971729) in Subjects with Type 2 Diabetes Mellitus and Established Vascular Disease, The VERTIS CV Study; National Library of Medicine: Bethesda, MD, USA, 2022; clinicaltrials.gov.
  124. Cannon, C.P.; Pratley, R.; Dagogo-Jack, S.; Mancuso, J.; Huyck, S.; Masiukiewicz, U.; Charbonnel, B.; Frederich, R.; Gallo, S.; Cosentino, F.; et al. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N. Engl. J. Med. 2020, 383, 1425–1435. [Google Scholar] [CrossRef] [PubMed]
  125. Cherney, D.Z.I.; Dagogo-Jack, S.; Cosentino, F.; Pratley, R.E.; Frederich, R.; Maldonado, M.; Liu, C.-C.; Cannon, C.P. Heart and Kidney Outcomes With Ertugliflozin in People with Non-Albuminuric Diabetic Kidney Disease: A Post Hoc Analysis from the Randomized VERTIS CV Trial. Kidney Int. Rep. 2022, 7, 1782–1792. [Google Scholar] [CrossRef] [PubMed]
  126. Dagogo-Jack, S.; Cannon, C.P.; Cherney, D.Z.I.; Cosentino, F.; Liu, J.; Pong, A.; Gantz, I.; Frederich, R.; Mancuso, J.P.; Pratley, R.E. Cardiorenal Outcomes with Ertugliflozin Assessed According to Baseline Glucose-lowering Agent: An Analysis from VERTIS CV. Diabetes Obes. Metab. 2022, 24, 1245–1254. [Google Scholar] [CrossRef]
  127. AstraZeneca. Study to Evaluate the Effect of Dapagliflozin on the Incidence of Worsening Heart Failure or Cardiovascular Death in Patients with Chronic Heart Failure with Reduced Ejection Fraction; National Library of Medicine: Bethesda, MD, USA, 2020; clinicaltrials.gov.
  128. Docherty, K.F.; Jhund, P.S.; Bengtsson, O.; Demets, D.L.; Inzucchi, S.E.; Kober, L.; Kosiborod, M.N.; Langkilde, A.M.; Lindholm, D.; Martinez, F.A.; et al. The Effect of Dapagliflozin across the Spectrum of Baseline Risk: A Post-Hoc Analysis of DAPA-HF. Eur. Heart J. 2020, 41, ehaa946.0931. [Google Scholar] [CrossRef]
  129. Butt, J.H.; Dewan, P.; Merkely, B.; Belohlávek, J.; Drożdż, J.; Kitakaze, M.; Inzucchi, S.E.; Kosiborod, M.N.; Martinez, F.A.; Tereshchenko, S.; et al. Efficacy and Safety of Dapagliflozin According to Frailty in Heart Failure With Reduced Ejection Fraction: A Post Hoc Analysis of the DAPA-HF Trial. Ann. Intern. Med. 2022, 175, 820–830. [Google Scholar] [CrossRef]
  130. Ingelheim, B. A Phase III Randomised, Double-Blind Trial to Evaluate Efficacy and Safety of Once Daily Empagliflozin 10 Mg Compared to Placebo, in Patients With Chronic Heart Failure With Reduced Ejection Fraction (HFrEF); National Library of Medicine: Bethesda, MD, USA, 2021; clinicaltrials.gov.
  131. Verma, S.; Dhingra, N.K.; Butler, J.; Anker, S.D.; Ferreira, J.P.; Filippatos, G.; Januzzi, J.L.; Lam, C.S.P.; Sattar, N.; Peil, B.; et al. Empagliflozin in the Treatment of Heart Failure with Reduced Ejection Fraction in Addition to Background Therapies and Therapeutic Combinations (EMPEROR-Reduced): A Post-Hoc Analysis of a Randomised, Double-Blind Trial. Lancet Diabetes Endocrinol. 2022, 10, 35–45. [Google Scholar] [CrossRef]
  132. Empagliflozin for HFrEF—Post Hoc Analysis of EMPEROR-Reduced. Available online: https://www.jwatch.org/na54543/2022/02/08/empagliflozin-hfref-post-hoc-analysis-emperor-reduced (accessed on 23 September 2023).
  133. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Pocock, S.J.; Carson, P.; Anand, I.; Doehner, W.; Haass, M.; et al. Effect of Empagliflozin on the Clinical Stability of Patients With Heart Failure and a Reduced Ejection Fraction. Circulation 2021, 143, 326–336. [Google Scholar] [CrossRef]
  134. 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. [Google Scholar] [CrossRef]
  135. Huang, K.; Luo, X.; Liao, B.; Li, G.; Feng, J. Insights into SGLT2 Inhibitor Treatment of Diabetic Cardiomyopathy: Focus on the Mechanisms. Cardiovasc. Diabetol. 2023, 22, 86. [Google Scholar] [CrossRef]
  136. Banerjee, S.K.; McGaffin, K.R.; Pastor-Soler, N.M.; Ahmad, F. SGLT1 Is a Novel Cardiac Glucose Transporter That Is Perturbed in Disease States. Cardiovasc. Res. 2009, 84, 111–118. [Google Scholar] [CrossRef] [PubMed]
  137. Li, Y.; Xu, G. Sodium Glucose Cotransporter 1 (SGLT1) Inhibitors in Cardiovascular Protection: Mechanism Progresses and Challenges. Pharmacol. Res. 2022, 176, 106049. [Google Scholar] [CrossRef] [PubMed]
  138. Zhao, M.; Li, N.; Zhou, H. SGLT1: A Potential Drug Target for Cardiovascular Disease. DDDT 2023, 17, 2011–2023. [Google Scholar] [CrossRef]
  139. Kondo, H.; Akoumianakis, I.; Badi, I.; Akawi, N.; Kotanidis, C.P.; Polkinghorne, M.; Stadiotti, I.; Sommariva, E.; Antonopoulos, A.S.; Carena, M.C.; et al. Effects of Canagliflozin on Human Myocardial Redox Signalling: Clinical Implications. Eur. Heart J. 2021, 42, 4947–4960. [Google Scholar] [CrossRef] [PubMed]
  140. Sayour, A.A.; Ruppert, M.; Oláh, A.; Benke, K.; Barta, B.A.; Zsáry, E.; Merkely, B.; Radovits, T. Effects of SGLT2 Inhibitors beyond Glycemic Control—Focus on Myocardial SGLT1. Int. J. Mol. Sci. 2021, 22, 9852. [Google Scholar] [CrossRef] [PubMed]
  141. Pitt, B.; Bhatt, D.L.; Metra, M. Does SGLT1 Inhibition Add to the Benefits of SGLT2 Inhibition in the Prevention and Treatment of Heart Failure? Eur. Heart J. 2022, 43, 4754–4757. [Google Scholar] [CrossRef] [PubMed]
  142. Seidelmann, S.B.; Feofanova, E.; Yu, B.; Franceschini, N.; Claggett, B.; Kuokkanen, M.; Puolijoki, H.; Ebeling, T.; Perola, M.; Salomaa, V.; et al. Genetic Variants in SGLT1, Glucose Tolerance, and Cardiometabolic Risk. J. Am. Coll. Cardiol. 2018, 72, 1763–1773. [Google Scholar] [CrossRef]
  143. Di Franco, A.; Cantini, G.; Tani, A.; Coppini, R.; Zecchi-Orlandini, S.; Raimondi, L.; Luconi, M.; Mannucci, E. Sodium-Dependent Glucose Transporters (SGLT) in Human Ischemic Heart: A New Potential Pharmacological Target. Int. J. Cardiol. 2017, 243, 86–90. [Google Scholar] [CrossRef]
  144. Unity Health Toronto. Effects of Empagliflozin on Cardiac Structure, Function, and Circulating Biomarkers in Patients With Type 2 Diabetes; National Library of Medicine: Bethesda, MD, USA, 2018; clinicaltrials.gov.
  145. Unity Health Toronto. Empagliflozin and Cardiac Remodelling in People Without Diabetes; National Library of Medicine: Bethesda, MD, USA, 2023; clinicaltrials.gov.
  146. Getz Pharma. Safety And Efficacy Of Empagliflozin In Pakistani Muslim Population With Type Ii Diabetes Mellitus; National Library of Medicine: Bethesda, MD, USA, 2020; clinicaltrials.gov.
  147. Liang, Y.; Arakawa, K.; Ueta, K.; Matsushita, Y.; Kuriyama, C.; Martin, T.; Du, F.; Liu, Y.; Xu, J.; Conway, B.; et al. Effect of Canagliflozin on Renal Threshold for Glucose, Glycemia, and Body Weight in Normal and Diabetic Animal Models. PLoS ONE 2012, 7, e30555. [Google Scholar] [CrossRef]
  148. Li, X.; Lu, Q.; Qiu, Y.; do Carmo, J.M.; Wang, Z.; da Silva, A.A.; Mouton, A.; Omoto, A.C.M.; Hall, M.E.; Li, J.; et al. Direct Cardiac Actions of the Sodium Glucose Co-Transporter 2 Inhibitor Empagliflozin Improve Myocardial Oxidative Phosphorylation and Attenuate Pressure-Overload Heart Failure. J. Am. Heart Assoc. 2021, 10, e018298. [Google Scholar] [CrossRef]
  149. Filippatos, T.D.; Liontos, A.; Papakitsou, I.; Elisaf, M.S. SGLT2 Inhibitors and Cardioprotection: A Matter of Debate and Multiple Hypotheses. Postgrad. Med. 2019, 131, 82–88. [Google Scholar] [CrossRef] [PubMed]
  150. Trum, M.; Riechel, J.; Wagner, S. Cardioprotection by SGLT2 Inhibitors—Does It All Come Down to Na+? Int. J. Mol. Sci. 2021, 22, 7976. [Google Scholar] [CrossRef] [PubMed]
  151. Ion Channels in the Heart—Bartos—Major Reference Works—Wiley Online Library. Available online: https://onlinelibrary.wiley.com/doi/10.1002/cphy.c140069 (accessed on 24 September 2023).
  152. Bertero, E.; Prates Roma, L.; Ameri, P.; Maack, C. Cardiac Effects of SGLT2 Inhibitors: The Sodium Hypothesis. Cardiovasc. Res. 2018, 114, 12–18. [Google Scholar] [CrossRef] [PubMed]
  153. Bell, R.M.; Yellon, D.M. SGLT2 Inhibitors: Hypotheses on the Mechanism of Cardiovascular Protection. Lancet Diabetes Endocrinol. 2018, 6, 435–437. [Google Scholar] [CrossRef]
  154. Mascolo, A.; Di Napoli, R.; Balzano, N.; Cappetta, D.; Urbanek, K.; De Angelis, A.; Scisciola, L.; Di Meo, I.; Sullo, M.G.; Rafaniello, C.; et al. Safety Profile of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors: A Brief Summary. Front. Cardiovasc. Med. 2022, 9, 1010693. [Google Scholar] [CrossRef]
  155. Chen, S.; Coronel, R.; Hollmann, M.W.; Weber, N.C.; Zuurbier, C.J. Direct Cardiac Effects of SGLT2 Inhibitors. Cardiovasc. Diabetol. 2022, 21, 45. [Google Scholar] [CrossRef]
  156. Baartscheer, A.; Schumacher, C.A.; van Borren, M.M.G.J.; Belterman, C.N.W.; Coronel, R.; Opthof, T.; Fiolet, J.W.T. Chronic Inhibition of Na+/H+-Exchanger Attenuates Cardiac Hypertrophy and Prevents Cellular Remodeling in Heart Failure. Cardiovasc. Res. 2005, 65, 83–92. [Google Scholar] [CrossRef]
  157. Inhibiting Mitochondrial Na+/Ca2+ Exchange Prevents Sudden Death in a Guinea Pig Model of Heart Failure|Circulation Research. Available online: https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.115.303062 (accessed on 24 September 2023).
  158. Baartscheer, A.; Schumacher, C.A.; Wüst, R.C.I.; Fiolet, J.W.T.; Stienen, G.J.M.; Coronel, R.; Zuurbier, C.J. Empagliflozin Decreases Myocardial Cytoplasmic Na+ through Inhibition of the Cardiac Na+/H+ Exchanger in Rats and Rabbits. Diabetologia 2017, 60, 568–573. [Google Scholar] [CrossRef]
  159. Uthman, L.; Baartscheer, A.; Bleijlevens, B.; Schumacher, C.A.; Fiolet, J.W.T.; Koeman, A.; Jancev, M.; Hollmann, M.W.; Weber, N.C.; Coronel, R.; 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. [Google Scholar] [CrossRef]
  160. SGLT2 Inhibitors and the Cardiac Na+/H+ Exchanger-1: The Plot Thickens | Cardiovascular Research|Oxford Academic. Available online: https://academic.oup.com/cardiovascres/article/117/14/2702/6288488?login=false (accessed on 24 September 2023).
  161. The Sodium–Glucose Cotransporter 2 Inhibitor Dapagliflozin Prevents Cardiomyopathy in a Diabetic Lipodystrophic Mouse Model|Diabetes|American Diabetes Association. Available online: https://diabetesjournals.org/diabetes/article/66/4/1030/16012/The-Sodium-Glucose-Cotransporter-2-Inhibitor (accessed on 24 September 2023).
  162. Hammoudi, N.; Jeong, D.; Singh, R.; Farhat, A.; Komajda, M.; Mayoux, E.; Hajjar, R.; Lebeche, D. Empagliflozin Improves Left Ventricular Diastolic Dysfunction in a Genetic Model of Type 2 Diabetes. Cardiovasc. Drugs Ther. 2017, 31, 233–246. [Google Scholar] [CrossRef]
  163. Cumhur Cure, M.; Cure, E. Effects of the Na+/H+ Ion Exchanger on Susceptibility to COVID-19 and the Course of the Disease. J. Renin-Angiotensin-Aldosterone Syst. 2021, 2021, e4754440. [Google Scholar] [CrossRef] [PubMed]
  164. Boedtkjer, E.; Aalkjaer, C. Intracellular pH in the Resistance Vasculature: Regulation and Functional Implications. JVR 2012, 49, 479–496. [Google Scholar] [CrossRef] [PubMed]
  165. Kazyken, D.; Lentz, S.I.; Fingar, D.C. Alkaline Intracellular pH Activates AMPK-mTORC2 Signaling to Promote Cell Survival during Growth Factor Limitation. J. Biol. Chem. 2021, 297, 101100. [Google Scholar] [CrossRef]
  166. Baker, H.E.; Kiel, A.M.; Luebbe, S.T.; Simon, B.R.; Earl, C.C.; Regmi, A.; Roell, W.C.; Mather, K.J.; Tune, J.D.; Goodwill, A.G. Inhibition of Sodium–Glucose Cotransporter-2 Preserves Cardiac Function during Regional Myocardial Ischemia Independent of Alterations in Myocardial Substrate Utilization. Basic. Res. Cardiol. 2019, 114, 25. [Google Scholar] [CrossRef] [PubMed]
  167. Chang, C.; Su, H.; Zhang, D.; Wang, Y.; Shen, Q.; Liu, B.; Huang, R.; Zhou, T.; Peng, C.; Wong, C.C.L.; et al. AMPK-Dependent Phosphorylation of GAPDH Triggers Sirt1 Activation and Is Necessary for Autophagy upon Glucose Starvation. Mol. Cell 2015, 60, 930–940. [Google Scholar] [CrossRef] [PubMed]
  168. Ghosh, H.S.; McBurney, M.; Robbins, P.D. SIRT1 Negatively Regulates the Mammalian Target of Rapamycin. PLoS ONE 2010, 5, e9199. [Google Scholar] [CrossRef]
  169. Sun, X.; Han, F.; Lu, Q.; Li, X.; Ren, D.; Zhang, J.; Han, Y.; Xiang, Y.K.; Li, J. Empagliflozin Ameliorates Obesity-Related Cardiac Dysfunction by Regulating Sestrin2-Mediated AMPK-mTOR Signaling and Redox Homeostasis in High-Fat Diet–Induced Obese Mice. Diabetes 2020, 69, 1292–1305. [Google Scholar] [CrossRef]
  170. Pan, C.; Chen, Z.; Li, C.; Han, T.; Liu, H.; Wang, X. Sestrin2 as a Gatekeeper of Cellular Homeostasis: Physiological Effects for the Regulation of Hypoxia-related Diseases. J. Cell Mol. Med. 2021, 25, 5341–5350. [Google Scholar] [CrossRef]
  171. Gong, L.; Wang, Z.; Wang, Z.; Zhang, Z. Sestrin2 as a Potential Target for Regulating Metabolic-Related Diseases. Front. Endocrinol. 2021, 12, 751020. [Google Scholar] [CrossRef]
  172. Zhang, L.; Zhang, H.; Xie, X.; Tie, R.; Shang, X.; Zhao, Q.; Xu, J.; Jin, L.; Zhang, J.; Ye, P. Empagliflozin Ameliorates Diabetic Cardiomyopathy via Regulated Branched-Chain Amino Acid Metabolism and mTOR/p-ULK1 Signaling Pathway-Mediated Autophagy. Diabetol. Metab. Syndr. 2023, 15, 93. [Google Scholar] [CrossRef]
  173. Feng, B.; Yu, P.; Yu, H.; Qian, B.; Li, Y.; Sun, K.; Shi, B.; Zhang, N.; Xu, G. Therapeutic Effects on the Development of Heart Failure with Preserved Ejection Fraction by the Sodium-Glucose Cotransporter 2 Inhibitor Dapagliflozin in Type 2 Diabetes. Diabetol. Metab. Syndr. 2023, 15, 141. [Google Scholar] [CrossRef] [PubMed]
  174. Lahnwong, S.; Chattipakorn, S.C.; Chattipakorn, N. Potential Mechanisms Responsible for Cardioprotective Effects of Sodium–Glucose Co-Transporter 2 Inhibitors. Cardiovasc. Diabetol. 2018, 17, 101. [Google Scholar] [CrossRef] [PubMed]
  175. 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. [Google Scholar] [CrossRef] [PubMed]
  176. Shi, X.; Verma, S.; Yun, J.; Brand-Arzamendi, K.; Singh, K.K.; Liu, X.; Garg, A.; Quan, A.; Wen, X.-Y. Effect of Empagliflozin on Cardiac Biomarkers in a Zebrafish Model of Heart Failure: Clues to the EMPA-REG OUTCOME Trial? Mol. Cell Biochem. 2017, 433, 97–102. [Google Scholar] [CrossRef]
  177. Ye, Y.; Bajaj, M.; Yang, H.-C.; Perez-Polo, J.R.; 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. [Google Scholar] [CrossRef] [PubMed]
  178. Takimoto, E.; Kass, D.A. Role of Oxidative Stress in Cardiac Hypertrophy and Remodeling. Hypertension 2007, 49, 241–248. [Google Scholar] [CrossRef]
  179. Habibi, J.; Aroor, A.R.; Sowers, J.R.; Jia, G.; Hayden, M.R.; Garro, M.; Barron, B.; Mayoux, E.; Rector, R.S.; Whaley-Connell, A.; 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. [Google Scholar] [CrossRef]
  180. Zhou, Y.; Wu, W. The Sodium-Glucose Co-Transporter 2 Inhibitor, Empagliflozin, Protects against Diabetic Cardiomyopathy by Inhibition of the Endoplasmic Reticulum Stress Pathway. Cell. Physiol. Biochem. 2017, 41, 2503–2512. [Google Scholar] [CrossRef]
  181. Rapamycin Attenuates High Glucose-Induced Inflammation Through Modulation of mTOR/NF-κB Pathways in Macrophages —PMC. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6831745/ (accessed on 24 September 2023).
  182. Gong, H.; Tai, H.; Huang, N.; Xiao, P.; Mo, C.; Wang, X.; Han, X.; Zhou, J.; Chen, H.; Tang, X.; et al. Nrf2-SHP Cascade-Mediated STAT3 Inactivation Contributes to AMPK-Driven Protection Against Endotoxic Inflammation. Front. Immunol. 2020, 11, 414. [Google Scholar] [CrossRef]
  183. Costello, R.A.; Nicolas, S.; Shivkumar, A. Sulfonylureas. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  184. Corcoran, C.; Jacobs, T.F. Metformin. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  185. Kasina, S.V.S.K.; Baradhi, K.M. Dipeptidyl Peptidase IV (DPP IV) Inhibitors. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  186. Collins, L.; Costello, R.A. Glucagon-Like Peptide-1 Receptor Agonists. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  187. Filion, K.B.; Lix, L.M.; Yu, O.H.; Dell’Aniello, S.; Douros, A.; Shah, B.R.; St-Jean, A.; Fisher, A.; Tremblay, E.; Bugden, S.C.; et al. Sodium Glucose Cotransporter 2 Inhibitors and Risk of Major Adverse Cardiovascular Events: Multi-Database Retrospective Cohort Study. BMJ 2020, 370, m3342. [Google Scholar] [CrossRef]
  188. Chen, T.-H.; Li, Y.-R.; Chen, S.-W.; Lin, Y.-S.; Sun, C.-C.; Chen, D.-Y.; Mao, C.-T.; Wu, M.; Chang, C.-H.; Chu, P.-H.; et al. Sodium-Glucose Cotransporter 2 Inhibitor versus Metformin as First-Line Therapy in Patients with Type 2 Diabetes Mellitus: A Multi-Institution Database Study. Cardiovasc. Diabetol. 2020, 19, 189. [Google Scholar] [CrossRef] [PubMed]
  189. Xie, Y.; Bowe, B.; Gibson, A.K.; McGill, J.B.; Maddukuri, G.; Al-Aly, Z. Comparative Effectiveness of Sodium-Glucose Cotransporter 2 Inhibitors vs Sulfonylureas in Patients With Type 2 Diabetes. JAMA Intern. Med. 2021, 181, 1043–1053. [Google Scholar] [CrossRef] [PubMed]
  190. Borghi, C.; Bragagni, A. The New Type 2 Diabetes Mellitus Therapy: Comparison between the Two Classes of Drugs GLPR (Glucagon-like Peptide Receptor) Agonists and SGLT2 (Sodium–Glucose Cotransporter 2) Inhibitors. Eur. Heart J. Suppl. 2020, 22, L28–L32. [Google Scholar] [CrossRef] [PubMed]
  191. Zelniker, T.A.; Wiviott, S.D.; Raz, I.; Im, K.; Goodrich, E.L.; Bonaca, M.P.; Mosenzon, O.; Kato, E.T.; Cahn, A.; Furtado, R.H.M.; et al. SGLT2 Inhibitors for Primary and Secondary Prevention of Cardiovascular and Renal Outcomes in Type 2 Diabetes: A Systematic Review and Meta-Analysis of Cardiovascular Outcome Trials. Lancet 2019, 393, 31–39. [Google Scholar] [CrossRef] [PubMed]
  192. Primary Prevention of Cardiovascular and Heart Failure Events With SGLT2 Inhibitors, GLP-1 Receptor Agonists, and Their Combination in Type 2 Diabetes|Diabetes Care|American Diabetes Association. Available online: https://diabetesjournals.org/care/article/45/4/909/141051/Primary-Prevention-of-Cardiovascular-and-Heart (accessed on 25 September 2023).
  193. Wu, H.; Xiao, C.; Zhao, Y.; Yin, H.; Yu, M. Liraglutide Improves Endothelial Function via the mTOR Signaling Pathway. J. Diabetes Res. 2021, 2021, 2936667. [Google Scholar] [CrossRef]
  194. Li, C.; Luo, J.; Jiang, M.; Wang, K. The Efficacy and Safety of the Combination Therapy With GLP-1 Receptor Agonists and SGLT-2 Inhibitors in Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2022, 13, 838277. [Google Scholar] [CrossRef]
  195. Fünfstück, R.; Nicolle, L.E.; Hanefeld, M.; Naber, K.G. Urinary Tract Infection in Patients with Diabetes Mellitus. Clin. Nephrol. 2012, 77, 40–48. [Google Scholar] [CrossRef]
  196. Wang, M.-C.; Tseng, C.-C.; Wu, A.-B.; Lin, W.-H.; Teng, C.-H.; Yan, J.-J.; Wu, J.-J. Bacterial Characteristics and Glycemic Control in Diabetic Patients with Escherichia Coli Urinary Tract Infection. J. Microbiol. Immunol. Infect. 2013, 46, 24–29. [Google Scholar] [CrossRef]
  197. Akkus, E. Urinary Tract Infection Risk in Women with Asymptomatic Bacteriuria or Urinalysis Abnormality at the Initiation of SGLT2 Inhibitors; National Library of Medicine: Bethesda, MD, USA, 2022; clinicaltrials.gov.
  198. Inventiva Pharma. A Placebo-Controlled, Proof-of-Concept Study to Evaluate the Safety and Efficacy of Lanifibranor Alone and in Combination With the Sodium-Glucose Transport Protein 2 (SGLT2) Inhibitor EmpaGliflozin in patiEnts With Non-Alcoholic Steatohepatitis (NASH) and Type 2 Diabetes Mellitus (T2DM); National Library of Medicine: Bethesda, MD, USA, 2023; clinicaltrials.gov.
  199. Shing, C.K. Effect of Empagliflozin on Liver Fat in Non-Alcoholic Fatty Liver Disease Patients Without Diabetes Mellitus: A Randomized, Double-Blind, Placebo-Controlled Trial; National Library of Medicine: Bethesda, MD, USA, 2022; clinicaltrials.gov.
  200. Yonsei University. Comparison of The Effects of Thiazolidinediones(TZD), Sodium- Glucose Cotransporter 2 Inhibitors(SGLT2i) Alone and TZD/SGLT2i Combination Therapy on Non-Alcoholic Fatty Liver Disease in Type 2 Diabetic Patients With Fatty Liver; National Library of Medicine: Bethesda, MD, USA, 2020; clinicaltrials.gov.
  201. Mahmud, F. Adolescent Type 1 Diabetes Treatment With SGLT2i for hyperglycEMia & hyPerfilTration Trial; National Library of Medicine: Bethesda, MD, USA, 2022; clinicaltrials.gov.
  202. Alaa, N. Evaluation of the Effect of Sodium-Glucose Cotransporter-2 (SGLT2) Inhibitors on Diabetic Retinopathy in Patients With Type 2 Diabetes Mellitus; National Library of Medicine: Bethesda, MD, USA, 2022; clinicaltrials.gov.
Figure 1. Schematic representation of diabetic cardiomyopathy pathophysiology. Insulin resistance in type 2 diabetes mellitus mediates systemic hyperglycemia and hyperlipidemia. These conditions induce metabolic changes in the heart and endothelial system, leading to mitochondrial dysfunction causing calcium imbalance and oxidative stress. As a result, other insults like inflammation, hypertrophy, and fibrosis arise as interdependent factors and culminate into cardiac dysfunction and progressive heart failure.
Figure 1. Schematic representation of diabetic cardiomyopathy pathophysiology. Insulin resistance in type 2 diabetes mellitus mediates systemic hyperglycemia and hyperlipidemia. These conditions induce metabolic changes in the heart and endothelial system, leading to mitochondrial dysfunction causing calcium imbalance and oxidative stress. As a result, other insults like inflammation, hypertrophy, and fibrosis arise as interdependent factors and culminate into cardiac dysfunction and progressive heart failure.
Ijms 24 15078 g001
Figure 2. mTORC1 and mTORC2 complexes and downstream cellular functions. mTORC1 is composed of a core complex of mTOR, Raptor, and mLST8, which is inhibited by DEPTOR and PRAS40. mTORC2 comprises a core of mTOR, Rictor, and mLST8 that is inhibited by DEPTOR and regulated by Protor1/2 and mSin1. mTORC1 versus mTORC2 activation affects diverse cellular functions.
Figure 2. mTORC1 and mTORC2 complexes and downstream cellular functions. mTORC1 is composed of a core complex of mTOR, Raptor, and mLST8, which is inhibited by DEPTOR and PRAS40. mTORC2 comprises a core of mTOR, Rictor, and mLST8 that is inhibited by DEPTOR and regulated by Protor1/2 and mSin1. mTORC1 versus mTORC2 activation affects diverse cellular functions.
Ijms 24 15078 g002
Figure 3. The cardiac mTORC1 signaling network. In normal conditions, IR-mediated PI3K/Akt activation leads to activated mTORC1, which inhibits autophagy via ULK1 and promotes protein synthesis of inflammatory and proliferative markers via S6K1 and eIF4E. In diabetic cardiomyopathy, mTORC1 is hyperactivated due to AMPK inhibition by hyperglycemia-mediated high APT:AMP ratio and mTORC1 hyperactivation induces a negative feedback loop to inhibit Akt, resulting in insulin resistance. Besides insulin resistance, mTORC1 hyperactivation also leads to dysregulated lipid synthesis and mitochondrial biogenesis, resulting in ROS upregulation and cardiac dysfunction. eIF4E—eukaryotic initiation factor 4E; IR—insulin receptor; IRS1—insulin receptor substrate 1; GLUT—glucose transporter; SLC—solute carrier group of membrane transporters; Rag—Ras-related GTPase; Rheb—Ras homolog enriched in brain; ULK—Unc-51-like kinase; ATG—autophagy-related protein.
Figure 3. The cardiac mTORC1 signaling network. In normal conditions, IR-mediated PI3K/Akt activation leads to activated mTORC1, which inhibits autophagy via ULK1 and promotes protein synthesis of inflammatory and proliferative markers via S6K1 and eIF4E. In diabetic cardiomyopathy, mTORC1 is hyperactivated due to AMPK inhibition by hyperglycemia-mediated high APT:AMP ratio and mTORC1 hyperactivation induces a negative feedback loop to inhibit Akt, resulting in insulin resistance. Besides insulin resistance, mTORC1 hyperactivation also leads to dysregulated lipid synthesis and mitochondrial biogenesis, resulting in ROS upregulation and cardiac dysfunction. eIF4E—eukaryotic initiation factor 4E; IR—insulin receptor; IRS1—insulin receptor substrate 1; GLUT—glucose transporter; SLC—solute carrier group of membrane transporters; Rag—Ras-related GTPase; Rheb—Ras homolog enriched in brain; ULK—Unc-51-like kinase; ATG—autophagy-related protein.
Ijms 24 15078 g003
Figure 4. Canonical and non-canonical (cardiac) mechanism for SGLT2i function in diabetic cardiomyopathy. Luminal glucose in the renal glomerular filtrate is reabsorbed (80–90%) into the proximal tubular epithelial cells via SGLT2 in the S1 segment of proximal convoluted tubule (PCT) of nephrons, and subsequently transported to the basolateral interstitial fluid through GLUT2. In diabetic cardiomyopathy therapy, SGLT2is act in a canonical manner by preventing renal glucose reabsorption to reduce overall hyperglycemia but studies indicate a non-canonical mechanism of SGLT2is in the heart. Owing to the absence of SGLT2 channels in the heart, selective SGLT2is can act on prolonged activated NHE to reduce intracellular sodium ions and subsequently promote an alkaline intracellular pH, which has been reported to activate AMPK and inhibit mTORC1. SGLT2 inhibitors have also been reported to mimic an energy-deprived environment that might promote Sestrin2-mediated AMPK and SIRT1 activation, leading to mTORC1 inactivation and cardiovascular benefits. PCT—proximal convoluted tubule; GLUT2—glucose transporter 2; NHE—sodium–hydrogen exchanger; NCX—sodium–calcium exchanger. ↑Nai+/↑Cai2+/↑mTORC1/↑ROS indicate an increase in intracellular Na+, Ca2+, mTORC1, and ROS levels; faded tail → through the NHE/GLUT2/SGLT2 channels indicates the flow of molecules/ions; dotted – → indicates indirect effects and solid → indicates direct activation/induction; blunt Ijms 24 15078 i001 indicates inhibition.
Figure 4. Canonical and non-canonical (cardiac) mechanism for SGLT2i function in diabetic cardiomyopathy. Luminal glucose in the renal glomerular filtrate is reabsorbed (80–90%) into the proximal tubular epithelial cells via SGLT2 in the S1 segment of proximal convoluted tubule (PCT) of nephrons, and subsequently transported to the basolateral interstitial fluid through GLUT2. In diabetic cardiomyopathy therapy, SGLT2is act in a canonical manner by preventing renal glucose reabsorption to reduce overall hyperglycemia but studies indicate a non-canonical mechanism of SGLT2is in the heart. Owing to the absence of SGLT2 channels in the heart, selective SGLT2is can act on prolonged activated NHE to reduce intracellular sodium ions and subsequently promote an alkaline intracellular pH, which has been reported to activate AMPK and inhibit mTORC1. SGLT2 inhibitors have also been reported to mimic an energy-deprived environment that might promote Sestrin2-mediated AMPK and SIRT1 activation, leading to mTORC1 inactivation and cardiovascular benefits. PCT—proximal convoluted tubule; GLUT2—glucose transporter 2; NHE—sodium–hydrogen exchanger; NCX—sodium–calcium exchanger. ↑Nai+/↑Cai2+/↑mTORC1/↑ROS indicate an increase in intracellular Na+, Ca2+, mTORC1, and ROS levels; faded tail → through the NHE/GLUT2/SGLT2 channels indicates the flow of molecules/ions; dotted – → indicates indirect effects and solid → indicates direct activation/induction; blunt Ijms 24 15078 i001 indicates inhibition.
Ijms 24 15078 g004
Table 1. Therapeutic targets of diabetic cardiomyopathy and associated clinical trials.
Table 1. Therapeutic targets of diabetic cardiomyopathy and associated clinical trials.
Therapeutic TargetDrug TreatmentStudyDesign of StudyStudy Outcomes
PPARαBlinded fenofibrate or placebo plus simvastatinACCORD (1999–2012) [24,25]Randomized, double-blind,
placebo-controlled
phase III trial in T2D patients (actual enrollment—10251)
For the primary outcome, cardiovascular risk was lower in the intense glycemia and blood pressure (BP) groups, compared to combined standard BP and glycemia treatment. For secondary outcomes, myocardial infarction and stroke were significantly reduced by intensive glycemia and BP treatment. There were more adverse effects but no statistically significant benefit or harm in terms of total mortality and cardiovascular disease mortality for any intensively treated groups compared to standard.
PDE5ASildenafil or placeboCECSID (2008–2009) [26,27]Randomized, double-blind,
placebo-controlled
phase IV trial in male T2D patients (Actual Enrollment- 59)
The study showed an improved ratio of left ventricular mass to end diastolic volume and LV contraction, besides reducing TGFβ levels and demonstrating an anti-remodeling effect. Endothelial function or cardiac metabolism were not affected, and no significant differences were found in glycemia, insulin, c-peptide, or lipid profile.
GLP1RLiraglutide or placeboLEADER (2010–2015) [28,29]Multi-center, randomized,
double-blind, placebo-controlled
phase III trial
in T2D patients (Actual Enrollment- 9341)
Liraglutide significantly reduced cardiovascular (CV) outcomes in patients with myocardial infarction (MI)/stroke history or having atherosclerotic CV diseases without MI/stroke history, but no improvement was reported in patients with only CV risk. In all the three groups, the percentage of adverse gastrointestinal events ranged from 55–65%.
IL-1βCanakinumab or placebo or standard of careCANTOS (2011–2019) [30,31]Randomized, double-blind,
placebo-controlled,
event-driven phase III
trial in patients with myocardial infarction and elevated hsCRP levels with/without
T2D (actual enrollment—10066)
Canakinumab reduced hsCRP and IL6 levels in patients with or without diabetes, thereby reducing recurrent cardiovascular events and heart failure hospitalizations, but did not reduce new-onset diabetes. Furthermore, the treatment had no long-term benefits on HbA1c or fasting plasma glucose.
NRF2Sulforaphane or placeboClinical trial with broccoli sprout extract to patients with type 2 diabetes (2015–2020) [32,33]Randomized, double-blind,
placebo-controlled
phase II trial in T2D patients (actual enrollment—103)
Sulforaphane improved HbA1c and fasting glucose levels in
patients with obesity and T2D but the study was not focused on cardiovascular health or outcomes. No severe adverse effects were observed.
Table 2. Phase III clinical trials of SGLT2is for diabetic cardiac dysfunction.
Table 2. Phase III clinical trials of SGLT2is for diabetic cardiac dysfunction.
Study and DurationTreatmentTotal EnrollmentKey Inclusion CriteriaStudy OutcomesAdverse/Side EffectsStudy Limitations
EMPA-REG OUTCOME
2010–2015 [100,105,109,110]
Empagliflozin vs. placebo7064Patients with T2D and high-risk/established cardiovascular disorders.Reduction in cardiovascular death and non-fatal myocardial infarction.
For EMPA-REG post hoc analysis, refer to [111,112].
Moderate benign mycotic genital infections.The study lacked adjustment for background medications in-trial and had a controversial post hoc nature [113,114].
CANVAS
2009–2017 [30,115]
Canagliflozin vs. placebo4330Patients with T2D and high cardiovascular risk; enrolled women population post-menopausal or on a birth-control regime.Reduction in the composite of cardiovascular deaths, non-fatal myocardial infarction, and non-fatal stroke.
For CANVAS post hoc analysis, refer to [116,117].
Risk of amputation (metatarsal) and moderate risk of genital infections.The program had a relatively small participant proportion, indicating moderate number of events for health outcomes and increasing the risk of false positive findings [115].
DECLARE-TIMI 58
2013–2018 [118,119]
Dapagliflozin vs. placebo17,190Patients with diabetes mellitus and non-insulin-dependent cardiovascular risk. Lower glycated hemoglobin along with lower rates of cardiovascular diseases and hospitalization.
For DECLARE-TIMI post hoc analysis, refer to [120].
Moderate genital infections.Low African American and Hispanic study population precludes any definite understanding of ethnicity-based treatment outcomes. Moreover, blood pressure subanalysis categories were not prespecified in the study [121,122].
VETRIS CV
2013–2019 [123,124]
Ertugliflozin vs. placebo with background glycemic rescue8246Patients with T2D and established cardiovascular diseases.Incidence of cardiovascular deaths and heart failure hospitalizations did not differ significantly between the ertugliflozin and placebo groups.
For VETRIS CV post hoc analysis, refer to [125].
Amputation risk in ~2% patients of the ertugliflozin groups. The study population was predominantly white and male, limiting ethnicity and sex-based study interpretation. Moreover, differences in baseline characteristics in some subgroups might affect the influence of background medications on observed Ertugliflozin effects [126].
DAPA-HF
2017–2019 [106,127]
Dapagliflozin vs. placebo4744Patients with <40% ejection fraction and symptomatic heart failure; 50% of patients with T2D. Reduction in cardiovascular deaths and heart failure hospitalizations for both diabetic and non-diabetic patients.
For DAPA-HF post hoc analysis, refer to [128,129].
No significant excess of genital infection or amputations observed between the dapagliflozin and placebo groups.The main limitation of the DAPA-HF trial included a reduced population of Black patients (<5%), elderly patients with co-morbidities (>66 years), and patients with sacubitril-valsartan at baseline [106].
EMPEROR-Reduced
2017–2020 [108,130]
Empagliflozin vs. placebo3730Patients with <40% ejection fraction and chronic heart failure risk; 50% of the patients with T2D.Reduction in cardiovascular risk and heart failure hospitalizations in both diabetic and non-diabetic patients.
For EMPEROR-Reduced post hoc analysis, refer to [131,132].
Uncomplicated genital tract infections observed in the empagliflozin group.The median follow-up duration was very limited (16 months) and outpatient events were not adjudicated or reviewed [133].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Saha, S.; Fang, X.; Green, C.D.; Das, A. mTORC1 and SGLT2 Inhibitors—A Therapeutic Perspective for Diabetic Cardiomyopathy. Int. J. Mol. Sci. 2023, 24, 15078. https://doi.org/10.3390/ijms242015078

AMA Style

Saha S, Fang X, Green CD, Das A. mTORC1 and SGLT2 Inhibitors—A Therapeutic Perspective for Diabetic Cardiomyopathy. International Journal of Molecular Sciences. 2023; 24(20):15078. https://doi.org/10.3390/ijms242015078

Chicago/Turabian Style

Saha, Sumit, Xianjun Fang, Christopher D. Green, and Anindita Das. 2023. "mTORC1 and SGLT2 Inhibitors—A Therapeutic Perspective for Diabetic Cardiomyopathy" International Journal of Molecular Sciences 24, no. 20: 15078. https://doi.org/10.3390/ijms242015078

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop