Next Article in Journal
Influence of Fetomaternal Microchimerism on Maternal NK Cell Reactivity against the Child’s Leukemic Blasts
Next Article in Special Issue
Melatonin Supplement Plus Exercise Effectively Counteracts the Challenges of Isoproterenol-Induced Cardiac Injury in Rats
Previous Article in Journal
Infigratinib (BGJ 398), a Pan-FGFR Inhibitor, Targets P-Glycoprotein and Increases Chemotherapeutic-Induced Mortality of Multidrug-Resistant Tumor Cells
Previous Article in Special Issue
Ceramides and Cardiovascular Risk Factors, Inflammatory Parameters and Left Ventricular Function in AMI Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Reactive Oxygen Species Induced Pathways in Heart Failure Pathogenesis and Potential Therapeutic Strategies

by
Aušra Mongirdienė
1,*,
Laurynas Skrodenis
2,
Leila Varoneckaitė
2,
Gerda Mierkytė
2 and
Justinas Gerulis
2
1
Department of Biochemistry, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, LT-50161 Kaunas, Lithuania
2
Medical Academy, Lithuanian University of Health Sciences, Mickevičiaus str. 9, LT-44307 Kaunas, Lithuania
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(3), 602; https://doi.org/10.3390/biomedicines10030602
Submission received: 14 January 2022 / Revised: 25 February 2022 / Accepted: 2 March 2022 / Published: 3 March 2022
(This article belongs to the Special Issue Oxidative Stress and Antioxidants in Cardiovascular Diseases)

Abstract

:
With respect to structural and functional cardiac disorders, heart failure (HF) is divided into HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Oxidative stress contributes to the development of both HFrEF and HFpEF. Identification of a broad spectrum of reactive oxygen species (ROS)-induced pathways in preclinical models has provided new insights about the importance of ROS in HFrEF and HFpEF development. While current treatment strategies mostly concern neuroendocrine inhibition, recent data on ROS-induced metabolic pathways in cardiomyocytes may offer additional treatment strategies and targets for both of the HF forms. The purpose of this article is to summarize the results achieved in the fields of: (1) ROS importance in HFrEF and HFpEF pathophysiology, and (2) treatments for inhibiting ROS-induced pathways in HFrEF and HFpEF patients. ROS-producing pathways in cardiomyocytes, ROS-activated pathways in different HF forms, and treatment options to inhibit their action are also discussed.

1. Introduction

With respect to structural and functional cardiac disorders, chronic heart failure (CHF) is classified into heart failure (HF) with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). The most common cause of HFrEF is cardiomyocyte loss due to ischemia. HFpEF is a heterogeneous syndrome with multiple different conditions that can contribute differently to the syndrome [1]. Patients with HFpEF make up more than 50% of all HF patients [1,2]. In recent decades, understanding of the pathophysiology and treatment of HFrEF has increased [3,4]. However, treatment options for patients with HFpEF are few [1]. The main therapeutic target for patients with HFrEF is the neuroendocrine chain with therapies including inhibitors of the renin angiotensin-aldosterone system, mineralocorticoid receptor antagonists, β-receptor blockers, and medications that increase the half-life of natriuretic peptides [5]. HFpEF treatment relies on addressing the reasons for the observed syndrome, including treating the underlying disease, blood pressure control, use of diuretics and addressing other factors that contribute to development of HFpEF [1].
Oxidative stress (an imbalance between the increased formation of reactive oxygen species (ROS) and the elimination or neutralization of ROS by an antioxidant system) plays an important role in the development of CHF [6] and correlates with left ventricle (LV) dysfunction and hypertrophy in the failing heart [7]. Therefore, pharmacologically targeting specific ROS and pathways induced by them, could be beneficial for CHF patients. Broad and detailed knowledge of the particular sources and formation of ROS, as well as their elimination in the cell, is required in order to better understand the ROS induced pathways. Therefore, we have sought to summarize results achieved in the fields of: (1) ROS significance for HFrEF and HFpEF pathophysiology, and (2) treatment options in the management of ROS-induced pathways in the human heart for both HFrEF and HFpEF patients.

2. ROS Sources, Importance and Danger in Human Cells

ROS are chemically reactive molecules that belong to a group of nine major types of free radicals. These molecules have an unpaired electron in the superoxide anion O2, which is unstable. Several compounds are termed ROS, including: free radicals (superoxide anion (O2)), the hydroxyl radical (.OH)) and oxidative agents (e.g., hydrogen peroxide (H2O2), peroxynitrite (ONOO), hypochlorite (OCl)) [8,9]. H2O2 is involved in the Fenton reaction in the presence of Fe2+ to produce –OH. O2 and H2O2 can produce –OH through the Haber–Weiss reaction [10]. ONOO is produced in the reaction of .NO with O2 [11] (Figure 1) and is known to contribute to chronic heart failure (CHF) pathogenesis [12].
The agents referred to are produced in the cells by the mitochondria and enzymes, such as lipoxygenases and cyclooxygenases, under normal conditions [13]. Some processes, such as apoptosis, immune system reactions, differentiation, activation of several transcriptional factors, cellular signaling pathways and induction of a mitogenic response require the presence of some ROS [14]. ROS signaling is either reversible and oxidative or produces reactive nitrogen species. O2•− takes part in signal transmission by: (1) causing post-translation redox modifications of proteins [15], (2) hydroxylation (addition of an HO group) [16], and (3) S-nitrosylation (oxidation of cysteine by NO) [17]. By these means, the reactivity, stability and conformation of the affected molecules is altered [17]. The superoxide anion and hydrogen peroxide are the main ROS that participate in redox signaling [18] (Figure 1). Hydroxyl radicals are more reactive and less specific and reversible [19]. Cell antioxidant enzymes, including superoxide dismutase, catalase, and glutathione peroxidase, protect the cell from ROS excess [20]. ROS excess is known to give rise to oxidative stress, which affects subcellular organelles, changes intracellular enzyme activity, creates intracellular Ca2+ overload and modulates gene expression [21,22]. In turn, cell lipids, proteins and deoxyribonucleic acid (DNA) are damaged, leading to impairment of normal cell function [22] (Figure 2).
ROS, as reported in the literature, activate the signal kinase and transcription factors that modify the function of intracellular proteins and signaling pathways in the heart and in this way contribute to the hypertrophic remodeling of the heart [23,24]. Additionally, ROS damage mitochondrial phospholipid membranes and, as a consequence, induce mitochondrial oxidative stress, leading to molecular mechanisms that contribute to the development and progression of heart failure [25].

3. ROS in the Pathogenesis of CHF Development

Being a by-product of aerobic metabolism, ROS are abundant in the cells of the myocardium and, if the balance between ROS production and antioxidant systems is impaired, they can greatly contribute to, or worsen, HF [26].
The proteins involved in redox signaling are protein kinase G (PKG) [27], the small G protein Ras [28], Ca/calmodulin-dependent protein kinase II (CaMKII) [29], protein kinase A (PKA) [30], class II histone deacetylases (HDACs) [31], matrix metalloproteinase (MMP) [32], protein kinase B/Akt [33], the extracellular signal-regulated kinase ½ (ERK1/2) [34], p38 MAP kinase [35], protein kinase C (PKC) [36], NF-kappa B [37], and transcription factors, including activated protein-1 [38].
Cardiomyocyte hypertrophy has been found to be associated with ROS activation of signaling kinases and transcription factors [23]. ROS also promotes post-translational modifications that change the function of specific proteins and signaling pathways, leading to hypertrophic remodeling [23,39]. ROS have been shown to be important in G protein-coupled receptor stimulation by angiotensin II, tumor necrosis factor-α (TNF-α), and α-adrenergic stimulation [23,40,41]. Angiotensin II may participate in myocardial hypertrophy by several intracellular pathways by activating: (1) protein kinase C, (2) c-Jun N-terminal kinase (JNK), (3) extracellular signal-regulated kinase, and (4) ROS formation [40,42]. Though the role of TNF-α in cardiomyocytes is not yet sufficiently known, TNF-α seems to play an autocrine or paracrine role in activating MMPs, which promote hypertrophic changes in the heart [41].
ROS affect different lipid membranes too, including the sarcolemma, mitochondrial membranes, nuclear membrane, and the sarcoplasmic reticulum, in which lipid radicals and lipid hydroperoxide (LPH) form [25,43,44]. As the lipid peroxidation cascade progresses, LPH reacts with fatty acids to form a more stable product, for example—malondialdehyde or 4-hydroxy-2-nonenal [45]. Destabilization of the phospholipid-rich inner mitochondrial membrane by peroxidation results in additional electron leakage and increase in ROS production intensity [43,45].
ROS activate the cardiac Na+/Ca2+ exchanger, which triggers cardiac hypertrophy through the Ca2+-dependent pathway [46] and contributes to Ca2+/calmodulin-dependent protein kinase II activation, leading to increase in cardiomyocyte death and CHF development [47]. The cardiac Na+/H+ exchanger (NHE1) was shown to be activated [48] and sarcolemmal Na+/K+ ATPase was found to be suppressed by ROS [49] and to be implicated in cardiac hypertrophy. It should be emphasized that the elevation of Na+ in cardiomyocytes may contribute to slower cardiac muscle relaxation and arrhythmias [50].
It should also be mentioned that heme oxygenase (HO) (an enzyme that catalyzes heme degradation) has been shown to reduce oxidative stress in cardiomyocytes by catalyzing the carbon monoxide (CO) producing reaction [50,51]. CO has been shown to act as an antioxidant and contribute to the anti-hypertrophic effect [51].
Additionally, ROS induced endothelial damage [52,53] and thrombosis development [54] are stated in the literature to take place in chronic HF development.

3.1. Enzymes Involved in ROS Production

NADPH oxidases (NOX) 2 and 4 [55], xanthine oxidoreductase (XOR) [56], and nitric oxide synthase (NOS) [57] are the common enzymes that produce ROS in cardiomyocytes (Figure 3).
Humans have seven NOX with a similar catalytic core, but different regulatory mechanisms [19]. NOX2 and NOX4 are abundantly expressed in cardiomyocytes, endothelium and fibroblasts. Every NOX produces the superoxide anion [62]. NOX2 and NOX4 activity is presented in Figure 3. The data presented in [69,70] implies that NOX4-derived ROS could contribute to overload-induced LV hypertrophy (LVH), and that NOX2 is produced in response to angiotensin II infusion. However, some studies suggest that LVH, as a response to chronic renin-angiotensin-aldosterone system activation, is not associated with NOX2 [71,72]. The enzymes involved in ROS production in cardiomyocytes and fibroblasts, and the pathways that they activate according to the literature [58,59,60,61,62,63,64,65,66,67,68], are presented in summary in Figure 3.
NOX has been shown to be involved in MMP activation in response to angiotensin II [73] and mechanical stretch [71] in the vessels. Experiments with mice and rats have demonstrated the role of NOX2 in the development of interstitial cardiac fibrosis, however, the NOX2-expressing cell type was not established [71,72,74]. NOX4 was shown to be expressed in cardiac fibroblasts [75,76] in animal models. Currently there are no in vivo experiments that could confirm the analogous case in humans.
The other ROS produced enzyme is XOR. XOR is involved in: (1) degradation of the purine nucleotides (AMP and GMP), in which it oxidizes hypoxanthine and xanthine to uric acid and H2O2 [77], (2) reduction of nitrite and nitrate, in which it produces NO and consequently promotes vasodilatation [78], or inflammation [78] and mitochondrial damage (as a result of overproduction) [79]. There are two forms of XOR: xanthine dehydrogenase and xanthine oxidase (XO). XO is involved in H2O2 production [80]. High levels of uric acid are found in patients with HF blood tests [77]; therefore, production of H2O2 is expected to be increased in these patients as well.
NOS catalyze NO production in a reaction where L-arginine is converted to L-citruline [81]. There are three isoforms of NOS. Two isoforms of NOS (endothelial (eNOS) and neuronal (nNOS)) are expressed more intensely in cardiomyocytes [82]. However, iNOS can contribute to contractile damage in CHF as well [83].
Increased ROS production is related both to myosin-activated protein kinase (MAPK) [64] (Figure 3) and adenosine monophosphate activated protein kinase (AMPK) activation [84]. AMPK activation leads to an increase in the antioxidants SOD and catalase (CAT) and uncoupling of protein 2 (UCP2) gene expression, leading to weakened apoptosis and NOX expression decrease [84] (not shown in Figure 3).
Another group of enzymes involved in CHF development through ROS production. is a family of NAD+ dependent class III histone deacetylases called sirtuins (SIRT) [85]. There are seven members of this enzyme group in different cell departments. SIRT3 is found in the mitochondria [86] and is involved in ATP production and ROS detoxication [87,88]. Some studies have found that SIRT3 is involved in cardiac muscle hypertrophy and fibrosis, leading eventually to CHF development [89,90]. Additionally, some studies have found that SIRT3 is involved in oxidative stress-mediated cell death in cardiomyocytes through protein Ku70 deacetylation, leading to deacetylated Ku70 interaction with the apoptosis regulator bcl-2-like protein4 (Bax) [91]. However, other studies have highlighted several mechanisms through which SIRT3 exerts a cardioprotective effect: (1) SIRT3 activates the antioxidant enzyme superoxide dismutase (SOD) [92], and (2) activates isocitrate dehydrogenase 2 (IDH2) by deacetylation. IDH2 uses NADP+ for reduction. SIRT3, in this way, increases NADPH levels, and increases glutathione (GSH) levels, thereby, inhibiting ROS production [93].
It can be concluded that several redox-signaling pathways may be modulated by ROS-producing enzymes, leading to cardiomyocyte hypertrophy, interstitial fibrosis and apoptosis. The value of NOX seems to be far more important in comparison with the other enzymes in that its activation can be triggered by both neuroendocrine factors and pressure overload, or by inflammatory cytokines. NOS play an important role in redox alterations in CHF development, both with substrates and cofactors. Sirtuins are involved in the enzyme-antioxidant activation that protects the heart from hypertrophy. NOX-, XOR-, NOS- and SIRT-mediated pathways could, therefore, be potential treatment targets for CHF development suppression.

3.2. Mitochondria in ROS Production and Enzyme-Antioxidants

The most abundant source of ROS in cells is the mitochondrial electron transport chain (ETC). A total of 0.2–2% of the electrons in the ETC leak out of the chain and interact with oxygen to produce superoxide or hydrogen peroxide [47]. Additionally, H2O2 is produced in the reaction, catalyzed by SOD1. After it has diffused from mitochondria, H2O2 is involved in physiological and pathological pathways (damaging proteins and lipids) [47]. H2O2 damages mitochondrial DNA, interferes with the Krebs cycle, ATP production, and fatty acid metabolism [94] and can trigger the opening of ion channels or the inner membrane anion channel inside the mitochondria, leading to cell death [95]. Proton leakage in the mitochondria consists of: (1) basal leakage (not regulated and related to the inner mitochondrial membrane’s lipid bilayer and the adenine nucleotide translocase (ANT)), and (2) inducible leakage (regulated and catalyzed or suppressed by uncoupling proteins and ANT) [47]. UCP2 is involved in cardiovascular disease; therefore, drugs targeting UCP expression or activity might be a potential treatment option. Hypoxia is suggested to further increase ROS production in the mitochondria [47]. ETC complexes III (CIII), and especially I (CI), are found to be the main sites of ROS production [96]. Therefore, the regulation of ROS production in these complexes may yield significant results.
Mitochondrial ROS are also involved in different cell signaling pathways, involving apoptosis [97], autophagy [98], and necrosis [99].
Other ROS sources in the mitochondria are the enzymes monoamine oxidase A and B, both located within the outer mitochondrial membrane (OMM). They catalyze the oxidative deamination of neurotransmitters and biogenic amines, leading to H2O2 production [100]. Monoamine oxidase A (MAO-A) is specific to cardiomyocytes [101]. It was discovered that MAO-A-dependent ROS formation may impair autophagy, leading to the accumulation of autophagosomes and mitochondrial fusion, resulting in microtubule-associated protein light chain 3-phosphatidylethanolamine conjugate (recruited to autophagosomal membranes) formation, and autophagy receptor (p62) and ubiquitylated protein accumulation, causing cardiomyocyte death and CHF. Both MAO-A derived H2O2 and aldehydes are able to directly target mitochondrial function [102]. Additionally, MAO-A-generated ROS has been shown to inhibit sphingosine kinase, which leads to ceramide accumulation and, thereby, to cardiomyocyte apoptosis [103]. Contractile proteins (actin and tropomyosin) are also affected [104].
Excess of H2O2 in the cell is eliminated by glutathione peroxidase (GPX) and peroxiredoxin (PRX). Both of these require GSH and thioredoxin for regeneration [105], which requires NADPH as a cofactor [105,106]. Nicotinamide nucleotide transhydrogenase, NADP+-dependent isocitrate dehydrogenase (IDH) and malic enzyme are involved in NADP+ regeneration to NADPH [107]. These require Krebs cycle products—NADH, malate and isocitrate. With IDH being the most important for NADPH regeneration [108], the Krebs cycle is necessary for the antioxidant capacity within mitochondria [109] as well as in the cytosol [110]. Aldehyde dehydrogenase (ALDH2) is another mitochondrial enzyme involved in antioxidant activity and participates in the detoxication of lipid peroxidation products [111]. Moreover, ROS through intermediate links, activate AMPK, leading to an increase in antioxidant enzyme gene expression (SOD, CAT and (UCP2)) [84].
In sum, the mitochondria are an important ROS source in cardiomyocytes. The amount of ROS produced by mitochondria depends on the supply of oxygen to the cell and activity of the enzymes that produce ROS (especially SOD1 and MAO-A). Cardiomyocyte cytosol antioxidant GPX and PRX regeneration also depends on Krebs cycle action. AMPK activation is important for antioxidant enzyme function.

4. Differences in ROS-Induced Pathways between HFrEF and HFpEF

Abnormalities in antioxidant values and oxidative states are found in CHF of various etiologies. Oxidative stress is increased in both HFrEF and HFpEF and is related to the pathogenesis of myocardial remodeling [9,21,55,112,113]. Comparison of oxidative stress readings in ischemic cardiomyopathy and non-ischemic cardiomyopathy patients and the correlations between oxidative stress parameters and clinical readings have highlighted the possibility that the defense mechanisms against ROS could differ between these groups [93].

4.1. ROS in HFrEF

HFrEF is considered in patients with an LV ejection fraction lower than 40% [5]. HFrEF is mostly associated with large scale cardiomyocyte death and formation of eccentric hypertrophy as a result of myocardial infarction, cardiomyopathy or valvular heart disease [114]. Irreversible cardiomyopathic changes, subcellular abnormalities and, in turn, decreased heart systolic-diastolic function are suggested to be the causes of elevated ROS levels in HFrEF [115]. Excess of ROS in HFrEF can participate in: (1) subcellular abnormalities and contractile function damage by proteins involved in excitation-contraction and modification coupling [116], (2) myocardial fibroblast proliferation and MMP activation [115], (3) mitochondrial dysfunction due to upsurge of mitochondrial matrix calcium [117], as well as mitochondrial fragmentation, stimulating cardiomyocytes to undergo apoptosis [118] and decrease in oxidative capacity in HFrEF [119]. Lysosomes are known to take part in active mitophagy of clustered mitochondria [117]. Mitophagy is upregulated and intensified with progression to HFrEF [119] due to peroxidation of the lysosomal membranes and lipofuscin accumulation [120].
The molecular mechanisms leading to mitochondrial clustering have not yet been clarified. An mRNA-binding protein, involved in the proper cytoplasmic distribution of mitochondria, named clustered mitochondria protein homolog (Cluh), has been suggested as a participant in mitochondrial biogenesis and oxidative capacity [121]. Cluh has been found to be downregulated in an HFrEF rat model [122], however, it has not yet been investigated in humans.
Proteins in OMM (mitofusin 1 and 2) [123], the inner mitochondrial membrane (IMM) (optic atrophy 1 (OPA1)) [124], and mitochondrial fission proteins (dynamin-related protein1 (DRP1)) [125] and fission1 [122] are stated to be damaged by ROS [122]. OPA1 was suggested to take part in mitochondrial respiratory efficiency [126], mitochondrial fragmentation and apoptosis [127], and its expression was found to be decreased in HFrEF patients [119]. It was discovered that the mitochondrial mitophagy marker BNIP3 takes part in promoting mitochondrial fragmentation by binding to OPA1, leading to OPA1 inhibition [127]. Additionally, BNIP3 inhibition increases DRP1 phosphorylation leading to its cytoplasmic translocation [117]. DRP1 was also found to be increased in HFrEF patients [119]. It is worth paying attention to mitochondrial dynamic proteins (MDPs) that are regulated by some signaling pathways involving proteasome-dependent degradation and transcription and, therefore, modulating mitochondrial function [128]. It was shown that MAPK phosphorylates MFN2, leading to its degradation, and is therefore important in myocardial remodeling induced by mitochondrial related apoptosis [129,130] (Figure 4).
Troponin I, phospholamban [129] and DRP1 [131] have been shown to be target proteins in PKA-related initiation of remodeling and progression to HF.
Calcium uptake in mitochondria is managed by the voltage-dependent anion channel (VDAC1) at the OMM [132], and is regulated by the mitochondrial calcium uniporter (MCU) at the IMM [133]. Calcium efflux is managed by the sodium/calcium/lithium exchanger at the IMM through intermembranous space, and then through VDAC1 into the cytosol [134]. Mitochondrial matrix calcium overload has been suggested as a major cause of mitochondrial dysfunction and decrease in oxidative capacity in patients with CHF [134], but decrease in MCU expression in HFrEF has also been suggested [135]. Therefore, clarification is needed on how activation of signaling pathways affects calcium flux through the OMM and IMM.
Decrease in oxidative capacity was found to be linked to decreasing expression of ETC complexes, or post-translation modifications of mitochondrial proteins (mainly acetylation) [117,119,136]. Cytochrome C oxidase activity was shown to be reduced along with reduction of expression of ETC complexes I and IV [117]. Decrease in ETC complex expression [119] and increase in protein acetylation is present in HFrEF patients [136]. Acetylation of ETC complexes, fatty acid beta-oxidation and tricarbonic acid (TCA) cycle proteins lead to inhibition of its activity [136] (Figure 4). It is speculated that the reason behind mitochondrial protein hyperacetylation is related to reduced protein deacetylation by SIRT3 and SIRT5, and excess of acyl-CoA [137]. Both SIRT3 and SIRT5 were found to be downregulated in a rat HFrEF phenotype, but not in a moderate cardiac remodeling setting [122].
Elevated cytoplasmic Na+, abnormal mitochondrial Ca2+ regulation and impaired energy metabolism are additional causes of diminished mitochondrial function in cardiomyocytes. These, in turn, downgrade mitochondrial energy supply and increase mitochondrial ROS release. Increased cytoplasmic Na+ impels mitochondrial Ca2+ depletion, mediated by mitochondrial Na+/Ca2+ exchanger activity. Therefore, qualitatively different patterns of ROS emission across a similar range of Ca2+ concentrations are produced [138].
A positive correlation between serum levels of reactive oxidative metabolites (DROM) and high-sensitivity C-reactive protein (hs-CRP) was shown [139], suggesting interfaces between oxidative stress and inflammation in HFrEF [140].
Post-ischemic condition, hemorrhage, severe trauma or toxic necrosis are the cause of sterile inflammation in HFrEF patients. As a result, endogenous stimuli trigger sterile inflammation by activating receptors, such as cluster of differentiation 36A, initializing the pathogen recognition receptor (PRR), resulting in tissue injury and intracellular cytokine release [141]. Subsequently, PRR triggers type I interferon (IFN), mitogen-activated kinase (MAPK) and nuclear factor kappa-B (NFκB), as a result, increasing pro-inflammatory chemokine and cytokine levels [142]. Therefore, granulopoiesis is induced by hematopoietic stem cells (HSC) that upregulate the production of neutrophils and monocytes [143]. Released neutrophils are conveyed through the blood into the heart where they phagocytose damaged cells. After infiltrating the heart, monocytes produce growth factors (IL-10, TGFβ) and cytokines to reduce inflammatory triggering and promote endothelial and smooth muscle cells to initiate scar formation [144]. This pathological event sequence causes cardiac fibroblasts to migrate and proliferate in the injury site where they are transformed into myofibroblasts. Stress fibers are produced by myofibroblasts, which cause interference and propagation of electrical signaling, as well as secretion of profibrotic signaling factors TNF-α, TGF-β and angiotensin II (Ang II). Together, these factors can induce and modify cardiomyocyte hypertrophy [145]. Moreover, myofibroblasts also line the extracellular matrix, resulting in interstitial and perivascular fibrosis that stiffens the myocardium, inducing collagenous scar formation [145]. Additionally, TNF-α triggers apoptosis in cardiomyocytes through death receptors [146].
Taken together, mitochondrial damage, including enzymes involved in metabolic pathways (such as fatty acid oxidation and the TCA cycle), OMM and IMM proteins, ETC protein acetylation and Ca2+ channels were found to be more related to ROS-induced cardiomyocyte damage and CHF progression in HFrEF patients. Mitochondrial-dysfunction-causing pathways can act simultaneously or subsequently contributing to HFrEF worsening. The possibilities for mitochondrial function improvement will be discussed in the treatment options section.

4.2. ROS in HFpEF

HF is defined as impaired LV myocardial contractility, diminished right ventricle (RV) function and decreased left atrium (LA) volumetric and contractile function. Energetic imbalances, interstitial fibrosis, cardiomyocyte hypertrophy, and oxidative stress derived from mitochondrial dysfunction and endothelial dysfunction, are always present in the setting of HFpEF [4,147].
Pro-inflammatory status affects multiple organ systems and contributes to generalized microvascular inflammation with diminished cyclic guanosine monophosphate (cGMP) and nitric oxide (NO) bioavailability, and reduced protein kinase G (PKG) activity. In turn, lessened PKG activity contributes greatly to cardiomyocyte hypertrophy and increased resting tension because of impaired connectin (a protein responsible for elasticity of the muscle) phosphorylation [141]. In addition, increased peroxynitrite concentrations, together with scarce NO availability, facilitate fibroblast proliferation through epidermal growth factor, platelet-derived growth factor, phosphatidylinositol 3-kinase and janus kinase pathways. These processes in combination cause stiffened cardiomyocytes and increased collagen deposits, which eventually lead to diastolic dysfunction due to elevated LV pressures in HFpEF [148,149,150]. Matrix metallopeptidase 9 (MMP9), a tissue inhibitor of MMP1, is also increased in HFpEF [151]. In addition, measured cardiomyocyte length and width were significantly larger in an HFpEF patient group, when compared with an HFrEF patient group [152].
Comorbidities play an important role in the HFpEF setting. Diabetes mellitus, obesity, chronic kidney disease, hypertension and anemia generate a systemic inflammatory setting. For example, in obese HFpEF patients, macrophages in adipose tissue promote secretion of proinflammatory cytokines and, in CHF patients with anemia, low hemoglobin concentration fosters oxidative stress, caused by immune response to iron deficiency [153,154,155]. A systemic inflammatory state, induced by comorbidities, causes elevated levels of interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), soluble ST2 (a receptor that inhibits cardioprotective impact of IL-33), and pentraxin 3 (a complement activator and autoimmunity control agent) [151,156,157]. In turn, pro-inflammatory cytokines induce ROS production in the endothelium through NOX [158], causing oxidative and nitrosative stress in the myocardium of HFpEF patients [159,160]. Obesity-induced inflammatory cytokines activate ROS production through NOX activation [161]. Decrease in NO has been shown to be highly important in HFpEF development through ROS [141] (Figure 5).
Due to uncoupled eNOS, superoxide production increases. In turn, low levels of NO react with superoxide to generate peroxynitrite, leading to nitration of tyrosine residues and formation of nitrotyrosine [162]. NO seems also to be responsible for early LV relaxation and reduction of end-diastolic stiffness [163]. In addition, NO acts by stimulating cardiac soluble guanylate cyclase (sGC) receptors, leading to cGMP synthesis [164]. Additionally, brain natriuretic peptide (BNP) stimulates particulate guanylate cyclase (pGC) [165]. cGMP is a secondary messenger, operating cGMP-gated cation channels, through cGMP regulated phosphodiesterases (PDEs) and cGMP-dependent protein kinases (PKG) [166]. In HFpEF, NO bioavailability is low due to inflammation and oxidative stress. cGMP concentration and PKG activity reduction was also observed in an HFpEF model. Moreover, oxidative stress shifts sGC towards an oxidized, heme-free form which is unresponsive to endogenous and exogenous NO [167]. Titin hypophosphorylation associated with stiffened cardiomyocytes was also observed in animal models [168,169]. Therefore, ROS are involved in the NO-sGC-cGMP-PKG pathway associated with titin hypophosphorylation and myocardial diastolic stiffness in HFpEF (Figure 5).
Structural vascular abnormalities are another important component in HFpEF pathogenesis. Stiffened arteries contribute greatly to increased pulse pressure and mean arterial and systolic blood pressures [170]. Mellisa A. Lyle et al. investigated contractile protein expression in HFpEF, HFrEF and control group patients and found that the HFpEF group had decreased myosin phosphatase target subunit 1-protein (responsible for NO mediated vasodilation) concentration. The authors, therefore, speculated that this was a possible reason why NO, cGMP or PKG signaling-pathway-targeted pharmacotherapies result in poor clinical benefits [171].
Inducible nitric oxide synthase (iNOS) upregulation is known to be an important factor in the development of HFpEF [172]. iNOS-related nitrosative stress increases s-nitrosylation of inositol-requiring protein 1α (IRE1 α) and decreases transcription factors involved in cellular stress response-spliced X-box-binding protein1 (XBP1s) levels [173]. It should also be mentioned that overexpression of XBP1s in cardiomyocytes weakened the HFpEF cardiac phenotype. It seems that iNOS-mediated nitrosylation of IRE1 α interferes with the XBP1 connection, which is required for the stress response [173]. Therefore, iNOS inhibition can be considered as a therapeutic strategy in the HFpEF setting.
Mitochondria play an important role in CHF, but are less understood in HFpEF pathophysiology [174]. Oxidative metabolism in mitochondria shifts towards a compensatory response through increased glycolysis; however, hypertensive HFpEF models do not seem to have correlation between increased glycolysis and, as a result, increased pyruvate oxidation rates, although it results in significant increase in proton production [175,176]. The same process was shown in an HFpEF-induced rat model, which displayed reduced pyruvate dehydrogenase activity, diminished glucose oxidation and increased pyruvate dehydrogenase kinase (PDK4) expression [177]. In obese models, mitochondrial ETC is affected through peak oxidative phosphorylation and NADH-associated respiration. NADH-associated dysfunction can be explained by diminished NADH-linked mitochondrial respiration due to calcium overload [178,179]. Declining energy reserves in the myocardium eventually lead to systolic dysfunction, even if it is “hidden” under a preserved ejection fraction [180].
Phosphodiesterase (PDE) 5 and 9 were shown to be upregulated in hypertrophy and CHF. In addition, PDE9-mediated natriuretic peptide (NPs) regulation in cardiomyocytes was more efficient than with NO-stimulated cGMP regulation [181]. PDE expression in the myocardium of HFpEF was increased and both PDE9 and PDE5 were involved in regulating cGMP-PKG activity [181]. Therefore, PDE9 and PDE5 could be potential treatment targets in patients with HFpEF.
Taken together, the NO-sGC-cGMP-PKG pathway appears to be the most investigated and important one in HFpEF pathogenesis. This pathway involves overproduction of hydrogen peroxide then initiates titin phosphorylation, leading to cardiomyocyte hypertrophy. Additionally, eNOS inhibition results in fibroblast proliferation. PDE is also involved in the cGMP induced pathway in HFpEF pathogenesis. The NO-sGC pathway has been investigated as a potential treatment target and will be discussed in the treatment strategy section of this article.

5. ROS-Induced Pathways as a Treatment Target in HFrEF and HFpEF

There are two interactive elements, related to other factors, that are implicit in the pathogenesis of CHF: inflammation and oxidative stress [182]. Some groups of compounds were investigated for their effect in oxidative stress-induced myocardial damage/remodeling reduction in CHF development including: (1) adenosine monophosphate-activated protein kinase (AMPK) activators, (2) renin-angiotensin system inhibitors (RAAS inhibitors), (3) inhibitors of ROS-producing enzymes, (4) antioxidants, (5) MAO inhibitors, (6) medications that improve mitochondrial function, and (7) substances that increase cGMP-PKG signaling. Each of these groups will be discussed below.

5.1. AMPK Activators

AMPK is an enzyme that has a pleiotropic cardioprotective impact and is important in the progression of CHF [84]. There are two AMPK isoforms: α1 and α2, and AMPK α2 seems to dominate in cardiomyocytes [183]. In normal conditions low amounts of ROS activate AMPK through different pathways [184], leading to antioxidant enzyme SOD, CAT and UCP2 gene expression (Figure 4), activation of pathways that produce adenosine triphosphate (ATP), suppression of apoptosis, inhibition of NOX expression and, by these pathways, protection from cardiac hypertrophy [146]. According to data presented above in the section on ROS in HFrEF, it appears that AMPK pathway action could be more feasible for HFrEF. Some drugs, such as metformin, statins, trimetazidine and resveratrol, were reported to have effects on AMPK activation and may prove to be beneficial in the clinical setting for ROS reduction and HF progression dampening [146]. For example, atorvastatin is suggested to activate the eNOS signaling pathway via AMPK, which, in turn controls NO bioavailability, maintains cardiovascular homeostasis and activates AMPK by altering the AMP/ATP ratio or increasing ROS-dependent PKC activity [146]. This process has been shown to attenuate heart dysfunction, fibrosis, and hypertrophy in a post-MI rat model [185]. Trimetazidine activates AMPK by influencing ATP levels in cardiomyocytes, thus improving heart function, New York Heart Association (NYHA) functional class, exercise tolerance and patient’s quality of life [186]. Resveratrol is stated to be able to inhibit cardiomyocyte hypertrophy through the AMPK-dependent pathway via two mechanisms: (1) at a high concentration (50–100 µM), it can activate AMPK by increasing the AMP/ATP ratio [187]; (2) the SIRT1-LKB1 (one of AMPK upstream activators) pathway [188]. Resveratrol in animal models decreases oxidative stress [189], but clinically it has not yet been thoroughly investigated [187,188]. AMPK not only improves energy supply to increase heart function, but also improves heart function by mediating various intracellular physiological functions, delaying myocardial fibrosis, and reducing heart damage in animal models. The benefits of these compounds in clinical studies with CHF patients appear to be worth exploring.

5.2. Renin-Angiotensin System Inhibitors

Angiotensin II (ANG II) is stated to promote excess accumulation of collagen [190] and is involved in ROS production in cardiomyocytes, leading to myocardial remodeling and chronic HF (Figure 4). Cardiomyocyte hypertrophy (induced by ANG II), therefore, could be inhibited by AT1R inhibition. Several potential drugs (e.g., valsartan, candesartan and kaempherol), affecting chronic HF pathogenesis through the ANG II pathway, have been investigated.
Treatment with valsartan reduced mRNA expression levels of NOX2 and NOX4, as well as the myocardial protein expression levels of NOX2 and NOX4, in rats with doxorubicin-induced myocardial injury [64]. The authors noted that ANG II increased the protein expression levels of NOX2 and NOX3 and the production of ROS, and that protein expression levels of ERK, JNK and P38, which lie downstream of MAPK, were increased as well. It was emphasized that pre-treatment with valsartan reduced the expression of AT1R, NOX2, NIX4 and ROS, therefore, activity of the MAPK signaling pathway was decreased [64]. Clinical studies have confirmed the valsartan effect on oxidative stress reduction through NOX2 in the human myocardium [191] (Table 1).
Neprilizine is an endopeptidase that cleaves the natriuretic peptides (NPs), bradykinin and adrenomedullin [195]. Inhibition of neprilysin is a main act due to an enhanced effect on biologically active NP. This inhibition increases the plasma concentrations of other vasoactive peptides, including vasodilators, such as adrenomedullin (a peptide associated with the calcitonin gene), bradykinin, and vasoconstrictor peptides, including endothelin-1 and angiotensin I and II [195]. Therefore, the AT1R inhibitor valsartan was added to neprilizine (sacubitril). It was discovered that sacubitril/valsartan (LCZ696—combination 1:1 of valsartan and sacubitril) reduced the risk of hospitalization for cardiac failure or death from cardiovascular disease in patients with HFrEF [196]. LCZ696 was also found to reduce plasma N-terminal pro b-type natriuretic peptide (NT-proBNP) concentration in HFpEF and to reduce the risk of death and hospitalization in HFrEF with EF ≤ 40% [192]; however, outcomes for HFpEF are not yet established. Therefore, LCZ696 provides a greater protection of target organs than AT1R therapy alone, including cardiovascular protection. This drug is superior in targeting the renin-angiotensin-aldosterone system (RAAS) in patients with HFrEF who can tolerate AT1R inhibitors, with a better safety and efficacy profile [190,192,197]. LCZ696 was well tolerated in a Phase II large HFrEF population [192], produced lower levels of NTproBNP (NCT01920711) and improved NYHA functional class [198,199].
Researchers recently investigated the usefulness of the ATR1 locator candesartan in HFpEF treatment and discovered that candesartan improved outcomes to a similar degree as for HFrEF patients [194].
Current guidelines strongly recommend neurohormonal antagonist treatment for HFrEF [200]. Despite HFpEF patients representing a majority of those with chronic HF in the general population, there are no recommendations for HFpEF treatment with sacubitril/valsartan [201].
Kaempherol (KFP, 3,4’,5,7-tetrahydroxyflavone) is a flavonoid, found abundantly in plant foods [202], and prevents oxidative stress [203]. KFP was suggested to inhibit cardiac remodeling through deactivation of mitogen-activated protein kinases (MAPKs) [204]. The activation of MAPK is known to promote fibrosis (Figure 3). Additionally, treatment aimed at cardiac fibroblasts with KFP resulted in decreased expression of pro-inflammatory cytokines [204], supplementing the cardioprotective effect. Further clinical studies are needed to determine the suitability of this medication in patients.

5.3. Inhibitors of ROS-Producing Enzymes

The mitochondrial and cytosol enzymes NOX, NOS and XOR were discussed as primarily ROS-produced enzymes in cardiomyocytes (Section 3.1 and Section 3.2). ETC proteins were suggested as ROS sources as well. One of the compounds related to mitochondrial ETC is Mito-Q—a combination of the triphenylphosphonium cation (TPP) and Q10. This remedy demonstrated promising antioxidative effects in several human studies [205,206,207,208]. Mito-Q is Q10 coupled to lipophilic TPP+ and it accumulates on the IMM [205]. Q10, when reduced to ubiquinol by the ETC, acts as an antioxidant, preventing mitochondrial oxidative damage [206] and resulting in heart hypertrophy reduction in an HF rat model [208]. MitoQ is stored in mitochondria in vivo and is a part of the redox system, together with the reduced hydroquinone MitoQuinol form. The pivotal aim of MitoQ is to protect and prevent cellular damage, triggered by mitochondrial ROS overproduction and oxidative stress [209,210]. It is important to mention that MitoQ is bound to the mitochondrial IMM, mostly in the hydrophobic membrane core, which is determined by the membrane potential, while the respiratory chain complex II is continuously processed into ubiquinol [211]. The active part of MitoQ is ubiquinone (coenzyme Q10) [212]. MitoQ was shown to protect against oxidative damage in animal models with HF by reducing hydrogen peroxide formation [207]. Further clinical studies are needed to confirm the analogous effect in larger humans.
NOX-, XOR- and NOS-induced pathway inhibition may be one of the ways to reduce ROS damage in the heart. In small clinical studies, it has been shown that myocardial O2 consumption is lowered and mechanical efficiency of the LV is improved by XOR inhibition [56] due to increased ATP flux through creatine kinase (CK) [213]. XOR inhibitors are known to improve LVEF [214], endothelial function [215], and to decrease BNP levels [216] in HF patients. However, larger studies did not show the same amount of benefit in an HFrEF patient group [217,218,219].
Inhibition of eNOS and combined treatment with BH4 also reduced ROS production in animal models with HF [63]. Overexpression of the enzyme catalyzing BH4 biosynthesis (GTP-cyclohydrolase 1) or oral consumption of BH4 shielded from harmful Ca2+ pathways and contractile dysfunction in isolated cardiomyocytes in vivo mice through nNOS action [220]. However, few clinical studies have investigated the benefits of BH4 usage and the results of these were rather poor.
Taken together, XOR inhibitors have been shown not to be effective in chronic HF patient treatment. Management of NOS activity is still at the research stages. We have not found any studies that investigated the benefits of NOX inhibition or Mito-Q effect in CHF patients.

5.4. Antioxidants

Administration of antioxidants (AOx) has been expected to be a simple and effective way to reduce oxidative harm in the myocardium. Vitamin-antioxidants, Mito-TENPO, enzyme-scavengers of ROS and elamipretide were investigated (Table 2).
Some small studies evaluated vitamin-antioxidants combinations in HFpEF [216] and HFrEF [221] and discovered improvement in peripheral vascular function and decrease in oxidative stress (Table 2). In the first study, reactive hyperemia (RH), a measure of microvascular function, did not change after OC (combination of α-lipoic acid, vitamin C and vitamin E) administration. Improvement in flow-mediated dilation was accompanied by significant increase in plasma nitrite and decrease in CRP, but additional biomarkers of oxidative stress, plasma concentrations of free radicals and antioxidant capacity were not altered by AOx. These findings confirm the efficacy of an over-the-counter OC combination in achieving systemic anti-inflammatory effects and improving peripheral vascular function, regardless of changes in global markers of oxidative stress in HFpEF, providing new insight into the potential therapeutic effect of AOx [216]. The second study of HFrEF patients was characterized by macrovascular endothelial dysfunction, which may be due, at least in part, to a change in redox balance, leading to increased oxidative stress and decreased endogenous AOx protection. The results of this small study showed that chronic AOx administration is a simple way to improve macrovascular function, reduce oxidative stress, and increase AOx capacity in patients with HFrEF [221] (Table 2). However, larger sample studies are needed to properly investigate the beneficial effects of this compound.
Mito-TENPO is a mitochondria-targeted chemical with superoxide-scavenging properties [208]. MitoTENPO was given to prevent and reverse HF [222] and improved LV contraction [223] in a mouse model. Despite these promising results, we did not find any studies performed with CHF patients.
One more area of AOx application in chronic HF therapy is enhancing ROS scavenging capacity through GSH [224], SOD [225] and catalase [226]. Different chemicals were investigated to increase SIRT3 activity as well [204]. Studies in these fields demonstrated beneficial results, but further investigations are still needed.
Despite increasing interest in oxidative stress management possibilities in CHF patients, most antioxidant therapies are not successful [6,227].

5.5. MAO Inhibitors

MAO are enzymes located on the OMM. They catalyze deamination of biogenic amines and neurotransmitters [102]. The mechanisms of MAO toxicity have been commonly associated with excessive H2O2 production, due to MAO appearing to be one of the major ROS sources within the mitochondria [100]. Studies in animal models suggested that activation of MAO-A/B plays a crucial role in progression from cardiac hypertrophy to cardiac failure, establishing a clear association between MAO-induced ROS production and mitochondrial dysfunction. However, MAO is suggested as a promising new therapeutic target in chronic diseases [228,229,230,231,232,233,234,235]. Despite the clinical relevance of these findings, and the possible indications for MAO inhibitors in the treatment of chronic HF, little is known about the activity of MAO in HF patients and its association with redox imbalance [236].

5.6. Mitochondrial Function Improvement

Mitochondrial function depends mainly on ETC and membrane integrity. Coenzyme Q (CoQ10) is one of the ETC components [237]. Currently, only one clinical study is being conducted to address this aspect of the pathophysiology of HFpEF. A study on CoQ10 in diastolic heart failure patients (NCT03133793) is investigating the efficacy of ubiquinol, a reduced form of CoQ10, which acts to reduce the severity of HFpEF symptoms and improve cardiac function [238].
CoQ10 is an essential cofactor of the ETC from complexes I and II to complex III. It maintains mitochondrial membrane potential, supports ATP synthesis and inhibits ROS generation [239,240]. Plasma levels of CoQ10 are decreased in patients with chronic HF, and correspond to the severity of a disease. Doses of CoQ10 have been reported to increase the incidence of adverse reactions at doses above 1200 mg/day, with doses of 22 to 400 mg/day being considered safe [241]. An analysis of small studies suggested that CoQ10 can improve LVEF in HFrEF [242] and can reduce cardiovascular mortality by 50% [243]. However, it was not sufficiently powerful due to poor prognostic effect [237] for it to be recommended in guidelines [5]. CoQ10 is also needed for eNOS management [235]. CoQ10 benefits in HFrEF patients are well discussed by A. Sharma and co-authors [237]. It remains unclear if prescribing of CoQ10 is useful due to mitochondrial function improvement, or due to its involvement in eNOS action. The utility of CoQ10 for HFpEF patients requires further investigation.
Elamipretide is a compound that accumulates in the IMM by binding to cardiolipin [244], a phospholipid, required for proper ETC function and other IMM proteins [244,245]. Cardiolipin can be oxidized by elevated ROS [246] and disturbs interaction of ETC complexes, leading to O2− increase and apoptosis initiation [247]. Elamipretide improved mitochondrial function in a dog model [248] and isolated cardiomyocytes [249,250]. Despite favorable effects in animal models and isolated cardiomyocytes, the clinical effects in humans with HF are rather modest (Table 3).

5.7. Chemicals Increasing cGMP-PKG Signaling

Constantijn Franssen, with co-authors, reviewed studies conducted before the year 2014 regarding medications that increased cGMP-PKG signaling [253]. The medicines reviewed were nitroxyl (HNO), enalapril, LCZ696 and sGC activators (e.g., cinaciguat, riociguat, vericiguat). HNO was shown to increase cGMP and to suppress NOX, resulting in an anti-hypertrophic effect in rat cardiomyocytes [254] and HFrEF patients [255], but clinical studies of the HNO effect in HFpEF were absent. LCZ696 was discussed in the chapter, “Renin-angiotensin system inhibitors”. Cinaciguat did not show any effect on cardiac index [256]. Riociguat improved symptoms and NT-proBNP levels [257]. Vericiguat was evaluated in a Phase II trial study in HFrEF and HFpEF [258].
Knowledge about medications that increase cGMP-PKG signaling has grown in the last decade. Sodium-glucose cotransporter 2 inhibitor (SLGT2), soluble guanylate cyclase (sGC) activators, PDE inhibitors, NO donors, and the vasodilator hydralazine were investigated.
Empagliflozin (sodium-glucose cotransporter 2 inhibitor) and sGC activator were suggested to have antioxidant and anti-inflammatory features in the myocardium of HF rats and HFpEF patients [259]. The results revealed that empagliflozin reduced cardiovascular mortality, all-cause mortality, and the number of hospitalizations for HFrEF. Moreover, both empagliflozin and sGC activator improved cardiomyocyte function by enhancing the phosphorylation of titin and other myofilament proteins, presumably due to improved signaling pathways, such as the nitric oxide (NO)/soluble guanylyl cyclase (sGC)/cGMP-dependent protein kinase (PKG) signaling pathway (NO-sGC-cGMP-PKG pathway) and the CaMKII-mediated hypertrophic pathway, PKC, ERK2, in addition to the PKA pathway [259] (Figure 5).
The PDE5 inhibitor sildenafil can inhibit guanosine 3′,5′-cyclic monophosphate (cGMP) breakdown, improve cardiac relaxation and LV remodeling [260]. The catalytic site of PDE5 generally degrades cGMP, and sildenafil potentiates the endogenous increase in cGMP by inhibiting its degradation [261]. Sildenfil reduced pulmonary vascular resistance and right heart pressure in patients with HFrEF, who had secondary pulmonary hypertension, and long-term treatment improved exercise tolerance, functional capacity, LV diastolic function and cardiac geometry [262,263,264]. In a large, long-term (24 weeks) trial of sildenafil (RELAX), PDE5 inhibitor did not improve LV diastolic function and did not reduce hypertrophy and pulmonary pressures. In this study, sildenafil did not increase plasma cGMP concentrations, therefore, exercise capacity and clinical status did not improve [265].
NO donors were also investigated for their capacity to improve heart function. Some of these donors, such as Angeli’s salt [266] and Piloty’s acid [267], appeared to be unstable. In turn, pure NO donors, such as the congener of Piloty’s acid, CXL-1020 [255], and the pro-drug of CXL-1020, cimlanod (BMS-986231), were generated [268]. The HNO donor BMS-986231 in animal models improved myocardial contractility and relaxation without increasing heart rate or oxygen consumption [269]. One study was performed with HFrEF, in which patients received intravenous infusions (i.v.) of BMS-986231 at various doses, and information about the safety and tolerability of medicine was provided [270]. However, the poor solubility of BMS-986231 limited its clinical use as an i.v. agent, and its oral bioavailability is still being investigated [268].
Major studies were performed with the protonated form of NO—nitroxyl (HNO). The action of HNO is preserved during oxidative stress because HNO does not react with superoxides [271], and undergoes moderate oxidative reactions through the formation of hydroxyl radicals [272]. HNO inhibits mitochondrial respiration by inhibiting complexes I and II, most likely by modifying specific cysteine residues in ETC proteins [273]. HNO increased cGMP levels and had NADPH oxidase (NOX2) inhibitory and antihypertrophic effects in rat cardiomyocytes [255]. HNO improved myocardial function due to direct positive lusitropic and inotropic effects, independent of cyclic adenosine monophosphate (cAMP), and due to combined venous and arterial dilation [274,275,276,277,278]. In addition, HNO modifies sarcomeric proteins to increase their Ca2+ sensitivity resulting in systolic force generation [270]. HNO also causes vasodilatation through endothelial soluble guanylate cyclase [266,279]. A recent study showed that HNO reduced left and right ventricle filling pressure and systemic vascular resistance in both animal and CHF patient models [255]. It was concluded that nitroxyl was well tolerated, reduced diastolic filling pressure and systemic vascular resistance, and raised cardiac output and stroke volume with unaltered heart rate [255]. Taken together, the novel cardio-protective properties of HNO show the therapeutic potential of HNO donors, particularly in situations where NO signaling is impaired (in HFpEF), but more detailed studies are required.
The vasodilator hydralazine has a beneficial effect on the balance between NO and O2, which is disturbed in patients with HF [280]. Clinical treatment with nitrates resulted in eventual tolerance to its vascular and hemodynamic effects, mainly due to endothelial dysfunction [281]. However, in combination with hydralazine, nitrate tolerance was avoided due to hydralazine inhibition on nitroglycerin-induced vascular O2, and peroxynitrite (ONOO) formation in vitro [282] and in vivo [283]. Thus, the antioxidant effect of hydralazine and the prevention of the development of its tolerance in response to isosorbide dinitrate (ISDN) [284] may at least partially explain why this combination improves morbidity and mortality in patients with chronic congestive HF [285]. The African-American Heart Failure (A-HeFT) trial demonstrated that ISDN and hydralazine combination has a large effect on survival in patients with HFrEF [286].
Recently, new classes of drugs that increase cGMP production by targeting guanylate cyclase at the NO receptor (sGC) have been discovered. These were designed in order to generate cGMO independently of NO and to target signaling cascades in the cardiovascular system [287]. Enzymes with a unique mode of action activate the oxidized, heme-free form of sGC, which does not react with NO. The oxidation or absence of the heme moiety increases the effect of cinaciguat on the sGC, causing a significant cGMP increase [288]. These compounds are called sGC stimulators and sGC activators. They differ in that sGC stimulators are targeted to bind to the regulatory domain and trigger cGMP production by binding the heme-containing non-oxidized form of the sGC regulatory domain [289].
In recent years, the soluble stimulant sGC vericiguate has attracted the attention of the medical community following reports of reduced clinical outcomes in patients with chronic heart failure. The NO-sGC-cGMP pathway is mediated by a different mechanism that complements current drug therapy for cardiovascular disease. cGMP deficiency is a characteristic trait of both HFrEF and HFpEF [258]. Vericiguate acts synergistically with endogenous NO [285,290], which is considered a nitroconstrictor that produces cGMP at low levels of NO (Figure 5). By increasing cGMP, vericiguate has also been shown to promote vascular relaxation and improve vascular tone regulation and myocardial dysfunction [285,291,292,293]. This would also attenuate left ventricular remodeling by inducing PKG-induced phosphorylation of titin after activation of PKG by cGMP [292] (Figure 5). sGC activators are well discussed by Chien Y.T. et all [294]. Vericiguat is currently in phase 3 clinical trials for HFrEF (BAY 1021189) and praliciguat is now in phase 2, in HFpEF (IW-1973, IWP-121) [294]. Drugs that alter cardiomyocyte homeostasis by increasing cGMP-PKG signaling in HF patients are summarized in Table 4.
To conclude, AMPK activators are suggested to improve heart function in animal models, and, with further research, have potential to be beneficial for patients with chronic HF. The AT1R and neprilysin inhibitor valsartan/sacubitril has been investigated in most detail of all Ang II inhibitors and was included in guidelines for HFrEF treatment. Kaempherol displayed cardioprotective effects in cell culture [203,204]; however, further clinical studies are needed in order to assess its suitability and beneficial effects for patients. The utility of antioxidants, mitochondrial-function-affecting drugs and MAO inhibitors have been poorly studied to date. Both the SLGT2 inhibitor, sGC activator empagliflozin and vericiguat produced gratifying treatment results in patients with HFpEF.

6. Conclusions

Mitochondrial damage, inflammation and enzyme-oxidants (NOX, XOR, NOS), as well as decreased activity of enzyme-antioxidants (GPX and PRX), can be assumed to be the main triggers for excess amounts of ROS in cardiomyocytes. AT1R is involved in NOX activation in both cardiomyocytes and fibroblasts. Enzyme-oxidants act through MAPK and NO synthesis inhibition pathways, leading to cardiomyocyte hypertrophy and interstitial fibrosis. Pro-inflammatory cytokines trigger ROS overproduction, leading to mitochondrial structural damage due to membrane protein, ion channel protein and protein acetylation in HFrEF mitochondria. Therefore, damaged mitochondria are suggested to be the main ROS source in HFrEF, while NO decrease, NO-sGC-cGMP signaling inhibition by ROS and enzymes iNOS, eNOS, PDE are understood to be the most important factors in HFpEF development.
The pathways involving both AMPK and MAPK protein kinases, AT1R and cGMP-PKG, are considered as treatment targets for halting chronic HF development. Therefore, some compound groups for oxidative stress-induced myocardial damage/remodeling reduction in HF development include: (1) activators of AMPK, (2) RAAS inhibitors, (3) inhibitors of ROS producing enzymes, (4) antioxidants, (5) MAO inhibitors, (6) medications that improve mitochondrial function, and (7) substances that increase cGMP-PKG signaling.
Achievements in reducing ROS-induced harmful pathways in chronic HF can be summarized as follows: AMPK activators are suggested to improve heart function in animal models, therefore, exploratory studies with patients afflicted with chronic HF could prove to be of great value. AT1R and the neprilysin inhibitor valsartan/sacubitril have been investigated in the most detail of all. Ang II inhibitors have yielded favorable results and been included in HFrEF treatment guidelines. Kaempherol displayed cardioprotective effects in animal models, but clinical studies are still needed to verify the suitability and treatment benefits in humans. XOR inhibitors and management of the NOS activity in chronic HF patient treatment are still in research stages. Further studies that investigate the benefits of NOX inhibition or Mito-Q effect in CHF patients are needed. The usefulness of antioxidants, mitochondrial-function-affecting drugs and MAO inhibitors are still poorly studied and understood. Both the SLGT2 inhibitor and the sGC activator empagliflozin and vericiguat displayed gratifying results in HFpEF treatment; however, their effects still require to be confirmed in randomized studies.

7. Perspectives

ROS were shown to be involved in both HFrEF and HFpEF pathogenesis through different pathways. Although treatment with unselective antioxidative treatment failed to demonstrate better outcomes in HFrEF and HFpEF patients, oxidative stress remains the focus of intensive research. It appears that selective antioxidant treatment ought to give more favorable results. Treatment regarding mitochondrial function improvement and cGMP-PKG signaling appear to need deeper investigation for both CHF patient groups. In HFrEF, patient usage of AMPK activators should be evaluated.

Author Contributions

Conceptualization, A.M.; writing—original draft preparation, A.M., L.S., L.V., G.M., J.G.; writing—review and editing, L.S., L.V., G.M., J.G.; visualization, L.S., J.G. All authors contributed equally. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbrevations

ALDH2aldehyde dehydrogenase
AMPadenosine monophosphate
AMPKadenosine monophosphate activated protein kinase
ANGIIangiotensin II
AOantioxidant
AOxvitamin C, vitamin E, α-lipoic acid (antioxidants)
AT1Rangiotensin II receptor
ATPadenosine triphosphate
BH4dihydrobiopterin-4
BNIP3mitochondrial mitophagy marker
BNPbrain natriuretic peptide
Baxbcl-2-like protein 4
CATchloramphenicol acetyltransferase
CHF–chronic heart failure
CIETC complex I
CIIIETC complex III
CKcreatine kinase
CRP–C-reactive protein
Cluhclustered mitochondria protein homolog
CoenzymeQ-10–ubiquinone
DM–diabetes mellitus
DROMreactive oxidative metabolites
DRP1dynamin related protein1
ERKextracellular signal-regulated kinase
ERK1/2extracellular signal-regulated kinase 1/2
ETCelectron transport chain
GTPguanosine triphosphate
GMPguanosine monophosphate
GPXglutathione peroxidase
GSHglutathione
H2O2hydrogen peroxide
HFheart failure
HFpEFheart failure with preserved ejection fraction
HFrEFheart failure with reduced ejection fraction
HNOnytroxyl
HSChematopoietic stem cells
IDHisocitrate dehydrogenase
IDH2isocitrate dehydrogenase
IL-10interleukin-10
IL-6interleukin-6
IMMinner mitochondrial membrane
IRE1αinositol-requiring protein 1α
ISDNisosorbide dinitrate
JNK-cJun N-terminal kinase
KFPKaempherol
LAleft atrium
LKB1liver kinase B1
LPHslipid hydroperoxide
LVleft ventricle
LVEDVleft ventricle end-diastolic volume
LVESVleft ventricle end-systolic volume
LVHleft ventricular hypertrophy
LVIDleft ventricular internal dimension
MAOmonoamine oxidase
MAO-Amonoamine oxidase A
MAPKmitogen-activated protein kinase
MCUmitochondrial calcium uniporter
MMPsmatrix metalloproteinase
NAPDH-nicotinamide adenine dinucleotide phosphate
NFκBnuclear factor kappa-B
NHE-1Na+/Ca2+ exchanger
NOnitric oxide
NO-2plasma nitrite
NOSnitric oxide synthase
NOXnicotinamide adenine dinucleotide phosphate oxidase
NPnatriuretic peptide
NT-proBNPN-terminal pro b-type natriuretic peptide
NYHA-New York Heart Association
OMMouter mitochondrial membrane
ONOOperoxynitrite
OPA1optic atrophy 1
P38a focal point of interactions of the serine/threonine kinases
PDEphosphodiesterases
PKAprotein kinase A
PKBprotein kinase B
PKCprotein kinase C
PKGprotein kinase G
PKSprotein kinase S
PRRpathogen recognition receptor
PRXperoxiredoxin
RAASrenin-angiotensin-aldosterone system
ROSreactive oxygen species
Rac-1GTP-binding protein
SGLT2sodium-glucose cotransporter
SIRTNAD+ dependent class III histone deacetylases
SODsuperoxide dismutase
TGFβtumor growth factor β
TNF-αtumor necrosis factor-α
TPPtriphenylphosphonium cation
TXNIPthioredoxin-interacting protein
UCP2uncoupling protein 2
VDAC1voltage-dependent anion channel 1
XBP1sX-box-binding protein1
XOxanthine oxidase
XORxanthine oxidoreductase
cAMPcyclic adenosine monophosphate
cGMPcyclic guanosine monophosphate
eNOSendothelial nitric oxide synthase
hs-CRPhigh-sensitivity C-reactive protein
iNOSinducible nitric oxide synthase
nNOSneuronal nitric oxide synthase
pGCparticulate guanylyl cyclase
sGCsoluble guanylate cyclase
β-OXbeta-oxidation

References

  1. Holland, D.; Kumbhani, D.J.; Ahmed, S.H.; Marwick, T.H. Effects of Treatment on Exercise Tolerance, Cardiac Function, and Mortality in Heart Failure with Preserved Ejection Fraction: A Meta-Analysis. J. Am. Coll. Cardiol. 2011, 57, 1676–1686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Henning, R.J. Diagnosis and treatment of heart failure with preserved left ventricular ejection fraction. World J. Cardiol. 2020, 12, 7–25. [Google Scholar] [CrossRef]
  3. Shah, A.M.; Mann, D. In search of new therapeutic targets and strategies for heart failure: Recent advances in basic science. Lancet 2011, 378, 704–712. [Google Scholar] [CrossRef] [Green Version]
  4. Shah, S.; Kitzman, D.W.; Borlaug, B.; Van Heerebeek, L.; Zile, M.; Kass, D.A.; Paulus, W.J. Phenotype-Specific Treatment of Heart Failure with Preserved Ejection Fraction. Circulation 2016, 134, 73–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Ponikowski, P.; Voors, A.A.; Anker, S.D.; Bueno, H.; Cleland, J.G.F.; Coats, A.J.S. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of. Eur. Heart J. 2016, 37, 2129–2200. [Google Scholar] [CrossRef]
  6. Bertero, E.; Maack, C. Calcium Signaling and Reactive Oxygen Species in Mitochondria. Circ. Res. 2018, 122, 1460–1478. [Google Scholar] [CrossRef]
  7. Sorescu, D.; Griendling, K.; Sorescu, D.; Griendling, K. Reactive Oxygen Species, Mitochondria, and NAD(P)H Oxidases in the Development and Progression of Heart Failure. Congest. Heart Fail. 2002, 8, 132–140. [Google Scholar] [CrossRef]
  8. Collin, F. Chemical Basis of Reactive Oxygen Species Reactivity and Involvement in Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 2407. [Google Scholar] [CrossRef] [Green Version]
  9. Panth, N.; Paudel, K.R.; Parajuli, K. Reactive Oxygen Species: A Key Hallmark of Cardiovascular Disease. Adv. Med. 2016, 2016, 9152732. [Google Scholar] [CrossRef] [Green Version]
  10. Wardman, P.; Candeias, L.P. Fenton chemistry: An introduction. Radiat. Res. 1996, 145, 523–531. [Google Scholar] [CrossRef]
  11. Münzel, T.; Gori, T.; Keaney, J.; Maack, C.; Daiber, A. Pathophysiological role of oxidative stress in systolic and diastolic heart failure and its therapeutic implications. Eur. Heart J. 2015, 36, 2555–2564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ferdinandy, P.; Danial, H.; Ambrus, I.; Rothery, R.; Schulz, R. Peroxynitrite Is a Major Contributor to Cytokine-Induced Myocardial Contractile Failure. Circ. Res. 2000, 87, 241–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hansen, J.M.; Go, Y.-M.; Jones, D.P. Nuclear and mitochondrial compartmentation of oxidative stress and redox signaling. Annu. Rev. Pharmacol. Toxicol. 2006, 46, 215–234. [Google Scholar] [CrossRef] [PubMed]
  14. Glasauer, A.; Chandel, N.S. Targeting antioxidants for cancer therapy. Biochem. Pharmacol. 2014, 92, 90–101. [Google Scholar] [CrossRef] [PubMed]
  15. Santos, C.X.; Raza, S.; Shah, A.M. Redox signaling in the cardiomyocyte: From physiology to failure. Int. J. Biochem. Cell Biol. 2016, 74, 145–151. [Google Scholar] [CrossRef] [Green Version]
  16. Zurlo, G.; Guo, J.; Takada, M.; Wei, W.; Zhang, Q. New Insights into Protein Hydroxylation and Its Important Role in Human Diseases. Biochim. Biophys. Acta 2016, 1866, 208–220. [Google Scholar] [CrossRef] [Green Version]
  17. Stomberski, C.; Hess, D.T.; Stamler, J.S. Protein S-Nitrosylation: Determinants of Specificity and Enzymatic Regulation of S-Nitrosothiol-Based Signaling. Antioxid. Redox Signal. 2019, 30, 1331–1351. [Google Scholar] [CrossRef]
  18. Turrens, J.F. Mitochondrial formation of reactive oxygen species. J. Physiol. 2003, 552, 335–344. [Google Scholar] [CrossRef]
  19. Burgoyne, J.R.; Mongue-Din, H.; Eaton, P.; Shah, A.M. Redox signaling in cardiac physiology and pathology. Circ. Res. 2012, 111, 1091–1106. [Google Scholar] [CrossRef]
  20. Deponte, M. Glutathione catalysis and the reaction mechanisms of glutathione-dependent enzymes. Biochim. Biophys. Acta Gen. Subj. 2013, 1830, 3217–3266. [Google Scholar] [CrossRef] [Green Version]
  21. Dhalla, N.S.; Temsah, R.M.; Netticadan, T. Role of oxidative stress in cardiovascular diseases. J. Hypertens. 2000, 18, 655–673. [Google Scholar] [CrossRef] [PubMed]
  22. Kotiadis, V.; Duchen, M.R.; Osellame, L.D. Mitochondrial quality control and communications with the nucleus are important in maintaining mitochondrial function and cell health. Biochim. Biophys. Acta Gen. Subj. 2013, 1840, 1254–1265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Takimoto, E.; Kass, D.A. Role of Oxidative Stress in Cardiac Hypertrophy and Remodeling. Hypertension 2007, 49, 241–248. [Google Scholar] [CrossRef] [PubMed]
  24. Sawyer, D.B.; Siwik, D.A.; Xiao, L.; Pimentel, D.R.; Singh, K.; Colucci, W.S. Role of Oxidative Stress in Myocardial Hypertrophy and Failure. J. Mol. Cell. Cardiol. 2002, 34, 379–388. [Google Scholar] [CrossRef]
  25. Christiansen, L.B.; Dela, F.; Koch, J.; Hansen, C.N.; Leifsson, P.S.; Yokota, T. Impaired cardiac mitochondrial oxidative phosphorylation and enhanced mitochondrial oxidative stress in feline hypertrophic cardiomyopathy. Am. J. Physiol. Circ. Physiol. 2015, 308, H1237–H1247. [Google Scholar] [CrossRef] [Green Version]
  26. Weissman, D.; Maack, C. Redox signaling in heart failure and therapeutic implications. Free Radic. Biol. Med. 2021, 171, 345–364. [Google Scholar] [CrossRef]
  27. Burgoyne, J.R.; Madhani, M.; Cuello, F.; Charles, R.L.; Brennan, J.P.; Schröder, E. Cysteine redox sensor in PKGIa enables oxidant-induced activation. Science 2007, 317, 1393–1397. [Google Scholar] [CrossRef]
  28. Kuster, G.M.; Siwik, D.A.; Pimentel, D.R.; Colucci, W.S. Role of Reversible, Thioredoxin-Sensitive Oxidative Protein Modifications in Cardiac Myocytes. Antioxid. Redox Signal. 2006, 8, 2153–2159. [Google Scholar] [CrossRef]
  29. Erickson, J.R.; Joiner, M.-L.A.; Guan, X.; Kutschke, W.; Yang, J.; Oddis, C.V.; Bartlett, R.K.; Lowe, J.S.; O’Donnell, S.E.; Aykin-Burns, N.; et al. A Dynamic Pathway for Calcium-Independent Activation of CaMKII by Methionine Oxidation. Cell 2008, 133, 462–474. [Google Scholar] [CrossRef] [Green Version]
  30. Brennan, J.P.; Bardswell, S.C.; Burgoyne, J.R.; Fuller, W.; Schröder, E.; Wait, R. Oxidant-induced activation of type I protein kinase A is mediated by RI subunit interprotein disulfide bond formation. J. Biol. Chem. 2006, 281, 21827–21836. [Google Scholar] [CrossRef] [Green Version]
  31. Ago, T.; Liu, T.; Zhai, P.; Chen, W.; Li, H.; Molkentin, J.; Vatner, S.F.; Sadoshima, J. A Redox-Dependent Pathway for Regulating Class II HDACs and Cardiac Hypertrophy. Cell 2008, 133, 978–993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Sood, H.S.; Cox, M.J.; Tyagi, S.C. Generation of Nitrotyrosine Precedes Activation of Metalloproteinase in Myocardium of Hyperhomocysteinemic Rats. Antioxid. Redox Signal. 2002, 4, 799–804. [Google Scholar] [CrossRef] [PubMed]
  33. Yasukawa, T.; Tokunaga, E.; Ota, H.; Sugita, H.; Martyn, J.A.J.; Kaneki, M. S-Nitrosylation-dependent Inactivation of Akt/Protein Kinase B in Insulin Resistance. J. Biol. Chem. 2005, 280, 7511–7518. [Google Scholar] [CrossRef] [Green Version]
  34. Kehat, I.; Molkentin, J.D. Extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in cardiac hypertrophy. Ann. N. Y. Acad. Sci. 2010, 1188, 96–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Meijles, D.N.; Cull, J.J.; Markou, T.; Cooper, S.T.; Haines, Z.H.; Fuller, S.J.; O’Gara, P.; Sheppard, M.N.; Harding, S.; Sugden, P.H.; et al. Redox Regulation of Cardiac ASK1 (Apoptosis Signal-Regulating Kinase 1) Controls p38-MAPK (Mitogen-Activated Protein Kinase) and Orchestrates Cardiac Remodeling to Hypertension. Hypertension 2020, 76, 1208–1218. [Google Scholar] [CrossRef] [PubMed]
  36. Newton, A.C.; Antal, C.E.; Steinberg, S.F. Protein kinase C mechanisms that contribute to cardiac remodelling. Clin. Sci. 2016, 130, 1499–1510. [Google Scholar] [CrossRef] [Green Version]
  37. Gao, G.; Dudley, S.C. Redox regulation, NF-kappaB, and atrial fibrillation. Antioxid. Redox Signal 2009, 11, 2265–2277. [Google Scholar] [CrossRef] [Green Version]
  38. Sasaki, H.; Galang, N.; Maulik, N. Redox regulation of NF-kappaB and AP-1 in ischemic reperfused heart. Antioxid. Redox Signal 1999, 1, 317–324. [Google Scholar] [CrossRef]
  39. Ho, E.; Karimi Galougahi, K.; Liu, C.-C.; Bhindi, R.; Figtree, G.A. Biological markers of oxidative stress: Applications to cardiovascular research and practice. Redox Biol. 2013, 1, 483–491. [Google Scholar] [CrossRef] [Green Version]
  40. Nakamura, K.; Fushimi, K.; Kouchi, H.; Mihara, K.; Miyazaki, M.; Ohe, T.; Namba, M. Inhibitory Effects of Antioxidants on Neonatal Rat Cardiac Myocyte Hypertrophy Induced by Tumor Necrosis Factor-α and Angiotensin II. Circulation 1998, 98, 794–799. [Google Scholar] [CrossRef] [Green Version]
  41. Yokoyama, T.; Nakano, M.; Bednarczyk, J.L.; McIntyre, B.W.; Entman, M.; Mann, U.L. Tumor Necrosis Factor-α Provokes a Hypertrophic Growth Response in Adult Cardiac Myocytes. Circulation 1997, 95, 1247–1252. [Google Scholar] [CrossRef]
  42. Yamazaki, T.; Komuro, I.; Zou, Y.; Kudoh, S.; Shiojima, I.; Hiroi, Y. Norepinephrine induces the raf-1 kinase/mitogen-activated protein kinase cascade through both alpha 1- and beta-adrenoceptors. Circulation 1997, 95, 1260–1268. [Google Scholar] [CrossRef] [PubMed]
  43. Balaban, R.S.; Nemoto, S.; Finkel, T. Mitochondria, oxidants, and aging. Cell 2005, 120, 483–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Wijnker, P.J.; Sequeira, V.; Kuster, D.W.; Van Der Velden, J. Hypertrophic Cardiomyopathy: A Vicious Cycle Triggered by Sarcomere Mutations and Secondary Disease Hits. Antioxid. Redox Signal. 2019, 31, 318–358. [Google Scholar] [CrossRef]
  45. Del Rio, D.; Stewart, A.J.; Pellegrini, N. A review of recent studies on malondialdehyde as toxic molecule and biological marker of oxidative stress. Nutr. Metab. Cardiovasc. Dis. 2005, 15, 316–328. [Google Scholar] [CrossRef] [PubMed]
  46. Horacio, E.C.; Cingolani, H.E.; Perez, N.G.; Aiello, E.A.; Ennis, I.L.; Garciarena, C.D.; Villa-Abrille, M.C.; Dulce, R.A.; Caldiz, C.I.; Yeves, A.M.; et al. Early signals after stretch leading to cardiac hypertrophy. Key role of NHE-1. Front. Biosci. 2008, 13, 7096–7114. [Google Scholar] [CrossRef] [Green Version]
  47. Palomeque, J.; Rueda, O.V.; Sapia, L.; Valverde, C.A.; Salas, M.; Petroff, M.V.; Mattiazzi, A. Angiotensin II–Induced Oxidative Stress Resets the Ca2+ Dependence of Ca2+–Calmodulin Protein Kinase II and Promotes a Death Pathway Conserved Across Different Species. Circ. Res. 2009, 105, 1204–1212. [Google Scholar] [CrossRef] [Green Version]
  48. Zhao, R.Z.; Jiang, S.; Zhang, L.; Yu, Z.-B. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int. J. Mol. Med. 2019, 44, 3–15. [Google Scholar] [CrossRef] [Green Version]
  49. Marck, P.; Pessoa, M.; Xu, Y.; Kutz, L.; Collins, D.; Yan, Y.; King, C.; Wang, X.; Duan, Q.; Cai, L.; et al. Cardiac Oxidative Signaling and Physiological Hypertrophy in the Na/K-ATPase α1s/sα2s/s Mouse Model of High Affinity for Cardiotonic Steroids. Int. J. Mol. Sci. 2021, 22, 3462. [Google Scholar] [CrossRef]
  50. Pieske, B.; Maier, L.S.; PiacentinoIII, V.; Weisser, J.; Hasenfuss, G.; Houser, S. Rate Dependence of [Na+] i and Contractility in Nonfailing and Failing Human Myocardium. Circulation 2002, 106, 447–453. [Google Scholar] [CrossRef] [Green Version]
  51. Liu, Y.; Wang, H.; Wang, J.; Wei, B.; Zhang, X.; Zhang, M.; Cao, D.; Dai, J.; Wang, Z.; Nyirimigabo, E.; et al. A positive feedback regulation of Heme oxygenase 1 by CELF1 in cardiac myoblast cells. Biochim. Biophys. Acta Gene Regul. Mech. 2019, 1862, 209–218. [Google Scholar] [CrossRef]
  52. AlHayaza, R.; Haque, E.; Karbasiafshar, C.; Sellke, F.W.; Abid, M.R. The Relationship Between Reactive Oxygen Species and Endothelial Cell Metabolism. Front. Chem. 2020, 8, 592688. [Google Scholar] [CrossRef] [PubMed]
  53. Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc. Pharmacol. 2018, 100, 1–19. [Google Scholar] [CrossRef]
  54. Cimmino, G.; Cirillo, P.; Ragni, M.; Conte, S.; Uccello, G.; Golino, P. Reactive oxygen species induce a procoagulant state in endothelial cells by inhibiting tissue factor pathway inhibitor. J. Thromb. Thrombolysis 2015, 40, 186–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhang, M.; Perino, A.; Ghigo, A.; Hirsch, E.; Shah, A.M. NADPH Oxidases in Heart Failure: Poachers or Gamekeepers? Antioxid. Redox Signal 2013, 18, 1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ekelund, U.E.G.; Harrison, R.W.; Shokek, O.; Thakkar, R.N.; Tunin, R.S.; Senzaki, H.; Kass, D.A.; Marbaán, E.; Hare, J.M. Intravenous Allopurinol Decreases Myocardial Oxygen Consumption and Increases Mechanical Efficiency in Dogs with Pacing-Induced Heart Failure. Circ. Res. 1999, 85, 437–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Jankovic, A.; Korac, A.; Buzadzic, B.; Stancic, A.; Otasevic, V.; Ferdinandy, P.; Daiber, A.; Korac, B. Targeting the NO/superoxide ratio in adipose tissue: Relevance to obesity and diabetes management. J. Cereb. Blood Flow Metab. 2016, 174, 1570–1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Looi, Y.H.; Grieve, D.J.; Siva, A.; Walker, S.J.; Anilkumar, N.; Cave, A.C.; Marber, M.; Monaghan, M.J.; Shah, A.M. Involvement of Nox2 NADPH Oxidase in Adverse Cardiac Remodeling After Myocardial Infarction. Hypertension 2008, 51, 319–325. [Google Scholar] [CrossRef] [Green Version]
  59. Lassègue, B.; San Martín, A.; Griendling, K.K. Biochemistry, Physiology, and Pathophysiology of NADPH Oxidases in the Cardiovascular System. Circ. Res. 2012, 110, 1364–1390. [Google Scholar] [CrossRef]
  60. Prosser, B.L.; Ward, C.W.; Lederer, W.J. X-ROS Signaling: Rapid Mechano-Chemo Transduction in Heart. Science 2011, 333, 1440–1445. [Google Scholar] [CrossRef] [Green Version]
  61. Izu, L.T.; Kohl, P.; Boyden, P.A.; Miura, M.; Banyasz, T.; Chiamvimonvat, N. Mechano-Electric and Mechano-Chemo-Transduction in Cardiomyocytes. J. Physiol. 2020, 598, 1285. [Google Scholar] [CrossRef] [PubMed]
  62. Murdoch, C.E.; Zhang, M.; Cave, A.C.; Shah, A.M. NADPH oxidase-dependent redox signalling in cardiac hypertrophy, remodelling and failure. Cardiovasc. Res. 2006, 71, 208–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Takimoto, E.; Champion, H.C.; Li, M.; Ren, S.; Rodriguez, E.R.; Tavazzi, B.; Lazzarino, G.; Paolocci, N.; Gabrielson, K.L.; Wang, Y.; et al. Oxidant stress from nitric oxide synthase–3 uncoupling stimulates cardiac pathologic remodeling from chronic pressure load. J. Clin. Investig. 2005, 115, 1221–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cheng, D.; Chen, L.; Tu, W.; Wang, H.; Wang, Q.; Meng, L. Protective effects of valsartan administration on doxorubicin-induced myocardial injury in rats and the role of oxidative stress and NOX2/NOX4 signaling. Mol. Med. Rep. 2020, 22, 4151–4162. [Google Scholar] [CrossRef]
  65. 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. Circ. Physiol. 2003, 285, H2290–H2297. [Google Scholar] [CrossRef] [Green Version]
  66. Nakagami, H.; Takemoto, M.; Liao, J.K. NADPH oxidase-derived superoxide anion mediates angiotensin II-induced cardiac hypertrophy. J. Mol. Cell. Cardiol. 2003, 35, 851–859. [Google Scholar] [CrossRef]
  67. Higuchi, Y.; Otsu, K.; Nishida, K.; Hirotani, S.; Nakayama, H.; Yamaguchi, O. The Small GTP-binding Protein Rac1 Induces Cardiac Myocyte Hypertrophy through the Activation of Apoptosis Signal-regulating Kinase 1 and Nuclear Factor-κB*. J. Biol. Chem. 2003, 278, 20770–20777. [Google Scholar] [CrossRef] [Green Version]
  68. Xiao, L.; Pimental, D.R.; Amin, J.K.; Singh, K.; Sawyer, D.B.; Colucci, W.S. MEK1/2-ERK1/2 mediates alpha1-adrenergic receptor-stimulated hypertrophy in adult rat ventricular myocytes. J. Mol. Cell Cardiol. 2001, 33, 779–787. [Google Scholar] [CrossRef]
  69. Byrne, J.A.; Grieve, D.J.; Bendall, J.K.; Li, J.-M.; Gove, C.; Lambeth, J.D.; Cave, A.C.; Shah, A.M. Contrasting Roles of NADPH Oxidase Isoforms in Pressure-Overload Versus Angiotensin II–Induced Cardiac Hypertrophy. Circ. Res. 2003, 93, 802–805. [Google Scholar] [CrossRef]
  70. Bendall, J.K.; Cave, A.C.; Heymes, C.; Gall, N.; Shah, A.M. Pivotal Role of a gp91 phox -Containing NADPH Oxidase in Angiotensin II-Induced Cardiac Hypertrophy in Mice. Circulation 2002, 105, 293–296. [Google Scholar] [CrossRef] [Green Version]
  71. Grote, K.; Flach, I.; Luchtefeld, M.; Akin, E.; Holland, S.M.; Drexler, H.; Schieffer, B. Mechanical Stretch Enhances mRNA Expression and Proenzyme Release of Matrix Metalloproteinase-2 (MMP-2) via NAD(P)H Oxidase–Derived Reactive Oxygen Species. Circ. Res. 2003, 92, e80–e86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Sun, Y.; Zhang, J.; Lu, L.; Chen, S.S.; Quinn, M.T.; Weber, K.T. Aldosterone-induced inflammation in the rat heart: Role of oxidative stress. Am. J. Pathol. 2002, 161, 1773–1781. [Google Scholar] [CrossRef]
  73. Luchtefeld, M.; Grote, K.; Grothusen, C.; Bley, S.; Bandlow, N.; Selle, T.; Strüber, M.; Haverich, A.; Bavendiek, U.; Drexler, H.; et al. Angiotensin II induces MMP-2 in a p47phox-dependent manner. Biochem. Biophys. Res. Commun. 2005, 328, 183–188. [Google Scholar] [CrossRef]
  74. Johar, S.; Cave, A.C.; Narayanapanicker, A.; Grieve, D.J.; Shah, A.M. Aldosterone mediates angiotensin II-induced interstitial cardiac fibrosis via a Nox2-containing NADPH oxidase. FASEB J. 2006, 20, 1546–1548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Colston, J.T.; De La Rosa, S.D.; Strader, J.R.; Anderson, M.A.; Freeman, G.L. H2O2 activates Nox4 through PLA2-dependent arachidonic acid production in adult cardiac fibroblasts. FEBS Lett. 2005, 579, 2533–2540. [Google Scholar] [CrossRef] [Green Version]
  76. Cucoranu, I.; Clempus, R.; Dikalova, A.; Phelan, P.J.; Ariyan, S.; Dikalov, S. NAD(P)H oxidase 4 mediates transforming growth factor-beta1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ. Res. 2005, 97, 900–907. [Google Scholar] [CrossRef] [Green Version]
  77. Harrison, R. Structure and function of xanthine oxidoreductase: Where are we now? Free Radic. Biol. Med. 2002, 33, 774–797. [Google Scholar] [CrossRef]
  78. Harrison, R. Physiological Roles of Xanthine Oxidoreductase. Drug Metab. Rev. 2004, 36, 363–375. [Google Scholar] [CrossRef]
  79. Gladden, J.D.; Zelickson, B.R.; Wei, C.-C.; Ulasova, E.; Zheng, J.; Ahmed, M.I.; Chen, Y.; Bamman, M.; Ballinger, S.; Darley-Usmar, V.; et al. Novel insights into interactions between mitochondria and xanthine oxidase in acute cardiac volume overload. Free Radic. Biol. Med. 2011, 51, 1975–1984. [Google Scholar] [CrossRef] [Green Version]
  80. Watanabe, K.; Watanabe, T.; Otaki, Y.; Shishido, T.; Murase, T.; Nakamura, T.; Kato, S.; Tamura, H.; Nishiyama, S.; Takahashi, H.; et al. Impact of plasma xanthine oxidoreductase activity in patients with heart failure with preserved ejection fraction. ESC Heart Fail. 2020, 7, 1735–1743. [Google Scholar] [CrossRef]
  81. Semenza, G.L. Hypoxia-Inducible Factor 1 and Cardiovascular Disease. Annu. Rev. Physiol. 2014, 76, 39–56. [Google Scholar] [CrossRef] [Green Version]
  82. Massion, P.B.; Balligand, J.-L. Relevance of nitric oxide for myocardial remodeling. Curr. Heart Fail. Rep. 2007, 4, 18–25. [Google Scholar] [CrossRef]
  83. Soskić, S.S.; Dobutović, B.D.; Sudar, E.M.; Obradović, M.M.; Nikolić, D.M.; Djordjevic, J.D.; Radak, D.J.; Mikhailidis, D.P.; Isenović, E.R. Regulation of Inducible Nitric Oxide Synthase (iNOS) and its Potential Role in Insulin Resistance, Diabetes and Heart Failure. Open Cardiovasc. Med. J. 2011, 5, 153–163. [Google Scholar] [CrossRef] [Green Version]
  84. Marino, A.; Hausenloy, D.J.; Andreadou, I.; Horman, S.; Bertrand, L.; Beauloye, C. AMP-activated protein kinase: A remarkable contributor to preserve a healthy heart against ROS injury. Free Radic. Biol. Med. 2021, 166, 238–254. [Google Scholar] [CrossRef] [PubMed]
  85. North, B.J.; Sinclair, D.A. The Intersection Between Aging and Cardiovascular Disease. Circ. Res. 2012, 110, 1097–1108. [Google Scholar] [CrossRef]
  86. Finkel, T.; Deng, C.-X.; Mostoslavsky, R. Recent progress in the biology and physiology of sirtuins. Nature 2009, 460, 587–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wang, T.; Cao, Y.; Zheng, Q.; Tu, J.; Zhou, W.; He, J.; Zhong, J.; Chen, Y.; Wang, J.; Cai, R.; et al. SENP1-Sirt3 Signaling Controls Mitochondrial Protein Acetylation and Metabolism. Mol. Cell 2019, 75, 823–834.e5. [Google Scholar] [CrossRef]
  88. Koentges, C.; Cimolai, M.C.; Pfeil, K.; Wolf, D.; Marchini, T.O.; Tarkhnishvili, A.; Hoffmann, M.M.; Odening, K.E.; Diehl, P.; Mühlen, C.V.Z.; et al. Impaired SIRT3 activity mediates cardiac dysfunction in endotoxemia by calpain-dependent disruption of ATP synthesis. J. Mol. Cell. Cardiol. 2019, 133, 138–147. [Google Scholar] [CrossRef] [PubMed]
  89. Sundaresan, N.R.; Gupta, M.; Kim, G.; Rajamohan, S.B.; Isbatan, A.; Gupta, M.P. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J. Clin. Investig. 2009, 119, 2758–2771. [Google Scholar] [CrossRef] [Green Version]
  90. Khalil, H.; Kanisicak, O.; Prasad, V.; Correll, R.N.; Fu, X.; Schips, T. Fibroblast-specific TGF-β-Smad2/3 signaling underlies cardiac fibrosis. J. Clin. Investig. 2017, 127, 3770–3783. [Google Scholar] [CrossRef]
  91. Luo, K.; Huang, W.; Tang, S. Sirt3 enhances glioma cell viability by stabilizing Ku70–BAX interaction. OncoTargets Ther. 2018, 11, 7559–7567. [Google Scholar] [CrossRef] [Green Version]
  92. Tseng, A.H.; Shieh, S.-S.; Wang, D.L. SIRT3 deacetylates FOXO3 to protect mitochondria against oxidative damage. Free Radic. Biol. Med. 2013, 63, 222–234. [Google Scholar] [CrossRef] [PubMed]
  93. Bergaggio, E.; Riganti, C.; Garaffo, G.; Vitale, N.; Mereu, E.; Bandini, C.; Pellegrino, E.; Pullano, V.; Omedè, P.; Todoerti, K.; et al. IDH2 inhibition enhances proteasome inhibitor responsiveness in hematological malignancies. Blood 2019, 133, 156–167. [Google Scholar] [CrossRef] [Green Version]
  94. Peoples, J.N.; Saraf, A.; Ghazal, N.; Pham, T.T.; Kwong, J.Q. Mitochondrial dysfunction and oxidative stress in heart disease. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [PubMed]
  95. Anderson, E.J.; Rodriguez, E.; Anderson, C.A.; Thayne, K.; Chitwood, W.R.; Kypson, A.P. Increased propensity for cell death in diabetic human heart is mediated by mitochondrial-dependent pathways. Am. J. Physiol. Circ. Physiol. 2011, 300, H118–H124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Brand, M.D. The sites and topology of mitochondrial superoxide production. Exp. Gerontol. 2010, 45, 466–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Sinha, K.; Das, J.; Pal, P.B.; Sil, P.C. Oxidative stress: The mitochondria-dependent and mitochondria-independent pathways of apoptosis. Arch. Toxicol. 2013, 87, 1157–1180. [Google Scholar] [CrossRef] [PubMed]
  98. Bensaad, K.; Cheung, E.C.; Vousden, K.H. Modulation of intracellular ROS levels by TIGAR controls autophagy. EMBO J. 2009, 28, 3015–3026. [Google Scholar] [CrossRef] [Green Version]
  99. Morgan, M.J.; Liu, Z.G. Reactive oxygen species in TNFalpha-induced signaling and cell death. Mol. Cells 2010, 30, 1–12. [Google Scholar] [CrossRef]
  100. Anderson, E.J.; Efird, J.T.; Davies, S.W.; O’Neal, W.T.; Darden, T.M.; Thayne, K.A.; Katunga, L.A.; Kindell, L.C.; Ferguson, T.B.; Anderson, C.A.; et al. Monoamine Oxidase is a Major Determinant of Redox Balance in Human Atrial Myocardium and is Associated with Postoperative Atrial Fibrillation. J. Am. Heart Assoc. 2014, 3, e000713. [Google Scholar] [CrossRef] [Green Version]
  101. Santin, Y.; Sicard, P.; Vigneron, F.; Guilbeau-Frugier, C.; Dutaur, M.; Lairez, O.; Couderc, B.; Manni, D.; Korolchuk, V.I.; Lezoualc’H, F.; et al. Oxidative Stress by Monoamine Oxidase-A Impairs Transcription Factor EB Activation and Autophagosome Clearance, Leading to Cardiomyocyte Necrosis and Heart Failure. Antioxid. Redox Signal. 2016, 25, 10–27. [Google Scholar] [CrossRef]
  102. Deshwal, S.; Di Sante, M.; Di Lisa, F.; Kaludercic, N. Emerging role of monoamine oxidase as a therapeutic target for cardiovascular disease. Curr. Opin. Pharmacol. 2017, 33, 64–69. [Google Scholar] [CrossRef] [PubMed]
  103. Pchejetski, D.; Kunduzova, O.; Dayon, A.; Calise, D.; Seguelas, M.-H.; Leducq, N.; Seif, I.; Parini, A.; Cuvillier, O. Oxidative Stress–Dependent Sphingosine Kinase-1 Inhibition Mediates Monoamine Oxidase A–Associated Cardiac Cell Apoptosis. Circ. Res. 2007, 100, 41–49. [Google Scholar] [CrossRef] [Green Version]
  104. Menazza, S.; Blaauw, B.; Tiepolo, T.; Toniolo, L.; Braghetta, P.; Spolaore, B.; Reggiani, C.; Di Lisa, F.; Bonaldo, P.; Canton, M. Oxidative stress by monoamine oxidases is causally involved in myofiber damage in muscular dystrophy. Hum. Mol. Genet. 2010, 19, 4207–4215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Molavian, H.; Tonekaboni, A.M.; Kohandel, M.; Sivaloganathan, S. The Synergetic Coupling among the Cellular Antioxidants Glutathione Peroxidase/Peroxiredoxin and Other Antioxidants and its Effect on the Concentration of H2O2. Sci. Rep. 2015, 5, srep13620. [Google Scholar] [CrossRef]
  106. Aon, M.A.; Stanley, B.A.; Sivakumaran, V.; Kembro, J.M.; O’Rourke, B.; Paolocci, N.; Cortassa, S. Glutathione/thioredoxin systems modulate mitochondrial H2O2 emission: An experimental-computational study. J. Gen. Physiol. 2012, 139, 479–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Ying, W. NAD+/NADH and NADP+/NADPH in cellular functions and cell death: Regulation and biological consequences. Antioxid. Redox Signal 2008, 10, 179–206. [Google Scholar] [CrossRef] [Green Version]
  108. Nickel, A.G.; Von Hardenberg, A.; Hohl, M.; Löffler, J.R.; Kohlhaas, M.; Becker, J.; Reil, J.-C.; Kazakov, A.; Bonnekoh, J.; Stadelmaier, M.; et al. Reversal of Mitochondrial Transhydrogenase Causes Oxidative Stress in Heart Failure. Cell Metab. 2015, 22, 472–484. [Google Scholar] [CrossRef] [Green Version]
  109. Kohlhaas, M.; Liu, T.; Knopp, A.; Zeller, T.; Ong, M.F.; Böhm, M.; O’Rourke, B.; Maack, C. Elevated Cytosolic Na+ Increases Mitochondrial Formation of Reactive Oxygen Species in Failing Cardiac Myocytes. Circulation 2010, 121, 1606–1613. [Google Scholar] [CrossRef] [Green Version]
  110. Schulz, E.; Wenzel, P.; Münzel, T.; Daiber, A. Mitochondrial Redox Signaling: Interaction of Mitochondrial Reactive Oxygen Species with Other Sources of Oxidative Stress. Antioxid. Redox Signal. 2014, 20, 308–324. [Google Scholar] [CrossRef]
  111. Wenzel, P.; Müller, J.; Zurmeyer, S.; Schuhmacher, S.; Schulz, E.; Oelze, M.; Pautz, A.; Kawamoto, T.; Wojnowski, L.; Kleinert, H.; et al. ALDH-2 deficiency increases cardiovascular oxidative stress—Evidence for indirect antioxidative properties. Biochem. Biophys. Res. Commun. 2008, 367, 137–143. [Google Scholar] [CrossRef] [PubMed]
  112. Bahoran, T.; Soobrattee, M.A.; Luximon-Ramma, V.; Aruoma, O.I. Free Radicals and Antioxidants in Cardiovascular Health and Disease. Internet J. Med. Updat. 2006, 1, 24–40. [Google Scholar] [CrossRef] [Green Version]
  113. Sverdlov, A.L.; Ngo, D.T.M.; Colucci, W.S. 8—Oxidative Stress in Heart Failure. In Heart Failure: A Companion to Braunwald’s Heart Disease; Elsevier: Philadelphia, PA, USA, 2020; pp. 115–126.e6. [Google Scholar]
  114. De Keulenaer, G.; Brutsaert, D.L. Diastolic and systolic heart failure are distinct phenotypes within the heart failure spectrum. Circulation 2011, 123, 2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Nishihara, T.; Tokitsu, T.; Sueta, D.; Oike, F.; Takae, M.; Fujisue, K.; Usuku, H.; Ito, M.; Kanazawa, H.; Araki, S.; et al. Clinical significance of reactive oxidative metabolites in patients with heart failure with reduced left ventricular ejection fraction. J. Card. Fail. 2020, 27, 57–66. [Google Scholar] [CrossRef]
  116. Hummel, S.L.; Seymour, E.M.; Brook, R.D.; Kolias, T.J.; Sheth, S.S.; Rosenblum, H.R.; Wells, J.M.; Weder, A.B. Low-Sodium Dietary Approaches to Stop Hypertension Diet Reduces Blood Pressure, Arterial Stiffness, and Oxidative Stress in Hypertensive Heart Failure with Preserved Ejection Fraction. Hypertension 2012, 60, 1200–1206. [Google Scholar] [CrossRef] [Green Version]
  117. Chaanine, A.H.; Kohlbrenner, E.; Gamb, S.I.; Guenzel, A.J.; Klaus, K.; Fayyaz, A.U.; Nair, K.S.; Hajjar, R.J.; Redfield, M.M. FOXO3a regulates BNIP3 and modulates mitochondrial calcium, dynamics, and function in cardiac stress. Am. J. Physiol. Circ. Physiol. 2016, 311, H1540–H1559. [Google Scholar] [CrossRef] [Green Version]
  118. Chaanine, A.H. Morphological Stages of Mitochondrial Vacuolar Degeneration in Phenylephrine-Stressed Cardiac Myocytes and in Animal Models and Human Heart Failure. Medicina 2019, 55, 239. [Google Scholar] [CrossRef] [Green Version]
  119. Chaanine, A.H.; Joyce, L.D.; Stulak, J.M.; Maltais, S.; Joyce, D.L.; Dearani, J.A.; Klaus, K.; Nair, K.S.; Hajjar, R.J.; Redfield, M.M. Mitochondrial Morphology, Dynamics, and Function in Human Pressure Overload or Ischemic Heart Disease with Preserved or Reduced Ejection Fraction. Circ. Heart Fail. 2019, 12, e005131. [Google Scholar] [CrossRef]
  120. Terman, A.; Kurz, T.; Gustafsson, B.; Brunk, U.T. The Involvement of Lysosomes in Myocardial Aging and Disease. Curr. Cardiol. Rev. 2008, 4, 107–115. [Google Scholar] [CrossRef] [Green Version]
  121. Leboucher, G.P.; Tsai, Y.C.; Yang, M.; Shaw, K.C.; Zhou, M.; Veenstra, T.D.; Glickman, M.H.; Weissman, A.M. Stress-Induced Phosphorylation and Proteasomal Degradation of Mitofusin 2 Facilitates Mitochondrial Fragmentation and Apoptosis. Mol. Cell 2012, 47, 547–557. [Google Scholar] [CrossRef] [Green Version]
  122. Chaanine, A.; LeJemtel, T.; Delafontaine, P. Mitochondrial Pathobiology and Metabolic Remodeling in Progression to Overt Systolic Heart Failure. J. Clin. Med. 2020, 9, 3582. [Google Scholar] [CrossRef]
  123. Chen, H.C.; Detmer, S.A.; Ewald, A.J.; Griffin, E.E.; Fraser, S.E.; Chan, D.C. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J. Cell Biol. 2003, 160, 189–200. [Google Scholar] [CrossRef] [PubMed]
  124. Cipolat, S.; De Brito, O.M.; Dal Zilio, B.; Scorrano, L. OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc. Natl. Acad. Sci. USA 2004, 101, 15927–15932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Chang, C.-R.; Blackstone, C. Dynamic regulation of mitochondrial fission through modification of the dynamin-related protein Drp1. Ann. N. Y. Acad. Sci. 2010, 1201, 34–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Varanita, T.; Soriano, M.E.; Romanello, V.; Zaglia, T.; Quintana-Cabrera, R.; Semenzato, M.; Menabò, R.; Costa, V.; Civiletto, G.; Pesce, P.; et al. The Opa1-Dependent Mitochondrial Cristae Remodeling Pathway Controls Atrophic, Apoptotic, and Ischemic Tissue Damage. Cell Metab. 2015, 21, 834–844. [Google Scholar] [CrossRef] [Green Version]
  127. Landes, T.; Emorine, L.J.; Courilleau, D.; Rojo, M.; Belenguer, P.; Arnauné-Pelloquin, L. The BH3-only Bnip3 binds to the dynamin Opa1 to promote mitochondrial fragmentation and apoptosis by distinct mechanisms. EMBO Rep. 2010, 11, 459–465. [Google Scholar] [CrossRef] [Green Version]
  128. Nan, J.; Zhu, W.; Rahman, M.; Liu, M.; Li, D.; Su, S.; Zhang, N.; Hu, X.; Yu, H.; Gupta, M.P.; et al. Molecular regulation of mitochondrial dynamics in cardiac disease. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1260–1273. [Google Scholar] [CrossRef]
  129. Chaanine, A.H.; Jeong, D.; Liang, L.; Chemaly, E.R.; Fish, K.; Gordon, R.E.; Hajjar, R.J. JNK modulates FOXO3a for the expression of the mitochondrial death and mitophagy marker BNIP3 in pathological hypertrophy and in heart failure. Cell Death Dis. 2012, 3, e265. [Google Scholar] [CrossRef] [Green Version]
  130. Aoki, H.; Kang, P.M.; Hampe, J.; Yoshimura, K.; Noma, T.; Matsuzaki, M.; Izumo, S. Direct Activation of Mitochondrial Apoptosis Machinery by c-Jun N-terminal Kinase in Adult Cardiac Myocytes. J. Biol. Chem. 2002, 277, 10244–10250. [Google Scholar] [CrossRef] [Green Version]
  131. Chang, C.-R.; Blackstone, C. Cyclic AMP-dependent Protein Kinase Phosphorylation of Drp1 Regulates Its GTPase Activity and Mitochondrial Morphology. J. Biol. Chem. 2007, 282, 21583–21587. [Google Scholar] [CrossRef] [Green Version]
  132. Pan, X.; Liu, J.; Nguyen, T.; Liu, C.; Sun, J.; Teng, Y. The physiological role of mitochondrial calcium revealed by mice lacking the mitochondrial calcium uniporter (MCU). Nat. Cell Biol. 2013, 15, 1464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wu, Y.; Rasmussen, T.P.; Koval, O.M.; Joiner, M.L.A.; Hall, D.D.; Chen, B. The mitochondrial uniporter controls fight or flight heart rate increases. Nat. Commun. 2015, 6, 6081. [Google Scholar] [CrossRef] [Green Version]
  134. Finkel, T.; Menazza, S.; Holmström, K.M.; Parks, R.J.; Liu, J.; Sun, J. The ins and outs of mitochondrial calcium. Circ. Res. 2015, 116, 1810–1819. [Google Scholar] [CrossRef] [Green Version]
  135. Chaanine, A.H.; Gordon, R.E.; Kohlbrenner, E.; Benard, L.; Jeong, D.; Hajjar, R.J. Potential role of BNIP3 in cardiac remodeling, myocardial stiffness, and endoplasmic reticulum: Mitochondrial calcium homeostasis in diastolic and systolic heart failure. Circ. Heart Fail. 2013, 6, 572–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Horton, J.L.; Martin, O.J.; Lai, L.; Riley, N.M.; Richards, A.L.; Vega, R.B.; Leone, T.C.; Pagliarini, D.J.; Muoio, D.M.; Bedi, K.C., Jr.; et al. Mitochondrial protein hyperacetylation in the failing heart. JCI Insight 2016, 1, e84897. [Google Scholar] [CrossRef] [PubMed]
  137. Chen, T.; Liu, J.; Li, N.; Wang, S.; Liu, H.; Li, J. Mouse SIRT3 Attenuates Hypertrophy-Related Lipid Accumulation in the Heart through the Deacetylation of LCAD. PLoS ONE 2015, 10, e0118909. [Google Scholar] [CrossRef] [PubMed]
  138. Cortassa, S.; Juhaszova, M.; Aon, M.A.; Zorov, D.B.; Sollott, S.J. Mitochondrial Ca2+, redox environment and ROS emission in heart failure: Two sides of the same coin? J. Mol. Cell Cardiol. 2021, 151, 113–125. [Google Scholar] [CrossRef] [PubMed]
  139. Nagai, T.; Anzai, T.; Kaneko, H.; Mano, Y.; Anzai, A.; Maekawa, Y. C-reactive protein overexpression exacerbates pressure overload-induced cardiac remodeling through enhanced inflammatory response. Hypertension 2011, 57, 208–215. [Google Scholar] [CrossRef]
  140. Kobayashi, S.; Inoue, N.; Ohashi, Y.; Terashima, M.; Matsui, K.; Mori, T. Interaction of oxidative stress and inflammatory response in coronary plaque instability: Important role of C-reactive protein. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1398–1404. [Google Scholar] [CrossRef] [Green Version]
  141. Paulus, W.J.; Tschöpe, C. A novel paradigm for heart failure with preserved ejection fraction: Comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J. Am. Coll. Cardiol. 2013, 62, 263–271. [Google Scholar] [CrossRef] [Green Version]
  142. Simmonds, S.J.; Cuijpers, I.; Heymans, S.; Jones, E.A.V. Cellular and Molecular Differences between HFpEF and HFrEF: A Step Ahead in an Improved Pathological Understanding. Cells 2020, 9, 242. [Google Scholar] [CrossRef] [Green Version]
  143. Katayama, Y.; Battista, M.; Kao, W.-M.; Hidalgo, A.; Peired, A.; Thomas, S.A.; Frenette, P.S. Signals from the Sympathetic Nervous System Regulate Hematopoietic Stem Cell Egress from Bone Marrow. Cell 2006, 124, 407–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Dutta, P.; Nahrendorf, M. Monocytes in myocardial infarction. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 1066–1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Noll, N.A.; Lal, H.; Merryman, W.D. Mouse Models of Heart Failure with Preserved or Reduced Ejection Fraction. Am. J. Pathol. 2020, 190, 1596–1608. [Google Scholar] [CrossRef] [PubMed]
  146. Li, X.; Liu, J.; Lu, Q.; Ren, D.; Sun, X.; Rousselle, T.; Tan, Y.; Li, J. AMPK: A therapeutic target of heart failure—Not only metabolism regulation. Biosci. Rep. 2019, 39, BSR20181767. [Google Scholar] [CrossRef]
  147. Kanwar, M.; Walter, C.; Clarke, M.; Aponte, M. Targeting heart failure with preserved ejection fraction: Current status and future prospects. Vasc. Health Risk Manag. 2016, 12, 129–141. [Google Scholar] [CrossRef] [Green Version]
  148. Hamdani, N.; Franssen, C.; Lourenço, A.; Falca o-Pires, I.; Fontoura, D.; Leite, S. Myocardial titin hypophosphorylation importantly contributes to heart failure with preserved ejection fraction in a rat metabolic risk model. Circ. Heart Fail. 2013, 6, 1239–1249. [Google Scholar] [CrossRef] [Green Version]
  149. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric Oxide and Peroxynitrite in Health and Disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [Green Version]
  150. Ben-Nun, D.; Buja, L.M.; Fuentes, F. Prevention of heart failure with preserved ejection fraction (HFpEF): Reexamining microRNA-21 inhibition in the era of oligonucleotide-based therapeutics. Cardiovasc. Pathol. 2020, 49, 107243. [Google Scholar] [CrossRef]
  151. Collier, P.; Watson, C.J.; Voon, V.; Phelan, D.; Jan, A.; Mak, G. Can emerging biomarkers of myocardial remodelling identify asymptomatic hypertensive patients at risk for diastolic dysfunction and diastolic heart failure? Eur. J. Heart Fail. 2011, 13, 1087–1095. [Google Scholar] [CrossRef] [Green Version]
  152. Willis, M.S.; Patterson, C. Proteotoxicity and cardiac dysfunction—Alzheimer’s disease of the heart? N. Engl. J. Med. 2013, 368, 455–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Taube, A.; Schlich, R.; Sell, H.; Eckardt, K.; Eckel, J. Inflammation and metabolic dysfunction: Links to cardiovascular diseases. Am. J. Physiol. Circ. Physiol. 2012, 302, H2148–H2165. [Google Scholar] [CrossRef]
  154. Jelic, S.; Lederer, D.J.; Adams, T.; Padeletti, M.; Colombo, P.C.; Factor, P.H.; Le Jemtel, T.H. Vascular Inflammation in Obesity and Sleep Apnea. Circulation 2010, 121, 1014–1021. [Google Scholar] [CrossRef] [PubMed]
  155. MacDougall, I.C.; Canaud, B.; De Francisco, A.L.; Filippatos, G.; Ponikowski, P.; Silverberg, D.; Van Veldhuisen, D.J.; Anker, S.D. Beyond the cardiorenal anaemia syndrome: Recognizing the role of iron deficiency. Eur. J. Heart Fail. 2012, 14, 882–886. [Google Scholar] [CrossRef] [PubMed]
  156. Matsubara, J.; Sugiyama, S.; Nozaki, T.; Sugamura, K.; Konishi, M.; Ohba, K.; Matsuzawa, Y.; Akiyama, E.; Yamamoto, E.; Sakamoto, K.; et al. Pentraxin 3 Is a New Inflammatory Marker Correlated with Left Ventricular Diastolic Dysfunction and Heart Failure with Normal Ejection Fraction. J. Am. Coll. Cardiol. 2011, 57, 861–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Shah, K.B.; Kop, W.J.; Christenson, R.H.; Diercks, D.B.; Henderson, S.; Hanson, K.; Li, S.-Y.; Defilippi, C.R. Prognostic Utility of ST2 in Patients with Acute Dyspnea and Preserved Left Ventricular Ejection Fraction. Clin. Chem. 2011, 57, 874–882. [Google Scholar] [CrossRef]
  158. Griendling, K.K.; Sorescu, D.; Ushio-Fukai, M. NAD(P)H oxidase: Role in cardiovascular biology and disease. Circ. Res. 2000, 86, 494–501. [Google Scholar] [CrossRef] [Green Version]
  159. Van Heerebeek, L.; Hamdani, N.; Falcão-Pires, I.; Leite-Moreira, A.F.; Begieneman, M.P.V.; Bronzwaer, J.G.F.; van der Velden, J.; Stienen, G.J.; Laarman, G.J.; Somsen, A.; et al. Low myocardial protein kinase G activity in heart failure with preserved ejection fraction. Circulation 2012, 126, 830–839. [Google Scholar] [CrossRef] [Green Version]
  160. Westermann, D.; Lindner, D.; Kasner, M.; Zietsch, C.; Savvatis, K.; Escher, F.; von Schlippenbach, J.; Skurk, C.; Steendijk, P.; Riad, A.; et al. Cardiac Inflammation Contributes to Changes in the Extracellular Matrix in Patients with Heart Failure and Normal Ejection Fraction. Circ. Heart Fail. 2011, 4, 44–52. [Google Scholar] [CrossRef] [Green Version]
  161. Dorsch, M.F.; Lawrance, R.A.; Sapsford, R.J.; Durham, N.; Oldham, J.; Greenwood, D.C.; Jackson, B.M.; Morrell, C.; Robinson, M.B.; Hall, A.S. Poor prognosis of patients presenting with symptomatic myocardial infarction but without chest pain. Heart 2001, 86, 494–498. [Google Scholar] [CrossRef] [Green Version]
  162. Zhang, M.; Shah, A.M. ROS signalling between endothelial cells and cardiac cells. Cardiovasc. Res. 2014, 102, 249–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Paulus, W.J.; Vantrimpont, P.J.; Shah, A.M. Paracrine Coronary Endothelial Control of Left Ventricular Function in Humans. Circulation 1995, 92, 2119–2126. [Google Scholar] [CrossRef]
  164. Denninger, J.W.; Marletta, M.A. Guanylate cyclase and the .NO/cGMP signaling pathway. Biochim. Biophys. Acta 1999, 1411, 334–350. [Google Scholar] [CrossRef] [Green Version]
  165. Cerra, M.; Pellegrino, D. Cardiovascular cGMP-generating systems in physiological and pathological conditions. Curr. Med. Chem. 2007, 14, 585–599. [Google Scholar] [CrossRef]
  166. Tsai, E.J.; Kass, D.A. Cyclic GMP signaling in cardiovascular pathophysiology and therapeutics. Pharmacol. Ther. 2009, 122, 216–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Hobbs, A.J.; Stasch, J.P. Soluble Guanylate Cyclase: Allosteric Activation and Redox Regulation. Nitric Oxide 2010, 1, 301–326. [Google Scholar]
  168. Hamdani, N.; Bishu, K.G.; Von Frieling-Salewsky, M.; Redfield, M.M.; Linke, W.A. Deranged myofilament phosphorylation and function in experimental heart failure with preserved ejection fraction. Cardiovasc. Res. 2012, 97, 464–471. [Google Scholar] [CrossRef] [Green Version]
  169. Bishu, K.G.; Hamdani, N.; Mohammed, S.F.; Kruger, M.; Ohtani, T.; Ogut, O.; Brozovich, F.V.; Burnett, J.C., Jr.; Linke, W.A.; Redfield, M.M. Sildenafil and B-Type Natriuretic Peptide Acutely Phosphorylate Titin and Improve Diastolic Distensibility In Vivo. Circulation 2011, 124, 2882–2891. [Google Scholar] [CrossRef] [Green Version]
  170. Lyle, M.A.; Brozovich, F.V. HFpEF, a Disease of the Vasculature: A Closer Look at the Other Half. Mayo Clin. Proc. 2018, 93, 1305–1314. [Google Scholar] [CrossRef] [Green Version]
  171. Lyle, M.A.; Alabdaljabar, M.S.; Han, Y.S.; Brozovich, F.V. The vasculature in HFpEF vs HFrEF: Differences in contractile protein expression produce distinct phenotypes. Heliyon 2019, 6, e03129. [Google Scholar] [CrossRef] [Green Version]
  172. Schiattarella, G.G.; Altamirano, F.; Tong, D.; French, K.M.; Villalobos, E.; Kim, S.Y.; Luo, X.; Jiang, N.; May, H.I.; Wang, Z.V.; et al. Nitrosative stress drives heart failure with preserved ejection fraction. Nature 2019, 568, 351–356. [Google Scholar] [CrossRef] [PubMed]
  173. Jiang, D.; Niwa, M.; Koong, A.C. Targeting the IRE1α–XBP1 branch of the unfolded protein response in human diseases. Semin. Cancer Biol. 2015, 33, 48–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Ventura-Clapier, R.; Garnier, A.; Veksler, V.; Joubert, F. Bioenergetics of the failing heart. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 1360–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Lopaschuk, G.D.; Ussher, J.R.; Folmes, C.D.L.; Jaswal, J.S.; Stanley, W.C. Myocardial fatty acid metabolism in health and disease. Physiol. Rev. 2010, 90, 207–258. [Google Scholar] [CrossRef]
  176. Fillmore, N.; Levasseur, J.L.; Fukushima, A.; Wagg, C.S.; Wang, W.; Dyck, J.R.B.; Lopaschuk, G.D. Uncoupling of glycolysis from glucose oxidation accompanies the development of heart failure with preserved ejection fraction. Mol. Med. 2018, 24, 1–12. [Google Scholar] [CrossRef] [Green Version]
  177. Mori, J.; Basu, R.; McLean, B.A.; Das, S.K.; Zhang, L.; Patel, V.B. Agonist-induced hypertrophy and diastolic dysfunction are associated with selective reduction in glucose oxidation: A metabolic contribution to heart failure with normal ejection fraction. Circ. Heart Fail. 2012, 5, 493–503. [Google Scholar] [CrossRef] [Green Version]
  178. Briston, T.; Roberts, M.; Lewis, S.; Powney, B.; Staddon, J.M.; Szabadkai, G.; Duchen, M.R. Mitochondrial permeability transition pore: Sensitivity to opening and mechanistic dependence on substrate availability. Sci. Rep. 2017, 7, 10492. [Google Scholar] [CrossRef]
  179. Miranda-Silva, D.; Wüst, R.C.I.; Conceição, G.; Gonçalves-Rodrigues, P.; Gonçalves, N.; Gonçalves, A.; Kuster, D.W.D.; Leite-Moreira, A.F.; van der Velden, J.; Beleza, J.M.D.S.; et al. Disturbed cardiac mitochondrial and cytosolic calcium handling in a metabolic risk-related rat model of heart failure with preserved ejection fraction. Acta Physiol. 2019, 228, e13378. [Google Scholar] [CrossRef] [Green Version]
  180. Kraigher-Krainer, E.; Shah, A.M.; Gupta, D.K.; Santos, A.; Claggett, B.; Pieske, B.; Zile, M.; Voors, A.A.; Lefkowitz, M.P.; Packer, M.; et al. Impaired Systolic Function by Strain Imaging in Heart Failure with Preserved Ejection Fraction. J. Am. Coll. Cardiol. 2013, 63, 447–456. [Google Scholar] [CrossRef] [Green Version]
  181. Lee, D.I.; Zhu, G.; Sasaki, T.; Cho, G.-S.; Hamdani, N.; Holewinski, R.J.; Jo, S.-H.; Danner, T.; Zhang, M.; Rainer, P.P.; et al. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature 2015, 519, 472–476. [Google Scholar] [CrossRef]
  182. Wang, L.; Huang, Z.; Huang, W.; Chen, X.; Shan, P.; Zhong, P.; Khan, Z.; Wang, J.; Fang, Q.; Liang, G.; et al. Inhibition of epidermal growth factor receptor attenuates atherosclerosis via decreasing inflammation and oxidative stress. Sci. Rep. 2017, 7, 45917. [Google Scholar] [CrossRef] [Green Version]
  183. Zaha, V.; Young, L.H. AMP-Activated Protein Kinase Regulation and Biological Actions in the Heart. Circ. Res. 2012, 111, 800–814. [Google Scholar] [CrossRef] [Green Version]
  184. Jeon, S.-M. Regulation and function of AMPK in physiology and diseases. Exp. Mol. Med. 2016, 48, e245. [Google Scholar] [CrossRef]
  185. Reichert, K.; Pereira do Carmo, H.R.; Galluce Torina, A.; Diógenes de Carvalho, D.; Carvalho Sposito, A.; De Souza Vilarinho, K.A. Atorvastatin Improves Ventricular Remodeling after Myocardial Infarction by Interfering with Collagen Metabolism. PLoS ONE 2016, 11, e0166845. [Google Scholar] [CrossRef] [PubMed]
  186. Di Napoli, P.; Taccardi, A.A.; Barsotti, A. Long term cardioprotective action of trimetazidine and potential effect on the inflammatory process in patients with ischaemic dilated cardiomyopathy. Heart 2005, 91, 161–165. [Google Scholar] [CrossRef] [PubMed]
  187. Lan, F.; Weikel, K.A.; Cacicedo, J.M.; Ido, Y. Resveratrol-Induced AMP-Activated Protein Kinase Activation Is Cell-Type Dependent: Lessons from Basic Research for Clinical Application. Nutrients 2017, 9, 751. [Google Scholar] [CrossRef] [Green Version]
  188. Wang, L.; Quan, N.; Sun, W.; Chen, X.; Cates, C.; Rousselle, T.; Zhou, X.; Zhao, X.; Li, J. Cardiomyocyte-specific deletion of Sirt1 gene sensitizes myocardium to ischaemia and reperfusion injury. Cardiovasc. Res. 2018, 114, 805–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Li, H.; Xia, N.; Förstermann, U. Cardiovascular effects and molecular targets of resveratrol. Nitric Oxide 2012, 26, 102–110. [Google Scholar] [CrossRef]
  190. Senni, M.; Mcmurray, J.; Wachter, R.; McIntyre, H.F.; Reyes, A.; Majercak, I.; Andreka, P.; Shehova-Yankova, N.; Anand, I.; Yilmaz, M.B.; et al. Initiating sacubitril/valsartan (LCZ696) in heart failure: Results of TITRATION, a double-blind, randomized comparison of two uptitration regimens. Eur. J. Heart Fail. 2016, 18, 1193–1202. [Google Scholar] [CrossRef] [PubMed]
  191. Baruch, L.; Anand, I.; Cohen, I.S.; Ziesche, S.; Judd, D.; Cohn, J.N. Augmented Short- and Long-Term Hemodynamic and Hormonal Effects of an Angiotensin Receptor Blocker Added to Angiotensin Converting Enzyme Inhibitor Therapy in Patients with Heart Failure. Circulation 1999, 99, 2658–2664. [Google Scholar] [CrossRef] [Green Version]
  192. McMurray, J.J.; Packer, M.; Desai, A.S.; Gong, J.; Lefkowitz, M.P.; Rizkala, A.R.; Rouleau, J.L.; Shi, V.C.; Solomon, S.D.; Swedberg, K. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N. Engl. J. Med. 2014, 371, 132–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Mazza, A.; Torin, G.; Schiavon, L.; Rossetti, C.; Cuppini, S. Role of sacubitril/valsartan in the treatment of chronic heart failure with reduced ejection fraction in elderly hypertensives with comorbidity: From clinical trials to real world. J. Hypertens. 2021, 39 (Suppl. S1), e279. [Google Scholar] [CrossRef]
  194. Lund, L.H.; Claggett, B.; Liu, J.; Lam, C.S.; Jhund, P.; Rosano, G.M.; Swedberg, K.; Yusuf, S.; Granger, C.B.; Pfeffer, M.A.; et al. Heart failure with mid-range ejection fraction in CHARM: Characteristics, outcomes and effect of candesartan across the entire ejection fraction spectrum. Eur. J. Heart Fail. 2018, 20, 1230–1239. [Google Scholar] [CrossRef] [Green Version]
  195. D’Elia, E.; Iacovoni, A.; Vaduganathan, M.; Lorini, L.F.; Perlini, S.; Senni, M. Neprilysin inhibition in heart failure: Mechanisms and substrates beyond modulating natriuretic peptides. Eur. J. Heart Fail. 2017, 19, 710–717. [Google Scholar] [CrossRef]
  196. Solomon, S.D.; McMurray, J.J.V.; Anand, I.S.; Ge, J.; Lam, C.S.P.; Maggioni, A.P.; Martinez, F.; Packer, M.; Pfeffer, M.A.; Pieske, B.; et al. Angiotensin-Neprilysin Inhibition in Heart Failure with Preserved Ejection Fraction. N. Engl. J. Med. 2019, 381, 1609–1620. [Google Scholar] [CrossRef] [Green Version]
  197. Gori, M.; Volterrani, M.; Piepoli, M.; Senni, M. Angiotensin receptor–neprilysin inhibitor (ARNi): Clinical studies on a new class of drugs. Int. J. Cardiol. 2016, 226, 136–140. [Google Scholar] [CrossRef]
  198. Solomon, S.D.; Zile, M.; Pieske, B.; Voors, A.; Shah, A.; Kraigher-Krainer, E.; Shi, V.; Bransford, T.; Takeuchi, M.; Gong, J.; et al. The angiotensin receptor neprilysin inhibitor LCZ696 in heart failure with preserved ejection fraction: A phase 2 double-blind randomised controlled trial. Lancet 2012, 380, 1387–1395. [Google Scholar] [CrossRef]
  199. Jhund, P.S.; Claggett, B.; Packer, M.; Zile, M.; Voors, A.A.; Pieske, B.; Lefkowitz, M.; Shi, V.; Bransford, T.; McMurray, J.J.V.; et al. Independence of the blood pressure lowering effect and efficacy of the angiotensin receptor neprilysin inhibitor, LCZ696, in patients with heart failure with preserved ejection fraction: An analysis of the PARAMOUNT trial. Eur. J. Heart Fail. 2014, 16, 671–677. [Google Scholar] [CrossRef] [Green Version]
  200. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E.; Drazner, M.H. 2013 ACCF/AHA guideline for the management of heart failure: Executive summary: A report of the American College of Cardiology Foundation/American Heart Association Task Force on practice guidelines. Circulation 2013, 128, 1810–1852. [Google Scholar] [CrossRef]
  201. Tsao, C.W.; Lyass, A.; Enserro, D.; Larson, M.G.; Ho, J.E.; Kizer, J.R.; Gottdiener, J.S.; Psaty, B.M.; Vasan, R.S. Temporal Trends in the Incidence of and Mortality Associated with Heart Failure with Preserved and Reduced Ejection Fraction. JACC Heart Fail. 2018, 6, 678–685. [Google Scholar] [CrossRef]
  202. Calderon-Montano, J.M.; Burgos-Moron, E.; Perez-Guerrero, C.; Lopez-Lazaro, M. A review on the dietary flavonoid kaempferol. Mini Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef] [PubMed]
  203. Arias, A.K.S.; Martin-Romero, F.J.; Gutiérrez-Merino, C. Kaempferol blocks oxidative stress in cerebellar granule cells and reveals a key role for reactive oxygen species production at the plasma membrane in the commitment to apoptosis. Free Radic. Biol. Med. 2004, 37, 48–61. [Google Scholar] [CrossRef]
  204. Du, Y.; Han, J.; Zhang, H.; Xu, J.; Jiang, L.; Ge, W. Kaempferol Prevents Against Ang II-induced Cardiac Remodeling Through Attenuating Ang II-induced Inflammation and Oxidative Stress. J. Cardiovasc. Pharmacol. 2019, 74, 326–335. [Google Scholar] [CrossRef] [PubMed]
  205. Kelso, G.F.; Porteous, C.M.; Coulter, C.V.; Hughes, G.; Porteous, W.K.; Ledgerwood, E.C. Selective targeting of a redox-active ubiquinone to mitochondria within cells: Antioxidant and antiapoptotic properties. J. Biol. Chem. 2001, 276, 4588–4596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Murphy, M. Understanding and preventing mitochondrial oxidative damage. Biochem. Soc. Trans. 2016, 44, 1219–1226. [Google Scholar] [CrossRef]
  207. Ribeiro Junior, R.F.; Dabkowski, E.R.; Shekar, K.C.; O’Connell, K.A.; Hecker, P.A.; Murphy, M.P. MitoQ improves mitochondrial dysfunction in heart failure induced by pressure overload. Free Radic. Biol. Med. 2018, 117, 18–29. [Google Scholar] [CrossRef]
  208. Trnka, J.; Blaikie, F.H.; Smith, R.A.; Murphy, M.P. A mitochondria-targeted nitroxide is reduced to its hydroxylamine by ubiquinol in mitochondria. Free Radic. Biol. Med. 2008, 44, 1406–1419. [Google Scholar] [CrossRef]
  209. Tauskela, J.S. MitoQ—A mitochondria-targeted antioxidant. IDrugs 2007, 10, 399–412. [Google Scholar]
  210. Chen, W.; Guo, C.; Jia, Z.; Wang, J.; Xia, M.; Li, C.; Li, M.; Yin, Y.; Tang, X.; Chen, T.; et al. Inhibition of Mitochondrial ROS by MitoQ Alleviates White Matter Injury and Improves Outcomes after Intracerebral Haemorrhage in Mice. Oxid. Med. Cell. Longev. 2020, 2020, 8285065. [Google Scholar] [CrossRef]
  211. Fuentes, E.; Araya-Maturana, R.; Urra, F.A. Regulation of mitochondrial function as a promising target in platelet activation-related diseases. Free Radic. Biol. Med. 2019, 136, 172–182. [Google Scholar] [CrossRef]
  212. James, A.M.; Sharpley, M.S.; Manas, A.-R.B.; Frerman, F.E.; Hirst, J.; Smith, R.A.J.; Murphy, M.P. Interaction of the Mitochondria-targeted Antioxidant MitoQ with Phospholipid Bilayers and Ubiquinone Oxidoreductases. J. Biol. Chem. 2007, 282, 14708–14718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Hirsch, G.A.; Bottomley, P.A.; Gerstenblith, G.; Weiss, R.G. Allopurinol Acutely Increases Adenosine Triphospate Energy Delivery in Failing Human Hearts. J. Am. Coll. Cardiol. 2012, 59, 802–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Cingolani, H.E.; Plastino, J.A.; Escudero, E.M.; Mangal, B.; Brown, J.; Pérez, N.G. The Effect of Xanthine Oxidase Inhibition Upon Ejection Fraction in Heart Failure Patients: La Plata Study. J. Card. Fail. 2006, 12, 491–498. [Google Scholar] [CrossRef]
  215. Doehner, W.; Schoene, N.; Rauchhaus, M.; Leyva-Leon, F.; Pavitt, D.V.; Reaveley, D.A. Effects of xanthine oxidase inhibition with allopurinol on endothelial function and peripheral blood flow in hyperuricemic patients with chronic heart failure: Results from 2 placebo-controlled studies. Circulation 2002, 105, 2619–2624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Ratchford, S.M.; Clifton, H.L.; Gifford, J.R.; LaSalle, D.T.; Thurston, T.S.; Bunsawat, K. Impact of acute antioxidant administration on inflammation and vascular function in heart failure with preserved ejection fraction. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2019, 317, R607–R614. [Google Scholar] [CrossRef]
  217. Hare, J.M.; Mangal, B.; Brown, J.; Fisher, C.; Freudenberger, R.; Colucci, W.; Mann, D.; Liu, P.; Givertz, M.M.; Schwarz, R.P. Impact of Oxypurinol in Patients with Symptomatic Heart Failure: Results of the OPT-CHF Study. J. Am. Coll. Cardiol. 2008, 51, 2301–2309. [Google Scholar] [CrossRef] [Green Version]
  218. Givertz, M.M.; Anstrom, K.J.; Redfield, M.M.; Deswal, A.; Haddad, H.; Butler, J. Effects of Xanthine Oxidase Inhibition in Hyperuricemic Heart Failure Patients: The Xanthine Oxidase Inhibition for Hyperuricemic Heart Failure Patients (EXACT-HF) Study. Circulation 2015, 131, 1763–1771. [Google Scholar] [CrossRef] [Green Version]
  219. Yokota, T.; Fukushima, A.; Kinugawa, S.; Okumura, T.; Murohara, T.; Tsutsui, H. Randomized Trial of Effect of Urate-Lowering Agent Febuxostat in Chronic Heart Failure Patients with Hyperuricemia (LEAF-CHF). Int. Heart J. 2018, 59, 976–982. [Google Scholar] [CrossRef] [Green Version]
  220. Carnicer, R.; Duglan, D.; Ziberna, K.; Recalde, A.; Reilly, S.; Simon, J.N.; Mafrici, S.; Arya, R.; Roselló-Lletí, E.; Chuaiphichai, S.; et al. BH4 Increases nNOS Activity and Preserves Left Ventricular Function in Diabetes. Circ. Res. 2021, 128, 585–601. [Google Scholar] [CrossRef]
  221. Bunsawat, K.; Ratchford, S.M.; Alpenglow, J.K.; Park, S.H.; Jarrett, C.L.; Stehlik, J.; Drakos, S.G.; Richardson, R.S.; Wray, D.W. Chronic antioxidant administration restores macrovascular function in patients with heart failure with reduced ejection fraction. Exp. Physiol. 2020, 105, 1384–1395. [Google Scholar] [CrossRef]
  222. Dey, S.; DeMazumder, D.; Sidor, A.; Brian Foster, D.; O’Rourke, B. Mitochondrial ROS Drive Sudden Cardiac Death and Chronic Proteome Remodeling in Heart Failure. Circ. Res. 2018, 123, 356–371. [Google Scholar] [CrossRef]
  223. Hoshino, A.; Okawa, Y.; Ariyoshi, M.; Kaimoto, S.; Uchihashi, M.; Fukai, K.; Iwai-Kanai, E.; Matoba, S. Oxidative Post-Translational Modifications Develop LONP1 Dysfunction in Pressure Overload Heart Failure. Circ. Heart Fail. 2014, 7, 500–509. [Google Scholar] [CrossRef] [Green Version]
  224. Shiomi, T.; Tsutsui, H.; Matsusaka, H.; Murakami, K.; Hayashidani, S.; Ikeuchi, M.; Wen, J.; Kubota, T.; Utsumi, H.; Takeshita, A. Overexpression of Glutathione Peroxidase Prevents Left Ventricular Remodeling and Failure After Myocardial Infarction in Mice. Circulation 2004, 109, 544–549. [Google Scholar] [CrossRef] [Green Version]
  225. Wang, P.; Chen, H.; Qin, H.; Sankarapandi, S.; Becher, M.W.; Wong, P.C.; Zweier, J.L. Overexpression of human copper, zinc-superoxide dismutase (SOD1) prevents postischemic injury. Proc. Natl. Acad. Sci. USA 1998, 95, 4556–4560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Qin, F.; Lennon-Edwards, S.; Lancel, S.; Biolo, A.; Siwik, D.A.; Pimentel, D.R. Cardiac-specific overexpression of catalase identifies hydrogen peroxide-dependent and independent-phases of myocardial remodeling, and prevents the progression to overt heart failure in Gαq-overexpressing transgenic mice. Circ. Heart Fail. 2010, 3, 306–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Maack, C.; Eschenhagen, T.; Hamdani, N.; Heinze, F.R.; Lyon, A.R.; Manstein, D.J. Treatments targeting inotropy: A position paper of the Committees on Translational Research and Acute Heart Failure of the Heart Failure Association of the European Society of Cardiology. Eur. Heart J. 2019, 40, 3626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Kaludercic, N.; Carpi, A.; Menabò, R.; Di Lisa, F.; Paolocci, N. Monoamine oxidases (MAO) in the pathogenesis of heart failure and ischemia/reperfusion injury. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 1323–1332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Bianchi, P.; Pimentel, D.R.; Murphy, M.; Colucci, W.; Parini, A. A new hypertrophic mechanism of serotonin in cardiac myocytes: Receptor-independent ROS generation. FASEB J. 2005, 19, 1–15. [Google Scholar] [CrossRef]
  230. Kaludercic, N.; Takimoto, E.; Nagayama, T.; Feng, N.; Lai, E.W.; Bedja, D.; Chen, K.; Gabrielson, K.L.; Blakely, R.D.; Shih, J.C.; et al. Monoamine Oxidase A–Mediated Enhanced Catabolism of Norepinephrine Contributes to Adverse Remodeling and Pump Failure in Hearts with Pressure Overload. Circ. Res. 2010, 106, 193–202. [Google Scholar] [CrossRef] [PubMed]
  231. Di Lisa, F.; Kaludercic, N.; Carpi, A.; Menabò, R.; Giorgio, M. Mitochondrial pathways for ROS formation and myocardial injury: The relevance of p66(Shc) and monoamine oxidase. Basic Res. Cardiol. 2009, 104, 131–139. [Google Scholar] [CrossRef]
  232. Carpi, A.; Menabò, R.; Kaludercic, N.; Pelicci, P.; Di Lisa, F.; Giorgio, M. The cardioprotective effects elicited by p66Shc ablation demonstrate the crucial role of mitochondrial ROS formation in ischemia/reperfusion injury. Biochim. Biophys. Acta Bioenerg. 2009, 1787, 774–780. [Google Scholar] [CrossRef]
  233. Villeneuve, C.; Guilbeau-Frugier, C.; Sicard, P.; Lairez, O.; Ordener, C.; Duparc, T.; De Paulis, D.; Couderc, B.; Spreux-Varoquaux, O.; Tortosa, F.; et al. p53-PGC-1α Pathway Mediates Oxidative Mitochondrial Damage and Cardiomyocyte Necrosis Induced by Monoamine Oxidase-A Upregulation: Role in Chronic Left Ventricular Dysfunction in Mice. Antioxid. Redox Signal. 2013, 18, 5–18. [Google Scholar] [CrossRef] [Green Version]
  234. Umbarkar, P.; Singh, S.; Arkat, S.; Bodhankar, S.; Lohidasan, S.; Sitasawad, S.L. Monoamine oxidase-A is an important source of oxidative stress and promotes cardiac dysfunction, apoptosis, and fibrosis in diabetic cardiomyopathy. Free Radic. Biol. Med. 2015, 87, 263–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Kaludercic, N.; Carpi, A.; Nagayama, T.; Sivakumaran, V.; Zhu, G.; Lai, E.W.; Bedja, D.; De Mario, A.; Chen, K.; Gabrielson, K.L.; et al. Monoamine Oxidase B Prompts Mitochondrial and Cardiac Dysfunction in Pressure Overloaded Hearts. Antioxid. Redox Signal. 2014, 20, 267–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Manni, M.E.; Rigacci, S.; Borchi, E.; Bargelli, V.; Miceli, C.; Giordano, C.; Raimondi, L.; Nediani, C. Monoamine Oxidase Is Overactivated in Left and Right Ventricles from Ischemic Hearts: An Intriguing Therapeutic Target. Oxidative Med. Cell. Longev. 2016, 2016, 4375418. [Google Scholar] [CrossRef]
  237. Sharma, A.; Fonarow, G.C.; Butler, J.; Ezekowitz, J.A.; Felker, G.M. Coenzyme Q10 and Heart Failure: A State-of-the-Art Review. Circ. Heart Fail. 2016, 9, e002639. [Google Scholar] [CrossRef] [PubMed]
  238. Pierce, J.D.; Mahoney, D.E.; Hiebert, J.B.; Thimmesch, A.R.; Diaz, F.J.; Smith, C.; Shen, Q.; Mudaranthakam, D.P.; Clancy, R.L. Study protocol, randomized controlled trial: Reducing symptom burden in patients with heart failure with preserved ejection fraction using ubiquinol and/or D-ribose. BMC Cardiovasc. Disord. 2018, 18, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Bentinger, M.; Tekle, M.; Dallner, G. Coenzyme Q—Biosynthesis and functions. Biochem. Biophys. Res. Commun. 2010, 396, 74–79. [Google Scholar] [CrossRef]
  240. McCarthy, S.; Somayajulu, M.; Sikorska, M.; Borowy-Borowski, H.; Pandey, S. Paraquat induces oxidative stress and neuronal cell death; neuroprotection by water-soluble Coenzyme Q10. Toxicol. Appl. Pharmacol. 2004, 201, 21–31. [Google Scholar] [CrossRef]
  241. Hathcock, J.N.; Shao, A. Risk assessment for coenzyme Q10 (Ubiquinone). Regul. Toxicol. Pharmacol. 2006, 45, 282–288. [Google Scholar] [CrossRef]
  242. Von Hardenberg, A.; Maack, C. Mitochondrial Therapies in Heart Failure. Handb. Exp. Pharmacol. 2017, 243, 491–514. [Google Scholar] [PubMed]
  243. Mortensen, S.A.; Rosenfeldt, F.; Kumar, A.; Dolliner, P.; Filipiak, K.J.; Pella, D. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: Results from Q-SYMBIO: A randomized double-blind trial. JACC Heart Fail. 2014, 2, 641–649. [Google Scholar] [CrossRef]
  244. Szeto, H.H. First-in-class cardiolipin-protective compound as a therapeutic agent to restore mitochondrial bioenergetics. J. Cereb. Blood Flow Metab. 2014, 171, 2029–2050. [Google Scholar] [CrossRef] [Green Version]
  245. Bertero, E.; Kutschka, I.; Maack, C.; Dudek, J. Cardiolipin remodeling in Barth syndrome and other hereditary cardiomyopathies. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165803. [Google Scholar] [CrossRef] [PubMed]
  246. 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]
  247. Yin, H.; Zhu, M. Free radical oxidation of cardiolipin: Chemical mechanisms, detection and implication in apoptosis, mitochondrial dysfunction and human diseases. Free Radic. Res. 2012, 46, 959–974. [Google Scholar] [CrossRef]
  248. Sabbah, H.N.; Gupta, R.C.; Kohli, S.; Wang, M.; Hachem, S.; Zhang, K. Chronic Therapy with Elamipretide (MTP-131), a Novel Mitochondria-Targeting Peptide, Improves Left Ventricular and Mitochondrial Function in Dogs with Advanced Heart Failure. Circ. Heart Fail. 2016, 9, e002206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Chatfield, K.C.; Sparagna, G.C.; Chau, S.; Phillips, E.K.; Ambardekar, A.V.; Aftab, M.; Mitchell, M.B.; Sucharov, C.C.; Miyamoto, S.D.; Stauffer, B.L. Elamipretide Improves Mitochondrial Function in the Failing Human Heart. JACC Basic Transl. Sci. 2019, 4, 147–157. [Google Scholar] [CrossRef]
  250. Brown, D.A.; Hale, S.L.; Baines, C.; del Rio, C.; Hamlin, R.L.; Yueyama, Y.; Kijtawornrat, A.; Yeh, S.T.; Frasier, C.R.; Stewart, L.M.; et al. Reduction of Early Reperfusion Injury with the Mitochondria-Targeting Peptide Bendavia. J. Cardiovasc. Pharmacol. Ther. 2013, 19, 121–132. [Google Scholar] [CrossRef] [Green Version]
  251. Daubert, M.A.; Yow, E.; Dunn, G.; Marchev, S.; Barnhart, H.; Douglas, P.S.; O’Connor, C.; Goldstein, S.; Udelson, J.E.; Sabbah, H.N. Novel Mitochondria-Targeting Peptide in Heart Failure Treatment: A Randomized, Placebo-Controlled Trial of Elamipretide. Circ. Heart Fail. 2017, 10, e004389. [Google Scholar] [CrossRef] [PubMed]
  252. Butler, J.; Khan, M.S.; Anker, S.D.; Fonarow, G.C.; Kim, R.J.; Nodari, S.; O’Connor, C.M.; Pieske, B.; Pieske-Kraigher, E.; Sabbah, H.N.; et al. Effects of Elamipretide on Left Ventricular Function in Patients with Heart Failure with Reduced Ejection Fraction: The PROGRESS-HF Phase 2 Trial. J. Card. Fail. 2020, 26, 429–437. [Google Scholar] [CrossRef] [PubMed]
  253. Gevaert, A.B.; Boen, J.R.; Segers, V.F.; Van Craenenbroeck, E.M. Heart Failure with Preserved Ejection Fraction: A Review of Cardiac and Noncardiac Pathophysiology. Front. Physiol. 2019, 10, 638. [Google Scholar] [CrossRef] [PubMed]
  254. Lin, E.Q.; Irvine, J.C.; Cao, A.H.; Alexander, A.E.; Love, J.E.; Patel, R.; McMullen, J.R.; Kaye, D.M.; Kemp-Harper, B.K.; Ritchie, R.H. Nitroxyl (HNO) Stimulates Soluble Guanylyl Cyclase to Suppress Cardiomyocyte Hypertrophy and Superoxide Generation. PLoS ONE 2012, 7, e34892. [Google Scholar] [CrossRef] [Green Version]
  255. Sabbah, H.N.; Tocchetti, C.G.; Wang, M.; Daya, S.; Gupta, R.C.; Tunin, R.S.; Mazhari, R.; Takimoto, E.; Paolocci, N.; Cowart, D.; et al. Nitroxyl (HNO). Circ. Heart Fail. 2013, 6, 1250–1258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Gheorghiade, M.; Greene, S.J.; Filippatos, G.; Erdmann, E.; Ferrari, R.; Levy, P.D.; Maggioni, A.; Nowack, C.; Mebazaa, A.; COMPOSE Investigators and Coordinators. Cinaciguat, a soluble guanylate cyclase activator: Results from the randomized, controlled, phase IIb COMPOSE programme in acute heart failure syndromes. Eur. J. Heart Fail. 2012, 14, 1056–1066. [Google Scholar] [CrossRef] [PubMed]
  257. Ghofrani, H.-A.; Galiè, N.; Grimminger, F.; Grünig, E.; Humbert, M.; Jing, Z.-C. Riociguat for the treatment of pulmonary arterial hypertension. N. Engl. J. Med. 2013, 369, 330–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Pieske, B.; Butler, J.; Filippatos, G.; Lam, C.; Maggioni, A.P.; Ponikowski, P.; Shah, S.; Solomon, S.; Kraigher-Krainer, E.; Samano, E.T.; et al. Rationale and design of the SOluble guanylate Cyclase stimulatoR in heArT failurE Studies (SOCRATES). Eur. J. Heart Fail. 2014, 16, 1026–1038. [Google Scholar] [CrossRef]
  259. Zhazykbayeva, S.; Pabel, S.; Mügge, A.; Sossalla, S.; Hamdani, N. The molecular mechanisms associated with the physiological responses to inflammation and oxidative stress in cardiovascular diseases. Biophys. Rev. 2020, 12, 947–968. [Google Scholar] [CrossRef] [PubMed]
  260. Redfield, M.M.; Borlaug, B.A.; Lewis, G.D.; Mohammed, S.F.; Semigran, M.J.; Lewinter, M.M.; Deswal, A.; Hernandez, A.F.; Lee, K.L.; Braunwald, E.; et al. PhosphdiesteRasE-5 Inhibition to Improve CLinical Status and EXercise Capacity in Diastolic Heart Failure (RELAX) trial: Rationale and design. Circ. Heart Fail. 2012, 5, 653–659. [Google Scholar] [CrossRef] [Green Version]
  261. Corbin, J.D. Mechanisms of action of PDE5 inhibition in erectile dysfunction. Int. J. Impot. Res. 2004, 16, S4–S7. [Google Scholar] [CrossRef] [Green Version]
  262. Lewis, G.D.; Lachmann, J.; Camuso, J.; Lepore, J.J.; Shin, J.; Martinovic, M.E.; Systrom, D.M.; Bloch, K.D.; Semigran, M.J. Sildenafil Improves Exercise Hemodynamics and Oxygen Uptake in Patients with Systolic Heart Failure. Circulation 2007, 115, 59–66. [Google Scholar] [CrossRef] [PubMed]
  263. Guazzi, M.; Vicenzi, M.; Arena, R.; Guazzi, M.D. PDE5 inhibition with sildenafil improves left ventricular diastolic function, cardiac geometry, and clinical status in patients with stable systolic heart failure: Results of a 1-year, prospective, randomized, placebo-controlled study. Circ. Heart Fail. 2011, 4, 8–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Guazzi, M.; Vicenzi, M.; Arena, R. Phosphodiesterase 5 inhibition with sildenafil reverses exercise oscillatory breathing in chronic heart failure: A long-term cardiopulmonary exercise testing placebo-controlled study. Eur. J. Heart Fail. 2012, 14, 82–90. [Google Scholar] [CrossRef] [PubMed]
  265. Guazzi, M.; Vicenzi, M.; Arena, R.; Guazzi, M.D. Pulmonary hypertension in heart failure with preserved ejection fraction: A target of phosphodiesterase-5 inhibition in a 1-year study. Circulation 2011, 124, 164–174. [Google Scholar] [CrossRef]
  266. Pinilla-Vera, M.; Hahn, V.S.; Kass, D.A. Leveraging Signaling Pathways to Treat Heart Failure with Reduced Ejection Fraction. Circ. Res. 2019, 124, 1618–1632. [Google Scholar] [CrossRef]
  267. Tocchetti, C.G.; Stanley, B.A.; Murray, C.I.; Sivakumaran, V.; Donzelli, S.; Mancardi, D.; Pagliaro, P.; Gao, W.D.; Van Eyk, J.; Kass, D.A.; et al. Playing with cardiac “redox switches”: The “HNO way” to modulate cardiac function. Antioxid. Redox Signal 2011, 114, 1687–1698. [Google Scholar] [CrossRef] [Green Version]
  268. Tita, C.; Gilbert, E.M.; Van Bakel, A.B.; Grzybowski, J.; Haas, G.J.; Jarrah, M.; Dunlap, S.H.; Gottlieb, S.S.; Klapholz, M.; Patel, P.C.; et al. A Phase 2a dose-escalation study of the safety, tolerability, pharmacokinetics and haemodynamic effects of BMS-986231 in hospitalized patients with heart failure with reduced ejection fraction. Eur. J. Heart Fail. 2017, 19, 1321–1332. [Google Scholar] [CrossRef] [Green Version]
  269. Hartman, J.C.; del Rio, C.; Reardon, J.E.; Zhang, K.; Sabbah, H.N. Intravenous Infusion of the Novel HNO Donor BMS-986231 Is Associated with Beneficial Inotropic, Lusitropic, and Vasodilatory Properties in 2 Canine Models of Heart Failure. JACC Basic Transl. Sci. 2018, 3, 625–638. [Google Scholar] [CrossRef]
  270. Felker, G.M.; Borentain, M.; Cleland, J.G.; DeSouza, M.M.; Kessler, P.D.; O’Connor, C.M.; Seiffert, D.; Teerlink, J.R.; Voors, A.A.; McMurray, J.J. Rationale and design for the development of a novel nitroxyl donor in patients with acute heart failure. Eur. J. Heart Fail. 2019, 21, 1022–1031. [Google Scholar] [CrossRef]
  271. Kemp-Harper, B.; Schmidt, H.H.H.W. cGMP in the vasculature. Handb. Exp. Pharmacol. 2009, 191, 447–467. [Google Scholar]
  272. Ivanova, J.; Salama, G.; Clancy, R.M.; Schor, N.F.; Nylander, K.D.; Stoyanovsky, D.A. Formation of nitroxyl and hydroxyl radical in solutions of sodium trioxodinitrate: Effects of pH and cytotoxicity. J. Biol. Chem. 2003, 278, 42761–42768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Shiva, S.; Crawford, J.H.; Ramachandran, A.; Ceaser, E.K.; Hillson, T.; Brookes, P.; Patel, R.P.; Darley-Usmar, V.M. Mechanisms of the interaction of nitroxyl with mitochondria. Biochem. J. 2004, 379, 359–366. [Google Scholar] [CrossRef] [PubMed]
  274. Tocchetti, C.G.; Wang, W.; Froehlich, J.P.; Huke, S.; Aon, M.A.; Wilson, G.M.; Di Benedetto, G.; O’Rourke, B.; Gao, W.D.; Wink, D.A.; et al. Nitroxyl Improves Cellular Heart Function by Directly Enhancing Cardiac Sarcoplasmic Reticulum Ca2+ Cycling. Circ. Res. 2007, 100, 96–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Paolocci, N.; Saavedra, W.F.; Miranda, K.M.; Martignani, C.; Isoda, T.; Hare, J.M.; Espey, M.G.; Fukuto, J.M.; Feelisch, M.; Wink, D.A.; et al. Nitroxyl anion exerts redox-sensitive positive cardiac inotropy in vivo by calcitonin gene-related peptide signaling. Proc. Natl. Acad. Sci. USA 2001, 98, 10463–10468. [Google Scholar] [CrossRef] [Green Version]
  276. Paolocci, N.; Katori, T.; Champion, H.C.; St John, M.E.; Miranda, K.M.; Fukuto, J.M. Positive inotropic and lusitropic effects of HNO/NO- in failing hearts: Independence from beta-adrenergic signaling. Proc. Natl. Acad. Sci. USA 2003, 100, 5537–5542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Ziolo, M.T.; Kohr, M.J.; Kaludercic, N.; Tocchetti, C.G.; Gao, W.D.; A Kass, D.; Janssen, P.M.L.; Paolocci, N. Nitroxyl enhances myocyte Ca2 transients by exclusively targeting SR Ca2+-cycling. Front. Biosci. 2010, E2, 614–626. [Google Scholar] [CrossRef] [Green Version]
  278. Dai, T.; Tian, Y.; Tocchetti, C.G.; Katori, T.; Murphy, A.M.; Kass, D.A.; Paolocci, N.; Gao, W.D. Nitroxyl increases force development in rat cardiac muscle. J. Physiol. 2007, 580, 951–960. [Google Scholar] [CrossRef]
  279. Tocchetti, C.G.; Mercurio, V.; Maack, C. The multifaceted mechanisms of nitroxyl in heart failure: Inodilator or “only” vasodilator? Eur. J. Heart Fail. 2021, 123, 1156–1159. [Google Scholar] [CrossRef]
  280. Hare, J.M. Nitroso–Redox Balance in the Cardiovascular System. N. Engl. J. Med. 2004, 351, 2112–2114. [Google Scholar] [CrossRef] [Green Version]
  281. Münzel, T.; Daiber, A.; Gori, T. More answers to the still unresolved question of nitrate tolerance. Eur. Heart J. 2013, 34, 2666–2673. [Google Scholar] [CrossRef]
  282. Daiber, A.; Oelze, M.; Coldewey, M.; Kaiser, K.; Huth, C.; Schildknecht, S.; Bachschmid, M.M.; Nazirisadeh, Y.; Ullrich, V.; Mülsch, A.; et al. Hydralazine is a powerful inhibitor of peroxynitrite formation as a possible explanation for its beneficial effects on prognosis in patients with congestive heart failure. Biochem. Biophys. Res. Commun. 2005, 338, 1865–1874. [Google Scholar] [CrossRef]
  283. Münzel, T.; Kurz, S.; Rajagopalan, S.; Thoenes, M.; Berrington, W.R.; A Thompson, J.; A Freeman, B.; Harrison, D.G. Hydralazine prevents nitroglycerin tolerance by inhibiting activation of a membrane-bound NADH oxidase. A new action for an old drug. J. Clin. Investig. 1996, 98, 1465–1470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Münzel, T.; Steven, S.; Daiber, A. Organic nitrates: Update on mechanisms underlying vasodilation, tolerance and endothelial dysfunction. Vasc. Pharmacol. 2014, 63, 105–113. [Google Scholar] [CrossRef] [PubMed]
  285. Sandner, P. From molecules to patients: Exploring the therapeutic role of soluble guanylate cyclase stimulators. Biol. Chem. 2018, 399, 679–690. [Google Scholar] [CrossRef] [PubMed]
  286. Taylor, A.L.; Ziesche, S.; Yancy, C.; Carson, P.; D’Agostino, R.; Ferdinand, K.; Taylor, M.; Adams, K.; Sabolinski, M.; Worcel, M.; et al. Combination of Isosorbide Dinitrate and Hydralazine in Blacks with Heart Failure. N. Engl. J. Med. 2004, 351, 2049–2057. [Google Scholar] [CrossRef] [Green Version]
  287. Evgenov, O.V.; Pacher, P.; Schmidt, P.M.; Haskó, G.; Schmidt, H.H.H.W.; Stasch, J.P. NO-independent stimulators and activators of soluble guanylate cyclase: Discovery and therapeutic potential. Nat. Rev. Drug Discov. 2006, 5, 755–768. [Google Scholar] [CrossRef] [Green Version]
  288. Montfort, W.R.; Wales, J.A.; Weichsel, A. Structure and Activation of Soluble Guanylyl Cyclase, the Nitric Oxide Sensor. Antioxid. Redox Signal. 2017, 26, 107–121. [Google Scholar] [CrossRef]
  289. Breitenstein, S.; Roessig, L.; Sandner, P.; Lewis, K.S. Novel sGC Stimulators and sGC Activators for the Treatment of Heart Failure. Handb. Exp. Pharmacol. 2017, 243, 225–247. [Google Scholar] [CrossRef]
  290. Follmann, M.; Ackerstaff, J.; Redlich, G.; Wunder, F.; Lang, D.; Kern, A.; Fey, P.; Griebenow, N.; Kroh, W.; Becker-Pelster, E.-M.; et al. Discovery of the Soluble Guanylate Cyclase Stimulator Vericiguat (BAY 1021189) for the Treatment of Chronic Heart Failure. J. Med. Chem. 2017, 60, 5146–5161. [Google Scholar] [CrossRef] [Green Version]
  291. Premer, C.; Kanelidis, A.J.; Hare, J.M.; Schulman, I.H. Rethinking Endothelial Dysfunction as a Crucial Target in Fighting Heart Failure. Mayo Clinic Proc. Innov. Qual. Outcomes 2019, 3, 1–13. [Google Scholar] [CrossRef] [Green Version]
  292. Kolijn, D.; Kovács, Á.; Herwig, M.; Lódi, M.; Sieme, M.; Alhaj, A.; Sandner, P.; Papp, Z.; Reusch, P.H.; Haldenwang, P.; et al. Enhanced Cardiomyocyte Function in Hypertensive Rats with Diastolic Dysfunction and Human Heart Failure Patients After Acute Treatment with Soluble Guanylyl Cyclase (sGC) Activator. Front. Physiol. 2020, 11, 345. [Google Scholar] [CrossRef]
  293. Archer, S.L.; Huang, J.M.; Hampl, V.; Nelson, D.P.; Shultz, P.J.; Weir, E.K. Nitric oxide and cGMP cause vasorelaxation by activation of a charybdotoxin-sensitive K channel by cGMP-dependent protein kinase. Proc. Natl. Acad. Sci. USA 1994, 91, 7583–7587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Buys, E.; Zimmer, D.; Chickering, J.; Graul, R.; Chien, Y.; Profy, A.; Hadcock, J.; Masferrer, J.; Milne, G. Discovery and development of next generation sGC stimulators with diverse multidimensional pharmacology and broad therapeutic potential. Nitric Oxide 2018, 78, 72–80. [Google Scholar] [CrossRef]
  295. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Rocca, H.-P.B.; 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]
  296. Lang, N.N.; Ahmad, F.A.; Cleland, J.G.; O’Connor, C.M.; Teerlink, J.R.; Voors, A.A.; Taubel, J.; Hodes, A.R.; Anwar, M.; Karra, R.; et al. Haemodynamic effects of the nitroxyl donor cimlanod (BMS -986231) in chronic heart failure: A randomized trial. Eur. J. Heart Fail. 2021, 23, 1147–1155. [Google Scholar] [CrossRef] [PubMed]
  297. Gheorghiade, M.; Greene, S.J.; Butler, J.; Filippatos, G.; Lam, C.S.P.; Maggioni, A.P.; Ponikowski, P.; Shah, S.J.; Solomon, S.D.; Kraigher-Krainer, E.; et al. Effect of vericiguat, a soluble guanylate cyclase stimulator, on natriuretic peptide levels in patients with worsening chronic heart failure and reduced ejection fraction: The SOCRATES-REDUCED randomized trial. JAMA 2015, 314, 2251–2262. [Google Scholar] [CrossRef] [PubMed]
  298. Pieske, B.; Maggioni, A.P.; Lam, C.S.; Pieske-Kraigher, E.; Filippatos, G.; Butler, J.; Ponikowski, P.; Shah, S.; Solomon, S.D.; Scalise, A.-V.; et al. Vericiguat in patients with worsening chronic heart failure and preserved ejection fraction: Results of the SOluble guanylate Cyclase stimulatoR in heArT failurE patientS with PRESERVED EF (SOCRATES-PRESERVED) study. Eur. Heart J. 2017, 38, 1119–1127. [Google Scholar] [CrossRef]
  299. Armstrong, P.W.; Lam, C.S.P.; Anstrom, K.J.; Ezekowitz, J.; Hernandez, A.F.; O’Connor, C.M. Effect of Vericiguat vs Placebo on Quality of Life in Patients with Heart Failure and Preserved Ejection Fraction: The VITALITY-HFpEF Randomized Clinical Trial. JAMA 2020, 324, 1512–1521. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Reactive oxygen species. O2•−-superoxide anion, OH-hydroxyl radical, H2O2-hydrogen peroxide, HOO-hydroperoxyle radical, H2O-water.
Figure 1. Reactive oxygen species. O2•−-superoxide anion, OH-hydroxyl radical, H2O2-hydrogen peroxide, HOO-hydroperoxyle radical, H2O-water.
Biomedicines 10 00602 g001
Figure 2. Harmful effects of ROS in cardiomyocytes (created with BioRender.com on 11 February 2022).
Figure 2. Harmful effects of ROS in cardiomyocytes (created with BioRender.com on 11 February 2022).
Biomedicines 10 00602 g002
Figure 3. Enzymes involved in ROS production in cardiomyocytes and fibroblasts (created with BioRender.com on 7 February 2022). NOX2 is presented to be activated by endothelin and angiotensin II [58], by cytokines [59] and mechanical stress [60]. Increased NOX2 activation leads to cytoskeletal dysfunction in patients with CHF [61]. It was discovered that superoxide anions, produced by NOX, can oxidize and degrade hydrobiopterin-4 (BH4) leading to NOS uncoupling [62]. Nitric oxide synthase 3 (NOS3) uncoupling was observed in myocardium exposed to chronic pressure load. NOS3 catalyzes nitric oxide (NO) synthesis under physiological conditions. NO has an antihypertrophic effect. However, NOS3 is uncoupled with pressure load, and this, in turn, leads to reduction in tetrahydrobiopterin-4 concentration, increase in ROS production, and, as a consequence, to cardiomyocyte hypertrophy [63]. It was also shown that increase in ROS activates MAPK, leading to increased expression of proteins, such as ERK, JNK and P38, which are related to cardiomyocyte hypertrophy [64] (Figure 3). What is more, NOX-derived ROS may activate XOR [65]. Additionally, angiotensin II-induced signaling and isolated cardiomyocyte hypertrophy are dependent on NOX2 [66]. GTP-binding protein Rac-1 (involved in NOX activation), as described in the literature, is involved in isolated myocyte hypertrophy, induced by endothelin I, phenylephrine, angiotensin II [67] and norepinephrine [68]. AMPK—adenosine monophosphate activated protein kinase; AT1R—angiotensin II receptor; NOX-NADPH oxidase; BH4—dihydrobiopterin-4; NOS—nitric oxide synthase; NO—nitric oxide; MAPK—mitogen-activated protein kinase; XOR—xanthine oxidoreductase; AMP—adenosine monophosphate; GMP—guanosine monophosphate; Rac-1—GTP-binding protein; NHE-1—sodium/hydrogen exchanger-1; ERK—extracellular signal-regulated kinase; JNK-c—Jun N-terminal kinase; p38—a focal point of interactions of the serine/threonine kinases), MMP—matrix metalloproteinase.
Figure 3. Enzymes involved in ROS production in cardiomyocytes and fibroblasts (created with BioRender.com on 7 February 2022). NOX2 is presented to be activated by endothelin and angiotensin II [58], by cytokines [59] and mechanical stress [60]. Increased NOX2 activation leads to cytoskeletal dysfunction in patients with CHF [61]. It was discovered that superoxide anions, produced by NOX, can oxidize and degrade hydrobiopterin-4 (BH4) leading to NOS uncoupling [62]. Nitric oxide synthase 3 (NOS3) uncoupling was observed in myocardium exposed to chronic pressure load. NOS3 catalyzes nitric oxide (NO) synthesis under physiological conditions. NO has an antihypertrophic effect. However, NOS3 is uncoupled with pressure load, and this, in turn, leads to reduction in tetrahydrobiopterin-4 concentration, increase in ROS production, and, as a consequence, to cardiomyocyte hypertrophy [63]. It was also shown that increase in ROS activates MAPK, leading to increased expression of proteins, such as ERK, JNK and P38, which are related to cardiomyocyte hypertrophy [64] (Figure 3). What is more, NOX-derived ROS may activate XOR [65]. Additionally, angiotensin II-induced signaling and isolated cardiomyocyte hypertrophy are dependent on NOX2 [66]. GTP-binding protein Rac-1 (involved in NOX activation), as described in the literature, is involved in isolated myocyte hypertrophy, induced by endothelin I, phenylephrine, angiotensin II [67] and norepinephrine [68]. AMPK—adenosine monophosphate activated protein kinase; AT1R—angiotensin II receptor; NOX-NADPH oxidase; BH4—dihydrobiopterin-4; NOS—nitric oxide synthase; NO—nitric oxide; MAPK—mitogen-activated protein kinase; XOR—xanthine oxidoreductase; AMP—adenosine monophosphate; GMP—guanosine monophosphate; Rac-1—GTP-binding protein; NHE-1—sodium/hydrogen exchanger-1; ERK—extracellular signal-regulated kinase; JNK-c—Jun N-terminal kinase; p38—a focal point of interactions of the serine/threonine kinases), MMP—matrix metalloproteinase.
Biomedicines 10 00602 g003
Figure 4. ROS in HFrEF pathogenesis (created with BioRender.com on 9 February 2022). Decrease in O2 and nutrient supply in cardiomyocytes results in ROS overproduction. Increase in ROS leads to MMP activation and consequently fibroblast proliferation, vacuolar degeneration, excitation-contraction coupled protein oxidation (consequently leading to decrease in contractile function of cardiomyocytes) and oxidation of mitochondrial OMM and IMM proteins. OMM protein damage results in MAPK pathway activation, leading to apoptosis. IMM protein OPA1 oxidation (resulting in inhibition) with BNIP3 inhibition by ROS causes mitochondrial fragmentation. Both mitochondrial and other cytosol protein oxidation by ROS lead to decrease in ETC protein expression, resulting in ATP production decrease, resulting in poor contraction. Hyperacetylation of ETC complexes, fatty acid beta-oxidation and TCA cycle proteins lead to inhibition of its activity. SIRT, in healthy cardiomyocytes, inhibits hyperacetylation and activates AMPK-induced pathway, leading to enzyme-antioxidant synthesis which leads to hypertrophy inhibition. Excess of ROS inhibits beneficial SIRT effects. (EF—ejection fraction; MMP—matrix metalloproteinase; ROS—reactive oxygen species; BNIP3—mitochondrial mitophagy marker; OPA1—optic atrophy 1 protein; DRP1—dynamin-related protein 1; MFN2—mitofusin 2; MAPK—mitogen-activated protein kinase; ETC—electron transport chain; β-OX—beta-oxidation; SIRT—sirtuin family of NAD+-dependent deacetylases; TCA—tricarboxylic acid cycle; LKB1—liver kinase B1; AMP—adenosine monophosphate; AMPK—AMP activated protein kinase; SOD—superoxide dismutase; CAT—chloramphenicol acetyltransferase; UCP2—uncoupling protein 2).
Figure 4. ROS in HFrEF pathogenesis (created with BioRender.com on 9 February 2022). Decrease in O2 and nutrient supply in cardiomyocytes results in ROS overproduction. Increase in ROS leads to MMP activation and consequently fibroblast proliferation, vacuolar degeneration, excitation-contraction coupled protein oxidation (consequently leading to decrease in contractile function of cardiomyocytes) and oxidation of mitochondrial OMM and IMM proteins. OMM protein damage results in MAPK pathway activation, leading to apoptosis. IMM protein OPA1 oxidation (resulting in inhibition) with BNIP3 inhibition by ROS causes mitochondrial fragmentation. Both mitochondrial and other cytosol protein oxidation by ROS lead to decrease in ETC protein expression, resulting in ATP production decrease, resulting in poor contraction. Hyperacetylation of ETC complexes, fatty acid beta-oxidation and TCA cycle proteins lead to inhibition of its activity. SIRT, in healthy cardiomyocytes, inhibits hyperacetylation and activates AMPK-induced pathway, leading to enzyme-antioxidant synthesis which leads to hypertrophy inhibition. Excess of ROS inhibits beneficial SIRT effects. (EF—ejection fraction; MMP—matrix metalloproteinase; ROS—reactive oxygen species; BNIP3—mitochondrial mitophagy marker; OPA1—optic atrophy 1 protein; DRP1—dynamin-related protein 1; MFN2—mitofusin 2; MAPK—mitogen-activated protein kinase; ETC—electron transport chain; β-OX—beta-oxidation; SIRT—sirtuin family of NAD+-dependent deacetylases; TCA—tricarboxylic acid cycle; LKB1—liver kinase B1; AMP—adenosine monophosphate; AMPK—AMP activated protein kinase; SOD—superoxide dismutase; CAT—chloramphenicol acetyltransferase; UCP2—uncoupling protein 2).
Biomedicines 10 00602 g004
Figure 5. NO–cGMP-PKG pathway in HFpEF development (created with BioRender.com on 9 February 2022). NO, produced in endothelium by eNOS in normal conditions, protects fibroblasts and cardiomyocytes from harmful proliferation. BH4 (hydrobiopterin-4) is required for eNOS action. NO acts via stimulation of cardiac sGC receptors (leading to cGMP synthesis). cGMP regulates phosphodiesterases (PDEs) and cGMP-dependent protein kinases (PKG). NO inhibits TXNIP, resulting in inhibition of apoptosis, however ROS inhibit this action. Oxidative stress shifts sGC towards an oxidized heme-free form which is unresponsive to endogenous and exogenous NO. Titin hypophosphorylation leads to hypertrophy of cardiomyocytes. Increased peroxynitrite concentrations, together with scarce NO availability, induce fibroblast proliferation. Due to uncoupled eNOS, superoxide production increases. In turn, low levels of NO react with superoxide to generate peroxynitrite. Peroxynitrite: (1) oxidizes BH4 to BH2 (BH2 inhibits eNOS), and (2) oxidizes Fe2+ to Fe3+ (Fe3+ inhibits cGMP production from GTP). Therefore, cGMP cannot activate PKG to phosphorylate titin, whereas titin phosphate prevents cardiomyocyte hypertrophy. For this reason, eNOS inhibition results in both fibroblast and cardiomyocyte proliferation. Neprylisin catalyzes NPs degradation, and PDE9 inhibits NPs. NPs acts through receptors in cardiomyocytes to modulate proliferation of cardiomyocytes. NO—nitric oxide, eNOS—endothelial nitric oxide synthase, BH4—dihydrobiopterin-4, NPs—natriuretic peptide, PKG—protein kinase G, a—activated, sGC—soluble guanylyl cyclase, pGC—particulate guanylyl cyclase, PDE-GMP—regulated phosphodiesterase, ROS—reactive oxygen species, titin-P—phosphorylated titin, GTP—guanosine triphosphate, TXNIP—thioredoxin-interacting protein.
Figure 5. NO–cGMP-PKG pathway in HFpEF development (created with BioRender.com on 9 February 2022). NO, produced in endothelium by eNOS in normal conditions, protects fibroblasts and cardiomyocytes from harmful proliferation. BH4 (hydrobiopterin-4) is required for eNOS action. NO acts via stimulation of cardiac sGC receptors (leading to cGMP synthesis). cGMP regulates phosphodiesterases (PDEs) and cGMP-dependent protein kinases (PKG). NO inhibits TXNIP, resulting in inhibition of apoptosis, however ROS inhibit this action. Oxidative stress shifts sGC towards an oxidized heme-free form which is unresponsive to endogenous and exogenous NO. Titin hypophosphorylation leads to hypertrophy of cardiomyocytes. Increased peroxynitrite concentrations, together with scarce NO availability, induce fibroblast proliferation. Due to uncoupled eNOS, superoxide production increases. In turn, low levels of NO react with superoxide to generate peroxynitrite. Peroxynitrite: (1) oxidizes BH4 to BH2 (BH2 inhibits eNOS), and (2) oxidizes Fe2+ to Fe3+ (Fe3+ inhibits cGMP production from GTP). Therefore, cGMP cannot activate PKG to phosphorylate titin, whereas titin phosphate prevents cardiomyocyte hypertrophy. For this reason, eNOS inhibition results in both fibroblast and cardiomyocyte proliferation. Neprylisin catalyzes NPs degradation, and PDE9 inhibits NPs. NPs acts through receptors in cardiomyocytes to modulate proliferation of cardiomyocytes. NO—nitric oxide, eNOS—endothelial nitric oxide synthase, BH4—dihydrobiopterin-4, NPs—natriuretic peptide, PKG—protein kinase G, a—activated, sGC—soluble guanylyl cyclase, pGC—particulate guanylyl cyclase, PDE-GMP—regulated phosphodiesterase, ROS—reactive oxygen species, titin-P—phosphorylated titin, GTP—guanosine triphosphate, TXNIP—thioredoxin-interacting protein.
Biomedicines 10 00602 g005
Table 1. Medicines affecting human cardiomyocytes via renin-angiotensin system in HF patients.
Table 1. Medicines affecting human cardiomyocytes via renin-angiotensin system in HF patients.
MedicinePatientsAppl., Dose and DurationResultsPathophysiological MechanismReference
ValsartanCHF, NYHA functional class II–IV, n = 836-week study, 80 or 160 mg bidProduced hemodynamic and hormonal effects.Blocks angiotensin AT1 receptor leading to NOX2 activity reduction.[191]
Sacubitril/valsartan (LCZ696)HFrEF (NYHA II–IV) and LVEF ≤ 40% n = 48225 yearsDecreased levels of NT-proBNP or improved left atrial volumes.Inhibits neprilysin/
ATR
[192]
Sacubitril/valsartanHFrEF, n = 54Twice a day 24/26, 49/51, 97/103 mgImproved NYHA class, decreased NT-pro BNP concentration, reduced mortality.Inhibits neprilysin/
ATR1
[193]
CandesartanHFpEF n = 1958, HFrEF, n = 19592.9 yearsImproved outcomes in both groups.ATR1 inhibitor[194]
Table 2. Antioxidants affecting human cardiomyocytes in chronic HF patients.
Table 2. Antioxidants affecting human cardiomyocytes in chronic HF patients.
MedicinePatientsAppl., Dose and DurationResultsPathophysiological MechanismReference
OTC OCsPatients with heart failure with preserved ejection fraction (HFpEF), n = 16.600 mg of α-lipoic acid, 1000 mg of vitamin C, and 600 IU of vitamin EImproved peripheral vascular function regardless of changes in global markers of oxidative stress in HFpEF.Alterations in redox balance as a result of attenuated endogenous AOx capacity and/or elevated oxidative stress might be an underlying mechanism.[216]
AOxPatients with HFrEF, n = 14.1 g of vitamin C, 600 IU of vitamin E, and 0 mg/day). 6 g α). -lipoic acidImproved macrovascular function, reduced oxidative stress, and increased AOx capacity in patients with HFrEFChanges redox balance; Increases oxidative stress;
Decreases endogenous AOx protection.
[221]
Table 3. Medicines affecting human cardiomyocyte mitochondria in chronic HF patients. (LVEDV- LV end-diastolic volume, LVESV-LV-end-systolic volume).
Table 3. Medicines affecting human cardiomyocyte mitochondria in chronic HF patients. (LVEDV- LV end-diastolic volume, LVESV-LV-end-systolic volume).
MedicinePatientsAppl., Dose and DurationResultsPathophysiological MechanismReference
Elamipretide (SS-31)HFrEF(EF ≤ 35%), n = 24 and placebo n = 12.i.v., 4-h infusion 0.25 mg × kg−1 × h−1↓LVESV, ↓LVEDVBy binding to cardiolipin, decreases ROS production.[251]
Elamipretide (SS-31)HFrEF(EF≤40%), n = 48 and placebo n = 23.p.o., 4 mg or 40 mg once daily for 28 days.Did not improve LVESV.By binding to cardiolipin, decreases ROS production.[252]
Coenzyme Q10Moderate to severe HFrEF, n = 420.p.o., 100 mg 3 times daily, 2 years.Significantly improved NYHA class, CV events ↓by 50%.Q10 is involved in eNOS regulation[243]
Table 4. Medicines affecting human cardiomyocytes by increasing cGMP-PKG signaling in HF patients. (LV-left ventricle, LA-left atrium).
Table 4. Medicines affecting human cardiomyocytes by increasing cGMP-PKG signaling in HF patients. (LV-left ventricle, LA-left atrium).
MedicinePatientsAppl., Dose and DurationResultsPathophysiological MechanismReference
EmpagliflozinHFpEF II–IV class (EF > 40%), n = 2997, placebo n = 2991.10 mg once daily or placebo 36 monthsReduced cardiovascular death and hospitalizationSLGT2 inhibitor and sGC activator[295]
SildenafilStable outpatient individuals with HFpEF, n = 160.24 weeksDid not improve exercise capacity and clinical status.Inhibits cGMP breakdown[265]
NitroxylHFpEF, n = 65.23 monthsHNO increased cGMP concentrations and had NOX inhibitory and antihypertrophic effects in rat cardiomyocytes.NO donor.[255]
CymlanodHFrEF, n = 455 h i.v infusion or placeboSlightly reduced LV and LA volumesNO donor[296]
HydralazineHFrEF, NYHA class III–IV (EF ≤ 35% or < 45% with LVIDd > 2.9 cm/m), n = 1050.18 monthsImproved survival.ROS scavenger;
Inhibitor of O2generation;
normalizes endogenous rates of vascular O2production [122].
[286]
CinaciguatHFrEF. n = 621 yearDid not significantly improve dyspnea or cardiac index.Increases cGMP production by targeting guanylate cyclase at the NO receptor (sGC).[256]
VericiguatHFrEF, (LVEF < 45%, history of decompensation within the last four weeks), n = 4561.25 mg, 2.5 mg, 5 mg, or 10 mg for 12 weeksWas well-tolerated and higher doses were associated with a greater reduction in NT-pro BNP level.Triggers cGMP production by binding the heme-containing non-oxidized form of sGC regulatory domain.[290]
HFpEF ( LVEF > 45% and a history of decompensation within the last four weeks), n = 477from 1.25 mg to 10 mg once daily for 12 weeksAppeared to be well-tolerated and improved patients with HFpEF quality of life; however, had no significant impact on NT-proBNP level.[297]
HFpEF (LVEF > 45%),
n = 789
15 mg or 10 mg dailyNo significant changes were observed.[298]
HFpEF (NYHA II to IV) with an LVEF < 45%, history of decompensation over the last six months, elevated NT-proBNP or BNP), n = 505010 mg once daily
for 10.8 months
Hospitalization for heart failure and death from cardiovascular causes were reduced compared to placebo.[299]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mongirdienė, A.; Skrodenis, L.; Varoneckaitė, L.; Mierkytė, G.; Gerulis, J. Reactive Oxygen Species Induced Pathways in Heart Failure Pathogenesis and Potential Therapeutic Strategies. Biomedicines 2022, 10, 602. https://doi.org/10.3390/biomedicines10030602

AMA Style

Mongirdienė A, Skrodenis L, Varoneckaitė L, Mierkytė G, Gerulis J. Reactive Oxygen Species Induced Pathways in Heart Failure Pathogenesis and Potential Therapeutic Strategies. Biomedicines. 2022; 10(3):602. https://doi.org/10.3390/biomedicines10030602

Chicago/Turabian Style

Mongirdienė, Aušra, Laurynas Skrodenis, Leila Varoneckaitė, Gerda Mierkytė, and Justinas Gerulis. 2022. "Reactive Oxygen Species Induced Pathways in Heart Failure Pathogenesis and Potential Therapeutic Strategies" Biomedicines 10, no. 3: 602. https://doi.org/10.3390/biomedicines10030602

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