Next Article in Journal
The Role of Vitamin D in Kidney Transplantation Outcomes: A Systematic Review
Next Article in Special Issue
Metformin Acutely Mitigates Oxidative Stress in Human Atrial Tissue: A Pilot Study in Overweight Non-Diabetic Cardiac Patients
Previous Article in Journal
Evaluation of the Antimicrobial Activity of ZnO Nanoparticles against Enterotoxigenic Staphylococcus aureus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oxidative Stress Management in Cardiorenal Diseases: Focus on Novel Antidiabetic Agents, Finerenone, and Melatonin

by
Panagiotis Theofilis
,
Aikaterini Vordoni
and
Rigas G. Kalaitzidis
*
Center for Nephrology “G. Papadakis”, General Hospital of Nikaia-Piraeus Agios Panteleimon, 18454 Piraeus, Greece
*
Author to whom correspondence should be addressed.
Life 2022, 12(10), 1663; https://doi.org/10.3390/life12101663
Submission received: 3 October 2022 / Revised: 15 October 2022 / Accepted: 19 October 2022 / Published: 20 October 2022
(This article belongs to the Special Issue Research Updates on Oxidative Stress and Cardiovascular Disease)

Abstract

:
Oxidative stress is characterized by excessive production of reactive oxygen species together with exhausted antioxidant defenses. This constitutes a main pathophysiologic process that is implicated in cardiovascular and renal diseases. In particular, enhanced oxidative stress may lead to low-density lipoprotein accumulation and oxidation, endothelial cell activation, adhesion molecule overexpression, macrophage activation, and foam cell formation, promoting the development and progression of atherosclerosis. The deleterious kidney effects of oxidative stress are numerous, including podocytopathy, mesangial enlargement, renal hypertrophy, tubulointerstitial fibrosis, and glomerulosclerosis. The prominent role of oxidative mechanisms in cardiorenal diseases may be counteracted by recently developed pharmacotherapies such as novel antidiabetic agents and finerenone. These agents have demonstrated significant antioxidant activity in preclinical and clinical studies. Moreover, the use of melatonin as a treatment in this field has been experimentally investigated, with large-scale clinical studies being awaited. Finally, clinical implications and future directions in this field are presented.

1. Introduction

Cardiovascular and renal diseases are leading causes of morbidity and mortality worldwide. The epidemiological trends of these non-communicable diseases are worrisome, with an increasing incidence and prevalence, especially in developing countries [1,2,3]. Moreover, owing to shared risk factors, these entities frequently coexist. Their pathophysiology is complex, comprising multiple pathways. Among them, oxidative stress is crucial in the development of atherosclerotic diseases, heart failure (HF), and chronic kidney disease (CKD) (Figure 1). Pharmacotherapies with pleiotropic effects, such as sodium-glucose cotransporter-2 (SGLT2) inhibitors, glucagon-like peptide-1 receptor agonists (GLP1-RA), and finerenone have recently been found to reduce the burden associated with these pathologic states. In this narrative review, we discuss the role of oxidative stress in cardiovascular diseases and CKD. Moreover, we present the available preclinical and clinical evidence regarding the antioxidant potential of agents with pleiotropic effects, such as SGLT2 inhibitors, GLP1-RA, and finerenone in cardiac and renal pathologies. Finally, we discuss the importance of melatonin (MT), an endogenous hormone acting mainly through antioxidant mechanisms to ameliorate cardiorenal diseases, according to a plethora of preclinical evidence.

2. Oxidative Stress in Cardiorenal Diseases

In live cells, during regular metabolic activities, reactive chemicals such as reactive oxygen species (ROS) and reactive nitrogen species are constantly produced by oxidation reactions, being either enzymatic or nonenzymatic. Free or primary radicals are independent chemical entities containing one or more unpaired electrons. These radicals are extremely reactive while looking for another unpaired electron. A new nonradical molecule is created when two unpaired electrons interact to form a covalent link; but more frequently, when free radicals interact with nonradical molecules, they produce new (secondary) radical molecules that start the chain reaction. Multiple tissues and organs eventually sustain oxidative damage as a result of this main and secondary radical chain reaction.
Numerous ROS-generating systems exist and are associated with the development and progression of cardiorenal diseases. To begin with, NADPH oxidases are among the main representatives. Being present in infiltrating macrophages and vascular wall cells, they comprise two membrane-bound subunits (p22phox and a Nox homolog) and a few cytosolic regulatory subunits [4]. They act by producing superoxide (or hydrogen peroxide in the case of Nox4) from molecular oxygen using NADPH as the electron donor. Experimental evidence in mice suggests the presence of three Nox isoforms in vascular smooth muscle cells (Nox1, Nox4) and endothelial cells (Nox2, Nox4) [5,6,7,8]. The role of Nox in atherosclerosis is diverse, with Nox1 and Nox2 being proatherogenic [9,10,11], while Nox4 is antiatherogenic [12,13,14]. NAPDH may be considered an orchestrator of oxidative stress as it can promote endothelial nitric oxide synthase (eNOS) uncoupling, xanthine oxidase (XO) activity, and mitochondrial ROS production. Moving to XO, its prooxidant effect is based on the generation of superoxide and hydrogen peroxide by using molecular oxygen as an electron acceptor. Among the known triggers of XO production are angiotensin II and oscillatory shear stress [15,16]. XO ultimately adheres to endothelial cells through endothelial glycosaminoglycans [17]. The XO-formed superoxide may be responsible for endothelial cell dysfunction, the initial step of atherosclerosis [17]. Next, mitochondria are responsible for generating physiological levels of superoxide through oxidative phosphorylation. This is subsequently converted to hydrogen peroxide and, ultimately, to water. However, mitochondrial oxidative stress may occur due to the upregulated ROS production and impaired ROS degradation [18]. Finally, eNOS uncoupling is prevalent in cases of oxidative stress, leading to superoxide and peroxynitrite production instead of NO [19]. Those oxidative substances may further augment eNOS uncoupling through tetrahydrobiopterin inactivation, which is a known eNOS cofactor [19]. NO is a crucial mediator of cellular homeostasis as it can regulate vascular tone, prevent platelet activation and aggregation, attenuate leukocyte migration and adhesion, and inhibit vascular smooth muscle cell proliferation [19].
This protective mechanism functions by repeatedly preventing the initial formation of scavenging oxidants and free radicals. Through this, oxidants are changed into less hazardous chemicals, and the secondary generation of harmful metabolites is prevented. The defense system then seeks to fix the molecular damage or strengthen the body’s natural antioxidant defenses, which are made up of nonenzymatic and enzymatic antioxidants. The presence of exhausted antioxidant mechanisms is another point of concern when discussing the effect of oxidative stress on the cardiovascular and renal systems. Among the well-studied antioxidants are superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), nicotinamide adenine dinucleotide (NAD+), glutathione (GSH), paraoxonases (POX), and thioredoxins. It should be noted that although their upregulated expression induces anti-atherosclerotic effects, extreme overexpression of these antioxidant molecules may lead to proatherogenic effects, as in the case of SOD [20]. Certain polymorphisms in the genes encoding SOD1 (rs9974610, rs10432782, rs1041740) [21], SOD2 (rs4880) [22,23,24], SOD3 (rs1799895, rs7655372) [25,26,27], and GPx1 (rs1050450) [26,28,29,30] may also be responsible for atherosclerotic manifestations. Polymorphisms in the genes encoding antioxidant enzymes have been implicated in the development of nephropathy such as with SOD1 (rs17880135, rs202446, rs9974610, rs204732, rs17880135, rs17881180, rs1041740) [31,32], SOD2 (rs4880, rs2758329, rs8031) [33,34,35], and GPx1 (rs1050450) [33].
Cardiovascular and renal risk factors, namely, diabetes mellitus, arterial hypertension, smoking, and dyslipidemia, are known inducers of augmented oxidative stress. As a result, a series of deleterious sequelae that are associated with atherosclerosis, cardiac and renal dysfunction occur. Starting with atherosclerosis, oxidative stress may initially promote LDL uptake in the vessel wall, possibly due to impaired NO bioavailability [36]. Moving to oxidized LDL (oxLDL), oxidized phospholipids play a major role in mediating many of its proatherogenic traits. Lipid peroxidation can happen by enzymatic or nonenzymatic processes, such as ROS produced by NADPH oxidase or uncoupled eNOS, or by myeloperoxidases, lipoxygenases, cyclooxygenases, and cytochrome P450 [37]. Malondialdehyde, 4-hydroxynonenal, phosphocholine of oxidized phospholipid, and 2-(-carboxyethyl) pyrrole are among the extremely reactive byproducts of lipid peroxidation. They promote the production of structural neoepitopes known as oxidation-specific epitopes (OSEs) [38]. On the surface of apoptotic cells and oxLDL molecules, OSEs have been identified, including oxidized phospholipids and amino groups changed by malondialdehyde. OSEs are recognized by receptors (toll-like receptors (TLRs), scavenger receptors) on endothelial cells and macrophages [38]. This is important for human physiology since tissue homeostasis is maintained by removing dying cells, cellular debris, and damaged molecules [38]. However, when produced in excess, chronic inflammation through proinflammatory molecule secretion is promoted. Sensing of OSEs by endothelial cells leads to oxLDL uptake by lectin-like oxidized LDL receptor-1 (LOX1), TLR2, and TLR4 [38]. As a result, a reduced NO biosynthesis, smooth muscle cell proliferation, and upregulation of adhesion and prothrombotic molecules are noted. Other than OSE overexpression, ROS may mediate shear stress-induced adhesion molecule overexpression [39]. ROS also contribute to macrophage activation and foam cell formation through OSE generation and binding to scavenger receptors or LOX1 [38,40].
In the context of CKD, cellular oxidative stress is an important contributing factor by causing apoptosis, senescence, decreased cell regeneration, and fibrosis in the kidney cells. Extracellular matrix protein buildup, podocyte destruction, mesangial enlargement, renal hypertrophy, endothelial dysfunction, tubulointerstitial fibrosis, and glomerulosclerosis are all effects of oxidative stress [41]. Thus, the decline in renal function and the course of the disease are both further impacted by oxidative stress. Moreover, mitochondrial dysregulation in CKD patients leads to excessive ROS production as a result of abnormal oxidative phosphorylation, and intensifies oxidative stress. Patients with CKD have been discovered to have elevated levels of many oxidative phosphorylation-related genes [42]. Other enzymes that start the creation of ROS, such as Nox, XO, and lipoxygenases, are increased in CKD [43]. XO activity is also higher in CKD, while NO bioavailability is diminished [44,45]. As a result, the increased vascular resistance in renal arteries may promote hypertensive nephropathy [45]. Furthermore, CKD-induced vitamin D deficiency may also promote oxidative stress and aid the progression of CKD [46].
Importantly, chronic inflammation in CKD is mostly attributed to oxidative stress. It has been suggested that chronic low-grade inflammation contributes to the pathogenesis of CKD. Inflammation induced by kidney damage attracts leucocytes and macrophages, resulting in ROS overproduction. Accumulation of ROS engages macrophages and releases cytokines, chemokines, and eicosanoids, which in turn sets off a series of inflammatory responses. The modulation of glomerular filtration rate, renal blood flow, and sodium excretion by cytokines and inflammatory mediators like tumor necrosis factor (TNF), transforming growth factor, and interleukins (ILs) represents a downstream effect [47]. Nuclear factor (NF)-κB, a transcription factor that controls the expression of genes for inflammatory mediators, is also activated by oxidative stress [48]. I-κB, an inhibitory protein that keeps NF-κB in an inactive state, is phosphorylated and degraded by oxidative stress, which causes NF-κB to become active. Antioxidants prevent ROS from activating the NF-κB pathway [49]. The relationship between inflammation and oxidative stress in disease pathogenesis is supported by the high levels of inflammatory markers present in patients with advanced-stage CKD, including C-reactive protein, TNF-α, and IL-6, as well as oxidative stress markers, such as plasma protein carbonyls and F2-isoprostanes [50,51].

3. Antioxidant Pharmacotherapies in Cardiorenal Diseases

Many agents have been found with putative antioxidant effects in cardiorenal diseases. However, in this review, we will summarize the evidence on recently established, efficacious cardiorenal pharmacotherapies such as SGLT2 inhibitors, GLP1 receptor agonists, and finerenone. At the same time, we touch upon MT, a well-known endogenous substance which is recently being investigated preclinically in cardiorenal disease. Due to its antioxidant potential, it may end up being a useful addition against cardiorenal diseases’ pathophysiology. Crucially, although these agents act through different pathways, they have all been found to decrease ROS formation and NOX, while enhancing the antioxidant defenses, as discussed below.

3.1. SGLT2 Inhibitors

SGLT2 inhibitors have been initially introduced for the treatment of type 2 diabetes mellitus, by inhibiting the reabsorption of glucose in the proximal convoluted tubule. In the trials aimed to establish their cardiovascular safety, these agents were proven cardioprotective by significantly reducing HF hospitalizations, among others. Subsequent trials such as CREDENCE [52], DAPA-CKD [53], and SCORED [54] have documented their efficacy in CKD. Importantly, those findings were also validated in individuals without type 2 diabetes mellitus. Their evaluation in HF populations in the EMPEROR-REDUCED [55] and DAPA-HF [56] trials led to the inclusion of this drug category as a main therapy in the treatment of HF with a reduced left ventricular ejection fraction [57]. As we have shown, SGLT2 inhibitors can lead to the improvement of imaging indices of both systolic and diastolic cardiac function [58]. Lately, the results of SOLOIST-WHF [59], EMPEROR-PRESERVED [60], and DELIVER trials [61] made them the only drug class with positive results in HF with mildly reduced or preserved ejection fraction. Other than cardiorenal protection, these agents may improve fatty liver disease [62].
It is obvious that their mechanism of action is pleiotropic and is independent of the modest glucose-lowering effect. Among the putative mechanisms of action is the restoration of autophagy, the reduction of inflammation, the prevention of endothelial dysfunction, and the downregulation of fibrotic and apoptotic pathways [63,64]. At the level of the kidney, specifically, SGLT2 inhibitors may induce a lowering of intraglomerular pressure, prevent podocytopathy, interact with the sympathetic nervous system, and lower arterial blood pressure. Several reports of recent experimental studies have presented an antioxidant effect of these agents in vitro and in vivo. A reduction of renal ROS, together with upregulation of renal antioxidant mechanisms, have been noted in experimental models of diabetes mellitus, H2O2-induced renal injury, ischemia-reperfusion injury, and inflammation (Table 1). These observations further support the notion that the effect of SGLT2 inhibitors is not based on glucose lowering. Moreover, cardiac ROS production and antioxidant systems may be modulated through the use of SGLT2 inhibitors. Again, these effects were irrespective of the experimental disease model, being present in diabetic cardiomyopathy, doxorubicin-induced cardiac injury, isoproterenol-induced cardiomyopathy, cardiac ischemia-reperfusion injury, and models of HF (Table 2). An antioxidant effect has also been detected in human umbilical vein endothelial cells and human coronary artery endothelial cells that were stimulated by tumor necrosis factor-α [65].
Scarce clinical evidence exists regarding the antioxidant potential of SGLT2 inhibitors. Lambadiari et al. were the first to report the effect of SGLT2 inhibitors on markers of oxidative stress (thiobarbituric acid reactive substances, malondialdehyde, reducing power, 2,2¢-azino-bis-(3-ethylbenzthiazoline-6-sulphonic acid) radical, and total antioxidant capacity) in 160 participants randomized to SGLT2 inhibitors, GLP1-RA, their combination, or insulin [88]. Individuals on SGLT2 inhibitors alone exhibited only decreases in 2,2¢-azino-bis-(3-ethylbenzthiazoline-6-sulphonic acid) radical after 12 months of follow-up, with no major differences being noted in the rest of the examined markers [88]. In another pilot study, SGLT2 inhibitors induced a reduction in urinary SOD and MnSOD activity, as well as in total antioxidant capacity, in individuals with type 2 diabetes mellitus [89]. The addition of SGLT2 inhibitors to angiotensin-converting enzyme inhibitor ramipril resulted in the lowering of urinary 8-isoprostane concentration compared to the addition of placebo to ramipril in the randomized, double-blind, placebo-controlled, crossover trial of Lytvyn et al. [90]. Moving to a study of 14 non-albuminuric patients with diabetes mellitus, no differences in the urinary oxidative stress marker, 8-hydroxydeoxyguanosine, were noted during the 1-, 3-, and 6-month follow-up [91]. Last but not least, in a circulating proteomics analysis of 1134 patients included in the EMPEROR-PRESERVED and EMPEROR-REDUCED trials, a differential expression of proteins associated with oxidative stress (angiopoietin-related protein 4, insulin-like growth factor-binding protein 4) was documented [92].

3.2. GLP1 Receptor Agonists

GLP1-RA constitute an antidiabetic drug category which has been extensively studied in cardiorenal medicine. Their efficacy is mostly centered around atherosclerotic complications prevention, namely, ischemic stroke, as documented in a recent meta-analysis of 6 double-blind, randomized placebo-control trials with 52821 type 2 diabetes mellitus patients [93]. When compared with SGLT2 inhibitors, GLP1-RA were also associated with a lower risk of major adverse limb events within the first two years after initiation [94]. However, there was no difference between those two drug classes in major adverse cardiovascular events in the study of Fu et al. [95], with the exception of a lower risk of ischemic stroke. GLP1-RA may additionally offer renal protection according to meta-analytic evidence [96]. However, their ability to lower the rates of HF hospitalizations is of lesser magnitude compared to SGLT2 inhibitors [97].
Although being solely used in patients with diabetes mellitus, these agents possess several pleiotropic properties. Focusing on their antioxidant effect (Table 3), liraglutide administration in male 129SV mice with streptozocin-induced diabetes promoted an increase in antioxidant molecules (catalase, GPx) in kidney specimens [98]. Low-dose lixisenatide reduced renal malondialdehyde and total Nox, paired with an increase in total antioxidant capacity [99]. This ultimately prevented early diabetic nephropathy development in diabetic Wistar rats [99]. GLP1-RA could also prevent podocyte apoptosis partially through a reduction in oxidative stress [100,101]. Moving to cardiac antioxidant effects, a recent study has shown that liraglutide administration in diabetic Sprague–Dawley rats attenuated the expression of Nox2 in atrial and ventricular tissue compared to placebo [102]. The antioxidant effect of liraglutide in a similar experimental model has been replicated on top of an anti-inflammatory action, leading to diminished cardiac injury [83]. Liraglutide also prevented high glucose-induced neonatal cardiomyocyte apoptosis through the downregulation of malondialdehyde and upregulation of antioxidant SOD [103]. In vitro evidence from H9C2 cardiomyoblasts treated with H2O2 suggests an increase in antioxidant potential (GPx, catalase, heme oxygenase-1) after administration of a GLP1 analog [104]. Antioxidant effects have been observed in endothelial cells cultured under high glucose and inflammatory settings that were treated with GLP1 analogs or GLP1-RA [105,106,107]. Dual glucagon-like peptide-1/glucose-dependent insulinotropic polypeptide receptor agonists deserve an honorable mention due to their astonishing results in the management of hyperglycemia and obesity, as recently shown [108,109,110,111]. These agents may also act via pleiotropic mechanisms, including attenuation of oxidative stress and inflammation, thus improving diabetes-induced cardiac dysfunction [112].
Although limited data are available from human studies, an antioxidant effect of GLP1-based therapeutics can be suggested. To begin with, Ceriello et al. showed that in both hypoglycemia and hyperglycemia, GLP1 infusion could prevent the increase in the oxidative plasma biomarkers nitrotyrosine and 8-iso prostaglandin F2alpha [119]. Lambadiari et al. showed that 12-month liraglutide administration led to a decrease in malondialdehyde and thiobarbituric acid reactive substances [88]. Liraglutide treatment for 12 weeks in patients with diabetic nephropathy was further associated with a drop in malondialdehyde and an increase in the antioxidant GPx [120]. The antioxidant potential of GLP1-RA, through changes in respective biomarkers, was additionally confirmed in a recently reported systematic review and meta-analysis [121].

3.3. Finerenone

Finerenone is a novel, selective, nonsteroidal mineralocorticoid receptor antagonist that has been recently introduced in the treatment of cardiorenal diseases. Landmark trials on patients with diabetic kidney disease have shown an 18% reduction in the incidence of the primary outcome (kidney failure, sustained decrease ≥ 40% in the eGFR, or death from renal causes), 14% reduction in the incidence of the secondary outcome (death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for HF), and 31% reduction in albuminuria, compared to placebo [122,123]. In patients with HF, finerenone was as efficacious as eplerenone, and may be associated with lesser a lesser increase in potassium levels [124].
Regarding its antioxidant potential (Table 3), an early experimental study by Gueret et al. in mice with coronary artery ligation-induced myocardial infarction initially documented such an effect. Specifically, finerenone abrogated oxidative stress in coronary arteries from noninfarcted mice incubated with low-dose angiotensin-II [125]. At the level of the kidney, finerenone prevented the increase in oxidative stress parameters (malondialdehyde, 8-hydroxy-guanosine) induced by acute bilateral renal ischemia/reperfusion [126]. This was later accompanied by prevention of CKD development (reduced albuminuria, renal vascular resistance, and tubular injury markers) [126]. Finerenone was also found to decrease myocardial ROS production and increase NO bioavailability after short-term administration in Zucker fa/fa rats [113]. This finding may have been responsible for the improvement in diastolic cardiac dysfunction, proteinuria, and tubular injury in the long term [113]. Finally, in Munich Wistar Frömter rats treated with finerenone, aortic ring protein expression of Mn-SOD and Cu/Zn-SOD was enhanced [114]. Total SOD activity was also augmented in the kidneys of those rats treated with finerenone [114]. Collectively, these findings suggest that finerenone’s antioxidant properties may be partly responsible for the cardiorenal benefits seen in randomized clinical trials.

3.4. Melatonin

MT is an amphiphilic tryptophan-derived indoleamine that is stimulated in response to darkness. Other than circadian rhythm regulation, MT has potent antioxidant properties, which are exerted either directly or indirectly through binding to its receptors (MT1 and MT2) [127]. Despite being a well-known molecule for decades, its therapeutic potential in the context of cardiorenal diseases is now beginning to be unveiled. Since its receptors are distributed across many organ systems, its beneficial actions may extend to various pathologies. MT has the potential to be an effective therapy for arterial hypertension through modulating endothelial function, oxidative stress, the autonomic nervous system, and the renin-angiotensin system [127]. Additionally, MT may improve beta-cell and insulin sensitivity [127].
By ameliorating those crucial cardiorenal risk factors, along with potent antioxidant properties, MT could emerge as a safe and effective treatment approach in this regard. Numerous preclinical research has been done in this area to investigate the role of antioxidant pathways in mediating the nephroprotective and cardioprotective effects of MT (Table 3). In human renal proximal tubule epithelial cell lines cultured under high glucose conditions, MT improved their antioxidant capacity, evidenced by upregulated catalase and total SOD activity [115]. Transforming growth factor-β1-treated NRK-49F cells treated with MT exhibited lower levels of intracellular ROS and malondialdehyde, as well as ameliorated reductions of the glutathione/oxidized glutathione ratio [128]. Moreover, the addition of MT treatment in diabetic Wistar rats receiving insulin resulted in downregulated expression of GSH, GSH reductase, glucose-6-phosphate dehydrogenase, and GSH-S-transferase in the renal cortex [129]. MT also improved DN in albino rats by suppressing renal malondialdehyde and stimulating antioxidant systems such as GSH, SOD, and catalase [116].
Several studies have also been performed in various experimental models of cardiovascular disease, assessing the antioxidant properties of MT. In rats with doxorubicin- or carbon tetrachloride-induced cardiotoxicity, MT decreased cardiac malondialdehyde [130,131,132]. In hypercholesterolemic mice with air pollution-provoked cardiac dysfunction, MT alleviated mitochondrial oxidative stress by regulating sirtuin 3-mediated SOD2 deacetylation [133]. Moving to H2O2-induced injury in H9C2 cells, MT abrogated the increases in ROS production by increasing the activity of antioxidant systems (GSH, GPx, SOD), through the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway [117]. Antioxidant effects were reported in Sprague–Dawley rats with myocardial ischemia/reperfusion injury, where MT decreased cardiac malondialdehyde while increasing cardiac SOD and GPx by activating the JAK2/STAT3 signaling pathway [134]. Similar results have been documented in other preclinical studies of cardiac ischemia/reperfusion injury [135,136,137,138]. Activation of cardiac MT2, but not MT1 receptors, may be responsible for this effect [139]. Next, diabetic cardiomyopathy was ameliorated in diabetic Wistar rats treated with MT through an antioxidant response, as shown by Kandemir et al. [118]. Such an effect may be mediated by the 5′ AMP-activated protein kinase/sirtuin 1 pathway [140]. The antioxidant properties of MT have also been observed in experimental models of angiotensin-II-induced cardiac hypertrophy [141], exhaustive exercise-induced cardiac injury [142], and myocardial infarction [143].
Although contemporary research on the cardiorenal benefits of MT is extensive at the preclinical level, strong evidence is limited from clinical studies. Beginning with the renal effects, nocturnal MT 10 mg resulted in a better glycemic profile and oxidative stress indicators in a recently published randomized controlled study of 60 patients with diabetic kidney disease [144]. Similar results were shown in diabetic patients on maintenance hemodialysis, who also had an improvement in inflammatory markers [145]. In a recently published randomized controlled study, kidney transplant recipients were given either MT or a placebo; the MT group showed decreased neutrophil gelatinase-associated lipocalin levels as well as lower levels of inflammatory and oxidative stress indicators [146]. However, no research has been done to determine how MT affects the course of CKD and significant cardiorenal consequences in CKD patients. Examining the function of MT throughout the various CKD phases would be intriguing because CKD is a disorder that causes premature aging. Moving to cardiac effects, in a small scale, placebo-controlled double-blinded randomized clinical trial of 92 patients with HF and a reduced ejection fraction, nighttime MT at a dose of 10 mg decreased the concentration of natriuretic peptides and improved the quality of life of the patients, compared to placebo [147]. Moreover, in recently reported systematic reviews and meta-analyses, MT conferred cardiac protection and improved cardiac function [148,149]. At the same time, it reduced the level of cardiac injury markers, inflammatory cytokines, and oxidative markers while it increased the concentration of antioxidant factors [148]. As with renal studies, evidence is lacking regarding the prevention or prognosis of cardiovascular disease, and future studies are needed in this regard.

4. Clinical Implications and Future Directions

Regarding clinical implications, SGLT2 inhibitors have been established in the treatment of cardiorenal diseases due to their overwhelming clinical benefit. However, the burden of side effects, physician inertia, and insurance issues may lead to higher than expected discontinuation rates [150], which should be further assessed in future real-world clinical practice studies. Moreover, the importance of SGLT2 receptor selectivity or dual SGLT1:SGLT2 inhibition deserves further validation, both in its antioxidant potential and its potentially incremental clinical benefit. Head-to-head trials could provide additional evidence in this direction.
Moving to GLP1-RA, another class with proven cardiorenal benefits used solely in patients with type 2 diabetes mellitus, we should state that there may be significant within class variations regarding efficacy, weight loss, and tolerability [151]. While semaglutide may provide the greatest benefit in glycemia management and weight loss, it is accompanied by a high burden of gastrointestinal adverse events, which could result in discontinuation. A lower risk of adverse events could be attributed to exenatide, and lixisenatide, at the loss of efficacy, however. Finally, liraglutide may possess the greatest balance between efficacy and safety. Patient satisfaction should also be taken into account, being higher in cases of once weekly injections compared to twice daily. Although discontinuation rates have been below 10% in clinical trials, this number is believed to be significantly higher in real-life clinical practice. Therefore, selection of the most appropriate agent should be individualized to accommodate cardiorenal benefit (seen with dulaglutide, liraglutide, and injectable semaglutide), glycemia management, weight loss, and tolerability in the most optimal way.
Finerenone, although beneficial in the completed clinical trials, has been examined only in the setting of diabetic CKD. The clinical spectrum of its efficacy may extend to non-diabetic CKD and HF should the ongoing randomized clinical trials (FIND-CKD/NCT05047263 and FINEARTS-HF/NCT04435626) provide positive results. In addition, further preclinical and clinical evidence is needed to determine whether the antioxidant effect is the driving force of its clinical benefit. Finally, despite being a promising therapeutic tool with minimal adverse effects according to the evidence mentioned above, MT’s optimal timing and dosage have not been clarified and represent an existing gap in evidence. Going forward, MT should be tested in adequately designed trials of individuals at high risk for developing cardiorenal diseases such as the elderly (NCT04631341), as well as those with established atherosclerotic disease, HF, and CKD.
Concerning future directions, several agents are in experimental phase. To begin with, activators of endogenous antioxidant systems may be an interesting option, namely, Nrf2 activators. Dimethyl fumarate, an example of this category, led to reduction in myocardial infarct size in animal models of myocardial ischemia/reperfusion injury and to prevention of atherosclerosis in apoE−/− mice [152,153]. This agent may ameliorate nephrotoxicity due to various causes in in vivo animal models [154,155,156,157]. Moving to Nox inhibition, the dual Nox1/4 inhibitor setanaxib (GKT137831) has been investigated in the experimental setting of doxorubicin-, angiotensin II-, and hypertension-induced cardiac remodeling, displaying cardioprotective effects based on antioxidant and antifibrotic action [158,159,160]. At the level of the kidney setanaxib rescued diabetic nephropathy by abrogating glomerular hypertrophy, mesangial matrix expansion, albuminuria, and podocytopathy [161,162,163,164]. Anti-atherosclerotic effects have also been reported [161,165].
Finally, the role of nanoparticles deserves an honorable mention. Loading of nanoparticles with antioxidants that can directly target the area of excessive ROS production is significant, as shown experimentally. H2O2-responsive nanoparticles have demonstrated efficacy in the preclinical setting of renal and myocardial ischemia/reperfusion injury, by releasing vanillyl alcohol [166,167]. Other antioxidants may also be loaded, such as SOD1, promoting cardioprotective actions [168,169]. Recently, Choi et al. developed nanomicelles able to sense ROS and loaded them with catalase-mimicking 1-dodecanethiol stabilized Mn3O4. Ultimately, the authors noted that inflammation and apoptosis were attenuated in the renal ischemia/reperfusion injury mouse model [170].
While the reduction of oxidative stress may be beneficial in the setting of cardiorenal pathology, it should be stressed that the non-specific suppression of ROS may account for the lack of benefit observed in clinical studies, since it could disrupt important ROS-mediated cellular signaling. Accordingly, targeted therapies such as nanoparticles may pave the way for selective antioxidant treatment at sites of ROS overexpression, such as the failing heart and kidneys.

5. Conclusions

Oxidative stress is a deleterious process that is involved in the pathophysiology of cardiorenal diseases, promoting their development and progression. Recently established pharmacotherapies possess pleiotropic effects that include antioxidant mechanisms, which may partly contribute to the positive effects seen in trials of populations with cardiovascular or renal disease. Research in this field is continuous, and future adequately-sized, randomized studies should further define the importance of these antioxidant effects in the management of cardiorenal diseases.

Author Contributions

Conceptualization, P.T.; methodology, P.T.; software, P.T.; validation, P.T., A.V. and R.G.K.; formal analysis, P.T.; investigation, P.T. and A.V.; data curation, P.T.; writing—original draft preparation, P.T.; writing—review and editing, A.V. and R.G.K.; visualization, P.T.; supervision, R.G.K.; project administration, P.T. and R.G.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

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. Kovesdy, C.P. Epidemiology of chronic kidney disease: An update 2022. Kidney Int. Suppl. 2022, 12, 7–11. [Google Scholar] [CrossRef]
  2. Townsend, N.; Kazakiewicz, D.; Lucy Wright, F.; Timmis, A.; Huculeci, R.; Torbica, A.; Gale, C.P.; Achenbach, S.; Weidinger, F.; Vardas, P. Epidemiology of cardiovascular disease in Europe. Nat. Rev. Cardiol. 2022, 19, 133–143. [Google Scholar] [CrossRef] [PubMed]
  3. Zhao, D. Epidemiological Features of Cardiovascular Disease in Asia. JACC 2021, 1, 1–13. [Google Scholar] [CrossRef]
  4. Drummond, G.R.; Selemidis, S.; Griendling, K.K.; Sobey, C.G. Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat. Rev. Drug Discov. 2011, 10, 453–471. [Google Scholar] [CrossRef] [Green Version]
  5. Ago, T.; Kitazono, T.; Ooboshi, H.; Iyama, T.; Han, Y.H.; Takada, J.; Wakisaka, M.; Ibayashi, S.; Utsumi, H.; Iida, M. Nox4 as the major catalytic component of an endothelial NAD(P)H oxidase. Circulation 2004, 109, 227–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Gorlach, A.; Brandes, R.P.; Nguyen, K.; Amidi, M.; Dehghani, F.; Busse, R. A gp91phox containing NADPH oxidase selectively expressed in endothelial cells is a major source of oxygen radical generation in the arterial wall. Circ. Res. 2000, 87, 26–32. [Google Scholar] [CrossRef] [Green Version]
  7. Ellmark, S.H.; Dusting, G.J.; Fui, M.N.; Guzzo-Pernell, N.; Drummond, G.R. The contribution of Nox4 to NADPH oxidase activity in mouse vascular smooth muscle. Cardiovasc. Res. 2005, 65, 495–504. [Google Scholar] [CrossRef] [Green Version]
  8. Lassegue, B.; Sorescu, D.; Szocs, K.; Yin, Q.; Akers, M.; Zhang, Y.; Grant, S.L.; Lambeth, J.D.; Griendling, K.K. Novel gp91(phox) homologues in vascular smooth muscle cells: Nox1 mediates angiotensin II-induced superoxide formation and redox-sensitive signaling pathways. Circ. Res. 2001, 88, 888–894. [Google Scholar] [CrossRef] [Green Version]
  9. Sobey, C.G.; Judkins, C.P.; Rivera, J.; Lewis, C.V.; Diep, H.; Lee, H.W.; Kemp-Harper, B.K.; Broughton, B.R.; Selemidis, S.; Gaspari, T.A.; et al. NOX1 deficiency in apolipoprotein E-knockout mice is associated with elevated plasma lipids and enhanced atherosclerosis. Free Radic. Res. 2015, 49, 186–198. [Google Scholar] [CrossRef]
  10. Douglas, G.; Bendall, J.K.; Crabtree, M.J.; Tatham, A.L.; Carter, E.E.; Hale, A.B.; Channon, K.M. Endothelial-specific Nox2 overexpression increases vascular superoxide and macrophage recruitment in ApoE(-)/(-) mice. Cardiovasc. Res. 2012, 94, 20–29. [Google Scholar] [CrossRef]
  11. Violi, F.; Carnevale, R.; Loffredo, L.; Pignatelli, P.; Gallin, J.I. NADPH Oxidase-2 and Atherothrombosis: Insight from Chronic Granulomatous Disease. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 218–225. [Google Scholar] [CrossRef] [Green Version]
  12. Gray, S.P.; Di Marco, E.; Kennedy, K.; Chew, P.; Okabe, J.; El-Osta, A.; Calkin, A.C.; Biessen, E.A.; Touyz, R.M.; Cooper, M.E.; et al. Reactive Oxygen Species Can Provide Atheroprotection via NOX4-Dependent Inhibition of Inflammation and Vascular Remodeling. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 295–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Schurmann, C.; Rezende, F.; Kruse, C.; Yasar, Y.; Lowe, O.; Fork, C.; van de Sluis, B.; Bremer, R.; Weissmann, N.; Shah, A.M.; et al. The NADPH oxidase Nox4 has anti-atherosclerotic functions. Eur. Heart J. 2015, 36, 3447–3456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Langbein, H.; Brunssen, C.; Hofmann, A.; Cimalla, P.; Brux, M.; Bornstein, S.R.; Deussen, A.; Koch, E.; Morawietz, H. NADPH oxidase 4 protects against development of endothelial dysfunction and atherosclerosis in LDL receptor deficient mice. Eur. Heart J. 2016, 37, 1753–1761. [Google Scholar] [CrossRef] [Green Version]
  15. Landmesser, U.; Spiekermann, S.; Preuss, C.; Sorrentino, S.; Fischer, D.; Manes, C.; Mueller, M.; Drexler, H. Angiotensin II induces endothelial xanthine oxidase activation: Role for endothelial dysfunction in patients with coronary disease. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 943–948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. McNally, J.S.; Davis, M.E.; Giddens, D.P.; Saha, A.; Hwang, J.; Dikalov, S.; Jo, H.; Harrison, D.G. Role of xanthine oxidoreductase and NAD(P)H oxidase in endothelial superoxide production in response to oscillatory shear stress. Am. J. Physiol. Heart Circ. Physiol. 2003, 285, H2290–H2297. [Google Scholar] [CrossRef] [Green Version]
  17. Battelli, M.G.; Polito, L.; Bolognesi, A. Xanthine oxidoreductase in atherosclerosis pathogenesis: Not only oxidative stress. Atherosclerosis 2014, 237, 562–567. [Google Scholar] [CrossRef] [Green Version]
  18. Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef]
  19. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, C.; Tousoulis, D. Inflammatory Mechanisms Contributing to Endothelial Dysfunction. Biomedicines 2021, 9, 781. [Google Scholar] [CrossRef]
  20. Tribble, D.L.; Gong, E.L.; Leeuwenburgh, C.; Heinecke, J.W.; Carlson, E.L.; Verstuyft, J.G.; Epstein, C.J. Fatty streak formation in fat-fed mice expressing human copper-zinc superoxide dismutase. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 1734–1740. [Google Scholar] [CrossRef]
  21. Neves, A.L.; Mohammedi, K.; Emery, N.; Roussel, R.; Fumeron, F.; Marre, M.; Velho, G. Allelic variations in superoxide dismutase-1 (SOD1) gene and renal and cardiovascular morbidity and mortality in type 2 diabetic subjects. Mol. Genet. Metab. 2012, 106, 359–365. [Google Scholar] [CrossRef] [PubMed]
  22. Mollsten, A.; Jorsal, A.; Lajer, M.; Vionnet, N.; Tarnow, L. The V16A polymorphism in SOD2 is associated with increased risk of diabetic nephropathy and cardiovascular disease in type 1 diabetes. Diabetologia 2009, 52, 2590–2593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Fujimoto, H.; Taguchi, J.; Imai, Y.; Ayabe, S.; Hashimoto, H.; Kobayashi, H.; Ogasawara, K.; Aizawa, T.; Yamakado, M.; Nagai, R.; et al. Manganese superoxide dismutase polymorphism affects the oxidized low-density lipoprotein-induced apoptosis of macrophages and coronary artery disease. Eur. Heart J. 2008, 29, 1267–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Kakko, S.; Paivansalo, M.; Koistinen, P.; Kesaniemi, Y.A.; Kinnula, V.L.; Savolainen, M.J. The signal sequence polymorphism of the MnSOD gene is associated with the degree of carotid atherosclerosis. Atherosclerosis 2003, 168, 147–152. [Google Scholar] [CrossRef]
  25. Marklund, S.L.; Nilsson, P.; Israelsson, K.; Schampi, I.; Peltonen, M.; Asplund, K. Two variants of extracellular-superoxide dismutase: Relationship to cardiovascular risk factors in an unselected middle-aged population. J. Intern. Med. 1997, 242, 5–14. [Google Scholar] [CrossRef] [Green Version]
  26. Nemoto, M.; Nishimura, R.; Sasaki, T.; Hiki, Y.; Miyashita, Y.; Nishioka, M.; Fujimoto, K.; Sakuma, T.; Ohashi, T.; Fukuda, K.; et al. Genetic association of glutathione peroxidase-1 with coronary artery calcification in type 2 diabetes: A case control study with multi-slice computed tomography. Cardiovasc. Diabetol. 2007, 6, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Yang, X.; Yang, S.; Xu, H.; Liu, D.; Zhang, Y.; Wang, G. Superoxide Dismutase Gene Polymorphism is Associated with Ischemic Stroke Risk in the China Dali Region Han Population. Neurologist 2021, 26, 27–31. [Google Scholar] [CrossRef] [PubMed]
  28. Heslop, C.L.; Tebbutt, S.J.; Podder, M.; Ruan, J.; Hill, J.S. Combined polymorphisms in oxidative stress genes predict coronary artery disease and oxidative stress in coronary angiography patients. Ann. Hum. Genet. 2012, 76, 435–447. [Google Scholar] [CrossRef]
  29. Hamanishi, T.; Furuta, H.; Kato, H.; Doi, A.; Tamai, M.; Shimomura, H.; Sakagashira, S.; Nishi, M.; Sasaki, H.; Sanke, T.; et al. Functional variants in the glutathione peroxidase-1 (GPx-1) gene are associated with increased intima-media thickness of carotid arteries and risk of macrovascular diseases in japanese type 2 diabetic patients. Diabetes 2004, 53, 2455–2460. [Google Scholar] [CrossRef] [Green Version]
  30. Zhang, J.X.; Wang, Z.M.; Zhang, J.J.; Zhu, L.L.; Gao, X.F.; Chen, S.L. Association of glutathione peroxidase-1 (GPx-1) rs1050450 Pro198Leu and Pro197Leu polymorphisms with cardiovascular risk: A meta-analysis of observational studies. J. Geriatr. Cardiol. 2014, 11, 141–150. [Google Scholar] [CrossRef]
  31. Al-Kateb, H.; Boright, A.P.; Mirea, L.; Xie, X.; Sutradhar, R.; Mowjoodi, A.; Bharaj, B.; Liu, M.; Bucksa, J.M.; Arends, V.L.; et al. Multiple superoxide dismutase 1/splicing factor serine alanine 15 variants are associated with the development and progression of diabetic nephropathy: The Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Genetics study. Diabetes 2008, 57, 218–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Mohammedi, K.; Maimaitiming, S.; Emery, N.; Bellili-Munoz, N.; Roussel, R.; Fumeron, F.; Hadjadj, S.; Marre, M.; Velho, G. Allelic variations in superoxide dismutase-1 (SOD1) gene are associated with increased risk of diabetic nephropathy in type 1 diabetic subjects. Mol. Genet. Metab. 2011, 104, 654–660. [Google Scholar] [CrossRef] [PubMed]
  33. Jerotic, D.; Matic, M.; Suvakov, S.; Vucicevic, K.; Damjanovic, T.; Savic-Radojevic, A.; Pljesa-Ercegovac, M.; Coric, V.; Stefanovic, A.; Ivanisevic, J.; et al. Association of Nrf2, SOD2 and GPX1 Polymorphisms with Biomarkers of Oxidative Distress and Survival in End-Stage Renal Disease Patients. Toxins 2019, 11, 431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Chao, C.T.; Huang, J.W.; Chiang, C.K.; Chen, Y.C.; Fang, C.C.; Hu, F.C.; Chang, C.C.; Yen, C.J. Diabetes mellitus, superoxide dismutase and peroxisome proliferator activated receptor gamma polymorphisms modify the outcome of end-stage renal disease patients of Han Chinese origin. Nephrology 2018, 23, 117–125. [Google Scholar] [CrossRef] [PubMed]
  35. Mohammedi, K.; Bellili-Munoz, N.; Driss, F.; Roussel, R.; Seta, N.; Fumeron, F.; Hadjadj, S.; Marre, M.; Velho, G. Manganese superoxide dismutase (SOD2) polymorphisms, plasma advanced oxidation protein products (AOPP) concentration and risk of kidney complications in subjects with type 1 diabetes. PLoS ONE 2014, 9, e96916. [Google Scholar] [CrossRef] [Green Version]
  36. Cardona-Sanclemente, L.E.; Born, G.V. Effect of inhibition of nitric oxide synthesis on the uptake of LDL and fibrinogen by arterial walls and other organs of the rat. Br. J. Pharmacol. 1995, 114, 1490–1494. [Google Scholar] [CrossRef] [Green Version]
  37. Su, L.J.; Zhang, J.H.; Gomez, H.; Murugan, R.; Hong, X.; Xu, D.; Jiang, F.; Peng, Z.Y. Reactive Oxygen Species-Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis. Oxid. Med. Cell. Longev. 2019, 2019, 5080843. [Google Scholar] [CrossRef] [Green Version]
  38. Binder, C.J.; Papac-Milicevic, N.; Witztum, J.L. Innate sensing of oxidation-specific epitopes in health and disease. Nat. Rev. Immunol. 2016, 16, 485–497. [Google Scholar] [CrossRef]
  39. Lehoux, S. Redox signalling in vascular responses to shear and stretch. Cardiovasc. Res. 2006, 71, 269–279. [Google Scholar] [CrossRef] [Green Version]
  40. Moore, K.J.; Sheedy, F.J.; Fisher, E.A. Macrophages in atherosclerosis: A dynamic balance. Nat. Rev. Immunol. 2013, 13, 709–721. [Google Scholar] [CrossRef] [Green Version]
  41. Jha, J.C.; Banal, C.; Chow, B.S.; Cooper, M.E.; Jandeleit-Dahm, K. Diabetes and Kidney Disease: Role of Oxidative Stress. Antioxid. Redox Signal. 2016, 25, 657–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Tanner, R.M.; Brown, T.M.; Muntner, P. Epidemiology of obesity, the metabolic syndrome, and chronic kidney disease. Curr. Hypertens. Rep. 2012, 14, 152–159. [Google Scholar] [CrossRef] [PubMed]
  43. Choi, B.H.; Kang, K.S.; Kwak, M.K. Effect of redox modulating NRF2 activators on chronic kidney disease. Molecules 2014, 19, 12727–12759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Gondouin, B.; Jourde-Chiche, N.; Sallee, M.; Dou, L.; Cerini, C.; Loundou, A.; Morange, S.; Berland, Y.; Burtey, S.; Brunet, P.; et al. Plasma Xanthine Oxidase Activity Is Predictive of Cardiovascular Disease in Patients with Chronic Kidney Disease, Independently of Uric Acid Levels. Nephron 2015, 131, 167–174. [Google Scholar] [CrossRef]
  45. Schmidt, R.J.; Baylis, C. Total nitric oxide production is low in patients with chronic renal disease. Kidney Int. 2000, 58, 1261–1266. [Google Scholar] [CrossRef] [Green Version]
  46. Norris, K.C.; Olabisi, O.; Barnett, M.E.; Meng, Y.X.; Martins, D.; Obialo, C.; Lee, J.E.; Nicholas, S.B. The Role of Vitamin D and Oxidative Stress in Chronic Kidney Disease. Int. J. Environ. Res. Public Health 2018, 15, 2701. [Google Scholar] [CrossRef] [Green Version]
  47. Noronha, I.L.; Fujihara, C.K.; Zatz, R. The inflammatory component in progressive renal disease—Are interventions possible? Nephrol. Dial. Transplant. 2002, 17, 363–368. [Google Scholar] [CrossRef] [Green Version]
  48. Li, N.; Karin, M. Is NF-kappaB the sensor of oxidative stress? FASEB J. 1999, 13, 1137–1143. [Google Scholar] [CrossRef]
  49. Schreck, R.; Rieber, P.; Baeuerle, P.A. Reactive oxygen intermediates as apparently widely used messengers in the activation of the NF-kappa B transcription factor and HIV-1. EMBO J. 1991, 10, 2247–2258. [Google Scholar] [CrossRef]
  50. Simmons, E.M.; Langone, A.; Sezer, M.T.; Vella, J.P.; Recupero, P.; Morrow, J.D.; Ikizler, T.A.; Himmelfarb, J. Effect of renal transplantation on biomarkers of inflammation and oxidative stress in end-stage renal disease patients. Transplantation 2005, 79, 914–919. [Google Scholar] [CrossRef]
  51. Cachofeiro, V.; Goicochea, M.; de Vinuesa, S.G.; Oubina, P.; Lahera, V.; Luno, J. Oxidative stress and inflammation, a link between chronic kidney disease and cardiovascular disease. Kidney Int. Suppl. 2008, 74, S4–S9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Perkovic, V.; Jardine, M.J.; Neal, B.; Bompoint, S.; Heerspink, H.J.L.; Charytan, D.M.; Edwards, R.; Agarwal, R.; Bakris, G.; Bull, S.; et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2019, 380, 2295–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Heerspink, H.J.L.; Stefansson, B.V.; Correa-Rotter, R.; Chertow, G.M.; Greene, T.; Hou, F.F.; Mann, J.F.E.; McMurray, J.J.V.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef] [PubMed]
  54. Bhatt, D.L.; Szarek, M.; Pitt, B.; Cannon, C.P.; Leiter, L.A.; McGuire, D.K.; Lewis, J.B.; Riddle, M.C.; Inzucchi, S.E.; Kosiborod, M.N.; et al. Sotagliflozin in Patients with Diabetes and Chronic Kidney Disease. N. Engl. J. Med. 2021, 384, 129–139. [Google Scholar] [CrossRef] [PubMed]
  55. 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] [PubMed]
  56. Jhund, P.S.; Solomon, S.D.; Docherty, K.F.; Heerspink, H.J.L.; Anand, I.S.; Bohm, M.; Chopra, V.; de Boer, R.A.; Desai, A.S.; Ge, J.; et al. Efficacy of Dapagliflozin on Renal Function and Outcomes in Patients with Heart Failure with Reduced Ejection Fraction: Results of DAPA-HF. Circulation 2021, 143, 298–309. [Google Scholar] [CrossRef]
  57. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Bohm, M.; Burri, H.; Butler, J.; Celutkiene, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef]
  58. Theofilis, P.; Antonopoulos, A.S.; Katsimichas, T.; Oikonomou, E.; Siasos, G.; Aggeli, C.; Tsioufis, K.; Tousoulis, D. The impact of SGLT2 inhibition on imaging markers of cardiac function: A systematic review and meta-analysis. Pharmacol. Res. 2022, 180, 106243. [Google Scholar] [CrossRef]
  59. Bhatt, D.L.; Szarek, M.; Steg, P.G.; Cannon, C.P.; Leiter, L.A.; McGuire, D.K.; Lewis, J.B.; Riddle, M.C.; Voors, A.A.; Metra, M.; et al. Sotagliflozin in Patients with Diabetes and Recent Worsening Heart Failure. N. Engl. J. Med. 2021, 384, 117–128. [Google Scholar] [CrossRef]
  60. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Bohm, M.; Brunner-La Rocca, H.P.; Choi, D.J.; Chopra, V.; Chuquiure-Valenzuela, E.; et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021, 385, 1451–1461. [Google Scholar] [CrossRef]
  61. Solomon, S.D.; McMurray, J.J.V.; Claggett, B.; de Boer, R.A.; DeMets, D.; Hernandez, A.F.; Inzucchi, S.E.; Kosiborod, M.N.; Lam, C.S.P.; Martinez, F.; et al. Dapagliflozin in Heart Failure with Mildly Reduced or Preserved Ejection Fraction. N. Engl. J. Med. 2022, 387, 1089–1098. [Google Scholar] [CrossRef] [PubMed]
  62. Ala, M. SGLT2 Inhibition for Cardiovascular Diseases, Chronic Kidney Disease, and NAFLD. Endocrinology 2021, 162, bqab157. [Google Scholar] [CrossRef] [PubMed]
  63. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, K.; Tousoulis, D. The impact of SGLT2 inhibitors on inflammation: A systematic review and meta-analysis of studies in rodents. Int. Immunopharmacol. 2022, 111, 109080. [Google Scholar] [CrossRef]
  64. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, K.; Tousoulis, D. Pleiotropic effects of SGLT2 inhibitors and heart failure outcomes. Diabetes Res. Clin. Pract. 2022, 188, 109927. [Google Scholar] [CrossRef]
  65. Uthman, L.; Li, X.; Baartscheer, A.; Schumacher, C.A.; Baumgart, P.; Hermanides, J.; Preckel, B.; Hollmann, M.W.; Coronel, R.; Zuurbier, C.J.; et al. Empagliflozin reduces oxidative stress through inhibition of the novel inflammation/NHE/[Na(+)]c/ROS-pathway in human endothelial cells. Biomed. Pharmacother. 2022, 146, 112515. [Google Scholar] [CrossRef]
  66. Ashrafi Jigheh, Z.; Ghorbani Haghjo, A.; Argani, H.; Roshangar, L.; Rashtchizadeh, N.; Sanajou, D.; Nazari Soltan Ahmad, S.; Rashedi, J.; Dastmalchi, S.; Mesgari Abbasi, M. Empagliflozin alleviates renal inflammation and oxidative stress in streptozotocin-induced diabetic rats partly by repressing HMGB1-TLR4 receptor axis. Iran. J. Basic Med. Sci. 2019, 22, 384–390. [Google Scholar] [CrossRef]
  67. Kimura, Y.; Kuno, A.; Tanno, M.; Sato, T.; Ohno, K.; Shibata, S.; Nakata, K.; Sugawara, H.; Abe, K.; Igaki, Y.; et al. Canagliflozin, a sodium-glucose cotransporter 2 inhibitor, normalizes renal susceptibility to type 1 cardiorenal syndrome through reduction of renal oxidative stress in diabetic rats. J. Diabetes Investig. 2019, 10, 933–946. [Google Scholar] [CrossRef]
  68. Das, N.A.; Carpenter, A.J.; Belenchia, A.; Aroor, A.R.; Noda, M.; Siebenlist, U.; Chandrasekar, B.; DeMarco, V.G. Empagliflozin reduces high glucose-induced oxidative stress and miR-21-dependent TRAF3IP2 induction and RECK suppression, and inhibits human renal proximal tubular epithelial cell migration and epithelial-to-mesenchymal transition. Cell. Signal. 2020, 68, 109506. [Google Scholar] [CrossRef]
  69. Zaibi, N.; Li, P.; Xu, S.Z. Protective effects of dapagliflozin against oxidative stress-induced cell injury in human proximal tubular cells. PLoS ONE 2021, 16, e0247234. [Google Scholar] [CrossRef]
  70. Ahmed, A.S.; Mona, M.M.; Abdel-Kareem, M.A.; Elsisy, R.A. SGLT2 inhibitor empagliflozin monotherapy alleviates renal oxidative stress in albino Wistar diabetic rats after myocardial infarction induction. Biomed. Pharmacother. 2021, 139, 111624. [Google Scholar] [CrossRef]
  71. Hudkins, K.L.; Li, X.; Holland, A.L.; Swaminathan, S.; Alpers, C.E. Regression of diabetic nephropathy by treatment with empagliflozin in BTBR ob/ob mice. Nephrol. Dial. Transplant. 2022, 37, 847–859. [Google Scholar] [CrossRef] [PubMed]
  72. Ala, M.; Khoshdel, M.R.F.; Dehpour, A.R. Empagliflozin Enhances Autophagy, Mitochondrial Biogenesis, and Antioxidant Defense and Ameliorates Renal Ischemia/Reperfusion in Nondiabetic Rats. Oxid. Med. Cell. Longev. 2022, 2022, 1197061. [Google Scholar] [CrossRef] [PubMed]
  73. Malinska, H.; Huttl, M.; Markova, I.; Miklankova, D.; Hojna, S.; Papousek, F.; Silhavy, J.; Mlejnek, P.; Zicha, J.; Hrdlicka, J.; et al. Beneficial Effects of Empagliflozin Are Mediated by Reduced Renal Inflammation and Oxidative Stress in Spontaneously Hypertensive Rats Expressing Human C-Reactive Protein. Biomedicines 2022, 10, 2066. [Google Scholar] [CrossRef] [PubMed]
  74. Ye, T.; Zhang, J.; Wu, D.; Shi, J.; Kuang, Z.; Ma, Y.; Xu, Q.; Chen, B.; Kan, C.; Sun, X.; et al. Empagliflozin Attenuates Obesity-Related Kidney Dysfunction and NLRP3 Inflammasome Activity through the HO-1-Adiponectin Axis. Front. Endocrinol. 2022, 13, 907984. [Google Scholar] [CrossRef] [PubMed]
  75. Xing, Y.J.; Liu, B.H.; Wan, S.J.; Cheng, Y.; Zhou, S.M.; Sun, Y.; Yao, X.M.; Hua, Q.; Meng, X.J.; Cheng, J.H.; et al. A SGLT2 Inhibitor Dapagliflozin Alleviates Diabetic Cardiomyopathy by Suppressing High Glucose-Induced Oxidative Stress in vivo and in vitro. Front. Pharmacol. 2021, 12, 708177. [Google Scholar] [CrossRef] [PubMed]
  76. Hsieh, P.L.; Chu, P.M.; Cheng, H.C.; Huang, Y.T.; Chou, W.C.; Tsai, K.L.; Chan, S.H. Dapagliflozin Mitigates Doxorubicin-Caused Myocardium Damage by Regulating AKT-Mediated Oxidative Stress, Cardiac Remodeling, and Inflammation. Int. J. Mol. Sci. 2022, 23, 10146. [Google Scholar] [CrossRef] [PubMed]
  77. Bugga, P.; Mohammed, S.A.; Alam, M.J.; Katare, P.; Meghwani, H.; Maulik, S.K.; Arava, S.; Banerjee, S.K. Empagliflozin prohibits high-fructose diet-induced cardiac dysfunction in rats via attenuation of mitochondria-driven oxidative stress. Life Sci. 2022, 307, 120862. [Google Scholar] [CrossRef]
  78. Li, X.; Flynn, E.R.; do Carmo, J.M.; Wang, Z.; da Silva, A.A.; Mouton, A.J.; Omoto, A.C.M.; Hall, M.E.; Hall, J.E. Direct Cardiac Actions of Sodium-Glucose Cotransporter 2 Inhibition Improve Mitochondrial Function and Attenuate Oxidative Stress in Pressure Overload-Induced Heart Failure. Front. Cardiovasc. Med. 2022, 9, 859253. [Google Scholar] [CrossRef]
  79. Tsai, K.L.; Hsieh, P.L.; Chou, W.C.; Cheng, H.C.; Huang, Y.T.; Chan, S.H. Dapagliflozin attenuates hypoxia/reoxygenation-caused cardiac dysfunction and oxidative damage through modulation of AMPK. Cell Biosci. 2021, 11, 44. [Google Scholar] [CrossRef]
  80. Rosa, C.M.; Campos, D.H.S.; Reyes, D.R.A.; Damatto, F.C.; Kurosaki, L.Y.; Pagan, L.U.; Gomes, M.J.; Correa, C.R.; Fernandes, A.A.H.; Okoshi, M.P.; et al. Effects of the SGLT2 Inhibition on Cardiac Remodeling in Streptozotocin-Induced Diabetic Rats, a Model of Type 1 Diabetes Mellitus. Antioxidants 2022, 11, 982. [Google Scholar] [CrossRef]
  81. Wang, J.; Huang, X.; Liu, H.; Chen, Y.; Li, P.; Liu, L.; Li, J.; Ren, Y.; Huang, J.; Xiong, E.; et al. Empagliflozin Ameliorates Diabetic Cardiomyopathy via Attenuating Oxidative Stress and Improving Mitochondrial Function. Oxid. Med. Cell. Longev. 2022, 2022, 1122494. [Google Scholar] [CrossRef] [PubMed]
  82. Kolijn, D.; Pabel, S.; Tian, Y.; Lodi, M.; Herwig, M.; Carrizzo, A.; Zhazykbayeva, S.; Kovacs, A.; Fulop, G.A.; Falcao-Pires, I.; et al. Empagliflozin improves endothelial and cardiomyocyte function in human heart failure with preserved ejection fraction via reduced pro-inflammatory-oxidative pathways and protein kinase Galpha oxidation. Cardiovasc. Res. 2021, 117, 495–507. [Google Scholar] [CrossRef]
  83. El-Shafey, M.; El-Agawy, M.S.E.; Eldosoky, M.; Ebrahim, H.A.; Elsherbini, D.M.A.; El-Sherbiny, M.; Asseri, S.M.; Elsherbiny, N.M. Role of Dapagliflozin and Liraglutide on Diabetes-Induced Cardiomyopathy in Rats: Implication of Oxidative Stress, Inflammation, and Apoptosis. Front. Endocrinol. 2022, 13, 862394. [Google Scholar] [CrossRef] [PubMed]
  84. Li, C.; Zhang, J.; Xue, M.; Li, X.; Han, F.; Liu, X.; Xu, L.; Lu, Y.; Cheng, Y.; Li, T.; et al. SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovasc. Diabetol. 2019, 18, 15. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, F.Z.; Wei, W.B.; Li, X.; Huo, J.Y.; Jiang, W.Y.; Wang, H.Y.; Qian, P.; Li, Z.Z.; Zhou, Y.B. The cardioprotective effect of the sodium-glucose cotransporter 2 inhibitor dapagliflozin in rats with isoproterenol-induced cardiomyopathy. Am. J. Transl. Res. 2021, 13, 10950–10961. [Google Scholar]
  86. Yurista, S.R.; Sillje, H.H.W.; Oberdorf-Maass, S.U.; Schouten, E.M.; Pavez Giani, M.G.; Hillebrands, J.L.; van Goor, H.; van Veldhuisen, D.J.; de Boer, R.A.; Westenbrink, B.D. Sodium-glucose co-transporter 2 inhibition with empagliflozin improves cardiac function in non-diabetic rats with left ventricular dysfunction after myocardial infarction. Eur. J. Heart Fail. 2019, 21, 862–873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Rahadian, A.; Fukuda, D.; Salim, H.M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Sata, M. Canagliflozin Prevents Diabetes-Induced Vascular Dysfunction in ApoE-Deficient Mice. J. Atheroscler. Thromb. 2020, 27, 1141–1151. [Google Scholar] [CrossRef] [Green Version]
  88. Lambadiari, V.; Thymis, J.; Kouretas, D.; Skaperda, Z.; Tekos, F.; Kousathana, F.; Kountouri, A.; Balampanis, K.; Parissis, J.; Andreadou, I.; et al. Effects of a 12-Month Treatment with Glucagon-like Peptide-1 Receptor Agonists, Sodium-Glucose Cotransporter-2 Inhibitors, and Their Combination on Oxidant and Antioxidant Biomarkers in Patients with Type 2 Diabetes. Antioxidants 2021, 10, 1379. [Google Scholar] [CrossRef]
  89. Nabrdalik-Lesniak, D.; Nabrdalik, K.; Sedlaczek, K.; Glowczynski, P.; Kwiendacz, H.; Sawczyn, T.; Hajzler, W.; Drozdz, K.; Hendel, M.; Irlik, K.; et al. Influence of SGLT2 Inhibitor Treatment on Urine Antioxidant Status in Type 2 Diabetic Patients: A Pilot Study. Oxid. Med. Cell. Longev. 2021, 2021, 5593589. [Google Scholar] [CrossRef]
  90. Lytvyn, Y.; Kimura, K.; Peter, N.; Lai, V.; Tse, J.; Cham, L.; Perkins, B.A.; Soleymanlou, N.; Cherney, D.Z.I. Renal and Vascular Effects of Combined SGLT2 and Angiotensin-Converting Enzyme Inhibition. Circulation 2022, 146, 450–462. [Google Scholar] [CrossRef]
  91. Shimohata, H.; Iwaki, Y.; Yamashita, M.; Ohgi, K.; Maruyama, H.; Takayasu, M.; Hirayama, K.; Kobayashi, M. The effect of sodium-glucose cotransporter 2 inhibitor (tofogliflozin) on renal tubular damage in diabetic patients without albuminuria. Int. Urol. Nephrol. 2022, 54, 1907–1914. [Google Scholar] [CrossRef] [PubMed]
  92. Zannad, F.; Ferreira, J.P.; Butler, J.; Filippatos, G.; Januzzi, J.L.; Sumin, M.; Zwick, M.; Saadati, M.; Pocock, S.J.; Sattar, N.; et al. Effect of Empagliflozin on Circulating Proteomics in Heart Failure: Mechanistic Insights from the EMPEROR Program. Eur. Heart J. 2022. [Google Scholar] [CrossRef] [PubMed]
  93. Qin, J.; Song, L. Glucagon-like peptide-1 (GLP-1) receptor agonists and cardiovascular events in patients with type 2 diabetes mellitus: A meta-analysis of double-blind, randomized, placebo-controlled clinical trials. BMC Endocr. Disord. 2022, 22, 125. [Google Scholar] [CrossRef] [PubMed]
  94. Lin, D.S.; Yu, A.L.; Lo, H.Y.; Lien, C.W.; Lee, J.K.; Chen, W.J. Major adverse cardiovascular and limb events in people with diabetes treated with GLP-1 receptor agonists vs SGLT2 inhibitors. Diabetologia 2022. [Google Scholar] [CrossRef] [PubMed]
  95. Fu, E.L.; Clase, C.M.; Janse, R.J.; Lindholm, B.; Dekker, F.W.; Jardine, M.J.; Carrero, J.J. Comparative effectiveness of SGLT2i versus GLP1-RA on cardiovascular outcomes in routine clinical practice. Int. J. Cardiol. 2022, 352, 172–179. [Google Scholar] [CrossRef]
  96. Li, X.; Song, Y.; Guo, T.; Xiao, G.; Li, Q. Effect of glucagon-like peptide 1 receptor agonists on the renal protection in patients with type 2 diabetes: A systematic review and meta-analysis. Diabetes Metab. 2022, 48, 101366. [Google Scholar] [CrossRef] [PubMed]
  97. Giugliano, D.; Longo, M.; Signoriello, S.; Maiorino, M.I.; Solerte, B.; Chiodini, P.; Esposito, K. The effect of DPP-4 inhibitors, GLP-1 receptor agonists and SGLT-2 inhibitors on cardiorenal outcomes: A network meta-analysis of 23 CVOTs. Cardiovasc. Diabetol. 2022, 21, 42. [Google Scholar] [CrossRef] [PubMed]
  98. Liljedahl, L.; Pedersen, M.H.; McGuire, J.N.; James, P. The impact of the glucagon-like peptide 1 receptor agonist liraglutide on the streptozotocin-induced diabetic mouse kidney proteome. Physiol. Rep. 2019, 7, e13994. [Google Scholar] [CrossRef] [Green Version]
  99. Abdel-Latif, R.G.; Ahmed, A.F.; Heeba, G.H. Low-dose lixisenatide protects against early-onset nephropathy induced in diabetic rats. Life Sci. 2020, 263, 118592. [Google Scholar] [CrossRef]
  100. Shi, J.X.; Huang, Q. Glucagonlike peptide1 protects mouse podocytes against high glucoseinduced apoptosis, and suppresses reactive oxygen species production and proinflammatory cytokine secretion, through sirtuin 1 activation in vitro. Mol. Med. Rep. 2018, 18, 1789–1797. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, S.S.; Wu, Z.; Zhang, Z.; Xiong, Z.Y.; Chen, H.; Huang, Q.B. Glucagon-like peptide-1 inhibits the receptor for advanced glycation endproducts to prevent podocyte apoptosis induced by advanced oxidative protein products. Biochem. Biophys. Res. Commun. 2017, 482, 1413–1419. [Google Scholar] [CrossRef] [PubMed]
  102. Baylan, U.; Korn, A.; Emmens, R.W.; Schalkwijk, C.G.; Niessen, H.W.M.; Krijnen, P.A.J.; Simsek, S. Liraglutide treatment attenuates inflammation markers in the cardiac, cerebral and renal microvasculature in streptozotocin-induced diabetic rats. Eur. J. Clin. Investig. 2022, 52, e13807. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, L.; Li, C.; Zhu, Q.; Li, N.; Zhou, H. Liraglutide, a glucagon-like peptide-1 analog, inhibits high glucose-induced oxidative stress and apoptosis in neonatal rat cardiomyocytes. Exp. Ther. Med. 2019, 17, 3734–3740. [Google Scholar] [CrossRef]
  104. Nuamnaichati, N.; Parichatikanond, W.; Mangmool, S. Cardioprotective Effects of Glucagon-like Peptide-1 (9-36) Against Oxidative Injury in H9c2 Cardiomyoblasts: Potential Role of the PI3K/Akt/NOS Pathway. J. Cardiovasc. Pharmacol. 2022, 79, e50–e63. [Google Scholar] [CrossRef]
  105. Shiraki, A.; Oyama, J.; Komoda, H.; Asaka, M.; Komatsu, A.; Sakuma, M.; Kodama, K.; Sakamoto, Y.; Kotooka, N.; Hirase, T.; et al. The glucagon-like peptide 1 analog liraglutide reduces TNF-alpha-induced oxidative stress and inflammation in endothelial cells. Atherosclerosis 2012, 221, 375–382. [Google Scholar] [CrossRef] [PubMed]
  106. Guo, L.; Qiao, Y.; Zhang, L.; Pan, Q. Protective Role of Glucagon-like Peptide-1 against High-Glucose-Induced Endothelial Oxidative Damage. Medicine 2015, 94, e2055. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, R.; Lu, L.; Guo, Y.; Lin, F.; Chen, H.; Chen, W.; Chen, M. Effect of Glucagon-like Peptide-1 on High-Glucose-induced Oxidative Stress and Cell Apoptosis in Human Endothelial Cells and Its Underlying Mechanism. J. Cardiovasc. Pharmacol. 2015, 66, 135–140. [Google Scholar] [CrossRef] [PubMed]
  108. Heise, T.; Mari, A.; DeVries, J.H.; Urva, S.; Li, J.; Pratt, E.J.; Coskun, T.; Thomas, M.K.; Mather, K.J.; Haupt, A.; et al. Effects of subcutaneous tirzepatide versus placebo or semaglutide on pancreatic islet function and insulin sensitivity in adults with type 2 diabetes: A multicentre, randomised, double-blind, parallel-arm, phase 1 clinical trial. Lancet Diabetes Endocrinol. 2022, 10, 418–429. [Google Scholar] [CrossRef]
  109. Karagiannis, T.; Avgerinos, I.; Liakos, A.; Del Prato, S.; Matthews, D.R.; Tsapas, A.; Bekiari, E. Management of type 2 diabetes with the dual GIP/GLP-1 receptor agonist tirzepatide: A systematic review and meta-analysis. Diabetologia 2022, 65, 1251–1261. [Google Scholar] [CrossRef]
  110. Jastreboff, A.M.; Aronne, L.J.; Ahmad, N.N.; Wharton, S.; Connery, L.; Alves, B.; Kiyosue, A.; Zhang, S.; Liu, B.; Bunck, M.C.; et al. Tirzepatide Once Weekly for the Treatment of Obesity. N. Engl. J. Med. 2022, 387, 205–216. [Google Scholar] [CrossRef]
  111. Inagaki, N.; Takeuchi, M.; Oura, T.; Imaoka, T.; Seino, Y. Efficacy and safety of tirzepatide monotherapy compared with dulaglutide in Japanese patients with type 2 diabetes (SURPASS J-mono): A double-blind, multicentre, randomised, phase 3 trial. Lancet Diabetes Endocrinol. 2022, 10, 623–633. [Google Scholar] [CrossRef]
  112. Wang, Y.; Cai, F.; Li, G.; Tao, Y. Novel dual glucagon-like peptide-1/glucose-dependent insulinotropic polypeptide receptor agonist attenuates diabetes and myocardial injury through inhibiting hyperglycemia, inflammation and oxidative stress in rodent animals. Bioengineered 2022, 13, 9184–9196. [Google Scholar] [CrossRef] [PubMed]
  113. Lachaux, M.; Barrera-Chimal, J.; Nicol, L.; Remy-Jouet, I.; Renet, S.; Dumesnil, A.; Wecker, D.; Richard, V.; Kolkhof, P.; Jaisser, F.; et al. Short- and long-term administration of the non-steroidal mineralocorticoid receptor antagonist finerenone opposes metabolic syndrome-related cardio-renal dysfunction. Diabetes Obes. Metab. 2018, 20, 2399–2407. [Google Scholar] [CrossRef] [PubMed]
  114. Gonzalez-Blazquez, R.; Somoza, B.; Gil-Ortega, M.; Martin Ramos, M.; Ramiro-Cortijo, D.; Vega-Martin, E.; Schulz, A.; Ruilope, L.M.; Kolkhof, P.; Kreutz, R.; et al. Finerenone Attenuates Endothelial Dysfunction and Albuminuria in a Chronic Kidney Disease Model by a Reduction in Oxidative Stress. Front. Pharmacol. 2018, 9, 1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Han, Y.S.; Yoon, Y.M.; Go, G.; Lee, J.H.; Lee, S.H. Melatonin Protects Human Renal Proximal Tubule Epithelial Cells against High Glucose-Mediated Fibrosis via the Cellular Prion Protein-TGF-beta-Smad Signaling Axis. Int. J. Med. Sci. 2020, 17, 1235–1245. [Google Scholar] [CrossRef]
  116. Ebaid, H.; Bashandy, S.A.E.; Abdel-Mageed, A.M.; Al-Tamimi, J.; Hassan, I.; Alhazza, I.M. Folic acid and melatonin mitigate diabetic nephropathy in rats via inhibition of oxidative stress. Nutr. Metab. 2020, 17, 6. [Google Scholar] [CrossRef] [Green Version]
  117. Li, P.; Hu, F.; Cao, X.; Luo, L.; Tu, Q. Melatonin receptor protects cardiomyocyte against oxidative stress-induced apoptosis through the MAPK-ERK signaling pathway. J. Recept. Signal Transduct. 2020, 40, 117–125. [Google Scholar] [CrossRef] [PubMed]
  118. Kandemir, Y.B.; Tosun, V.; Guntekin, U. Melatonin protects against streptozotocin-induced diabetic cardiomyopathy through the mammalian target of rapamycin (mTOR) signaling pathway. Adv. Clin. Exp. Med. 2019, 28, 1171–1177. [Google Scholar] [CrossRef] [Green Version]
  119. Ceriello, A.; Novials, A.; Ortega, E.; Canivell, S.; La Sala, L.; Pujadas, G.; Esposito, K.; Giugliano, D.; Genovese, S. Glucagon-like peptide 1 reduces endothelial dysfunction, inflammation, and oxidative stress induced by both hyperglycemia and hypoglycemia in type 1 diabetes. Diabetes Care 2013, 36, 2346–2350. [Google Scholar] [CrossRef] [Green Version]
  120. Liu, J.; Guo, S.; Li, H.; Liu, X.Y. Effects of glucagon-like peptide-1 receptor agonists (GLP-1RAs) on podocytes, inflammation, and oxidative stress in patients with diabetic nephropathy (DN). Pak. J. Med. Sci. 2022, 38, 1170–1174. [Google Scholar] [CrossRef]
  121. Bray, J.J.H.; Foster-Davies, H.; Salem, A.; Hoole, A.L.; Obaid, D.R.; Halcox, J.P.J.; Stephens, J.W. Glucagon-like peptide-1 receptor agonists improve biomarkers of inflammation and oxidative stress: A systematic review and meta-analysis of randomised controlled trials. Diabetes Obes. Metab. 2021, 23, 1806–1822. [Google Scholar] [CrossRef] [PubMed]
  122. Pitt, B.; Filippatos, G.; Agarwal, R.; Anker, S.D.; Bakris, G.L.; Rossing, P.; Joseph, A.; Kolkhof, P.; Nowack, C.; Schloemer, P.; et al. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N. Engl. J. Med. 2021, 385, 2252–2263. [Google Scholar] [CrossRef] [PubMed]
  123. Bakris, G.L.; Agarwal, R.; Anker, S.D.; Pitt, B.; Ruilope, L.M.; Rossing, P.; Kolkhof, P.; Nowack, C.; Schloemer, P.; Joseph, A.; et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2020, 383, 2219–2229. [Google Scholar] [CrossRef]
  124. Pitt, B.; Kober, L.; Ponikowski, P.; Gheorghiade, M.; Filippatos, G.; Krum, H.; Nowack, C.; Kolkhof, P.; Kim, S.Y.; Zannad, F. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: A randomized, double-blind trial. Eur. Heart J. 2013, 34, 2453–2463. [Google Scholar] [CrossRef] [PubMed]
  125. Gueret, A.; Harouki, N.; Favre, J.; Galmiche, G.; Nicol, L.; Henry, J.P.; Besnier, M.; Thuillez, C.; Richard, V.; Kolkhof, P.; et al. Vascular Smooth Muscle Mineralocorticoid Receptor Contributes to Coronary and Left Ventricular Dysfunction after Myocardial Infarction. Hypertension 2016, 67, 717–723. [Google Scholar] [CrossRef] [Green Version]
  126. Barrera-Chimal, J.; Andre-Gregoire, G.; Nguyen Dinh Cat, A.; Lechner, S.M.; Cau, J.; Prince, S.; Kolkhof, P.; Loirand, G.; Sauzeau, V.; Hauet, T.; et al. Benefit of Mineralocorticoid Receptor Antagonism in AKI: Role of Vascular Smooth Muscle Rac1. J. Am. Soc. Nephrol. 2017, 28, 1216–1226. [Google Scholar] [CrossRef] [Green Version]
  127. Theofilis, P.; Vordoni, A.; Kalaitzidis, R.G. The Role of Melatonin in Chronic Kidney Disease and Its Associated Risk Factors: A New Tool in Our Arsenal? Am. J. Nephrol. 2022, 53, 565–574. [Google Scholar] [CrossRef]
  128. Kim, J.Y.; Park, J.H.; Jeon, E.J.; Leem, J.; Park, K.K. Melatonin Prevents Transforming Growth Factor-beta1-Stimulated Transdifferentiation of Renal Interstitial Fibroblasts to Myofibroblasts by Suppressing Reactive Oxygen Species-Dependent Mechanisms. Antioxidants 2020, 9, 39. [Google Scholar] [CrossRef] [Green Version]
  129. Hajam, Y.A.; Rai, S.; Pandi-Perumal, S.R.; Brown, G.M.; Reiter, R.J.; Cardinali, D.P. Coadministration of Melatonin and Insulin Improves Diabetes-Induced Impairment of Rat Kidney Function. Neuroendocrinology 2021, 112, 807–822. [Google Scholar] [CrossRef]
  130. Arinno, A.; Maneechote, C.; Khuanjing, T.; Ongnok, B.; Prathumsap, N.; Chunchai, T.; Arunsak, B.; Kerdphoo, S.; Shinlapawittayatorn, K.; Chattipakorn, S.C.; et al. Cardioprotective effects of melatonin and metformin against doxorubicin-induced cardiotoxicity in rats are through preserving mitochondrial function and dynamics. Biochem. Pharmacol. 2021, 192, 114743. [Google Scholar] [CrossRef]
  131. Ciric Zdravkovic, S.; Kostic, T.; Marcetic, Z.P.; Sulovic, L.S.; Nedeljkovic, B.M.; Preljevic, A.; Toskic, D.; Sokolovic, D. Melatonin modulates acute cardiac muscle damage induced by carbon tetrachloride—Involvement of oxidative damage, glutathione, and arginine and nitric oxide metabolism. Can. J. Physiol. Pharmacol. 2021, 99, 360–367. [Google Scholar] [CrossRef] [PubMed]
  132. Durdagi, G.; Pehlivan, D.Y.; Oyar, E.O.; Bahceci, S.A.; Ozbek, M. Effects of Melatonin and Adrenomedullin in Reducing the Cardiotoxic Effects of Doxorubicin in Rats. Cardiovasc. Toxicol. 2021, 21, 354–364. [Google Scholar] [CrossRef] [PubMed]
  133. Jiang, J.; Liang, S.; Zhang, J.; Du, Z.; Xu, Q.; Duan, J.; Sun, Z. Melatonin ameliorates PM2.5-induced cardiac perivascular fibrosis through regulating mitochondrial redox homeostasis. J. Pineal Res. 2021, 70, e12686. [Google Scholar] [CrossRef]
  134. Lan, H.; Su, Y.; Liu, Y.; Deng, C.; Wang, J.; Chen, T.; Jules, K.E.D.; Masau, J.F.; Li, H.; Wei, X. Melatonin protects circulatory death heart from ischemia/reperfusion injury via the JAK2/STAT3 signalling pathway. Life Sci. 2019, 228, 35–46. [Google Scholar] [CrossRef]
  135. Ma, X.; Wang, S.; Cheng, H.; Ouyang, H.; Ma, X. Melatonin Attenuates Ischemia/Reperfusion-Induced Oxidative Stress by Activating Mitochondrial Fusion in Cardiomyocytes. Oxid. Med. Cell. Longev. 2022, 2022, 7105181. [Google Scholar] [CrossRef]
  136. Xu, C.; Wang, J.; Fan, Z.; Zhang, S.; Qiao, R.; Liu, Y.; Yang, J.; Yang, L.; Wang, H. Cardioprotective effects of melatonin against myocardial ischaemia/reperfusion injury: Activation of AMPK/Nrf2 pathway. J. Cell. Mol. Med. 2021, 25, 6455–6459. [Google Scholar] [CrossRef] [PubMed]
  137. Bai, Y.; Yang, Y.; Cui, B.; Lin, D.; Wang, Z.; Ma, J. Temporal effect of melatonin posttreatment on anoxia/reoxygenation injury in H9c2 cells. Cell Biol. Int. 2022, 46, 637–648. [Google Scholar] [CrossRef]
  138. Singhanat, K.; Apaijai, N.; Sumneang, N.; Maneechote, C.; Arunsak, B.; Chunchai, T.; Chattipakorn, S.C.; Chattipakorn, N. Therapeutic potential of a single-dose melatonin in the attenuation of cardiac ischemia/reperfusion injury in prediabetic obese rats. Cell. Mol. Life Sci. 2022, 79, 300. [Google Scholar] [CrossRef]
  139. Han, D.; Wang, Y.; Chen, J.; Zhang, J.; Yu, P.; Zhang, R.; Li, S.; Tao, B.; Wang, Y.; Qiu, Y.; et al. Activation of melatonin receptor 2 but not melatonin receptor 1 mediates melatonin-conferred cardioprotection against myocardial ischemia/reperfusion injury. J. Pineal Res. 2019, 67, e12571. [Google Scholar] [CrossRef]
  140. Wang, B.; Li, J.; Bao, M.; Chen, R.; Li, H.; Lu, B.; Chen, M.; Huang, D.; Zhang, Y.; Gao, F.; et al. Melatonin Attenuates Diabetic Myocardial Microvascular Injury through Activating the AMPK/SIRT1 Signaling Pathway. Oxid. Med. Cell. Longev. 2021, 2021, 8882130. [Google Scholar] [CrossRef]
  141. Yang, Y.; Du, J.; Xu, R.; Shen, Y.; Yang, D.; Li, D.; Hu, H.; Pei, H.; Yang, Y. Melatonin alleviates angiotensin-II-induced cardiac hypertrophy via activating MICU1 pathway. Aging 2020, 13, 493–515. [Google Scholar] [CrossRef] [PubMed]
  142. Ishihara, R.; Barros, M.P.; Silva, C.M.D.; Borges, L.D.S.; Hatanaka, E.; Lambertucci, R.H. Melatonin improves the antioxidant capacity in cardiac tissue of Wistar rats after exhaustive exercise. Free Radic. Res. 2021, 55, 776–791. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, Y.; Zhang, S.; Ma, Y.; Xiang, A.; Sun, H.; Song, J.; Yang, W.; Li, X.; Xu, H. Melatonin protected against myocardial infarction injury in rats through a Sirt6-dependent antioxidant pathway. Adv. Clin. Exp. Med. 2022, 31, 277–284. [Google Scholar] [CrossRef] [PubMed]
  144. Satari, M.; Bahmani, F.; Reiner, Z.; Soleimani, A.; Aghadavod, E.; Kheiripour, N.; Asemi, Z. Metabolic and Anti-inflammatory Response to Melatonin Administration in Patients with Diabetic Nephropathy. Iran. J. Kidney Dis. 2021, 1, 22–30. [Google Scholar]
  145. Ostadmohammadi, V.; Soleimani, A.; Bahmani, F.; Aghadavod, E.; Ramezani, R.; Reiter, R.J.; Mansournia, M.A.; Banikazemi, Z.; Soleimani, M.; Zaroudi, M.; et al. The Effects of Melatonin Supplementation on Parameters of Mental Health, Glycemic Control, Markers of Cardiometabolic Risk, and Oxidative Stress in Diabetic Hemodialysis Patients: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Ren. Nutr. 2020, 30, 242–250. [Google Scholar] [CrossRef]
  146. Panah, F.; Ghorbanihaghjo, A.; Argani, H.; Haiaty, S.; Rashtchizadeh, N.; Hosseini, L.; Dastmalchi, S.; Rezaeian, R.; Alirezaei, A.; Jabarpour, M.; et al. The effect of oral melatonin on renal ischemia-reperfusion injury in transplant patients: A double-blind, randomized controlled trial. Transpl. Immunol. 2019, 57, 101241. [Google Scholar] [CrossRef]
  147. Hoseini, S.G.; Heshmat-Ghahdarijani, K.; Khosrawi, S.; Garakyaraghi, M.; Shafie, D.; Mansourian, M.; Roohafza, H.; Azizi, E.; Sadeghi, M. Melatonin supplementation improves N-terminal pro-B-type natriuretic peptide levels and quality of life in patients with heart failure with reduced ejection fraction: Results from MeHR trial, a randomized clinical trial. Clin. Cardiol. 2022, 45, 417–426. [Google Scholar] [CrossRef]
  148. Dominguez-Rodriguez, A.; Abreu-Gonzalez, P.; Baez-Ferrer, N.; Reiter, R.J.; Avanzas, P.; Hernandez-Vaquero, D. Melatonin and Cardioprotection in Humans: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Cardiovasc. Med. 2021, 8, 635083. [Google Scholar] [CrossRef]
  149. Lv, T.; Yan, J.; Lou, Y.; Zhang, Z.; Ye, M.; Zhou, J.; Luo, F.; Bi, C.; Lin, H.; Zhang, J.; et al. Evaluation of Melatonin Therapy in Patients with Myocardial Ischemia-Reperfusion Injury: A Systematic Review and Meta-Analysis. Oxid. Med. Cell. Longev. 2022, 2022, 4610522. [Google Scholar] [CrossRef]
  150. Kim, H.; Lee, S.H.; Cho, J.H.; Lee, H.; Yim, H.W.; Yoon, K.H.; Kim, H.S. Discontinuation rate and reason for discontinuation after sodium-glucose cotransporter 2 inhibitor prescription in real clinical practice. J. Clin. Pharm. Ther. 2020, 45, 1271–1277. [Google Scholar] [CrossRef]
  151. Trujillo, J.M.; Nuffer, W.; Smith, B.A. GLP-1 receptor agonists: An updated review of head-to-head clinical studies. Ther. Adv. Endocrinol. Metab. 2021, 12, 2042018821997320. [Google Scholar] [CrossRef] [PubMed]
  152. Meili-Butz, S.; Niermann, T.; Fasler-Kan, E.; Barbosa, V.; Butz, N.; John, D.; Brink, M.; Buser, P.T.; Zaugg, C.E. Dimethyl fumarate, a small molecule drug for psoriasis, inhibits Nuclear Factor-kappaB and reduces myocardial infarct size in rats. Eur. J. Pharmacol. 2008, 586, 251–258. [Google Scholar] [CrossRef] [PubMed]
  153. Luo, M.; Sun, Q.; Zhao, H.; Tao, J.; Yan, D. The Effects of Dimethyl Fumarate on Atherosclerosis in the Apolipoprotein E-Deficient Mouse Model with Streptozotocin-Induced Hyperglycemia Mediated By the Nuclear Factor Erythroid 2-Related Factor 2/Antioxidant Response Element (Nrf2/ARE) Signaling Pathway. Med. Sci. Monit. 2019, 25, 7966–7975. [Google Scholar] [CrossRef] [PubMed]
  154. Ashari, S.; Naghsh, N.; Salari, Y.; Barghi, N.G.; Bagheri, A. Dimethyl Fumarate Attenuates Di-(2-Ethylhexyl) Phthalate-Induced Nephrotoxicity through the Nrf2/HO-1 and NF-kappaB Signaling Pathways. Inflammation 2022. [Google Scholar] [CrossRef]
  155. Zhen, X.; Jindong, L.; Yang, Z.; Yashi, R.; Wei, G.; Wei, J.; Wei, Z.; Sudong, L. Activation of Nrf2 Pathway by Dimethyl Fumarate Attenuates Renal Ischemia-Reperfusion Injury. Transplant. Proc. 2021, 53, 2133–2139. [Google Scholar] [CrossRef]
  156. Zhou, K.; Xie, M.; Yi, S.; Tang, Y.; Luo, H.; Xiao, Q.; Xiao, J.; Li, Y. Dimethyl fumarate ameliorates endotoxin-induced acute kidney injury against macrophage oxidative stress. Ren. Fail. 2021, 43, 1229–1239. [Google Scholar] [CrossRef] [PubMed]
  157. Takasu, C.; Vaziri, N.D.; Li, S.; Robles, L.; Vo, K.; Takasu, M.; Pham, C.; Liu, S.; Farzaneh, S.H.; Foster, C.E., 3rd; et al. Treatment with Dimethyl Fumarate Attenuates Calcineurin Inhibitor-induced Nephrotoxicity. Transplantation 2015, 99, 1144–1150. [Google Scholar] [CrossRef]
  158. Zeng, S.Y.; Yang, L.; Yan, Q.J.; Gao, L.; Lu, H.Q.; Yan, P.K. Nox1/4 dual inhibitor GKT137831 attenuates hypertensive cardiac remodelling associating with the inhibition of ADAM17-dependent proinflammatory cytokines-induced signalling pathways in the rats with abdominal artery constriction. Biomed. Pharmacother. 2019, 109, 1907–1914. [Google Scholar] [CrossRef]
  159. Zheng, H.; Xu, N.; Zhang, Z.; Wang, F.; Xiao, J.; Ji, X. Setanaxib (GKT137831) Ameliorates Doxorubicin-Induced Cardiotoxicity by Inhibiting the NOX1/NOX4/Reactive Oxygen Species/MAPK Pathway. Front. Pharmacol. 2022, 13, 823975. [Google Scholar] [CrossRef]
  160. Zhao, Q.D.; Viswanadhapalli, S.; Williams, P.; Shi, Q.; Tan, C.; Yi, X.; Bhandari, B.; Abboud, H.E. NADPH oxidase 4 induces cardiac fibrosis and hypertrophy through activating Akt/mTOR and NFkappaB signaling pathways. Circulation 2015, 131, 643–655. [Google Scholar] [CrossRef] [Green Version]
  161. Gray, S.P.; Jha, J.C.; Kennedy, K.; van Bommel, E.; Chew, P.; Szyndralewiez, C.; Touyz, R.M.; Schmidt, H.; Cooper, M.E.; Jandeleit-Dahm, K.A.M. Combined NOX1/4 inhibition with GKT137831 in mice provides dose-dependent reno- and atheroprotection even in established micro- and macrovascular disease. Diabetologia 2017, 60, 927–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Liang, Y.; Liu, H.; Fang, Y.; Lin, P.; Lu, Z.; Zhang, P.; Jiao, X.; Teng, J.; Ding, X.; Dai, Y. Salvianolate ameliorates oxidative stress and podocyte injury through modulation of NOX4 activity in db/db mice. J. Cell. Mol. Med. 2021, 25, 1012–1023. [Google Scholar] [CrossRef]
  163. Gorin, Y.; Cavaglieri, R.C.; Khazim, K.; Lee, D.Y.; Bruno, F.; Thakur, S.; Fanti, P.; Szyndralewiez, C.; Barnes, J.L.; Block, K.; et al. Targeting NADPH oxidase with a novel dual Nox1/Nox4 inhibitor attenuates renal pathology in type 1 diabetes. Am. J. Physiol. Renal. Physiol. 2015, 308, F1276–F1287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Jha, J.C.; Gray, S.P.; Barit, D.; Okabe, J.; El-Osta, A.; Namikoshi, T.; Thallas-Bonke, V.; Wingler, K.; Szyndralewiez, C.; Heitz, F.; et al. Genetic targeting or pharmacologic inhibition of NADPH oxidase nox4 provides renoprotection in long-term diabetic nephropathy. J. Am. Soc. Nephrol. 2014, 25, 1237–1254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Di Marco, E.; Gray, S.P.; Chew, P.; Koulis, C.; Ziegler, A.; Szyndralewiez, C.; Touyz, R.M.; Schmidt, H.H.; Cooper, M.E.; Slattery, R.; et al. Pharmacological inhibition of NOX reduces atherosclerotic lesions, vascular ROS and immune-inflammatory responses in diabetic Apoe(-/-) mice. Diabetologia 2014, 57, 633–642. [Google Scholar] [CrossRef] [PubMed]
  166. Shin, H.; Jeong, S.; Lee, Y.; Jeon, C.; Kwon, G.; Kim, S.; Lee, D. H2O2-Activatable Antioxidant Polymeric Prodrug Nanoparticles for the Prevention of Renal Ischemia/Reperfusion Injury. Biomacromolecules 2022, 23, 3810–3821. [Google Scholar] [CrossRef] [PubMed]
  167. Bae, S.; Park, M.; Kang, C.; Dilmen, S.; Kang, T.H.; Kang, D.G.; Ke, Q.; Lee, S.U.; Lee, D.; Kang, P.M. Hydrogen Peroxide-Responsive Nanoparticle Reduces Myocardial Ischemia/Reperfusion Injury. J. Am. Heart Assoc. 2016, 5, e003697. [Google Scholar] [CrossRef]
  168. Altshuler, P.J.; Schiazza, A.R.; Luo, L.; Helmers, M.R.; Chhay, B.; Han, J.J.; Hu, R.; Herbst, D.A.; Tsourkas, A.; Cheng, Z.; et al. Superoxide Dismutase-Loaded Nanoparticles Attenuate Myocardial Ischemia-Reperfusion Injury and Protect against Chronic Adverse Ventricular Remodeling. Adv. Ther. 2021, 4, 2100036. [Google Scholar] [CrossRef]
  169. Seshadri, G.; Sy, J.C.; Brown, M.; Dikalov, S.; Yang, S.C.; Murthy, N.; Davis, M.E. The delivery of superoxide dismutase encapsulated in polyketal microparticles to rat myocardium and protection from myocardial ischemia-reperfusion injury. Biomaterials 2010, 31, 1372–1379. [Google Scholar] [CrossRef] [Green Version]
  170. Choi, H.S.; Mathew, A.P.; Uthaman, S.; Vasukutty, A.; Kim, I.J.; Suh, S.H.; Kim, C.S.; Ma, S.K.; Graham, S.A.; Kim, S.W.; et al. Inflammation-sensing catalase-mimicking nanozymes alleviate acute kidney injury via reversing local oxidative stress. J. Nanobiotechnol. 2022, 20, 205. [Google Scholar] [CrossRef]
Figure 1. Agents with antioxidant effects, such as sodium-glucose cotransporter-2 (SGLT2) inhibitors, glucagon-like peptide-1 receptor agonists (GLP1-RA), finerenone, and melatonin may induce reductions in reactive oxygen species (ROS) and reinforce antioxidant system activity. Ultimately, the burden of diseases associated with oxidative stress such as heart failure (HF), atherosclerosis, and chronic kidney disease (CKD) is diminished. SOD: superoxide dismutase, GPx: glutathione peroxidase, NAD+: nicotinamide adenine dinucleotide, POX: paraoxonases, Nox: NADPH oxidase, XO: xanthine oxidase, MDA: malondialdehyde.
Figure 1. Agents with antioxidant effects, such as sodium-glucose cotransporter-2 (SGLT2) inhibitors, glucagon-like peptide-1 receptor agonists (GLP1-RA), finerenone, and melatonin may induce reductions in reactive oxygen species (ROS) and reinforce antioxidant system activity. Ultimately, the burden of diseases associated with oxidative stress such as heart failure (HF), atherosclerosis, and chronic kidney disease (CKD) is diminished. SOD: superoxide dismutase, GPx: glutathione peroxidase, NAD+: nicotinamide adenine dinucleotide, POX: paraoxonases, Nox: NADPH oxidase, XO: xanthine oxidase, MDA: malondialdehyde.
Life 12 01663 g001
Table 1. Preclinical evidence of renal antioxidant effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors.
Table 1. Preclinical evidence of renal antioxidant effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors.
StudyExperimental ModelDisease TypeSGLT2 InhibitorSGLT2 Inhibitor Effect
Ashrafi Jigheh et al. [66]Wistar ratsDMEmpagliflozin ↓ renal MDA
↑ renal SOD and GPx
Kimura et al. [67] OLETF rats DMCanagliflozin ↓ renal MDA, 4HNE, Nox2, and Nox4
Das et al. [68]Proximal tubular epithelial cellsHigh glucoseEmpagliflozin↓ O− 2 and H2O2 generation
Zaibi et al. [69]Human proximal tubular cellsH2O2-induced injuryDapagliflozin ↓ cytosolic ROS production
Ahmed et al. [70]Wistar ratsDM post-MIEmpagliflozin ↓ renal Nox2 and Nox4 mRNA
Hudkins et al. [71]BTBR ob/ob miceDNEmpagliflozin ↓ urinary markers of RNA/DNA damage
↓ carbonyl oxidation in situ
Ala et al. [72]Wistar ratsRenal IR injuryEmpagliflozin ↓ renal MDA
Malinska et al. [73]Spontaneously hypertensive ratsInflammationEmpagliflozin ↑ renal GPx, CAT, GSH
↓ renal CD, TBARS
Ye et al. [74]C57BL/6J miceObesityEmpagliflozin ↑ heme oxygenase-1
DM: diabetes mellitus, MDA: malondialdehyde, SOD: superoxide dismutase, GPx: glutathione peroxidase, OLETF: Otsuka Long-Evans Tokushima Fatty, 4HNE: 4-hydroxynonenal, Nox: nicotinamide adenine dinucleotide phosphate oxidase, ROS: reactive oxygen species, MI: myocardial infarction, DN: diabetic nephropathy, IR: ischemia-reperfusion, CAT: catalase, GSH: glutathione, CD: conjugated dienes, TBARS: thiobarbituric acid reactive substances. ↑ indicates an increase, ↓ indicates a decrease.
Table 2. Preclinical evidence of antioxidant effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors on the cardiovascular system.
Table 2. Preclinical evidence of antioxidant effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors on the cardiovascular system.
StudyExperimental ModelDisease TypeSGLT2 InhibitorSGLT2 Inhibitor Effect
Xing et al. [75]Sprague-Dawley rats
Cardiac myoblasts H9C2
DMDapagliflozin ↓ myocardial MDA, Cu/Zn SOD
↓ cardiomyoblast H2O2,
↑ cardiomyoblast Cu/Zn-SOD expression and total SOD activity
Hsieh et al. [76]Cardiac myoblast H9C2Doxorubicin-induced injuryDapagliflozin ↑ heme oxygenase-1 and NADPH quinone oxidoreductase
↑ SOD activity
Bugga et al. [77]Sprague-Dawley ratsDMEmpagliflozin ↓ total cellular and mitochondrial ROS
Li et al. [78]C57Bl/6JPressure Overload-Induced HFEmpagliflozin↑ heme oxygenase-1, NRF-2, catalase
↓ O2-, H2O2
Tsai et al. [79]Cardiac myoblasts H9C2
Primary cardiomyocytes
Cardiac IR injuryDapagliflozin ↓ NADPH activity
↓ ROS formation
Rosa et al. [80]Wistar ratsDMDapagliflozin ↓ lipid hydroperoxide
↑ SOD, GPx
Wang et al. [81]db/db mice
Cardiac myoblasts H9C2
DMEmpagliflozin ↓ cardiac 4HNE and 3-nitrotyrosine
↓ cardiac total cellular and mitochondrial ROS
↓ cardiomyoblast total cellular and mitochondrial ROS
Kolijn et al. [82]Human cardiomyocytes
ZDF rats
HFpEFEmpagliflozin ↓ H2O2, 3-nitrotyrosine
↑ glutathione
El-Shafey et al. [83]Sprague-Dawley ratsDMDapagliflozin ↓ myocardial MDA
↑ myocardial glutathione, catalase
Li et al. [84]KK-Ay miceDMEmpagliflozin ↓ myocardial lipid hydroperoxide, MDA, Nox4
↑ myocardial GPx, SOD
Wang et al. [85]Sprague-Dawley ratsIsoproterenol-induced cardiomyopathyDapagliflozin ↓ myocardial Nox2, MDA, ROS, NADPH activity,
Yurista et al. [86]Sprague-Dawley ratsPost-MI HFEmpagliflozin↓ AOPP, Nox2
Uthman et al. [65]HUVECs
HCAECs
TNFα-induced endothelial dysfunctionEmpagliflozin ↓ HUVEC and HCAEC ROS production
Rahadian et al. [87]ApoE−/− miceDMCanagliflozin ↓ aortic Nox2, p22phox
↓ urinary 8-hydroxydeoxyguanosine
MDA: malondialdehyde, SOD: superoxide dismutase, NADPH: nicotinamide adenine dinucleotide phosphate, ROS: reactive oxygen species, HF: heart failure, NRF-2: nuclear factor erythroid 2–related factor 2, GPx: glutathione peroxidase, 4HNE: 4-hydroxynonenal, HFpEF: HF with preserved ejection fraction, MI: myocardial infarction, Nox2: NADPH oxidase-2, HUVEC: human umbilical vein endothelial cell, HCAEC: human coronary artery endothelial cell, TNFα: tumor necrosis factor-α. ↑ indicates an increase, ↓ indicates a decrease.
Table 3. Selected preclinical evidence of cardiac and renal antioxidant effects of GLP1 receptor agonists, finerenone, and melatonin.
Table 3. Selected preclinical evidence of cardiac and renal antioxidant effects of GLP1 receptor agonists, finerenone, and melatonin.
StudyExperimental ModelDisease TypeAgentAntioxidant Effect
Liljedahl et al. [98]129SV miceDMLiraglutide ↑ renal catalase, GPx
Abdel-Latif et al. [99]Wistar ratsDMLixisenatide ↓ renal MDA and total Nox
↑ total antioxidant capacity
Baylan et al. [102]Sprague-Dawley ratsDMLiraglutide ↓ atrial and ventricular Nox2
Nuamnaichati et al. [104]H9C2 cellsH2O2GLP1 analogue ↑ GPx, catalase, heme oxygenase-1
Zhang et al. [103]Neonatal cardiomyocyteDM + inflammationLiraglutide ↓ MDA
↑ SOD activity
Lachaux et al. [113]Zucker fa/fa ratsMetabolic syndromeFinerenone ↓ myocardial ROS
↑ NO bioavailability
González-Blázquez et al. [114]Munich Wistar Frömter ratsCKDFinerenone ↑ aortic Mn-SOD and Cu/Zn-SOD
↑ renal total SOD activity
Han et al. [115]Human renal proximal tubule epithelial cellsDMMelatonin ↑ catalase and SOD activity
Ebaid et al. [116]Albino ratsDNMelatonin ↓ renal MDA
↑ GSH, SOD, catalase
Li et al. [117]H9C2 cellsH2O2 toxicityMelatonin ↑ GSH, GPx, SOD
Kandemir et al. [118]Wistar ratsDMMelatonin ↑ cardiac SOD, catalase, GPx
DM: diabetes mellitus, GPx: glutathione peroxidase, MDA: malondialdehyde, Nox: NADPH oxidase, SOD: superoxide dismutase, ROS: reactive oxygen species, NO: nitric oxide CKD: chronic kidney disease, DN: diabetic nephropathy, GSH: glutathione. ↑ indicates an increase, ↓ indicates a decrease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Theofilis, P.; Vordoni, A.; Kalaitzidis, R.G. Oxidative Stress Management in Cardiorenal Diseases: Focus on Novel Antidiabetic Agents, Finerenone, and Melatonin. Life 2022, 12, 1663. https://doi.org/10.3390/life12101663

AMA Style

Theofilis P, Vordoni A, Kalaitzidis RG. Oxidative Stress Management in Cardiorenal Diseases: Focus on Novel Antidiabetic Agents, Finerenone, and Melatonin. Life. 2022; 12(10):1663. https://doi.org/10.3390/life12101663

Chicago/Turabian Style

Theofilis, Panagiotis, Aikaterini Vordoni, and Rigas G. Kalaitzidis. 2022. "Oxidative Stress Management in Cardiorenal Diseases: Focus on Novel Antidiabetic Agents, Finerenone, and Melatonin" Life 12, no. 10: 1663. https://doi.org/10.3390/life12101663

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