Next Article in Journal
Beyond Quadruple Therapy and Current Therapeutic Strategies in Heart Failure with Reduced Ejection Fraction: Medical Therapies with Potential to Become Part of the Therapeutic Armamentarium
Next Article in Special Issue
Synergistic Effects of Weight Loss and Catheter Ablation: Can microRNAs Serve as Predictive Biomarkers for the Prevention of Atrial Fibrillation Recurrence?
Previous Article in Journal
Polyallylamine Binds to Aβ Amyloid and Inhibits Antibody Recognition
Previous Article in Special Issue
Early Impairment of Paracrine and Phenotypic Features in Resident Cardiac Mesenchymal Stromal Cells after Thoracic Radiotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin C as Scavenger of Reactive Oxygen Species during Healing after Myocardial Infarction

by
Huabo Zheng
1,2,
Yichen Xu
2,3,
Elisa A. Liehn
2,4,* and
Mihaela Rusu
5,*
1
Department of Cardiology, Angiology and Intensive Care, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany
2
Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
3
Department of Histology and Embryology, Medicine and Life Sciences, Hainan Medical University, Haikou 571199, China
4
National Institute of Pathology “Victor Babes”, Splaiul Independentei Nr. 99-101, 050096 Bucharest, Romania
5
Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(6), 3114; https://doi.org/10.3390/ijms25063114
Submission received: 17 November 2023 / Revised: 31 January 2024 / Accepted: 10 February 2024 / Published: 7 March 2024
(This article belongs to the Special Issue Current Research for Heart Disease Biology and Therapeutics)

Abstract

:
Currently, coronary artery bypass and reperfusion therapies are considered the gold standard in long-term treatments to restore heart function after acute myocardial infarction. As a drawback of these restoring strategies, reperfusion after an ischemic insult and sudden oxygen exposure lead to the exacerbated synthesis of additional reactive oxidative species and the persistence of increased oxidation levels. Attempts based on antioxidant treatment have failed to achieve an effective therapy for cardiovascular disease patients. The controversial use of vitamin C as an antioxidant in clinical practice is comprehensively systematized and discussed in this review. The dose-dependent adsorption and release kinetics mechanism of vitamin C is complex; however, this review may provide a holistic perspective on its potential as a preventive supplement and/or for combined precise and targeted therapeutics in cardiovascular management therapy.

1. Introduction

Myocardial infarction (MI) is one of the most severe clinical manifestations of heart disease, which is a leading cause of death worldwide [1] despite considerable clinical management improvements in the past decade [2,3]. The first recognized cause of MI is the chemo-mechanical disruption of vulnerable or unstable plaque, which leads to the formation of a thrombus and occlusion of the vessels [4]. Mechanical (balloon angioplasty) and pharmacological (thrombolysis) modern management therapies restore blood flow in patients with MI [5,6,7,8] by removing the plaque or occluded thrombus from the blocked coronary artery [9]. While MI leads to the recruitment and activation of inflammatory cells (e.g., neutrophils [10], macrophages [11], monocytes [12], regulatory T-cells [13], CD4+ T-cells [14], mast cells [15]), reperfusion results in a burst of free radical formation [16]. Thus, the increased oxidative stress and imbalanced levels of the production and accumulation of reactive oxygen species (ROS) will exaggerate tissular damage, known as reperfusion injury, which results in increased myocyte damage and later increased fibrosis and remodeling [17,18]. Despite tremendous progress towards reducing post-ischemic reperfusion events, patients undergoing MI still experience increased mortality and morbidity [2,3]. Understanding changes in physio-pathological parameters, including heart function [19], heart frequency [20], and vascular integrity [21], is important for establishing novel therapeutic strategies to prevent or minimize heart failure.
A key component of cardiomyocyte death is mitochondrial dysfunction during ischemic reperfusion injury [22]. At rest, the energy reservoir produced by mitochondrial activity represents 6–30 kg of adenosine triphosphate (ATP) daily [23]. This provides the heart with the amount of ATP necessary for a cardiac muscle demand of 100,000 beats daily [24]. Although the metabolism of the heart appears to be complex, studies show that three main mechanisms are highly involved in contraction: (i) glycolysis and β-oxidation of free fatty acids are the primary fuel sources; (ii) oxidative phosphorylation of adenosine diphosphate (ADP) will produce ATP; and (iii) high-energy transfer from ATP to the heart occurs via myofibrils.
Antioxidant therapies have been proposed to efficiently reduce oxidative stress and cellular damage during MI by boosting the body’s natural defense mechanisms. Polyphenols are known to have a broad spectrum of antioxidant benefits [25]. Vitamin E is particularly important in protecting cell membranes from oxidative damage [26]. Vitamin C is effective in neutralizing ‘harmful’ free radicals [27] and, when combined with vitamin E, it acts synergistically to reinforce the protective effects [28]. Moreover, the effects of vitamin C extend beyond the mere neutralization of free radicals [29,30]. By donating its electrons, vitamin C can protect important biomolecules (proteins, lipids, carbohydrates, and nucleic acids) from being damaged by oxidants, including ROS [31,32]
Previous studies of the association between vitamin C levels and MI seem to have conflicting results. Generally, fruit and vegetable intake is known to be reflected in plasma vitamin C and is inversely associated with the risk of heart failure [33]. The multivariant Cox model of a 6-year stratified cohort study (age, season, and examination year) showed that patients with a low plasma vitamin C concentration (<11.4 μM) had a significant association (3.5-fold) with an increased risk of MI [34]. Another study involving 180 male patients could not find any correlation related to MI risk, irrespective of smoking status [35]. Similarly, other studies have failed to demonstrate the beneficial effects of vitamin C intake on cardiovascular health and other complications [36], including MI [37,38]. However, a prospective study of 3919 older men followed up for a mean period of 11 years demonstrated the relationship between a reduced risk of heart failure and higher plasma vitamin C levels, independent of MI [39].
This review comprehensively revisits the role of vitamin C as an antioxidant during healing after MI, revealing its mechanisms and offering prospects for clinical use.

2. Metabolism of Vitamin C

Vitamin C is an important co-enzymatic factor for cellular function and survival. In vivo or in vitro, vitamin C undergoes oxidation to dehydroascorbic acid (DHA), which is promoted by oxidants like hydrogen peroxide (H2O2) or catalyzed by the enzyme vitamin C oxidase [40,41]. In vivo, DHA has a half-life of only a few minutes, and it will normally be reduced back to vitamin C by thioredoxin reductase, dehydroascorbate oxoreductase, and 3-α-hydroxysteroid dehydrogenase [41,42]. In the liver and the kidney, dehydroascorbic acid will be hydrolyzed to 2,3-diketo-L-gulonate and then decarboxylated to L-xylonate and L-lyxonate [43,44], both of which can enter the pentose phosphate pathway and be degraded [40]. Vitamin C autoxidizes easily to the superoxide anion (O2•−) and ascorbyl anion (ASC•−) in vitro (Figure 1) [45]. The degradation of vitamin C may be prevented by the phosphate moieties in the cell culture medium [46]. For example, the supplementation of 75 μM ascorbate with 500 μM 2-phosphoascorbate administration can maintain detectable levels of vitamin C for 72 h in the culture medium (1 ng/106 cells) [47,48].
Figure 1. Metabolism of vitamin C in vivo vs. in vitro. In vivo, vitamin C is oxidized to dehydroascorbic acid (DHA) when reacts with ROS. In vitro, ascorbate autoxidizes easily to O2•− and ASC•−.
Figure 1. Metabolism of vitamin C in vivo vs. in vitro. In vivo, vitamin C is oxidized to dehydroascorbic acid (DHA) when reacts with ROS. In vitro, ascorbate autoxidizes easily to O2•− and ASC•−.
Ijms 25 03114 g001

3. The Role of Vitamin C in Cardiovascular Pathologies

The molecular regulators, such as ROS, are continuously generated in response to various conditions and play a key role in the pathophysiology of the heart. For example, an optimum heart function requires an ideal balance between ROS-producing enzymes and antioxidants, yet our knowledge remains limited in this regard [49]. Therefore, the increased production of ROS and oxidative stress are critical modulators in the healing and remodeling processes after MI [50,51].

3.1. Role of ROS in the Heart

The causative role of ROS in cell pathology remains unknown. ROS appear to be generated as degradation products during cellular metabolism and respiration and differ in their origin. Novel methodologies need to be developed to allow comprehensive investigations of the mechanism and regulation of ROS production at the cellular level.
Low levels of ROS (e.g., 10 μM H2O2, 500 nM H2O2) can be considered potent redox signaling messengers responsible for normal metabolic processes [52,53]. On the contrary, abnormal accumulation levels of ROS are associated with the loss of signaling and cellular damage [54]. Low levels of O2•− and H2O2 can also increase the expression levels of hypoxia-inducible factor-1 (HIF-1) under hypoxia [54]. As a result, HIF-1 upregulates the transcription of genes responsible for angiogenesis, such as vascular endothelial growth factor [55]. Further, low levels of ROS play an important role in cardiomyocyte differentiation and proliferation [53]. However, chronically low levels of ROS are associated with prolonged cellular stress and cardiomyopathy [56].
Excessive ROS (1000 µM H2O2 [57], 100 µM H2O2 [58]) is reported to induce mutations of nuclear [59] and mitochondrial DNA [60], affecting the replication, proteins post-translation modifications [61], and membrane phospholipids [60] in the cardiac cells, accelerating aging and cell death [62].
Due to the higher diffusion rate of O2 in the myocardium, compared to other tissues, more ROS is produced under oxidative stress [63]. Indeed, in pathological conditions such as cardiac ischemia/reperfusion, cardiac hypertrophy, and heart failure, clinical data show the correlation of oxidative stress with an increased level of redox, and proinflammatory and profibrotic signaling [64,65].
Following ischemic injury, the reperfusion increases the expression of ROS, allowing large amounts of O2•−, H2O2, etc. to accumulate in the heart [66]. Concomitantly, under mechanical shear stress, a burst of both NO and O2•− is released in the mitochondria, generating peroxynitrite and decreasing the bioavailability of NO. In a failing heart, a large pool of ROS production leads to an increase in mitochondrial permeability and outer membrane rupture, releasing apoptotic signaling molecules, such as cytochrome c (cyt c) [67].
Recent studies have revealed a strong similarity between the deleterious effects caused by the generation of ROS in myocardial tissue following ischemia/reperfusion and a specific iron-dependent form of non-apoptotic cell death known as ferroptosis [68]. The critical relationship between ROS and ferroptosis is particularly relevant in the context of MI, where both increased oxidative stress and iron overload coincide. Ferroptosis leads to cellular death through iron-catalyzed lipid peroxidation. This process can be prevented by an antioxidant, selenoprotein glutathione peroxidase 4 (GPX4), a vital enzyme that detoxifies lipid peroxides and stops ferroptosis [69,70]. Reliable therapeutic strategies, concerning the ischemic heart, could target ferroptosis to reduce reperfusion injury by the manipulation of iron availability or lipid peroxidation.
Additionally, the interaction between ROS, iron balance, and lipid peroxidation in the myocardium opens the way for new therapeutic targets in the management of ischemic heart diseases. For example, the investigation of molecules and pathways of ferroptosis, including ferroportin [71], FOXO1 [72], and others, may provide useful insights into the causal relationships between iron metabolism, ROS, and myocardial viability following ischemia.
The usage of H2O2 outside the cells may exceed 100–5000 times the usage in the cells (10 nM) [73] and, in the chronic inflammatory tissue, these values may be even ten times higher [73]. Enzymatic antioxidants, such as superoxide dismutase (SOD) and glutathione peroxidase (GPx), or non-enzymatic antioxidants, such as vitamin E and N-acetylcysteine, were reported to play important roles in scavenging ROS [63]. Copper zinc superoxide dismutase (Cu/Zn-SOD), located in the cytosol and the mitochondrial intermembrane space [74], and manganese superoxide dismutase (Mn-SOD), located in the mitochondrial matrix [75], catalyze O2•− to H2O2 in the heart. Catalase (CAT) and GPx [76,77] are vital antioxidative enzymes, in both mitochondria and cytosol, that scavenge the excess H2O2 in the heart (Figure 2).
ROS, such as H2O2 and hydroxyl (OH•−), can be reduced by GSH, which is present in the cytosol, the nuclei, and the mitochondria [78] (Figure 2). Pathological conditions will tip the scale towards the failure of antioxidant clearance rather than the synthesis of abnormal amounts of ROS.

3.2. Cytosolic Sources of ROS (ctROS) in the Heart

At specific concentration levels, ROS has a detrimental or beneficial role in cell function (levels see Section 3.1) [79]. Oxidative stress is one of the main contributors to generating uncontrollably large amounts of ROS. For example, in the failing heart, oxidative stress is associated with significantly increased ctROS levels. ctROS signaling induces cardiomyocyte hypertrophy and cell death, influencing extracellular matrix remodeling and left ventricle chamber dilation [80,81]. It is important to address how ctROS signaling, from various sources, affects the heart, and how the heart finds reserves for the preservation of its function. For example, ctROS produced by NOX can lead to the activation of profibrotic genes such as NF-κB or matrix metalloproteinase (MMP)-2 [82] and, therefore, to increase cardiac fibrosis [83]. In the case of ischemia-reperfusion, the XO-associated abnormal ctROS synthesis leads to oxidative stress-mediated F-actin depolymerization, which induces mitochondrial dysfunction and fission, and apoptosis of the cardiac microvascular endothelial cells [84,85].
The major source of ctROS is the seven-NOX enzymatic complexes. In particular, NOX2 and NOX4 isoforms seem to have distinct physiological and pathophysiological roles in the heart [51,86]. For example, angiotensin II induces the activation of cytosolic subunits of NOX2 such as p47phox, p67phox, p40phox, and Rac1. These activated subunits bind to flavocytochrome and induce the synthesis of radical species such as O2•− and OH•− [87]. These reactive species stimulate calcium release from the sarcoplasmic reticulum in myocytes and excitation–contraction coupling [87], which can result in contractile dysfunction and myocyte death. Activated NOX2 can also modulate interstitial fibrosis via activating metalloproteinase activity following MI [88]. Although NOX4 activity is controversial, it has an important role in angiogenesis [89], mediating metabolic stress responses [90] and detoxifying responses [91]. NOX4, localized in the endoplasmic reticulum and mitochondria, generates large amounts of detrimental H2O2 [92]. Another cytosolic source of ROS is H2O2, which may derive from XO and uric acid [93].
Another important source of cytosolic ROS in myocytes is the uncoupling of endothelial nitric oxide synthase (eNOS) [94] and inducible nitric oxide synthase (iNOS) [95]. Their upregulation is associated with increased infarct size and cardiac dysfunction in ischemia/reperfusion injury in the diabetic rat heart [95] and chronic pressure-induced hypertrophy in wild-type mice, respectively [94].
Although cardiac cytochrome P450 enzymes have major cellular metabolic and homeostasis characteristics, they also represent a rich cytosolic source of ROS, as observed in cigarette smoking patients [96]. The inhibition of cardiac cytochrome P450 enzyme, as demonstrated in a rat heart model, may constitute a potential clinical approach to suppressing ctROS formation [97].
Extracellularly, a minor potential source of H2O2 production is the autoxidation of small molecules such as catecholamines [98]. In vivo experiments show that catecholamines-derived ROS cause oxidation of lipids in the rat heart and thus are associated with negative cardiac remodeling [99,100].
Among ctROS sources, NOX2 seems to be the most important in the pathological heart. A high level and activity of NOX2 produces large amounts of ROS, causing injury and initiating the fibrotic progress of the pathological condition, such as ischemia.

3.3. Mitochondrial Sources of ROS (mtROS) in Heart

Mitochondria are the “energy-producing motor” of cardiac cells, as the heart constantly demands the synthesis of high levels of ATP. The ATP synthesis undergoes a multistep process as follows: post-translational modifications (e.g., oxidative phosphorylation); electron transfer across the five protein complexes (complexes I–V); and two transporters (coenzyme Q and cyt c). Oxidative stress limits ATP synthesis [101], resulting in uncontrolled mtROS formation and accumulation, thus impairing the contractile function of cardiac cells [102,103]. Following the Krebs cycle, the oxidation of metabolic products leads to the formation of mtROS in the form of O2•− [63] and to reduced nicotinamide-adenine dinucleotide (NADH) [104] or reduced flavin adenine dinucleotide (FADH2) [105] (Figure 2). As a result of electron transport chain (ETC) across complexes I and III, mtROS production associates partially to O2 → O2•−, commonly described as the main sources of mtROS [106,107]. However, enzymes in the respiratory complex II can also reduce O2 levels under hypoxia settings, thereby impeding ATP production [108]. Several research findings and examples that explain the involvement of complexes I–V in regulating the remodeling of myocardial following injury are described below. Key research questions are identified and systematically reviewed such as: (i) what are the key factors responsible for the dysregulation of mtROS production; (ii) what are the effects of mtROS on the function of mitochondria and cardiac cells; and (iii) what is the role of mtROS in the progress of MI?
Complex I (NADH dehydrogenase) is the first enzyme of the mitochondria respiratory chain. Complex I is the most sensitive respiratory chain complex, being identified as the central site of ROS production in the mitochondria. Structurally, complex I has a hydrophobic unit immersed in the inner membrane and a hydrophilic unit faced in the matrix (Figure 2A). The hydrophobic unit of complex I, under critical thermodynamic conditions, favors the reverse electron transport and reduction of ubiquinone, dismutase of O2 to H2O2, and OH•− species formation. The hydrophilic unit participates in pumping H+ into the intermembrane space and mediates the transfer of high energy electrons from NADH to flavin mononucleotide to form the NAD+, then to coenzyme Q (ubiquinone) to form ubiquinol (QH2), supported by the Fe-S carrier [109] (Figure 2A). Both flavin mononucleotide (FMN)- and Q- binding sites may be subjected to electron leakage from ETC under pathological conditions, leading to the formation of O2•− species [110,111]. At least 40% of all mitochondrial disorders are associated directly with DNA mutations in subunits of complex I, because elevated levels of O2•− cannot be optimally regulated by SOD, CAT, and GPx [112] (Figure 2A). A rotenone toxin model revealed the increased release of mitochondrial O2•− in the A7r5 SMCs derived from rat aorta [113]. Presumably, a fraction of O2•− is dismutated to H2O2 by SOD and further reduced to H2O by antioxidants, such as CAT and GPx in the mitochondrial matrix [112]. Excessive mitochondrial H2O2 causes direct damage to the internal structures, including proteins, lipids, and DNA in the mitochondria of cardiomyocytes [114]. Another fraction of O2•− binds to NO and forms peroxynitrite, which may result in the upstream of rotenone binding sites and a progressive shift of redox potential [115], leading to the decrease of ATP production in the mitochondria and the dysfunction of cardiomyocytes [116].
Using the artificial acceptor hexaammineruthenium (III) reductase, Stepanova, A. et al. revealed that increased levels of covalently non-bound flavin mononucleotide and O2 depletion lead to reduced mitochondrial complex I activity and ATP content [117]. Moreover, to the best of our knowledge, the complex metabolic interplay among, e.g., fatty acids, glycero-3-dehydrogenase, electron-transferring flavoprotein, etc. and the mechanisms of mitochondrial O2•− release are yet not known in vivo. However, the redox potential shift evidenced either in the in vitro model [118] or in the acute brain ischemia/reperfusion mice model [119,120] can be associated with changes in cardiac function.
In contrast to complex I, in cardiac complex II (succinate dehydrogenase) in ischemia/reperfusion injury, higher mitochondrial oxygen consumption is detected [121]. In complex II, less H+ is generated and less ATP is formed from FADH2 than from NADH. In the presence of succinate dehydrogenase, succinate converts into fumarate and reduces FAD to FADH2, which donates electrons to the Fe-S clusters, to ultimately be transferred to the reduced ubiquinol form of coenzyme Q [122] (Figure 2B). The dicarboxylate-binding site of complex II (IIf site) is associated with electron leakage from ETC and ROS generation [123,124] (Figure 2B). In vitro research using isolated mitochondria suggested that the IIf sites were reduced after the inhibition of complex II by atpenin A5 in myocardial ischemia [123]. As a result, the reduced fraction of site IIf is inversely associated with ROS production in complex II, thereby impairing the mitochondrial activity and cardiac cell function [123]. Succinate is an important substrate of complex II that accumulates and is rapidly oxidized by complex II upon ischemia/reperfusion, thus leading to the accumulation of large amounts of O2•− in complex II [50,120]. Interestingly, at the tissue level, malonate, the competitive inhibitor of complex II, significantly reduces mtROS levels and infarct size in the heart after ischemia/reperfusion [125] by increasing the mitochondrial oxygen consumption rate [121]. In this context, complex II might serve as a metabolic target source within the respiratory chain.
Complex III (cyt c reductase) catalyzes the transfer of electrons from the readily formed ubiquinol to cyt c in the Q cycle [126] via two distinct redox paths [127,128], while H+ is pumped back into the intermembrane space [126] (Figure 2C). One path uses the Fe-S cluster group and heme group of cyt c1 to receive four high-energy electrons from two molecules of QH2. Two of the electrons are transferred to the water-soluble molecule of cyt c, and another two electrons are transferred to cytochrome b to participate in the second electron transfer path [128]. The second electron transfer path uses cyt b as electron—cargo to partially reduce the Q to a semiquinone radical anion (Q•−). Further, Q•− receives another electron from cyt b, generating QH2 at the end of the electron transfer chain [106,127]. In vitro, the decreased activity of mitochondrial complex III from aging rat hearts is associated directly with increased ROS production, thereby correlating to the increased infarction size after ischemia/reperfusion injury [129]. As a possible mechanism, aging increases the ionic strength around mitochondrial complex III, which decreases the activity of subunits VIII or X in the cyt c binding site of complex III [130]. Likewise, the mitochondrial complex III activity is reduced significantly in the ischemia/reperfusion rabbit model, which is associated with elevated ROS levels [123]. Similarly, studies in rats revealed that ischemia/reperfusion destroys the structures of hemes b, c, and c1, and thereby the activity of complex III in cardiac cells [131]. To regulate ROS production in the post-ischemic heart, the large quantities of O2•− accumulated in the reduction sites of Qo and Qi of complex III should be reduced [132]. The reduced O2•− synthesis in the Q sites of cyt c can be achieved pharmacologically by inhibiting the Qo and Qi sites with antimycin A [133] and myxothiazol [134], e.g., in rat hippocampal CA1 cells, with stigmatellin, e.g., in cremaster arterioles of wild type mice [135], and with Ginsenoside Rc, e.g., in rat cardiomyocytes after ischemia/reperfusion [136].
The complex IV (cyt c oxidase) transfers the electrons from reduced cyt c molecules to oxygen to generate H2O molecules [137]. Following the ETC, a H+ electrochemical gradient is established across the inner mitochondrial membrane [137] (Figure 2D). Cuprous ion (Cu+) is able to react with O2 and form a peroxide bridge between heme groups and cupric ion (Cu2+); thus, the electrons from cyt c are transferred to the heme groups [138]. The scission of the peroxide bridge occurs as a result of H+ extraction from the mitochondrial matrix and the reduction of Cu2+ to Cu+. H2O molecules are generated as reaction products [138]. The heme a3 site is the O2 binding site in complex IV (Figure 2D). This site can be competitively inhibited by molecules such as H2S, CO, and NO [139,140,141], thus blocking the electron transport from cyt c to O2 and generating increased levels of ROS [142]. The insufficient O2 supply during MI results in increased electron leakage, leading to the impairment of complex IV activity and the accumulation of large amounts of mROS [143,144]. As such, the overproduction of mROS reduces the oxidant scavenging capacity of mitochondrial antioxidants, such as MnSOD [75], Cu/ZnSOD [74], and GSH [78].
In the in vivo MI models, the increased levels of ROS (OH•− and O2•−) are associated with 30% to 50% activity of some subunits of complex I (e.g., ND1), such as cyt b in complex III and the whole IV, thereby explaining the impaired activity of these complexes [143,144,145].
Recently, it has been proved that the hyperphosphorylation of mitochondrial proteins in complex IV in myocardial cells is induced by activating protein kinase A (PKA) signaling pathways, which alters the function of mitochondrial proteins [143] and impairs mitochondrial activity [143]. Cyclic adenosine monophosphate (cAMP) in mitochondria is an important activator of PKA [146], causing a decrease in the complex IV activity of cardiac cells after ischemia/reperfusion injury [143,147] (Figure 2D).
Complex V (ATP synthase) uses the motor force of H+ ions, established in the forehead redox sequences, to synthesize ATP molecules by ADP or phosphate molecules, releasing them into the mitochondrial matrix [148] (Figure 2E). Although complex V does not generate ROS, under pathological conditions, the mitochondrial permeability transition pore (MPTP) is activated by the presence of a large accumulation of ROS synthesized by previous complexes. The main components of MPTP are the voltage-dependent anion channel and the adenine nucleotide translocator. Several other MPTP components, e.g., cyclophilin D may interact with the adenine nucleotide translocator, whereas complex V dimers bind to cyclophilin D from the mitochondrial matrix [149] (Figure 2E).
Also, the phosphorylation of complex V dimers subunit c leads to the opening of MPTP [150], subsequently uncoupling oxidative phosphorylation and the production of mtROS [151]. The opening of MPTP may also induce cell apoptosis or necrosis by releasing pro-apoptotic proteins, such as apoptosis regulator BAX [50,120] (Figure 2E), thus increasing the infarct size after ischemia/reperfusion [152]. In the in vivo animal experiments using dogs with microembolization-induced heart failure, the improved activity or expression of mitochondrial complex V is associated with a preserved ejection fraction of the heart [153].
Figure 2. Schematic diagram of various sources of ROS generation and its effect on mitochondrial activity in CVD (brown arrows point to the enzymatic sources of ROS formation). Oxidized ubiquinol receives two electrons from Fe-S clusters of complexes I (A) [154] and II (B) [122] to form ubiquinol; ubiquinol transports electrons to Fe-S clusters in complex III. Cyt c mediates the transport of electrons from complex III (C) to complex IV (D) [126]. Complex IV delivers the electrons to O2 and forms H2O. Hydrogen ions are pumped by complexes I [154], III [126], and IV [137] from the mitochondrial matrix to the mitochondrial intermembrane space to generate the H+ electrochemical gradient for ATP generation. MnSOD [75], Cu/Zn-SOD [74], CAT [76], GPx [76], TRx [77], and GSH are vital for the scavenging of ROS in the mitochondria. Cu/Zn-SOD, CAT, GPx, thioredoxins, and GSH also play important roles in scavenging ROS in the cytosol [74,76,78,155]. Complex V (ATP synthase) synthesizes ATP molecules [148] (E). Complex V dimers participate in the formation of MPTP, which also includes VDAC, ANT, and CypD. The overproduction of ROS in the mitochondria causes the opening of MPTP, producing a large amount of mtROS [156]. The opening of MPTP can even activate Bax and then lead to the release of cyt c, the activation of caspase 3 [50,120], and apoptotic cell death [152,157] (E).
Figure 2. Schematic diagram of various sources of ROS generation and its effect on mitochondrial activity in CVD (brown arrows point to the enzymatic sources of ROS formation). Oxidized ubiquinol receives two electrons from Fe-S clusters of complexes I (A) [154] and II (B) [122] to form ubiquinol; ubiquinol transports electrons to Fe-S clusters in complex III. Cyt c mediates the transport of electrons from complex III (C) to complex IV (D) [126]. Complex IV delivers the electrons to O2 and forms H2O. Hydrogen ions are pumped by complexes I [154], III [126], and IV [137] from the mitochondrial matrix to the mitochondrial intermembrane space to generate the H+ electrochemical gradient for ATP generation. MnSOD [75], Cu/Zn-SOD [74], CAT [76], GPx [76], TRx [77], and GSH are vital for the scavenging of ROS in the mitochondria. Cu/Zn-SOD, CAT, GPx, thioredoxins, and GSH also play important roles in scavenging ROS in the cytosol [74,76,78,155]. Complex V (ATP synthase) synthesizes ATP molecules [148] (E). Complex V dimers participate in the formation of MPTP, which also includes VDAC, ANT, and CypD. The overproduction of ROS in the mitochondria causes the opening of MPTP, producing a large amount of mtROS [156]. The opening of MPTP can even activate Bax and then lead to the release of cyt c, the activation of caspase 3 [50,120], and apoptotic cell death [152,157] (E).
Ijms 25 03114 g002
Among the various sources of ROS in the heart, mitochondrial complexes I and III are the key players in cardiac pathology. They produce a large amount of ROS, which induces damage to the cellular structures (e.g., the damage to DNA, proteins, and lipids) and even the death of the cardiomyocytes, particularly in diabetes [158]. In conclusion, ROS, which are mainly produced by mitochondrial complexes I and III in cardiomyocytes, play an important role in cardiac pathology.

3.4. Role of Vitamin C as a Scavenger of ROS

Vitamin C is the most known antioxidant that scavenges various types of radicals, e.g., OH•−, H2O2, and O2•− [159,160,161]. Vitamin C either donates one electron to O2•− to produce ASC•− or loses a second electron to form its oxidized form, DHA [159,160,161] (Figure 3A–E). Observational studies demonstrated the antioxidant capacity of vitamin C even when antioxidative enzymes, such as SOD and CAT, are decreased [162,163]. Further, the dietary supplementation of 1000 mg/day for 6 weeks, in the case of stroke-prone spontaneously hypertensive rats, downregulated the expression of O2•− in association with the reduced activity of NOX [164].
Interestingly, vitamin C increases the activity of antioxidative enzymes. For example, small supplementation amounts of 30 μM of vitamin C significantly reduced ROS amounts within 60 min in the isolated mitochondria [165] by increasing the activity of Mn-SOD and GPx [165] (Figure 3A–D).
As we have mentioned in Section 3.1, CAT and Cu/Zn- SOD are key antioxidative enzyme regulators of mitochondrial function and they can be stimulated by vitamin C. An in vivo experiment in diabetic rats showed that the supplementation of vitamin C (50 mg/kg per day for 4 weeks) increased the activity of CAT and Cu/Zn- SOD in the liver [166].
Glutathione (GSH) is a well-known nonenzymic antioxidant with a protective role in the cells. It can be decreased by oxidative stress in in vivo MI animal models [167], whereas its oxidized form (GSSG) is increased in isoproterenol-induced cardiac remodeling, which is also related to oxidative stress [168]. Vitamin C maintains cellular GSH levels within different sites [169,170] (Figure 3A–D). It is worth noting that a supplementation of vitamin C of 200 mg/day for patients aged 33–63 years with Type 2 diabetes was sufficient to significantly increase the activity of SOD by 3% and GPx by 52% in blood samples [171].
Vitamin C (1 μM) also prevents lipid peroxidation by clearing ROS and maintaining the levels of antioxidants [172,173]. Thus, the development of ferroptosis is inhibited [172], allowing us to speculate that the damage ferroptosis causes to cardiomyocytes after MI may be inhibited by vitamin C.
Moreover, vitamin C has an antioxidant role in the treatment and prognosis of MI in animal models. Pigs that received a vitamin C hydrogel after MI surgery showed improved cardiac function, increased vascular density, and reduced fibrosis compared to the untreated group. This improvement was attributed to the suppression of oxidative stress by vitamin C [174]. In another study, MI was induced in rats with isoproterenol, leading to enhanced oxidative stress and elevated levels of inflammatory factors such as tumor necrosis factor-α. The supplementation of vitamin C (250 mg/kg body weight, intragastric injection) reduced the oxidative stress, inflammation, and size of the infarction in the heart [175]. Likewise, under MI conditions, the oral supplementation of vitamin C (250 mg/kg body weight) in rats further confirmed its antioxidant role by preserving cardiac function [176]. This aligns with recent clinical evidence suggesting vitamin C’s potential in reducing oxidative stress and improving outcomes for patients with acute coronary syndromes and those undergoing cardiac surgeries [177].
Although the mechanisms for reducing the cardiovascular risk remain unclear, the role of vitamin C in scavenging ROS [178], enhancing NO production [179], and its potential benefits for microperfusion [177], myocardial protection [79], and hemodynamic stability [180]—especially in critically ill patients [181]—is increasingly recognized.
Thus, despite being controversial, preclinical and clinical studies have demonstrated that vitamin C plays beneficial roles in cardiovascular diseases (either in peri-operative or post-operative interventions) [182]. However, the effects of vitamin C seemed to be dose- and time-dependent. For example, overdoses of vitamin C (≥300 mg/day, Table 1) may increase cardiovascular mortality [183]. This highlights the importance of careful dosage management in therapeutic settings. Additionally, a meta-analysis has linked vitamin C intake levels with an elevated risk of developing kidney stones [184].
In conclusion, vitamin C can directly decrease ROS levels and protect the cardiovascular system from oxidative stress by improving the electron transport chain mitochondrial function. Vitamin C also scavenges mtROS in the cardiovascular system by maintaining the levels of intracellular antioxidants, including Mn-SOD, GPx, CAT, Cu/Zn-SOD, and glutathione. Although vitamin C may be considered a potential effector in disease management strategy, care should be taken concerning its dose and time-dependent administration, particularly at dosages exceeding 300 mg/day.
Figure 3. Overview of the effect of vitamin C on scavenging mitochondria-produced ROS in CVD. Vitamin C in the mitochondria reduces O2•− [159], OH•− [78], and H2O2 [78] produced by mitochondrial complexes I (A), II (B), III (C), IV (D), and V (E), and is oxidized to DHA or ASC•−. Vitamin C is also reported to increase the activity or levels of GSH [78], Mn-SOD [165,185], GPx [165], CAT [166,186], Cu/Zn-SOD [185,186] (AD), and therefore enhance the ability to scavenge ROS in the mitochondria in the CVD condition.
Figure 3. Overview of the effect of vitamin C on scavenging mitochondria-produced ROS in CVD. Vitamin C in the mitochondria reduces O2•− [159], OH•− [78], and H2O2 [78] produced by mitochondrial complexes I (A), II (B), III (C), IV (D), and V (E), and is oxidized to DHA or ASC•−. Vitamin C is also reported to increase the activity or levels of GSH [78], Mn-SOD [165,185], GPx [165], CAT [166,186], Cu/Zn-SOD [185,186] (AD), and therefore enhance the ability to scavenge ROS in the mitochondria in the CVD condition.
Ijms 25 03114 g003
Table 1. The outcome of vitamin C administration in clinical trials of cardiovascular pathology. PCI, percutaneous coronary intervention; MI, myocardial infarction; HF, heart failure; iv, intravenous; SOD, superoxide dismutase; XO, xanthine oxidase.
Table 1. The outcome of vitamin C administration in clinical trials of cardiovascular pathology. PCI, percutaneous coronary intervention; MI, myocardial infarction; HF, heart failure; iv, intravenous; SOD, superoxide dismutase; XO, xanthine oxidase.
PopulationStudy Size (n)Age (Years)Intervention
(Doses of Vitamin C)
Trial
Duration
OutcomesReference
Male with the first MI180<65Habitual3 monthsLow plasma concentration had no benefit on MI[35]
Patients with sepsis and ARDS present for less than 24 h167 50 mg/kg in dextrose 5% in water, n = 84 (iv)96 hNo improvement of organ dysfunction sores, inflammation and vascular injury[36]
Male with no prevalent HF391960–79Habitual11 yearsHigh plasma concentration reduced the risk of HF with and without myocardial infarction[39]
Postmenopausal women with
diabetes
192355–69≥300 mg/d (orally)a mean time of 15 yearsHigher cardiovascular
mortality
[183]
Healthy adults13421a mean of 61320–1110 mg/d (orally)a mean time of 11 yearsLower cardiovascular mortality[187]
Patients with clinically stable class I or II effort angina pectoris56a mean of 67
(50–84) years
16.6 mg/min over 1 h before PCI (iv)-Microcirculatory reperfusion improved; Oxidative stress decreased[188]
Patients with asymptomatic adults with elevated coronary
calcium scores
100550–70Combination of vitamin C 1 g, vitamin E 1000 U and atorvastatin 20 mg daily (orally)a mean duration of 4.3 yearsLevels of total cholesterol, low-density lipoprotein cholesterol and triglycerides reduced[189]
Patients with MI800A mean of 621000 mg in 12 h (iv) followed by 1200 mg/day combined with vitamin E 600 mg/day (orally)For 30 daysIn-hospital cardiac mortality, non-fatal new myocardial infarction, shock/pulmonary edema, etc. decreased.[190]
Adults without MI or stroke14641≥50500 mg/day (orally)For 8 yearsNo benefit on MI, stroke or cardiovascular mortality.[191]
Patients with MI8453.1 ± 11.2 in the antioxidant treatment group,
55.1 ± 8.4 years in the control group
250 mL 20% mannitol (iv) over the first hour, 1000 mg of vitamin C in 500 mL 5% glucose (iv) over the first 4 h-Short term (e.g., cardiogenic shock), and long term (e.g., left ventricular insufficiency) complications decreased.[192]
Patients with MI120-500 mg, twice per day after MI (orally)5 daysLevels of SOD and total thiols increased Levels of XO and malondialdehyde decreased.[193]
Male with MI2160.5 ± 6.82000 mg after MI (orally)-Oxidative stress decreased.[194]

4. Conclusions

The beneficial role of vitamin C in the cardiovascular system remains controversial, despite solid evidence demonstrating that vitamin C reduces excessive ROS formation. The non-linearity between vitamin C supplementation levels and variable ROS sources seems to shift the oxidative-redox events in the cardiac cell, which might impair mitochondrial function, highlighting the importance of further studies at the molecular level. The controversial effects of vitamin C on cell culture do not translate into side effects in clinical studies, thus polarizing the research outcome and making it difficult to draw clinical guidelines for vitamin C implementation as a therapeutic medicine. Although vitamin C demonstrates a strong antioxidant function, at both the cellular and organ levels, further studies are needed to provide cumulative time-dependent dose data to clarify the role of vitamin C in the prevention and/or cure of cardiovascular diseases. The controversial risks of vitamin C administration in human studies, particularly in terms of kidney stone formation at chronically high doses, underscore the necessity of developing future combination therapies for cardiovascular diseases.
An interesting future strategy concerns the establishment of an effective role of vitamin C in ferroptosis, since the interaction between ROS, iron balance, and lipid peroxidation in the myocardium is critical for regulating the fate of cells. This may provide useful insights into new therapeutic targets for the management of ischemic heart diseases.

Author Contributions

Conceptualization: M.R.; writing—original draft preparation: H.Z.; writing—review and editing, H.Z., Y.X., E.A.L. and M.R. All authors have read and agreed to the published version of the manuscript.

Funding

H.Z. (CSC no. 202108080145) and Y.X. (CSC no. 202108080025) received scholarship funding from the China Scholarship Council. Elisa Liehn was supported by the Ministry of Research, Innovation and Digitization, CNCS—UEFISCDI, project number PN-III-P4-PCE-2021-1680, within PNCDI III.

Acknowledgments

Special thanks to Charles Anthony Da Costa for reviewing and correcting the English language and grammar in our manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hartikainen, T.S.; Sörensen, N.A.; Haller, P.M.; Goßling, A.; Lehmacher, J.; Zeller, T.; Blankenberg, S.; Westermann, D.; Neumann, J.T. Clinical application of the 4th Universal Definition of Myocardial Infarction. Eur. Heart J. 2020, 41, 2209–2216. [Google Scholar] [CrossRef]
  2. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1736–1788. [CrossRef]
  3. Roth, G.A.; Johnson, C.; Abajobir, A.; Abd-Allah, F.; Abera, S.F.; Abyu, G.; Ahmed, M.; Aksut, B.; Alam, T.; Alam, K.; et al. Global, Regional, and National Burden of Cardiovascular Diseases for 10 Causes, 1990 to 2015. J. Am. Coll. Cardiol. 2017, 70, 1–25. [Google Scholar] [CrossRef]
  4. Barnett, R. Acute myocardial infarction. Lancet 2019, 393, 2580. [Google Scholar] [CrossRef]
  5. Fox, K.A.A.; Steg, P.G.; Eagle, K.A.; Goodman, S.G.; Anderson, F.A.; Granger, C.B.; Flather, M.D.; Budaj, A.; Quill, A.; Gore, J.M. Decline in rates of death and heart failure in acute coronary syndromes, 1999–2006. JAMA 2007, 297, 1892–1900. [Google Scholar] [CrossRef]
  6. Furman, M.I.; Dauerman, H.L.; Goldberg, R.J.; Yarzebski, J.; Lessard, D.; Gore, J.M. Twenty-two year (1975 to 1997) trends in the incidence, in-hospital and long-term case fatality rates from initial Q-wave and non-Q-wave myocardial infarction: A multi-hospital, community-wide perspective. J. Am. Coll. Cardiol. 2001, 37, 1571–1580. [Google Scholar] [CrossRef] [PubMed]
  7. Liew, R.; Sulfi, S.; Ranjadayalan, K.; Cooper, J.; Timmis, A.D. Declining case fatality rates for acute myocardial infarction in South Asian and white patients in the past 15 years. Heart 2006, 92, 1030–1034. [Google Scholar] [CrossRef]
  8. Mandelzweig, L.; Battler, A.; Boyko, V.; Bueno, H.; Danchin, N.; Filippatos, G.; Gitt, A.; Hasdai, D.; Hasin, Y.; Marrugat, J.; et al. The second Euro Heart Survey on acute coronary syndromes: Characteristics, treatment, and outcome of patients with ACS in Europe and the Mediterranean Basin in 2004. Eur. Heart J. 2006, 27, 2285–2293. [Google Scholar] [CrossRef] [PubMed]
  9. Latif, F.; Uyeda, L.; Edson, R.; Bhatt, D.L.; Goldman, S.; Holmes, D.R.; Rao, S.V.; Shunk, K.; Aggarwal, K.; Uretsky, B.; et al. Stent-Only Versus Adjunctive Balloon Angioplasty Approach for Saphenous Vein Graft Percutaneous Coronary Intervention: Insights from DIVA Trial. Circ. Cardiovasc. Interv. 2020, 13, e008494. [Google Scholar] [CrossRef] [PubMed]
  10. Niccoli, G.; Kharbanda, R.K.; Crea, F.; Banning, A.P. No-reflow: Again prevention is better than treatment. Eur. Heart J. 2010, 31, 2449–2455. [Google Scholar] [CrossRef]
  11. Fan, Q.; Tao, R.; Zhang, H.; Xie, H.; Lu, L.; Wang, T.; Su, M.; Hu, J.; Zhang, Q.; Chen, Q.; et al. Dectin-1 Contributes to Myocardial Ischemia/Reperfusion Injury by Regulating Macrophage Polarization and Neutrophil Infiltration. Circulation 2019, 139, 663–678. [Google Scholar] [CrossRef] [PubMed]
  12. Peet, C.; Ivetic, A.; Bromage, D.I.; Shah, A.M. Cardiac monocytes and macrophages after myocardial infarction. Cardiovasc. Res. 2020, 116, 1101–1112. [Google Scholar] [CrossRef] [PubMed]
  13. Sun, X.; Feng, Y.; Gong, C.; Bao, X.; Wei, Z.; Chang, L.; Chen, H.; Xu, B. Hypertension-Driven Regulatory T-Cell Perturbations Accelerate Myocardial Ischemia-Reperfusion Injury. Hypertension 2023, 80, 2046–2058. [Google Scholar] [CrossRef] [PubMed]
  14. Mathes, D.; Weirather, J.; Nordbeck, P.; Arias-Loza, A.-P.; Burkard, M.; Pachel, C.; Kerkau, T.; Beyersdorf, N.; Frantz, S.; Hofmann, U. CD4+ Foxp3+ T-cells contribute to myocardial ischemia-reperfusion injury. J. Mol. Cell Cardiol. 2016, 101, 99–105. [Google Scholar] [CrossRef] [PubMed]
  15. Xiong, W.; Zhou, R.; Qu, Y.; Yang, Y.; Wang, Z.; Song, N.; Liang, R.; Qian, J. Dexmedetomidine preconditioning mitigates myocardial ischemia/reperfusion injury via inhibition of mast cell degranulation. Biomed. Pharmacother. 2021, 141, 111853. [Google Scholar] [CrossRef] [PubMed]
  16. Maxwell, S.R. Anti-oxidant therapy: Does it have a role in the treatment of human disease? Expert Opin. Investig. Drugs 1997, 6, 211–236. [Google Scholar] [CrossRef]
  17. Lim, S.L.; Lam, C.S.P.; Segers, V.F.M.; Brutsaert, D.L.; De Keulenaer, G.W. Cardiac endothelium-myocyte interaction: Clinical opportunities for new heart failure therapies regardless of ejection fraction. Eur. Heart J. 2015, 36, 2050–2060. [Google Scholar] [CrossRef]
  18. 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]
  19. Guo, Y.; Pu, W.T. Cardiomyocyte Maturation: New Phase in Development. Circ. Res. 2020, 126, 1086–1106. [Google Scholar] [CrossRef]
  20. Wang, J.; Huang, P.; Yu, Q.; Lu, J.; Liu, P.; Yang, Y.; Feng, Z.; Cai, J.; Yang, G.; Yuan, H.; et al. Epilepsy and long-term risk of arrhythmias. Eur. Heart J. 2023, 44, 3374–3382. [Google Scholar] [CrossRef]
  21. Luxán, G.; Stewen, J.; Díaz, N.; Kato, K.; Maney, S.K.; Aravamudhan, A.; Berkenfeld, F.; Nagelmann, N.; Drexler, H.C.; Zeuschner, D.; et al. Endothelial EphB4 maintains vascular integrity and transport function in adult heart. Elife 2019, 8, e45863. [Google Scholar] [CrossRef]
  22. Neubauer, S. The failing heart—An engine out of fuel. N. Engl. J. Med. 2007, 356, 1140–1151. [Google Scholar] [CrossRef]
  23. Bisaccia, G.; Ricci, F.; Gallina, S.; Di Baldassarre, A.; Ghinassi, B. Mitochondrial Dysfunction and Heart Disease: Critical Appraisal of an Overlooked Association. Int. J. Mol. Sci. 2021, 22, 614. [Google Scholar] [CrossRef] [PubMed]
  24. Faust, O.; Hong, W.; Loh, H.W.; Xu, S.; Tan, R.-S.; Chakraborty, S.; Barua, P.D.; Molinari, F.; Acharya, U.R. Heart rate variability for medical decision support systems: A review. Comput. Biol. Med. 2022, 145, 105407. [Google Scholar] [CrossRef] [PubMed]
  25. Behl, T.; Bungau, S.; Kumar, K.; Zengin, G.; Khan, F.; Kumar, A.; Kaur, R.; Venkatachalam, T.; Tit, D.M.; Vesa, C.M.; et al. Pleotropic Effects of Polyphenols in Cardiovascular System. Biomed. Pharmacother. 2020, 130, 110714. [Google Scholar] [CrossRef]
  26. Miyazawa, T.; Burdeos, G.C.; Itaya, M.; Nakagawa, K.; Miyazawa, T. Vitamin E: Regulatory Redox Interactions. IUBMB Life 2019, 71, 430–441. [Google Scholar] [CrossRef]
  27. Berretta, M.; Quagliariello, V.; Maurea, N.; Di Francia, R.; Sharifi, S.; Facchini, G.; Rinaldi, L.; Piezzo, M.; Manuela, C.; Nunnari, G.; et al. Multiple Effects of Ascorbic Acid against Chronic Diseases: Updated Evidence from Preclinical and Clinical Studies. Antioxidants 2020, 9, 1182. [Google Scholar] [CrossRef]
  28. Salonen, R.M.; Nyyssönen, K.; Kaikkonen, J.; Porkkala-Sarataho, E.; Voutilainen, S.; Rissanen, T.H.; Tuomainen, T.-P.; Valkonen, V.-P.; Ristonmaa, U.; Lakka, H.-M.; et al. Six-year effect of combined vitamin C and E supplementation on atherosclerotic progression: The Antioxidant Supplementation in Atherosclerosis Prevention (ASAP) Study. Circulation 2003, 107, 947–953. [Google Scholar] [CrossRef]
  29. Bechara, N.; Flood, V.M.; Gunton, J.E. A Systematic Review on the Role of Vitamin C in Tissue Healing. Antioxidants 2022, 11, 1605. [Google Scholar] [CrossRef] [PubMed]
  30. Tomşa, A.M.; Răchişan, A.L.; Pandrea, S.L.; Benea, A.; Uifălean, A.; Toma, C.; Popa, R.; Pârvu, A.E.; Junie, L.M. Curcumin and Vitamin C Attenuate Gentamicin-Induced Nephrotoxicity by Modulating Distinctive Reactive Species. Metabolites 2022, 13, 49. [Google Scholar] [CrossRef]
  31. Steinberg, D.; Parthasarathy, S.; Carew, T.E.; Khoo, J.C.; Witztum, J.L. Beyond cholesterol. Modifications of low-density lipoprotein that increase its atherogenicity. N. Engl. J. Med. 1989, 320, 915–924. [Google Scholar]
  32. Carr, A.; Frei, B. Does vitamin C act as a pro-oxidant under physiological conditions? FASEB J. 1999, 13, 1007–1024. [Google Scholar] [CrossRef]
  33. Pfister, R.; Sharp, S.J.; Luben, R.; Wareham, N.J.; Khaw, K.T. Plasma vitamin C predicts incident heart failure in men and women in European Prospective Investigation into Cancer and Nutrition-Norfolk prospective study. Am. Heart J. 2011, 162, 246–253. [Google Scholar] [CrossRef]
  34. Nyyssonen, K.; Parviainen, M.T.; Salonen, R.; Tuomilehto, J.; Salonen, J.T. Vitamin C deficiency and risk of myocardial infarction: Prospective population study of men from eastern Finland. BMJ 1997, 314, 634–638. [Google Scholar] [CrossRef] [PubMed]
  35. Riemersma, R.A.; Carruthers, K.F.; Elton, R.A.; Fox, K.A. Vitamin C and the risk of acute myocardial infarction. Am. J. Clin. Nutr. 2000, 71, 1181–1186. [Google Scholar] [CrossRef]
  36. Fowler, A.A., 3rd; Truwit, J.D.; Hite, R.D.; Morris, P.E.; DeWilde, C.; Priday, A.; Fisher, B.; Thacker, L.R., 2nd; Natarajan, R.; Brophy, D.F.; et al. Effect of Vitamin C Infusion on Organ Failure and Biomarkers of Inflammation and Vascular Injury in Patients with Sepsis and Severe Acute Respiratory Failure: The CITRIS-ALI Randomized Clinical Trial. JAMA 2019, 322, 1261–1270. [Google Scholar] [CrossRef] [PubMed]
  37. Kushi, L.H.; Folsom, A.R.; Prineas, R.J.; Mink, P.J.; Wu, Y.; Bostick, R.M. Dietary antioxidant vitamins and death from coronary heart disease in postmenopausal women. N. Engl. J. Med. 1996, 334, 1156–1162. [Google Scholar] [CrossRef] [PubMed]
  38. Loria, C.M.; Klag, M.J.; Caulfield, L.E.; Whelton, P.K. Vitamin C status and mortality in US adults. Am. J. Clin. Nutr. 2000, 72, 139–145. [Google Scholar] [CrossRef] [PubMed]
  39. Wannamethee, S.G.; Bruckdorfer, K.R.; Shaper, A.G.; Papacosta, O.; Lennon, L.; Whincup, P.H. Plasma vitamin C, but not vitamin E, is associated with reduced risk of heart failure in older men. Circ. Heart Fail. 2013, 6, 647–654. [Google Scholar] [CrossRef] [PubMed]
  40. Bánhegyi, G.; Braun, L.; Csala, M.; Puskás, F.; Mandl, J. Ascorbate metabolism and its regulation in animals. Free Radic. Biol. Med. 1997, 23, 793–803. [Google Scholar] [CrossRef] [PubMed]
  41. Deutsch, J.C. Dehydroascorbic acid. J. Chromatogr. A 2000, 881, 299–307. [Google Scholar] [CrossRef] [PubMed]
  42. Bode, A.M.; Cunningham, L.; Rose, R.C. Spontaneous decay of oxidized ascorbic acid (dehydro-L-ascorbic acid) evaluated by high-pressure liquid chromatography. Clin. Chem. 1990, 36, 1807–1809. [Google Scholar] [CrossRef] [PubMed]
  43. Linster, C.L.; Van Schaftingen, E. Vitamin C. Biosynthesis, recycling and degradation in mammals. FEBS J. 2007, 274, 1–22. [Google Scholar] [CrossRef]
  44. Kanfer, J.; Ashwell, G.; Burns, J.J. Formation of L-lyxonic and L-xylonic acids from L-ascorbic acid in rat kidney. J. Biol. Chem. 1960, 235, 2518–2521. [Google Scholar] [CrossRef]
  45. Buettner, G.R. Ascorbate autoxidation in the presence of iron and copper chelates. Free Radic. Res. Commun. 1986, 1, 349–353. [Google Scholar] [CrossRef]
  46. Hata, R.; Senoo, H. L-ascorbic acid 2-phosphate stimulates collagen accumulation, cell proliferation, and formation of a three-dimensional tissuelike substance by skin fibroblasts. J. Cell Physiol. 1989, 138, 8–16. [Google Scholar] [CrossRef] [PubMed]
  47. Frikke-Schmidt, H.; Lykkesfeldt, J. Keeping the intracellular vitamin C at a physiologically relevant level in endothelial cell culture. Anal. Biochem. 2010, 397, 135–137. [Google Scholar] [CrossRef]
  48. Lykkesfeldt, J. Determination of ascorbic acid and dehydroascorbic acid in biological samples by high-performance liquid chromatography using subtraction methods: Reliable reduction with tris[2-carboxyethyl]phosphine hydrochloride. Anal. Biochem. 2000, 282, 89–93. [Google Scholar] [CrossRef]
  49. Nathan, C.; Cunningham-Bussel, A. Beyond oxidative stress: An immunologist’s guide to reactive oxygen species. Nat. Rev. Immunol. 2013, 13, 349–361. [Google Scholar] [CrossRef]
  50. Ramachandra, C.J.; Hernandez-Resendiz, S.; Crespo-Avilan, G.E.; Lin, Y.-H.; Hausenloy, D.J. Mitochondria in acute myocardial infarction and cardioprotection. EBioMedicine 2020, 57, 102884. [Google Scholar] [CrossRef]
  51. Lassegue, B.; Martin, A.S.; Griendling, K.K. Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system. Circ. Res. 2012, 110, 1364–1390. [Google Scholar] [CrossRef]
  52. Gurusamy, N.; Mukherjee, S.; Lekli, I.; Bearzi, C.; Bardelli, S.; Das, D.K. Inhibition of ref-1 stimulates the production of reactive oxygen species and induces differentiation in adult cardiac stem cells. Antioxid. Redox Signal. 2009, 11, 589–600. [Google Scholar] [CrossRef] [PubMed]
  53. Buggisch, M.; Ateghang, B.; Ruhe, C.; Strobel, C.; Lange, S.; Wartenberg, M.; Sauer, H. Stimulation of ES-cell-derived cardiomyogenesis and neonatal cardiac cell proliferation by reactive oxygen species and NADPH oxidase. J. Cell Sci. 2007, 120, 885–894. [Google Scholar] [CrossRef]
  54. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
  55. Manalo, D.J.; Rowan, A.; Lavoie, T.; Natarajan, L.; Kelly, B.D.; Ye, S.Q.; Garcia, J.G.N.; Semenza, G.L. Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. Blood 2005, 105, 659–669. [Google Scholar] [CrossRef]
  56. Yun, J.; Rocic, P.; Pung, Y.F.; Belmadani, S.; Carrao, A.C.R.; Ohanyan, V.; Chilian, W.M. Redox-dependent mechanisms in coronary collateral growth: The “redox window” hypothesis. Antioxid. Redox Signal. 2009, 11, 1961–1974. [Google Scholar] [CrossRef]
  57. Su, B.; Bu, S.D.; Kong, B.H.; Dai, R.X.; Su, Q. Cystatin C alleviates H2O2-induced H9c2 cell injury. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 6360–6370. [Google Scholar]
  58. Jiang, F.; Xu, X.R.; Li, W.M.; Xia, K.; Wang, L.F.; Yang, X.C. Monotropein alleviates H2O2-induced inflammation, oxidative stress and apoptosis via NF-kappaB/AP-1 signaling. Mol. Med. Rep. 2020, 22, 4828–4836. [Google Scholar] [CrossRef] [PubMed]
  59. Othmene, Y.B.; Monceaux, K.; Karoui, A.; Salem, I.B.; Belhadef, A.; Abid-Essefi, S.; Lemaire, C. Tebuconazole induces ROS-dependent cardiac cell toxicity by activating DNA damage and mitochondrial apoptotic pathway. Ecotoxicol. Environ. Saf. 2020, 204, 111040. [Google Scholar] [CrossRef]
  60. Elorza, A.A.; Soffia, J.P. mtDNA Heteroplasmy at the Core of Aging-Associated Heart Failure. An Integrative View of OXPHOS and Mitochondrial Life Cycle in Cardiac Mitochondrial Physiology. Front. Cell Dev. Biol. 2021, 9, 625020. [Google Scholar] [CrossRef]
  61. Francois, A.; Canella, A.; Marcho, L.M.; Stratton, M.S. Protein acetylation in cardiac aging. J. Mol. Cell Cardiol. 2021, 157, 90–97. [Google Scholar] [CrossRef]
  62. Moris, D.; Spartalis, M.; Tzatzaki, E.; Spartalis, E.; Karachaliou, G.-S.; Triantafyllis, A.S.; Karaolanis, G.I.; Tsilimigras, D.I.; Theocharis, S. The role of reactive oxygen species in myocardial redox signaling and regulation. Ann. Transl. Med. 2017, 5, 324. [Google Scholar] [CrossRef]
  63. 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]
  64. Montezano, A.C.; Dulak-Lis, M.; Tsiropoulou, S.; Harvey, A.; Briones, A.M.; Touyz, R.M. Oxidative stress and human hypertension: Vascular mechanisms, biomarkers, and novel therapies. Can. J. Cardiol. 2015, 31, 631–641. [Google Scholar] [CrossRef]
  65. Guzik, T.J.; Touyz, R.M. Oxidative Stress, Inflammation, and Vascular Aging in Hypertension. Hypertension 2017, 70, 660–667. [Google Scholar] [CrossRef] [PubMed]
  66. Bernal, M.E.; Varon, J.; Acosta, P.; Montagnier, L. Oxidative stress in critical care medicine. Int. J. Clin. Pract. 2010, 64, 1480–1488. [Google Scholar] [CrossRef]
  67. Weiss, J.N.; Korge, P.; Honda, H.M.; Ping, P. Role of the mitochondrial permeability transition in myocardial disease. Circ. Res. 2003, 93, 292–301. [Google Scholar] [CrossRef] [PubMed]
  68. Yu, Y.; Yan, Y.; Niu, F.; Wang, Y.; Chen, X.; Su, G.; Liu, Y.; Zhao, X.; Qian, L.; Liu, P.; et al. Ferroptosis: A cell death connecting oxidative stress, inflammation and cardiovascular diseases. Cell Death Discov. 2021, 7, 193. [Google Scholar] [CrossRef]
  69. Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol. 2016, 26, 165–176. [Google Scholar] [CrossRef]
  70. Park, T.-J.; Park, J.H.; Lee, G.S.; Lee, J.-Y.; Shin, J.H.; Kim, M.W.; Kim, Y.S.; Kim, J.-Y.; Oh, K.-J.; Han, B.-S.; et al. Quantitative proteomic analyses reveal that GPX4 downregulation during myocardial infarction contributes to ferroptosis in cardiomyocytes. Cell Death Dis. 2019, 10, 835. [Google Scholar] [CrossRef]
  71. Chung, B.; Wang, Y.; Thiel, M.; Rostami, F.; Rogoll, A.; Hirsch, V.G.; Malik, Z.; Bührke, A.; Bär, C.; Klintschar, M.; et al. Pre-emptive iron supplementation prevents myocardial iron deficiency and attenuates adverse remodelling after myocardial infarction. Cardiovasc. Res. 2023, 119, 1969–1980. [Google Scholar] [CrossRef]
  72. Ju, J.; Li, X.-M.; Zhao, X.-M.; Li, F.-H.; Wang, S.-C.; Wang, K.; Li, R.-F.; Zhou, L.-Y.; Liang, L.; Wang, Y.; et al. Circular RNA FEACR inhibits ferroptosis and alleviates myocardial ischemia/reperfusion injury by interacting with NAMPT. J. Biomed. Sci. 2023, 30, 45. [Google Scholar] [CrossRef] [PubMed]
  73. Forman, H.J.; Bernardo, A.; Davies, K.J.A. What is the concentration of hydrogen peroxide in blood and plasma? Arch. Biochem. Biophys. 2016, 603, 48–53. [Google Scholar] [CrossRef]
  74. Chen, Z.; Oberley, T.D.; Ho, Y.-S.; Chua, C.C.; Siu, B.; Hamdy, R.C.; Epstein, C.J.; Chua, B.H. Overexpression of CuZnSOD in coronary vascular cells attenuates myocardial ischemia/reperfusion injury. Free Radic. Biol. Med. 2000, 29, 589–596. [Google Scholar] [CrossRef] [PubMed]
  75. Long, B.; Gan, T.-Y.; Zhang, R.-C.; Zhang, Y.-H. miR-23a Regulates Cardiomyocyte Apoptosis by Targeting Manganese Superoxide Dismutase. Mol. Cells 2017, 40, 542–549. [Google Scholar] [CrossRef]
  76. Simmons, T.W.; Jamall, I.S. Relative importance of intracellular glutathione peroxidase and catalase in vivo for prevention of peroxidation to the heart. Cardiovasc. Res. 1989, 23, 774–779. [Google Scholar] [CrossRef] [PubMed]
  77. Huang, Q.; Zhou, H.J.; Zhang, H.; Huang, Y.; Hinojosa-Kirschenbaum, F.; Fan, P.; Yao, L.; Belardinelli, L.; Tellides, G.; Giordano, F.J.; et al. Thioredoxin-2 inhibits mitochondrial reactive oxygen species generation and apoptosis stress kinase-1 activity to maintain cardiac function. Circulation 2015, 131, 1082–1097. [Google Scholar] [CrossRef] [PubMed]
  78. Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef] [PubMed]
  79. Xu, Y.; Zheng, H.; Nilcham, P.; Bucur, O.; Vogt, F.; Slabu, I.; Liehn, E.A.; Rusu, M. Vitamin C Regulates the Profibrotic Activity of Fibroblasts in In Vitro Replica Settings of Myocardial Infarction. Int. J. Mol. Sci. 2023, 24, 8379. [Google Scholar] [CrossRef]
  80. 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]
  81. Kaludercic, N.; Mialet-Perez, J.; Paolocci, N.; Parini, A.; Di Lisa, F. Monoamine oxidases as sources of oxidants in the heart. J. Mol. Cell Cardiol. 2014, 73, 34–42. [Google Scholar] [CrossRef] [PubMed]
  82. Johar, S.; Cave, A.C.; Narayanapanicker, A.; Grieve, D.J.; Shah, A.M.; 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]
  83. Cau, S.B.; Guimaraes, D.A.; Rizzi, E.; Ceron, C.S.; Gerlach, R.F.; Tanus-Santos, J.E. The Nuclear Factor kappaB Inhibitor Pyrrolidine Dithiocarbamate Prevents Cardiac Remodelling and Matrix Metalloproteinase-2 Up-Regulation in Renovascular Hypertension. Basic Clin. Pharmacol. Toxicol. 2015, 117, 234–241. [Google Scholar] [CrossRef]
  84. Ryu, H.M.; Kim, Y.J.; Oh, E.J.; Oh, S.H.; Choi, J.Y.; Cho, J.H.; Kim, C.D.; Park, S.H.; Kim, Y.L. Hypoxanthine induces cholesterol accumulation and incites atherosclerosis in apolipoprotein E-deficient mice and cells. J. Cell. Mol. Med. 2016, 20, 2160–2172. [Google Scholar] [CrossRef] [PubMed]
  85. Zhou, H.; Wang, J.; Hu, S.; Zhu, H.; Toanc, S.; Ren, J. BI1 alleviates cardiac microvascular ischemia-reperfusion injury via modifying mitochondrial fission and inhibiting XO/ROS/F-actin pathways. J. Cell. Physiol. 2019, 234, 5056–5069. [Google Scholar] [CrossRef] [PubMed]
  86. Bedard, K.; Krause, K.-H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef]
  87. Zhang, M.; Prosser, B.L.; Bamboye, M.A.; Gondim, A.N.; Santos, C.X.; Martin, D.; Ghigo, A.; Perino, A.; Brewer, A.C.; Ward, C.W. Contractile function during angiotensin-II activation: Increased Nox2 activity modulates cardiac calcium handling via phospholamban phosphorylation. J. Am. Coll. Cardiol. 2015, 66, 261–272. [Google Scholar] [CrossRef]
  88. Zhang, M.; Perino, A.; Ghigo, A.; Hirsch, E.; Shah, A.M. NADPH oxidases in heart failure: Poachers or gamekeepers? Antioxid. Redox Signal. 2013, 18, 1024–1041. [Google Scholar] [CrossRef]
  89. Zhang, M.; Brewer, A.C.; Schröder, K.; Santos, C.X.; Grieve, D.J.; Wang, M.; Anilkumar, N.; Yu, B.; Dong, X.; Walker, S.J. NADPH oxidase-4 mediates protection against chronic load-induced stress in mouse hearts by enhancing angiogenesis. Proc. Natl. Acad. Sci. USA 2010, 107, 18121–18126. [Google Scholar] [CrossRef]
  90. Smyrnias, I.; Zhang, X.; Zhang, M.; Murray, T.V.; Brandes, R.P.; Schröder, K.; Brewer, A.C.; Shah, A.M. Nicotinamide adenine dinucleotide phosphate oxidase-4–dependent upregulation of nuclear factor erythroid–derived 2-like 2 protects the heart during chronic pressure overload. Hypertension 2015, 65, 547–553. [Google Scholar] [CrossRef]
  91. Brewer, A.C.; Murray, T.V.; Arno, M.; Zhang, M.; Anilkumar, N.P.; Mann, G.E.; Shah, A.M. Nox4 regulates Nrf2 and glutathione redox in cardiomyocytes in vivo. Free Radic. Biol. Med. 2011, 51, 205–215. [Google Scholar] [CrossRef] [PubMed]
  92. Sciarretta, S.; Volpe, M.; Sadoshima, J. NOX4 regulates autophagy during energy deprivation. Autophagy 2014, 10, 699–701. [Google Scholar] [CrossRef] [PubMed]
  93. Nomura, J.; Busso, N.; Ives, A.; Matsui, C.; Tsujimoto, S.; Shirakura, T.; Tamura, M.; Kobayashi, T.; So, A.; Yamanaka, Y. Xanthine oxidase inhibition by febuxostat attenuates experimental atherosclerosis in mice. Sci. Rep. 2014, 4, 4554. [Google Scholar] [CrossRef]
  94. 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]
  95. Okazaki, T.; Otani, H.; Shimazu, T.; Yoshioka, K.; Fujita, M.; Katano, T.; Ito, S.; Iwasaka, T. Reversal of inducible nitric oxide synthase uncoupling unmasks tolerance to ischemia/reperfusion injury in the diabetic rat heart. J. Mol. Cell. Cardiol. 2011, 50, 534–544. [Google Scholar] [PubMed]
  96. Gottlieb, R.A. Cytochrome P450: Major player in reperfusion injury. Arch. Biochem. Biophys. 2003, 420, 262–267. [Google Scholar] [PubMed]
  97. Granville, D.; Tashakkor, B.; Takeuchi, C.; Gustafsson, A.B.; Huang, C.; Sayen, M.R.; Wentworth, P., Jr.; Yeager, M.; Gottlieb, R.A. Reduction of ischemia and reperfusion-induced myocardial damage by cytochrome P450 inhibitors. Proc. Natl. Acad. Sci. USA 2004, 101, 1321–1326. [Google Scholar] [CrossRef]
  98. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef]
  99. Rupp, H.; Dhalla, K.S.; Dhalla, N.S. Mechanisms of cardiac cell damage due to catecholamines: Significance of drugs regulating central sympathetic outflow. J. Cardiovasc. Pharmacol. 1994, 24, S16–S24. [Google Scholar] [CrossRef]
  100. Zhang, G.-X.; Kimura, S.; Nishiyama, A.; Shokoji, T.; Rahman, M.; Yao, L.; Nagai, Y.; Fujisawa, Y.; Miyatake, A.; Abe, Y. Cardiac oxidative stress in acute and chronic isoproterenol-infused rats. Cardiovasc. Res. 2005, 65, 230–238. [Google Scholar] [CrossRef]
  101. Oknińska, M.; Zambrowska, Z.; Zajda, K.; Paterek, A.; Brodaczewska, K.; Mackiewicz, U.; Szczylik, C.; Torbicki, A.; Kieda, C.; Mączewski, M. Right ventricular myocardial oxygen tension is reduced in monocrotaline-induced pulmonary hypertension in the rat and restored by myo-inositol trispyrophosphate. Sci. Rep. 2021, 11, 18002. [Google Scholar] [CrossRef]
  102. Balachander, K.; Priyadharsini, J.V.; Paramasivam, A. Role of exosomal mitochondria in cardiovascular diseases. Hypertens. Res. 2023, 46, 812–813. [Google Scholar] [CrossRef]
  103. Ramaccini, D.; Montoya-Uribe, V.; Aan, F.J.; Modesti, L.; Potes, Y.; Wieckowski, M.R.; Krga, I.; Glibetić, M.; Pinton, P.; Giorgi, C.; et al. Mitochondrial Function and Dysfunction in Dilated Cardiomyopathy. Front. Cell Dev. Biol. 2020, 8, 624216. [Google Scholar] [CrossRef]
  104. Wagner, M.; Bertero, E.; Nickel, A.; Kohlhaas, M.; Gibson, G.E.; Heggermont, W.; Heymans, S.; Maack, C. Selective NADH communication from α-ketoglutarate dehydrogenase to mitochondrial transhydrogenase prevents reactive oxygen species formation under reducing conditions in the heart. Basic Res. Cardiol. 2020, 115, 53. [Google Scholar] [CrossRef]
  105. Edalat, A.; Schulte-Mecklenbeck, P.; Bauer, C.; Undank, S.; Krippeit-Drews, P.; Drews, G.; Düfer, M. Mitochondrial succinate dehydrogenase is involved in stimulus-secretion coupling and endogenous ROS formation in murine beta cells. Diabetologia 2015, 58, 1532–1541. [Google Scholar] [CrossRef]
  106. Quinlan, C.L.; Gerencser, A.A.; Treberg, J.R.; Brand, M.D. The mechanism of superoxide production by the antimycin-inhibited mitochondrial Q-cycle. J. Biol. Chem. 2011, 286, 31361–31372. [Google Scholar] [CrossRef]
  107. Quinlan, C.L.; Perevoschikova, I.V.; Goncalves, R.L.S.; Hey-Mogensen, M.; Brand, M.D. The determination and analysis of site-specific rates of mitochondrial reactive oxygen species production. Methods Enzym. 2013, 526, 189–217. [Google Scholar]
  108. Quinlan, C.L.; Orr, A.L.; Perevoshchikova, I.V.; Treberg, J.R.; Ackrell, B.A.; Brand, M.D. Mitochondrial complex II can generate reactive oxygen species at high rates in both the forward and reverse reactions. J. Biol. Chem. 2012, 287, 27255–27264. [Google Scholar] [CrossRef] [PubMed]
  109. Martínez-Reyes, I.; Cardona, L.R.; Kong, H.; Vasan, K.; McElroy, G.S.; Werner, M.; Kihshen, H.; Reczek, C.R.; Weinberg, S.E.; Gao, P.; et al. Mitochondrial ubiquinol oxidation is necessary for tumour growth. Nature 2020, 585, 288–292. [Google Scholar] [CrossRef] [PubMed]
  110. Murphy, P.D. The Four Elements and the Recovery of Referentiality in Ecocriticism, Introduction. In Ecocritical Exploration in Literary and Cultural Studies: Fences, Boundaries, and Fields; Rowman & Littlefield: New York, NY, USA, 2009; pp. 1–13. [Google Scholar]
  111. Kussmaul, L.; Hirst, J. The mechanism of superoxide production by NADH: Ubiquinone oxidoreductase (complex I) from bovine heart mitochondria. Proc. Natl. Acad. Sci. USA 2006, 103, 7607–7612. [Google Scholar] [CrossRef] [PubMed]
  112. Smeitink, J.; van den Heuvel, L.; DiMauro, S. The genetics and pathology of oxidative phosphorylation. Nat. Rev. Genet. 2001, 2, 342–352. [Google Scholar] [CrossRef]
  113. Ochi, R.; Dhagia, V.; Lakhkar, A.; Patel, D.; Wolin, M.S.; Gupte, S.A. Rotenone-stimulated superoxide release from mitochondrial complex I acutely augments L-type Ca2+ current in A7r5 aortic smooth muscle cells. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H1118–H1128. [Google Scholar] [CrossRef]
  114. Dikalov, S.; Itani, H.; Richmond, B.; Vergeade, A.; Rahman, S.M.J.; Boutaud, O.; Blackwell, T.; Massion, P.P.; Harrison, D.G.; Dikalova, A. Tobacco smoking induces cardiovascular mitochondrial oxidative stress, promotes endothelial dysfunction, and enhances hypertension. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H639–H646. [Google Scholar] [CrossRef]
  115. Vignais, P.; Colbeau, A. Molecular biology of microbial hydrogenases. Curr. Issues Mol. Biol. 2004, 6, 159–188. [Google Scholar]
  116. Sag, C.M.; Wagner, S.; Maier, L.S. Role of oxidants on calcium and sodium movement in healthy and diseased cardiac myocytes. Free Radic. Biol. Med. 2013, 63, 338–349. [Google Scholar] [CrossRef]
  117. Stepanova, A.; Sosunov, S.; Niatsetskaya, Z.; Konrad, C.; Starkov, A.A.; Manfredi, G.; Wittig, I.; Ten, V.; Galkin, A. Redox-dependent loss of flavin by mitochondrial complex I in brain ischemia/reperfusion injury. Antioxid. Redox Signal. 2019, 31, 608–622. [Google Scholar] [CrossRef] [PubMed]
  118. Brandt, U. Energy converting NADH: Quinone oxidoreductase (complex I). Annu. Rev. Biochem. 2006, 75, 69–92. [Google Scholar] [CrossRef]
  119. Quinlan, C.L.; Perevoshchikova, I.V.; Hey-Mogensen, M.; Orr, A.L.; Brand, M.D. Sites of reactive oxygen species generation by mitochondria oxidizing different substrates. Redox Biol. 2013, 1, 304–312. [Google Scholar] [CrossRef]
  120. Chouchani, E.T.; Pell, V.R.; Gaude, E.; Aksentijević, D.; Sundier, S.Y.; Robb, E.L.; Logan, A.; Nadtochiy, S.M.; Ord, E.N.; Smith, A.C. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 2014, 515, 431–435. [Google Scholar] [CrossRef] [PubMed]
  121. Zhu, S.C.; Chen, C.; Wu, Y.N.; Ahmed, M.; Kitmitto, A.; Greenstein, A.S.; Kim, S.J.; Shao, Y.F.; Zhang, Y.H. Cardiac complex II activity is enhanced by fat and mediates greater mitochondrial oxygen consumption following hypoxic re-oxygenation. Pflügers Arch.-Eur. J. Physiol. 2020, 472, 367–374. [Google Scholar] [CrossRef] [PubMed]
  122. Markevich, N.I.; Galimova, M.H.; Markevich, L.N. Hysteresis and bistability in the succinate-CoQ reductase activity and reactive oxygen species production in the mitochondrial respiratory complex II. Redox Biol. 2020, 37, 101630. [Google Scholar] [CrossRef]
  123. Korge, P.; Calmettes, G.; John, S.A.; Weiss, J.N. Reactive oxygen species production induced by pore opening in cardiac mitochondria: The role of complex III. J. Biol. Chem. 2017, 292, 9882–9895. [Google Scholar] [CrossRef]
  124. Dona, M.; Lamers, M.; Rohde, S.; Gorissen, M.; Timmers, H.J. Targeting the Redox Balance Pathway Using Ascorbic Acid in sdhb Zebrafish Mutant Larvae. Cancers 2021, 13, 5124. [Google Scholar] [CrossRef]
  125. Valls-Lacalle, L.; Barba, I.; Miró-Casas, E.; Alburquerque-Béjar, J.J.; Ruiz-Meana, M.; Fuertes-Agudo, M.; Rodríguez-Sinovas, A.; García-Dorado, D. Succinate dehydrogenase inhibition with malonate during reperfusion reduces infarct size by preventing mitochondrial permeability transition. Cardiovasc. Res. 2016, 109, 374–384. [Google Scholar] [CrossRef]
  126. Di Trani, J.M.; Liu, Z.; Whitesell, L.; Brzezinski, P.; Cowen, L.E.; Rubinstein, J.L. Rieske head domain dynamics and indazole-derivative inhibition of Candida albicans complex III. Structure 2022, 30, 129–138.e4. [Google Scholar] [CrossRef] [PubMed]
  127. Vennam, P.R.; Fisher, N.; Krzyaniak, M.D.; Kramer, D.M.; Bowman, M.K. A caged, destabilized, free radical intermediate in the q-cycle. Chembiochem 2013, 14, 1745–1753. [Google Scholar] [CrossRef] [PubMed]
  128. Fernandez-Vizarra, E.; Zeviani, M. Mitochondrial complex III Rieske Fe-S protein processing and assembly. Cell Cycle 2018, 17, 681–687. [Google Scholar] [CrossRef] [PubMed]
  129. Lesnefsky, E.J.; Gudz, T.I.; Moghaddas, S.; Migita, C.T.; Ikeda-Saito, M.; Turkaly, P.J.; Hoppel, C.L. Aging decreases electron transport complex III activity in heart interfibrillar mitochondria by alteration of the cytochrome c binding site. J. Mol. Cell. Cardiol. 2001, 33, 37–47. [Google Scholar] [CrossRef] [PubMed]
  130. Schmitt, M.E.; Trumpower, B.L. Subunit 6 regulates half-of-the-sites reactivity of the dimeric cytochrome bc1 complex in Saccharomyces cerevisiae. J. Biol. Chem. 1990, 265, 17005–17011. [Google Scholar] [CrossRef] [PubMed]
  131. Chen, C.-L.; Kang, P.T.; Zhang, L.; Xiao, K.; Zweier, J.L.; Chilian, W.M.; Chen, Y.-R. Reperfusion mediates heme impairment with increased protein cysteine sulfonation of mitochondrial complex III in the post-ischemic heart. J. Mol. Cell. Cardiol. 2021, 161, 23–38. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, Y.-R.; Zweier, J.L. Cardiac mitochondria and reactive oxygen species generation. Circ. Res. 2014, 114, 524–537. [Google Scholar] [CrossRef]
  133. Lai, B.; Zhang, L.; Dong, L.-Y.; Zhu, Y.-H.; Sun, F.-Y.; Zheng, P. Inhibition of Qi site of mitochondrial complex III with antimycin A decreases persistent and transient sodium currents via reactive oxygen species and protein kinase C in rat hippocampal CA1 cells. Exp. Neurol. 2005, 194, 484–494. [Google Scholar] [CrossRef]
  134. Lai, B.; Zhang, L.; Dong, L.-Y.; Zhu, Y.-H.; Sun, F.-Y.; Zheng, P. Impact of inhibition of Qo site of mitochondrial complex III with myxothiazol on persistent sodium currents via superoxide and protein kinase C in rat hippocampal CA1 cells. Neurobiol. Dis. 2006, 21, 206–216. [Google Scholar] [CrossRef] [PubMed]
  135. Baudry, N.; Laemmel, E.; Vicaut, E. In vivo reactive oxygen species production induced by ischemia in muscle arterioles of mice: Involvement of xanthine oxidase and mitochondria. Am. J. Physiol. Heart Circ. Physiol. 2008, 294, H821–H828. [Google Scholar] [CrossRef] [PubMed]
  136. Huang, Q.; Su, H.; Qi, B.; Wang, Y.; Yan, K.; Wang, X.; Li, X.; Zhao, D. A SIRT1 activator, ginsenoside Rc, promotes energy metabolism in cardiomyocytes and neurons. J. Am. Chem. Soc. 2021, 143, 1416–1427. [Google Scholar] [CrossRef] [PubMed]
  137. Lee, H.J.; Svahn, E.; Swanson, J.M.J.; Lepp, H.; Voth, G.A.; Brzezinski, P.; Gennis, R.B. Intricate role of water in proton transport through cytochrome c oxidase. J. Am. Chem. Soc. 2010, 132, 16225–16239. [Google Scholar] [CrossRef] [PubMed]
  138. Kolbe, F.; Safarian, S.; Piórek, Ż.; Welsch, S.; Müller, H.; Michel, H. Cryo-EM structures of intermediates suggest an alternative catalytic reaction cycle for cytochrome c oxidase. Nat. Commun. 2021, 12, 6903. [Google Scholar] [CrossRef] [PubMed]
  139. Bouillaud, F.; Blachier, F. Mitochondria and sulfide: A very old story of poisoning, feeding, and signaling? Antioxid. Redox Signal. 2011, 15, 379–391. [Google Scholar] [CrossRef] [PubMed]
  140. Timkovich, R.; Thrasher, J.S. Carbon monoxide oxygenase activity of cytochrome cd1. Biochemistry 1988, 27, 5383–5388. [Google Scholar] [CrossRef] [PubMed]
  141. Cooper, C.E. Nitric oxide and cytochrome oxidase: Substrate, inhibitor or effector? Trends Biochem. Sci. 2002, 27, 33–39. [Google Scholar] [CrossRef]
  142. Feng, X.; Zhang, H.; Shi, M.; Chen, Y.; Yang, T.; Fan, H. Toxic effects of hydrogen sulfide donor NaHS induced liver apoptosis is regulated by complex IV subunits and reactive oxygen species generation in rats. Environ. Toxicol. 2020, 35, 322–332. [Google Scholar] [CrossRef] [PubMed]
  143. Prabu, S.K.; Anandatheerthavarada, H.K.; Raza, H.; Srinivasan, S.; Spear, J.F.; Avadhani, N.G. Protein kinase A-mediated phosphorylation modulates cytochrome c oxidase function and augments hypoxia and myocardial ischemia-related injury. J. Biol. Chem. 2006, 281, 2061–2070. [Google Scholar] [CrossRef] [PubMed]
  144. Lee, H.-L.; Chen, C.-L.; Yeh, S.T.; Zweier, J.L.; Chen, Y.-R. Biphasic modulation of the mitochondrial electron transport chain in myocardial ischemia and reperfusion. Am. J. Physiol.-Heart Circ. Physiol. 2012, 302, H1410–H1422. [Google Scholar] [CrossRef]
  145. Ide, T.; Tsutsui, H.; Hayashidani, S.; Kang, D.; Suematsu, N.; Nakamura, K.-I.; Utsumi, H.; Hamasaki, N.; Takeshita, A. Mitochondrial DNA damage and dysfunction associated with oxidative stress in failing hearts after myocardial infarction. Circ. Res. 2001, 88, 529–535. [Google Scholar] [CrossRef]
  146. Rosca, M.; Minkler, P.; Hoppel, C.L. Cardiac mitochondria in heart failure: Normal cardiolipin profile and increased threonine phosphorylation of complex IV. Biochim. Biophys. Acta (BBA)-Bioenergy 2011, 1807, 1373–1382. [Google Scholar] [CrossRef]
  147. Fang, J.-K.; Prabu, S.K.; Sepuri, N.B.; Raza, H.; Anandatheerthavarada, H.K.; Galati, D.; Spear, J.; Avadhani, N.G. Site specific phosphorylation of cytochrome c oxidase subunits I, IVi1 and Vb in rabbit hearts subjected to ischemia/reperfusion. FEBS Lett. 2007, 581, 1302–1310. [Google Scholar] [CrossRef] [PubMed]
  148. Antoniel, M.; Jones, K.; Antonucci, S.; Spolaore, B.; Fogolari, F.; Petronilli, V.; Giorgio, V.; Carraro, M.; Di Lisa, F.; Forte, M.; et al. The unique histidine in OSCP subunit of F-ATP synthase mediates inhibition of the permeability transition pore by acidic pH. EMBO Rep. 2018, 19, 257–268. [Google Scholar] [CrossRef]
  149. Naoi, M.; Wu, Y.; Shamoto-Nagai, M.; Maruyama, W. Mitochondria in neuroprotection by phytochemicals: Bioactive polyphenols modulate mitochondrial apoptosis system, function and structure. Int. J. Mol. Sci. 2019, 20, 2451. [Google Scholar] [CrossRef]
  150. Azarashvili, T.; Odinokova, I.; Bakunts, A.; Ternovsky, V.; Krestinina, O.; Tyynelä, J.; Saris, N.-E.L. Potential role of subunit c of F0F1-ATPase and subunit c of storage body in the mitochondrial permeability transition. Effect of the phosphorylation status of subunit c on pore opening. Cell Calcium 2014, 55, 69–77. [Google Scholar]
  151. Hausenloy, D.J.; Yellon, D.M. The mitochondrial permeability transition pore: Its fundamental role in mediating cell death during ischaemia and reperfusion. J. Mol. Cell. Cardiol. 2003, 35, 339–341. [Google Scholar] [CrossRef]
  152. Murphy, E.; Steenbergen, C. Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol. Rev. 2008, 88, 581–609. [Google Scholar] [CrossRef]
  153. 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]
  154. Efremov, R.G.; Sazanov, L.A. Structure of the membrane domain of respiratory complex I. Nature 2011, 476, 414–420. [Google Scholar] [CrossRef] [PubMed]
  155. Oka, S.-I.; Byun, J.; Huang, C.-Y.; Imai, N.; Ralda, G.; Zhai, P.; Xu, X.; Kashyap, S.; Warren, J.S.; Maschek, J.A.; et al. Nampt Potentiates Antioxidant Defense in Diabetic Cardiomyopathy. Circ. Res. 2021, 129, 114–130. [Google Scholar] [CrossRef]
  156. Zorov, D.B.; Filburn, C.R.; Klotz, L.O.; Zweier, J.L.; Sollott, S.J. Reactive oxygen species (ROS)-induced ROS release: A new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes. J. Exp. Med. 2000, 192, 1001–1014. [Google Scholar] [CrossRef]
  157. Lesnefsky, E.J.; Chen, Q.; Moghaddas, S.; Hassan, M.O.; Tandler, B.; Hoppel, C.L. Blockade of electron transport during ischemia protects cardiac mitochondria. J. Biol. Chem. 2004, 279, 47961–47967. [Google Scholar] [CrossRef]
  158. Wu, Z.; Jankowski, V.; Jankowski, J. Irreversible post-translational modifications—Emerging cardiovascular risk factors. Mol. Asp. Med. 2022, 86, 101010. [Google Scholar] [CrossRef]
  159. Nanni, E.J., Jr.; Stallings, M.D.; Sawyer, D.T. Does superoxide ion oxidize catechol,. alpha.-tocopherol, and ascorbic acid by direct electron transfer? J. Am. Chem. Soc. 1980, 102, 4481–4485. [Google Scholar] [CrossRef]
  160. Sawyer, D.T.; Calderwood, T.S.; Johlman, C.L.; Wilkins, C.L. Oxidation by superoxide ion of catechols, ascorbic acid, dihydrophenazine, and reduced flavins to their respective anion radicals. A common mechanism via a combined proton-hydrogen atom transfer. J. Org. Chem. 1985, 50, 1409–1412. [Google Scholar] [CrossRef]
  161. Sawyer, D.T.; Chiericato, G., Jr.; Tsuchiya, T. Oxidation of ascorbic acid and dehydroascorbic acid by superoxide ion in aprotic media. J. Am. Chem. Soc. 1982, 104, 6273–6278. [Google Scholar] [CrossRef]
  162. Zhang, Z.; Zhao, X.; Gao, M.; Xu, L.; Qi, Y.; Wang, J.; Yin, L. Dioscin alleviates myocardial infarction injury via regulating BMP4/NOX1-mediated oxidative stress and inflammation. Phytomedicine 2022, 103, 154222. [Google Scholar] [CrossRef]
  163. Imran, M.; Hassan, M.Q.; Akhtar, M.S.; Rahman, O.; Akhtar, M.; Najmi, A.K. Sacubitril and valsartan protect from experimental myocardial infarction by ameliorating oxidative damage in Wistar rats. Clin. Exp. Hypertens. 2019, 41, 62–69. [Google Scholar] [CrossRef]
  164. Chen, X.; Touyz, R.M.; Park, J.B.; Schiffrin, E.L. Antioxidant effects of vitamins C and E are associated with altered activation of vascular NADPH oxidase and superoxide dismutase in stroke-prone SHR. Hypertension 2001, 38, 606–611. [Google Scholar] [CrossRef]
  165. Valdecantos, M.P.; Pérez-Matute, P.; Quintero, P.; Martínez, J.A. Vitamin C, resveratrol and lipoic acid actions on isolated rat liver mitochondria: All antioxidants but different. Redox Rep. 2010, 15, 207–216. [Google Scholar] [CrossRef]
  166. Sadi, G.; Yılmaz, Ö.; Güray, T. Effect of vitamin C and lipoic acid on streptozotocin-induced diabetes gene expression: mRNA and protein expressions of Cu–Zn SOD and catalase. Mol. Cell. Biochem. 2008, 309, 109–116. [Google Scholar] [CrossRef]
  167. Song, Y.; Wang, B.; Zhu, X.; Hu, J.; Sun, J.; Xuan, J.; Ge, Z. Human umbilical cord blood-derived MSCs exosome attenuate myocardial injury by inhibiting ferroptosis in acute myocardial infarction mice. Cell Biol. Toxicol. 2021, 37, 51–64. [Google Scholar] [CrossRef] [PubMed]
  168. Chen, X.; Wan, W.; Ran, Q.; Ye, T.; Sun, Y.; Liu, Z.; Liu, X.; Shi, S.; Qu, C.; Zhang, C.; et al. Pinocembrin mediates antiarrhythmic e ff ects in rats with isoproterenol-induced cardiac remodeling. Eur. J. Pharmacol. 2022, 920, 174799. [Google Scholar] [CrossRef]
  169. Ting, H.H.; Timimi, F.K.; Haley, E.A.; Roddy, M.-A.; Ganz, P.; Creager, M.A. Vitamin C improves endothelium-dependent vasodilation in forearm resistance vessels of humans with hypercholesterolemia. Circulation 1997, 95, 2617–2622. [Google Scholar] [CrossRef] [PubMed]
  170. Sabharwal, A.K.; May, J.M. α-Lipoic acid and ascorbate prevent LDL oxidation and oxidant stress in endothelial cells. Mol. Cell. Biochem. 2008, 309, 125–132. [Google Scholar] [CrossRef] [PubMed]
  171. Mahmoudabadi, M.M.S.; Rahbar, A.R. Effect of EPA and vitamin C on superoxide dismutase, glutathione peroxidase, total antioxidant capacity and malondialdehyde in type 2 diabetic patients. Oman Med. J. 2014, 29, 39. [Google Scholar] [CrossRef] [PubMed]
  172. Aguilera, A.; Berdun, F.; Bartoli, C.; Steelheart, C.; Alegre, M.; Bayir, H.; Tyurina, Y.Y.; Kagan, V.E.; Salerno, G.; Pagnussat, G.; et al. C-ferroptosis is an iron-dependent form of regulated cell death in cyanobacteria. J. Cell Biol. 2022, 221, e201911005. [Google Scholar] [CrossRef]
  173. Valgimigli, L. Lipid Peroxidation and Antioxidant Protection. Biomolecules 2023, 13, 1291. [Google Scholar] [CrossRef]
  174. Vu, T.D.; Pal, S.N.; Ti, L.-K.; Martinez, E.C.; Rufaihah, A.J.; Ling, L.H.; Lee, C.-N.; Richards, A.M.; Kofidis, T. An autologous platelet-rich plasma hydrogel compound restores left ventricular structure, function and ameliorates adverse remodeling in a minimally invasive large animal myocardial restoration model: A translational approach: Vu and Pal “Myocardial Repair: PRP, Hydrogel and Supplements”. Biomaterials 2015, 45, 27–35. [Google Scholar]
  175. Jing, L.; Wang, Y.; Zhao, X.-M.; Zhao, B.; Han, J.-J.; Qin, S.-C.; Sun, X.-J. Cardioprotective Effect of Hydrogen-rich Saline on Isoproterenol-induced Myocardial Infarction in Rats. Heart Lung Circ. 2015, 24, 602–610. [Google Scholar] [CrossRef]
  176. Patel, P.; Parikh, M.; Shah, H.; Gandhi, T. Inhibition of RhoA/Rho kinase by ibuprofen exerts cardioprotective effect on isoproterenol induced myocardial infarction in rats. Eur. J. Pharmacol. 2016, 791, 91–98. [Google Scholar] [CrossRef]
  177. Nabzdyk, C.S.; Bittner, E.A. Vitamin C in the critically ill—Indications and controversies. World J. Crit. Care Med. 2018, 7, 52–61. [Google Scholar] [CrossRef] [PubMed]
  178. Kaidow, A.; Ishii, N.; Suzuki, S.; Shiina, T.; Kasahara, H. Vitamin C Maintenance against Cell Growth Arrest and Reactive Oxygen Species Accumulation in the Presence of Redox Molecular Chaperone hslO Gene. Int. J. Mol. Sci. 2022, 23, 12786. [Google Scholar] [CrossRef]
  179. Tosato, M.; Calvani, R.; Picca, A.; Ciciarello, F.; Galluzzo, V.; Coelho-Júnior, H.J.; Di Giorgio, A.; Di Mario, C.; Gervasoni, J.; Gremese, E.; et al. Effects of l-Arginine Plus Vitamin C Supplementation on Physical Performance, Endothelial Function, and Persistent Fatigue in Adults with Long COVID: A Single-Blind Randomized Controlled Trial. Nutrients 2022, 14, 4984. [Google Scholar] [CrossRef] [PubMed]
  180. Reynolds, P.S.; Fisher, B.J.; McCarter, J.; Sweeney, C.; Martin, E.J.; Middleton, P.; Ellenberg, M.; Fowler, E.; Brophy, D.F.; Fowler, A.A.; et al. Interventional vitamin C: A strategy for attenuation of coagulopathy and inflammation in a swine multiple injuries model. J. Trauma Acute Care Surg. 2018, 85, S57–S67. [Google Scholar] [CrossRef] [PubMed]
  181. Szyller, J.; Jagielski, D.; Bil-Lula, I. Antioxidants in Arrhythmia Treatment-Still a Controversy? A Review of Selected Clinical and Laboratory Research. Antioxidants 2022, 11, 1109. [Google Scholar] [CrossRef] [PubMed]
  182. Morelli, M.B.; Gambardella, J.; Castellanos, V.; Trimarco, V.; Santulli, G. Vitamin C and Cardiovascular Disease: An Update. Antioxidants 2020, 9, 1227. [Google Scholar] [CrossRef] [PubMed]
  183. Lee, D.-H.; Folsom, A.R.; Harnack, L.; Halliwell, B.; Jacobs, D.R. Does supplemental vitamin C increase cardiovascular disease risk in women with diabetes? Am. J. Clin. Nutr. 2004, 80, 1194–1200. [Google Scholar] [CrossRef]
  184. O’Connor, E.A.; Evans, C.V.; Ivlev, I.; Rushkin, M.C.; Thomas, R.G.; Martin, A.; Lin, J.S. Vitamin and Mineral Supplements for the Primary Prevention of Cardiovascular Disease and Cancer: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 2022, 327, 2334–2347. [Google Scholar] [CrossRef] [PubMed]
  185. Yi, R.; Feng, M.; Chen, Q.; Long, X.; Park, K.-Y.; Zhao, X. The Effect of Lactobacillus plantarum CQPC02 on Fatigue and Biochemical Oxidation Levels in a Mouse Model of Physical Exhaustion. Front. Nutr. 2021, 8, 641544. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, J.; Chen, L.; Zhang, L.; Chen, Q.; Tan, F.; Zhao, X. Effect of Lactobacillus fermentum HFY03 on the Antifatigue and Antioxidation Ability of Running Exhausted Mice. Oxid. Med. Cell. Longev. 2021, 2021, 8013681. [Google Scholar] [CrossRef] [PubMed]
  187. Martín-Calvo, N.; Martínez-González, M.Á. Vitamin C Intake is Inversely Associated with Cardiovascular Mortality in a Cohort of Spanish Graduates: The SUN Project. Nutrients 2017, 9, 954. [Google Scholar] [CrossRef] [PubMed]
  188. Basili, S.; Tanzilli, G.; Mangieri, E.; Raparelli, V.; Di Santo, S.; Pignatelli, P.; Violi, F. Intravenous ascorbic acid infusion improves myocardial perfusion grade during elective percutaneous coronary intervention: Relationship with oxidative stress markers. JACC Cardiovasc. Interv. 2010, 3, 221–229. [Google Scholar] [CrossRef] [PubMed]
  189. Arad, Y.; Spadaro, L.A.; Roth, M.; Newstein, D.; Guerci, A.D. Treatment of asymptomatic adults with elevated coronary calcium scores with atorvastatin, vitamin C, and vitamin E: The St. Francis Heart Study randomized clinical trial. J. Am. Coll. Cardiol. 2005, 46, 166–172. [Google Scholar] [CrossRef]
  190. Jaxa-Chamiec, T.; Bednarz, B.; Drozdowska, D.; Gessek, J.; Gniot, J.; Janik, K.; Kawka-Urbanek, T.; Maciejewski, P.; Ogórek, M.; Szpajer, M. Antioxidant effects of combined vitamins C and E in acute myocardial infarction. The randomized, double-blind, placebo controlled, multicenter pilot Myocardial Infarction and VITamins (MIVIT) trial. Kardiol. Pol. 2005, 62, 344–350. [Google Scholar]
  191. Sesso, H.D.; Buring, J.E.; Christen, W.G.; Kurth, T.; Belanger, C.; MacFadyen, J.; Bubes, V.; Manson, J.E.; Glynn, R.J.; Gaziano, J.M. Vitamins E and C in the prevention of cardiovascular disease in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2008, 300, 2123–2133. [Google Scholar] [CrossRef]
  192. Laskowski, H.; Minczykowski, A.; Wysocki, H. Mortality and clinical course of patients with acute myocardial infarction treated with streptokinase and antioxidants: Mannitol and ascorbic acid. Int. J. Cardiol. 1995, 48, 235–237. [Google Scholar] [CrossRef] [PubMed]
  193. Bhakuni, P.; Chandra, M.; Misra, M.K. Effect of ascorbic acid supplementation on certain oxidative stress parameters in the post reperfusion patients of myocardial infarction. Mol. Cell. Biochem. 2006, 290, 153–158. [Google Scholar] [CrossRef] [PubMed]
  194. Kato, K.; Fukuma, N.; Kimura-Kato, Y.; Aisu, N.; Tuchida, T.; Mabuchi, K.; Takano, T. Improvement of sympathetic response to exercise by oral administration of ascorbic acid in patients after myocardial infarction. Int. J. Cardiol. 2006, 111, 240–246. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zheng, H.; Xu, Y.; Liehn, E.A.; Rusu, M. Vitamin C as Scavenger of Reactive Oxygen Species during Healing after Myocardial Infarction. Int. J. Mol. Sci. 2024, 25, 3114. https://doi.org/10.3390/ijms25063114

AMA Style

Zheng H, Xu Y, Liehn EA, Rusu M. Vitamin C as Scavenger of Reactive Oxygen Species during Healing after Myocardial Infarction. International Journal of Molecular Sciences. 2024; 25(6):3114. https://doi.org/10.3390/ijms25063114

Chicago/Turabian Style

Zheng, Huabo, Yichen Xu, Elisa A. Liehn, and Mihaela Rusu. 2024. "Vitamin C as Scavenger of Reactive Oxygen Species during Healing after Myocardial Infarction" International Journal of Molecular Sciences 25, no. 6: 3114. https://doi.org/10.3390/ijms25063114

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