Open access peer-reviewed chapter

Oxidative Stress in Cardiovascular Diseases

Written By

Laura Mourino-Alvarez, Tamara Sastre-Oliva, Nerea Corbacho-Alonso and Maria G. Barderas

Submitted: 16 May 2022 Reviewed: 14 June 2022 Published: 22 July 2022

DOI: 10.5772/intechopen.105891

From the Edited Volume

Importance of Oxidative Stress and Antioxidant System in Health and Disease

Edited by Suna Sabuncuoğlu and Ahmet Yalcinkaya

Chapter metrics overview

215 Chapter Downloads

View Full Metrics

Abstract

Cardiovascular diseases encompass a range of pathologies that affect the heart or blood vessels. Oxidative stress is an important factor that contributes to the development of these pathologies. Adverse effects due to oxidative stress manifest when there is an imbalance between the production and elimination of reactive oxygen species (ROS), or when physiological mechanisms of repair for oxidative injury are overburdened. This chapter focuses on ROS accumulation and antioxidant system deficiencies in the context of their influence on cardiovascular disease. We also discuss the importance of high throughput approaches, such as proteomics, with regard to their role in advancing the field of precision medicine for cardiovascular diseases, while keeping in mind the ultimate goal of improving patient care and quality of life.

Keywords

  • cardiovascular disease
  • endothelial dysfunction
  • reactive oxygen species
  • thiol compounds
  • oxidative stress

1. Introduction

Cardiovascular diseases (CVDs) constitute a major cause of global mortality and are an important source of rising healthcare costs; furthermore, they result in significant decreases in the quality of life (QoL) of those who suffer from these conditions [1, 2]. The great majority of CVDs are chronic diseases, and hence, rising incidence translates into higher prevalence, which leads to a healthcare burden that may persist over decades. Prevalence has long been on the rise in almost all countries, with reported increases from 271 million in 1990 to 523 million in 2019, while CVD-associated deaths have also followed suite, increasing from 12.1 million to 18.6 million in the same period [3]. CVDs encompass a range of pathologies that affect the heart or blood vessels, including ischemic heart disease, cerebrovascular disease (stroke), peripheral arterial disease, heart failure, and heart valve disease. Hypercholesterolemia, hypertension, and diabetes, which are associated with oxidative stress (OS) and inflammatory activation, are well-known risk factors for CVD [4, 5, 6, 7].

OS originates in cells and tissues when the balance between oxidative and antioxidative compounds (or mechanisms) is disrupted in favor of oxidation. This imbalance between ROS production and antioxidant defenses may be due to increased ROS formation (e.g., superoxide, hydroxyl, nitric oxide, or hydrogen peroxide) and/or insufficiency in antioxidants (e.g., ascorbate, alpha-tocopherol, or thiol-based redox compounds) [8]. The consequences of OS include lipid peroxidation, membrane damage, and the activation of proteases, nucleases, and protein kinases. With respect to vascular functions, excess ROS regulates the release of factors that drive vasoconstriction or vasodilation, leading to endothelial cell (EC) damage, vascular smooth muscle cell (VSMC) hyperplasia, and structural remodeling (Figure 1) [9, 10]. Regulation of vascular tone is critical for the maintenance of cardiovascular health. In this sense, OS plays an important role as an initiator of the EC dysfunction. In physiological conditions, maintenance of appropriate endothelial function provides vasorelaxant properties through the release of vasoactive substances. Nevertheless, under OS conditions, there exists an imbalance between the production of vasoprotective and vasorelaxant factors and vasoconstrictor substances by the endothelium [11]. In addition, oxidative stress could induce vascular inflammation and injury through activation of the transcription factors, upregulation of adhesion molecules, stimulation of chemokine production, and recruitment of inflammatory cells [9, 10].

Figure 1.

Cardiovascular risk factors disrupt oxidative metabolism and negatively affect the vascular system through lipid peroxidation, endothelial damage, immune cell activation, structural remodeling, or inflammation.

In the following sections, we focus primarily on the OS-related mechanisms that have been associated with CVD development. Firstly, we discuss how ROS accumulate and the importance of this accumulation with regard to endothelial dysfunction and the vascular system. Next, we concentrate on the role of thiols in the antioxidant system and the consequences of the suppression of this defense system. Finally, we highlight the importance of high throughput approaches, such as proteomics, with respect to their contributions to the advances in the field of precision medicine related to CVD, while keeping in mind the ultimate goal of improving patient care and QoL.

Advertisement

2. ROS accumulation: mitochondrial and ER-related stress

Endothelial damage, vascular dysfunction, cardiac remodeling, immune cell activation, and systemic inflammation are key processes implicated in CVDs, all of which are affected by ROS accumulation [12, 13, 14, 15]. The primary oxidase system underlying OS in vascular disease is the NADPH oxidase (NOX) system, the main function of which is to produce ROS [16]. Importantly, while ROS production was originally considered to incite harmful effects, and low levels of ROS are necessary for physiological processes, including cell proliferation, migration, differentiation, and cytoskeletal organization [17]. However, excessive activation of NOX also activates other oxidase systems to maintain OS (Figure 2). These secondary mechanisms include, but are not limited to, endothelial nitric oxide synthase (eNOS) uncoupling, mitochondrial stress, and endoplasmic reticulum (ER) stress, which also contribute to redox changes in CVD [18, 19, 20]. eNOS is primarily responsible for the generation of vasoprotective nitric oxide (NO). Endothelial-derived NO is an important vasodilator and a potent inhibitor of platelet aggregation and leukocyte adhesion. As such, NO is one of the most important anti-atherogenic factors in vasculature [21]. Under normal conditions, eNOS exists as a dimer that is stabilized by the essential cofactor, tetrahydrobiopterin (BH4). However, when BH4 is inactivated due to excess ROS, the dimer breaks down, resulting in the production of vaso-injurious superoxide (O2) as opposed to NO [22].

Figure 2.

Excessive activation of NADPH oxidase (NOX) increases the production of ROS to harmful levels. Subsequently, secondary mechanisms, such as endothelial nitric oxide synthase (eNOS) uncoupling, mitochondrial stress, and endoplasmic reticulum (ER) stress, are triggered, perpetuating oxidative stress and injurious effects in a vicious cycle.

The cross-talk between the NOX system and mitochondria may cause ROS-induced release of ROS, a positive feed-forward mechanism of ROS production. As a result, NOX-derived ROS increases the production of mitochondrial ROS, which in turn stimulates NOX activation [23, 24, 25]. One consequence of normal mitochondrial metabolism and homeostasis is the production of controlled levels of ROS caused by the reduction of O2 to O2 through the respiratory chain [26]. Under physiological conditions, these levels are safe and normal cellular activity is maintained through the protection of mitochondria by a multilayer network of antioxidant systems. One particular example is the manganese-dependent superoxide dismutase (SOD2) enzyme, which converts the O2 anion to hydrogen peroxide (H2O2), which is subsequently transformed to water by protective mechanisms, such as catalase, glutathione peroxidase, and peroxiredoxins [27]. Nevertheless, if ROS levels exceed the normal range, the resulting oxidative environment is harmful and cannot be compensated for by these protective mechanisms [23, 27, 28]. In relation to this, mitochondrial dysfunction also has important implications for CVD. The protective role of different antioxidant systems against atherosclerosis is related to H2O2 metabolisms, such as that driven by the catalase or paraoxonase family, as seen in mouse models [29, 30, 31]. Mitochondrial dysfunction is associated with the redox inactivation of the mitochondrial deacetylase Sirtuin 3 (also named Silent acting type information regulation 2 homolog 3, SIRT3) and the mitochondrial antioxidant SOD2 also contributes to the development of hypertension [32, 33]. These discoveries brought forth the use of mitochondrial-targeted interventions as therapeutic agents. Mitoquinone (MitoQ), a mitochondrial ROS scavenger, has been shown to reduce macrophage infiltration into atherosclerotic plaques in a mouse model of metabolic syndrome [34]. Moreover, MitoQ protects against the development of hypertension by improving endothelial NO bioavailability and reduces cardiac hypertrophy in models of hypertension [35]. Similar favorable vascular responses have been demonstrated in humans treated with MitoQ. After oral supplementation, MitoQ was associated with a decrease in plasma oxidized low-density lipoprotein (LDL), a circulating marker of OS, and the amelioration of endothelial function [36].

The ER is increasingly being recognized as an important player in the redox pathophysiology of the cardiovascular system [37, 38, 39]. Protein folding contributes to the generation of ROS since ER oxidoreductases serve as terminal electron acceptors with oxygen. When ER stress develops, demands on protein folding are excessively increased and oxygen is not completely reduced, leading to the generation of O2 anions with the consequent production of H2O2 or other ROS [40, 41]. The ER and mitochondria are closely associated as the protein folding process is energy-dependent. As a consequence, ATP depletion may occur during ER stress, which can stimulate oxidative phosphorylation in the mitochondria due to increased ATP need, subsequently resulting in greater ROS generation. In addition, mitochondrial ROS production will also increase if unfolded proteins accumulate in the ER via the release of Ca2+ or interactions with SOD [42, 43]. Of note, ER stress has been shown to play an important role in vascular cell phenotypic switching, de-differentiation, calcification, and apoptosis, contributing to endothelial dysfunction and vascular remodeling in hypertension and in atherosclerosis [44, 45].

With regard to these data, it has been inferred that compounds, which can alleviate ER stress could be pharmacological options to elicit antioxidant effects. One example is the antihypertensive drug guanabenz, which is demonstrated to confer protection against the detrimental accumulation of misfolded proteins in cardiac myocytes [46]. However, there is much controversy about this compound, since it has been reported to induce β-cell dysfunction in vitro and in vivo (in rodents) and it may lead to impaired glucose tolerance [47]. Other ER stress inhibitors, such as tauroursodeoxycholic acid (TUDCA) or 4-phenylbutyrate (4-PBA), also have beneficial effects on the cardiovascular system. Administration of TUDCA has been shown to alleviate myocardial contractile dysfunction and reduce blood pressure in rodents [48, 49, 50]. Likewise, 4-PBA protects against atherosclerotic lesion growth [51], prevents cardiac rupture and remodeling by inhibiting cardiac apoptotic and fibrotic signaling pathways [52], and attenuates interstitial fibrosis and cardiac hypertrophy caused by pressure overload [53]. These studies are among the studies that provide support to the proof of concept that modulation of ER stress can improve cardiovascular function.

Advertisement

3. Thiol-based redox compounds in CVD

Thiol-based redox compounds, such as thioredoxins (Trxs), glutaredoxins (Grxs), and peroxiredoxins (Prxs), are primary contributors to the intracellular redox state, thereby modulating metabolism, signaling, and cell survival pathways [54]. In proteins, the thiol groups of cysteine side chains are highly susceptible to reversible or irreversible oxidative modifications. Besides forming disulfide bonds between two different proteins, these protein thiols can also form disulfide bridges with low-molecular-weight thiols like glutathione, they can be oxidized to sulfenic, sulfinic, and sulfonic, or suffer S-nitrosylation [55]. These modifications can alter the activity of numerous proteins that contain cysteines (Cys) as their anionic form. As such, thiolate (RS) plays critical role in protein structure, function, and regulation. Although the Trxs and Grxs systems function differently, they both maintain a reduced intracellular redox state in mammalian cells by reducing protein thiols. Oxidized Grx is reduced by reduced glutathione (GSH), and the oxidized glutathione (GSSG) is then recycled by glutathione reductase (GR) at the expense of NADPH. The Grx system involves the GR, Grx, and the glutathione redox pair (GSH/GSSG). Under oxidative conditions, in which the GSH concentration decreases and GSSG increases, Grx is more likely to be oxidized [55]. Thus, the GSH-to-GSSG ratio (GSH/GSSG) in the cell is an important marker of the redox environment and a major determinant of the cellular redox potential. Meanwhile, Prxs are a family of conserved abundant Cys-based peroxidases that consist of six Prx isoforms (Prx1–6) in mammalian cells. This family plays an essential role in the detoxification of hydrogen peroxide, aliphatic and aromatic hydroperoxides, and peroxynitrite [56, 57].

Currently, the implications of these thiol-based redox compounds on CVDs are being widely studied [55, 58, 59]. Endogenous Trx has the potential to decrease reperfusion-induced arrhythmia [60] and is a central mediator of cardiomyocyte growth [61]. Upregulation of Trxs has been detected in ischemia [62, 63] and cardiac failure [64, 65], probably as a compensatory mechanism in response to myocardial injury. In fact, the role of Trx1, the cytosolic isoform of the Trx family, in myocardial hypertrophy is identified to be fundamental as it acts as a negative regulator under normal conditions but initiates hypertrophic signaling in the myocardium when it is oxidized due to stress [66, 67]. Moreover, the important role of Trx in hypertension has been demonstrated in mice, through experiments involving injection of human Trx and other studies in which Trx-overexpressing transgenic mice were assessed [68]. In these studies, Trx decreased age-related hypertension through different mechanisms, such as preservation of functional eNOS and NO release, and maintenance of relaxation responses. These researchers also found decreased arterial stiffness and improved vascular flow as a result of both Trx injection and overexpression. In another study, Trx1 overexpression was found to decrease infarct size and improve myocardial function after infarction [69].

The Grx superfamily has also been associated with enhanced cell survival and improved resistance to OS in CVD. In fact, overexpression of Grx1 has been shown to reduce ventricular remodeling and improve cardiac function [70], effects that were later proposed to be associated with the induction of angiogenesis. Nevertheless, Grx may offer protection to the ischemic heart by inhibiting apoptosis as demonstrated by an in vivo study in which Grx up-regulation inhibited EC migration and impaired hind limb revascularization [71]. The cardioprotective effect of Grx1 via a decrease in OS-mediated apoptosis is well documented [72, 73, 74]. In human coronary arteries, Grx was expressed (together with Trx) in ECs, fibroblasts, and smooth muscle cells. Specifically, the macrophages infiltrating fibrous plaques of atheroma that are known to produce ROS strongly express Grx and Trx [75]. In the year 2000, a Grx isoform located in the cytosol was discovered, defined as Grx3 or protein kinase C-interacting cousin of thioredoxin (PICOT). Although there is little data available about this oxidoreductase, it has a promising cardioprotective effect against ischemia/reperfusion-induced cardiac injury [76], as well as in aging and heart failure [77].

Apart from oxidoreductases, Prx proteins are essential for the regulation of OS and H2O2-mediated intracellular signaling. Some studies have shown that different isoforms of Prx protect against atherosclerosis. On the one hand, Prx1 scavenges H2O2 [78] and degrades lipids in macrophages, thereby reducing foam cell formation [79]. On the other hand, Prx2 and Prx4 modify the development of atherosclerotic plaques by altering immune cell infiltration [80, 81]. Moreover, Prx2 protects against neointimal thickening [82] and negatively regulates H2O2 generation and the formation of thrombosis [83, 84], whereas, its deficiency has been shown to promote the progression of the abdominal aortic aneurysm [85]. Indeed, Prx has an important influence on myocardial tissue and the overexpression of Prdx1 in cardiomyocytes was recently proposed to confer protection against cardiac hypertrophy and heart failure in the presence of pressure overload [86]. Prx3 ameliorates inflammation and protects the heart against left ventricular remodeling and failure after myocardial infarction [87], and both Prx3 and Prx6 may have a protective role during ischemia/reperfusion-induced heart injury [88]. Together these data suggest that Prx could be a potential target in therapeutic strategies to combat CVD.

Advertisement

4. Redox proteomics to get inside oxidative stress

The regulation of redox signaling commonly involves the post-translational modification (PTM) of proteins, in which different amino acids, including methionine and tyrosine, and most importantly Cys, are sensitive to oxidation and other modifications [89, 90]. As described above, the oxidation/reduction of thiol proteins has emerged as one of the major mechanisms through which reactive oxidants modulate cell signaling [91]. Accordingly, several proteomics-based techniques have been developed to enrich, identify, and characterize thiol-related redox modifications, referred to as redox proteomics [92]. The main aims of redox proteomics include (1) the identification of proteins that are modified or that interact with regulatory oxidoreductases, (2) the identification of residues susceptible to modification, including specific ROS-induced redox modifications, and (3) the quantification of the proportions of the modified protein(s) [93]. Crucially, RS is very reactive, and thus in order to study reversible Cys oxidations, it is essential to block this reactivity to capture the in vivo thiol-redox status. For this reason, one of the initial steps in thiol proteomics is to block free thiols in a sample with an alkylating reagent, such as iodoacetamide or derivatives of maleimide [94]. This also enables the reversibility of thiol modifications to be used to selectively label and/or purify redox-modified proteins. After alkylation, proteins can then be reduced using a thiol reducing agent, such as dithiothreitol or tris (2-carboxyethyl) phosphine, and, newly-formed free thiols can also be studied. Differential thiol isotopic labeling allows samples of different groups to be combined, reducing run-to-run experimental variation and permitting relative quantification between different samples (Figure 3) [95]. Although redox proteomics is still in its infancy, assessment of possible biomarkers through mass spectrometry (MS)-driven strategies may be promising for the study of OS in different disease states. In contrast to direct measurement of ROS levels, which is a complex task given the short half-life and high reactivity of these species, redox proteomics allows the detection of the resulting oxidative damage to proteins. Thus, it provides mechanistic insight into cellular toxicity mechanisms. For that reason, some studies employing this approach have been performed in CVDs.

Figure 3.

Essential workflow for redox proteomics analysis includes blocking or alkylation of free thiols and reduction to form new free thiols that can also be studied. This allows the identification of proteins that are modified and the quantification of the relevant proportions of the modified protein.

One of the first attempts to apply redox proteome analysis to heart proteins was performed using two different proteomic methods: gel-based (DIGE) and gel-free proteomics (ICAT) [96]. Heart tissue was analyzed with the purpose to identify and quantify redox-sensitive proteins, resulting in the elucidation of 50 proteins as potential targets of H2O2 oxidation through the ICAT method and 26 such proteins with the DIGE method, 13 of which were detected with both methods. Several years later, a different technique, named GELSILOX, was used to investigate the mechanisms of damage produced in the heart by ischemia/reperfusion injury and the effects of ischemic preconditioning in mitochondria purified from cardiomyocytes [97]. Several proteins were identified that had previously been associated with redox changes but had not been distinguished in the context of ischemia/reperfusion damage. Although these were mainly proof-of-concept studies of the methodology used, they provide useful information about the redox proteome of the cardiovascular system, including specific sites of oxidation. More recently, the plasma redox proteome was analyzed using FASILOX, a novel multiplexed proteomic strategy of isotope labeling, followed by liquid chromatography (LC)-tandem MS [98]. A plasma signature was proposed for the stratification of young individuals and to detect those with a higher probability of suffering a future cardiovascular event. The same approach was used to analyze aortic valve tissue obtained from patients with calcific aortic valve disease [99], which revealed different protein profiles in calcified valve tissue in patients with and without atherosclerosis. Differences in the redox status of the aortic valve tissue were also found despite their high degree of calcification and affectation. Additionally, two specific sites of cysteine oxidation in albumin that had not been described previously were also found. The results of these studies highlight the enormous potential of redox proteomics, an approach that is set to become a key tool to obtain new insights into CVD-related protein modifications.

Advertisement

5. Future perspectives: a focus on redox biology to improve patient management

Overall, excessive OS due to exaggerated ROS production or insufficient antioxidant defense has been closely associated with CVD. Improving our understanding of these pathways may permit the modulation of redox biology, which can be instrumental in reducing the systemic impact of these alterations. From a clinical point of view, the combination of standard clinical evaluation with results from high throughput approaches is essential to take a step forward in precision medicine, currently a highly desired goal in medicine. By definition, precision medicine focuses on selecting the appropriate treatment for each patient based on individual phenotypes. This includes a reliable and accurate risk stratification that would allow better screening of patients at high risk and avoiding the use of unnecessary treatments with regard to potential side effects. Since the disruption in OS balance leads to a vicious cycle, it is crucial to identify high-risk patients to avoid the damage caused by an uncontrolled redox milieu.

CVDs are multifactorial diseases that are usually accompanied by other comorbidities, and OS incites a systemic effect that can damage different organs and systems. From a pharmacological point of view, this implies that modification of the redox state may also affect distinct organs in different patients, which may have beneficial and detrimental effects. This kind of treatment can theoretically improve the function of different systems and may be considered an integral treatment, although this unfortunately leads to less control over potential side effects. The only way to overcome this issue is to intensify research focusing on OS and its relationship with disease. Although molecules related to OS are well known but their possible roles as cardioprotective molecules are still not fully understood. More information about this facet of their activity will be essential to design successful new pharmacological approaches. However, we must be aware that there is a long way to go in this regard. Indeed, it should not be forgotten that alterations that alter multiple metabolic pathways may be detrimental when applied at inappropriate times or doses.

Advertisement

Acknowledgments

This research was funded by the Junta de Comunidades de Castilla-La Mancha (JCCM, co-funded by the European Regional Development Fund, SBPLY/19/180501/000226), the Instituto de Salud Carlos III through the project PI18/00995, PI21/00384 (co-funded by European Regional Development Fund/European Social Fund—“Investing in your future”) Sociedad Española de Cardiología, 2020 and Grant PRB3 (IPT17/0019—ISCIII-SGEFI/ERDF). These results are aligned with the Spanish initiative on the Human Proteome Project (SpHPP).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Collaborators GDaI. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: A systematic analysis for the global burden of disease study 2019. Lancet. 2020;396(10258):1204-1222. DOI: 10.1016/s0140-6736(20)30925-9
  2. 2. Mensah GA, Roth GA, Fuster V. The global burden of cardiovascular diseases and risk factors: 2020 and beyond. Journal of the American College of Cardiology. 2019;74(20):2529-2532. DOI: 10.1016/j.jacc.2019.10.009
  3. 3. Roth GA, Mensah GA, Johnson CO, Addolorato G, Ammirati E, Baddour LM, et al. Global burden of cardiovascular diseases and risk factors, 1990-2019: Update from the GBD 2019 study. Journal of the American College of Cardiology. 2020;76(25):2982-3021. DOI: 10.1016/j.jacc.2020.11.010
  4. 4. Brunelli E, La Russa D, Pellegrino D. Impaired oxidative status is strongly associated with cardiovascular risk factors. Oxidative medicine and cellular longevity. 2017;2017:6480145. DOI: 10.1155/2017/6480145
  5. 5. Fuster V, Voûte J. MDGs: Chronic diseases are not on the agenda. Lancet. 2005;366(9496):1512-1514. DOI: 10.1016/s0140-6736(05)67610-6
  6. 6. Lepedda AJ, Formato M. Oxidative modifications in advanced atherosclerotic plaques: A focus on in situ protein sulfhydryl group oxidation. Oxidative medicine and cellular longevity. 2020;2020:6169825. DOI: 10.1155/2020/6169825
  7. 7. Pastore A, Piemonte F. Protein glutathionylation in cardiovascular diseases. International Journal of Molecular Sciences. 2013;14(10):20845-20876. DOI: 10.3390/ijms141020845
  8. 8. Halliwell B. The role of oxygen radicals in human disease, with particular reference to the vascular system. Haemostasis. 1993;23(Suppl. 1):118-126. DOI: 10.1159/000216921
  9. 9. Zhao S, Cheng CK, Zhang CL, Huang Y. Interplay between oxidative stress, cyclooxygenases, and prostanoids in cardiovascular diseases. Antioxidants & Redox Signaling. 2021;34(10):784-799. DOI: 10.1089/ars.2020.8105
  10. 10. Xu T, Ding W, Ji X, Ao X, Liu Y, Yu W, et al. Oxidative stress in cell death and cardiovascular diseases. Oxidative Medicine and Cellular Longevity. 2019;2019:9030563. DOI: 10.1155/2019/9030563
  11. 11. Sena CM, Pereira AM, Seiça R. Endothelial dysfunction—A major mediator of diabetic vascular disease. Biochimica et Biophysica Acta. 2013;2013(1832):2216-2231. DOI: 10.1016/j.bbadis.2013.08.006
  12. 12. Case AJ, Tian J, Zimmerman MC. Increased mitochondrial superoxide in the brain, but not periphery, sensitizes mice to angiotensin II-mediated hypertension. Redox Biology. 2017;11:82-90. DOI: 10.1016/j.redox.2016.11.011
  13. 13. Lopes RA, Neves KB, Tostes RC, Montezano AC, Touyz RM. Downregulation of nuclear factor erythroid 2-related factor and associated antioxidant genes contributes to redox-sensitive vascular dysfunction in hypertension. Hypertension. 2015;66(6):1240-1250. DOI: 10.1161/hypertensionaha.115.06163
  14. 14. Silva GC, Silva JF, Diniz TF, Lemos VS, Cortes SF. Endothelial dysfunction in DOCA-salt-hypertensive mice: Role of neuronal nitric oxide synthase-derived hydrogen peroxide. Clinical Science (London, England). 2016;130(11):895-906. DOI: 10.1042/cs20160062
  15. 15. Guzik TJ, Touyz RM. Oxidative stress, inflammation, and vascular aging in hypertension. Hypertension. 2017;70(4):660-667. DOI: 10.1161/hypertensionaha.117.07802
  16. 16. Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: The role of oxidant stress. Circulation Research. 2000;87(10):840-844. DOI: 10.1161/01.res.87.10.840
  17. 17. Brown DI, Griendling KK. Regulation of signal transduction by reactive oxygen species in the cardiovascular system. Circulation Research. 2015;116(3):531-549. DOI: 10.1161/circresaha.116.303584
  18. 18. Gao L, Siu KL, Chalupsky K, Nguyen A, Chen P, Weintraub NL, et al. Role of uncoupled endothelial nitric oxide synthase in abdominal aortic aneurysm formation: Treatment with folic acid. Hypertension. 2012;59(1):158-166. DOI: 10.1161/hypertensionaha.111.181644
  19. 19. Youn JY, Wang T, Blair J, Laude KM, Oak JH, McCann LA, et al. Endothelium-specific sepiapterin reductase deficiency in DOCA-salt hypertension. American Journal of Physiology. Heart and Circulatory Physiology. 2012;302(11):H2243-H2249. DOI: 10.1152/ajpheart.00835.2011
  20. 20. Siu KL, Miao XN, Cai H. Recoupling of eNOS with folic acid prevents abdominal aortic aneurysm formation in angiotensin II-infused apolipoprotein E null mice. PLoS One. 2014;9(2):e88899. DOI: 10.1371/journal.pone.0088899
  21. 21. Förstermann U, Closs EI, Pollock JS, Nakane M, Schwarz P, Gath I, et al. Nitric oxide synthase isozymes. Characterization, purification, molecular cloning, and functions. Hypertension. 1994;23(6 Pt 2):1121-1131. DOI: 10.1161/01.hyp.23.6.1121
  22. 22. Daiber A, Xia N, Steven S, Oelze M, Hanf A, Kröller-Schön S, et al. New therapeutic implications of endothelial nitric oxide synthase (eNOS) function/ dysfunction in cardiovascular disease. International Journal of Molecular Sciences. 2019;20(1):187. DOI: 10.3390/ ijms200101877
  23. 23. Zorov DB, Filburn CR, Klotz LO, Zweier JL, Sollott SJ. Reactive oxygen species (ROS)-induced ROS release: A new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes. The Journal of Experimental Medicine. 2000;192(7):1001-1014. DOI: 10.1084/jem.192.7.1001
  24. 24. Zinkevich NS, Gutterman DD. ROS-induced ROS release in vascular biology: Redox-redox signaling. American Journal of Physiology. Heart and Circulatory Physiology. 2011;301(3):H647-H653. DOI: 10.1152/ajpheart.01271.2010
  25. 25. Kim YM, Kim SJ, Tatsunami R, Yamamura H, Fukai T, Ushio-Fukai M. ROS-induced ROS release orchestrated by Nox4, Nox2, and mitochondria in VEGF signaling and angiogenesis. American Journal of Physiology Cell Physiology. 2017;312(6):C749-Cc64. DOI: 10.1152/ajpcell.00346.2016
  26. 26. Murphy MP. How mitochondria produce reactive oxygen species. The Biochemical Journal. 2009;417(1):1-13. DOI: 10.1042/bj20081386
  27. 27. Andreyev AY, Kushnareva YE, Starkov AA. Mitochondrial metabolism of reactive oxygen species. Biochemistry Biokhimiia. 2005;70(2):200-214. DOI: 10.1007/s10541-005-0102-7
  28. 28. Starkov AA. The role of mitochondria in reactive oxygen species metabolism and signaling. Annals of the New York Academy of Sciences. 2008;1147:37-52. DOI: 10.1196/annals.1427.015
  29. 29. Förstermann U, Xia N, Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circulation Research. 2017;120(4):713-735. DOI: 10.1161/circresaha.116.309326
  30. 30. Tward A, Xia YR, Wang XP, Shi YS, Park C, Castellani LW, et al. Decreased atherosclerotic lesion formation in human serum paraoxonase transgenic mice. Circulation. 2002;106(4):484-490. DOI: 10.1161/01.cir.0000023623.87083.4f
  31. 31. Yang H, Roberts LJ, Shi MJ, Zhou LC, Ballard BR, Richardson A, et al. Retardation of atherosclerosis by overexpression of catalase or both Cu/Zn-superoxide dismutase and catalase in mice lacking apolipoprotein E. Circulation Research. 2004;95(11):1075-1081. DOI: 10.1161/01.RES.0000149564.49410.0d
  32. 32. Dikalova AE, Itani HA, Nazarewicz RR, McMaster WG, Flynn CR, Uzhachenko R, et al. Sirt3 impairment and SOD2 hyperacetylation in vascular oxidative stress and hypertension. Circulation Research. 2017;121(5):564-574. DOI: 10.1161/circresaha.117.310933
  33. 33. Su H, Zeng H, Liu B, Chen JX. Sirtuin 3 is essential for hypertension-induced cardiac fibrosis via mediating pericyte transition. Journal of Cellular and Molecular Medicine. 2020;24(14):8057-8068. DOI: 10.1111/jcmm.15437
  34. 34. Mercer JR, Yu E, Figg N, Cheng KK, Prime TA, Griffin JL, et al. The mitochondria-targeted antioxidant MitoQ decreases features of the metabolic syndrome in ATM+/−/ApoE−/− mice. Free Radical Biology & Medicine. 2012;52(5):841-849. DOI: 10.1016/j.freeradbiomed.2011.11.026
  35. 35. Graham D, Huynh NN, Hamilton CA, Beattie E, Smith RA, Cochemé HM, et al. Mitochondria-targeted antioxidant MitoQ10 improves endothelial function and attenuates cardiac hypertrophy. Hypertension. 2009;54(2):322-328. DOI: 10.1161/hypertensionaha.109.130351
  36. 36. Rossman MJ, Santos-Parker JR, Steward CAC, Bispham NZ, Cuevas LM, Rosenberg HL, et al. Chronic supplementation with a mitochondrial antioxidant (MitoQ) improves vascular function in healthy older adults. Hypertension. 2018;71(6):1056-1063. DOI: 10.1161/hypertensionaha.117.10787
  37. 37. Young CN. Endoplasmic reticulum stress in the pathogenesis of hypertension. Experimental Physiology. 2017;102(8):869-884. DOI: 10.1113/ep086274
  38. 38. Zito E. ERO1: A protein disulfide oxidase and H2O2 producer. Free Radical Biology & Medicine. 2015;83:299-304. DOI: 10.1016/j.freeradbiomed.2015.01.011
  39. 39. Rainbolt TK, Saunders JM, Wiseman RL. Stress-responsive regulation of mitochondria through the ER unfolded protein response. Trends in Endocrinology and Metabolism: TEM. 2014;25(10):528-537. DOI: 10.1016/j.tem.2014.06.007
  40. 40. Malhotra JD, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress: A vicious cycle or a double-edged sword? Antioxidants & Redox Signaling. 2007;9(12):2277-2293. DOI: 10.1089/ars.2007.1782
  41. 41. Cao SS, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress in cell fate decision and human disease. Antioxidants & Redox Signaling. 2014;21(3):396-413. DOI: 10.1089/ars.2014.5851
  42. 42. Bravo R, Gutierrez T, Paredes F, Gatica D, Rodriguez AE, Pedrozo Z, et al. Endoplasmic reticulum: ER stress regulates mitochondrial bioenergetics. The International Journal of Biochemistry & Cell Biology. 2012;44(1):16-20. DOI: 10.1016/j.biocel.2011.10.012
  43. 43. Santos CX, Nabeebaccus AA, Shah AM, Camargo LL, Filho SV, Lopes LR. Endoplasmic reticulum stress and Nox-mediated reactive oxygen species signaling in the peripheral vasculature: Potential role in hypertension. Antioxidants & Redox Signaling. 2014;20(1):121-134. DOI: 10.1089/ars.2013.5262
  44. 44. Shanahan CM, Furmanik M. Endoplasmic reticulum stress in arterial smooth muscle cells: A novel regulator of vascular disease. Current Cardiology Reviews. 2017;13(2):94-105. DOI: 10.2174/1573403x12666161014094738
  45. 45. Ochoa CD, Wu RF, Terada LS. ROS signaling and ER stress in cardiovascular disease. Molecular Aspects of Medicine. 2018;63:18-29. DOI: 10.1016/j.mam.2018.03.002
  46. 46. Neuber C, Uebeler J, Schulze T, Sotoud H, El-Armouche A, Eschenhagen T. Guanabenz interferes with ER stress and exerts protective effects in cardiac myocytes. PLoS One. 2014;9(6):e98893. DOI: 10.1371/journal.pone.0098893
  47. 47. Abdulkarim B, Hernangomez M, Igoillo-Esteve M, Cunha DA, Marselli L, Marchetti P, et al. Guanabenz sensitizes pancreatic β cells to lipotoxic endoplasmic reticulum stress and apoptosis. Endocrinology. 2017;158(6):1659-1670. DOI: 10.1210/en.2016-1773
  48. 48. Choi SK, Lim M, Byeon SH, Lee YH. Inhibition of endoplasmic reticulum stress improves coronary artery function in the spontaneously hypertensive rats. Scientific Reports. 2016;6:31925. DOI: 10.1038/srep31925
  49. 49. Ceylan-Isik AF, Sreejayan N, Ren J. Endoplasmic reticulum chaperon tauroursodeoxycholic acid alleviates obesity-induced myocardial contractile dysfunction. Journal of Molecular and Cellular Cardiology. 2011;50(1):107-116. DOI: 10.1016/j.yjmcc.2010.10.023
  50. 50. Han S, Bal NB, Sadi G, Usanmaz SE, Tuglu MM, Uludag MO, et al. Inhibition of endoplasmic reticulum stress protected DOCA-salt hypertension-induced vascular dysfunction. Vascular Pharmacology. 2019;113:38-46. DOI: 10.1016/j.vph.2018.11.004
  51. 51. Lynn EG, Lhoták Š, Lebeau P, Byun JH, Chen J, Platko K, et al. 4-Phenylbutyrate protects against atherosclerotic lesion growth by increasing the expression of HSP25 in macrophages and in the circulation of Apoe(−/−) mice. The FASEB Journal. 2019;33(7):8406-8422. DOI: 10.1096/fj.201802293RR
  52. 52. Luo T, Kim JK, Chen B, Abdel-Latif A, Kitakaze M, Yan L. Attenuation of ER stress prevents post-infarction-induced cardiac rupture and remodeling by modulating both cardiac apoptosis and fibrosis. Chemico-Biological Interactions. 2015;225:90-98. DOI: 10.1016/j.cbi.2014.10.032
  53. 53. Luo T, Chen B, Wang X. 4-PBA prevents pressure overload-induced myocardial hypertrophy and interstitial fibrosis by attenuating endoplasmic reticulum stress. Chemico-Biological Interactions. 2015;242:99-106. DOI: 10.1016/j.cbi.2015.09.025
  54. 54. Holmgren A. Antioxidant function of thioredoxin and glutaredoxin systems. Antioxidants & Redox Signaling. 2000;2(4):811-820. DOI: 10.1089/ars.2000.2.4-811
  55. 55. Berndt C, Lillig CH, Holmgren A. Thiol-based mechanisms of the thioredoxin and glutaredoxin systems: Implications for diseases in the cardiovascular system. American Journal of Physiology. Heart and Circulatory Physiology. 2007;292(3):H1227-H1236. DOI: 10.1152/ajpheart.01162.2006
  56. 56. Rhee SG, Kang SW, Chang TS, Jeong W, Kim K. Peroxiredoxin, a novel family of peroxidases. IUBMB Life. 2001;52(1-2):35-41. DOI: 10.1080/15216540252774748
  57. 57. Szeliga M. Peroxiredoxins in neurodegenerative diseases. Antioxidants (Basel, Switzerland). 2020;9(12):1203. DOI: 10.3390/antiox9121203
  58. 58. Jeong SJ, Park JG, Oh GT. Peroxiredoxins as potential targets for cardiovascular disease. Antioxidants (Basel, Switzerland). 2021;10(8):1244. DOI: 10.3390/antiox10081244
  59. 59. Andreadou I, Efentakis P, Frenis K, Daiber A, Schulz R. Thiol-based redox-active proteins as cardioprotective therapeutic agents in cardiovascular diseases. Basic Research in Cardiology. 2021;116(1):44. DOI: 10.1007/s00395-021-00885-5
  60. 60. Aota M, Matsuda K, Isowa N, Wada H, Yodoi J, Ban T. Protection against reperfusion-induced arrhythmias by human thioredoxin. Journal of Cardiovascular Pharmacology. 1996;27(5):727-732. DOI: 10.1097/00005344-199605000-00016
  61. 61. Yoshioka J, Schulze PC, Cupesi M, Sylvan JD, MacGillivray C, Gannon J, et al. Thioredoxin-interacting protein controls cardiac hypertrophy through regulation of thioredoxin activity. Circulation. 2004;109(21):2581-2586. DOI: 10.1161/01.cir.0000129771.32215.44
  62. 62. Meuillet EJ, Mahadevan D, Berggren M, Coon A, Powis G. Thioredoxin-1 binds to the C2 domain of PTEN inhibiting PTEN's lipid phosphatase activity and membrane binding: A mechanism for the functional loss of PTEN's tumor suppressor activity. Archives of Biochemistry and Biophysics. 2004;429(2):123-133. DOI: 10.1016/j.abb.2004.04.020
  63. 63. Turoczi T, Chang VW, Engelman RM, Maulik N, Ho YS, Das DK. Thioredoxin redox signaling in the ischemic heart: An insight with transgenic mice overexpressing Trx1. Journal of Molecular and Cellular Cardiology. 2003;35(6):695-704. DOI: 10.1016/s0022-2828(03)00117-2
  64. 64. Jekell A, Hossain A, Alehagen U, Dahlström U, Rosén A. Elevated circulating levels of thioredoxin and stress in chronic heart failure. European Journal of Heart Failure. 2004;6(7):883-890. DOI: 10.1016/j.ejheart.2004.03.003
  65. 65. Kishimoto C, Shioji K, Nakamura H, Nakayama Y, Yodoi J, Sasayama S. Serum thioredoxin (TRX) levels in patients with heart failure. Japanese Circulation Journal. 2001;65(6):491-494. DOI: 10.1253/jcj.65.491
  66. 66. Ago T, Sadoshima J. Thioredoxin1 as a negative regulator of cardiac hypertrophy. Antioxidants & Redox Signaling. 2007;9(6):679-687. DOI: 10.1089/ars.2007.1529
  67. 67. Haendeler J, Popp R, Goy C, Tischler V, Zeiher AM, Dimmeler S. Cathepsin D and H2O2 stimulate degradation of thioredoxin-1: Implication for endothelial cell apoptosis. The Journal of Biological Chemistry. 2005;280(52):42945-42951. DOI: 10.1074/jbc.M506985200
  68. 68. Hilgers RH, Kundumani-Sridharan V, Subramani J, Chen LC, Cuello LG, Rusch NJ, et al. Thioredoxin reverses age-related hypertension by chronically improving vascular redox and restoring eNOS function. Science Translational Medicine. 2017;9(376):eaaf6094. DOI: 10.1126/scitranslmed.aaf6094
  69. 69. Koneru S, Penumathsa SV, Thirunavukkarasu M, Zhan L, Maulik N. Thioredoxin-1 gene delivery induces heme oxygenase-1 mediated myocardial preservation after chronic infarction in hypertensive rats. American Journal of Hypertension. 2009;22(2):183-190. DOI: 10.1038/ajh.2008.318
  70. 70. Adluri RS, Thirunavukkarasu M, Zhan L, Dunna NR, Akita Y, Selvaraju V, et al. Glutaredoxin-1 overexpression enhances neovascularization and diminishes ventricular remodeling in chronic myocardial infarction. PLoS One. 2012;7(3):e34790. DOI: 10.1371/journal.pone.0034790
  71. 71. Murdoch CE, Shuler M, Haeussler DJ, Kikuchi R, Bearelly P, Han J, et al. Glutaredoxin-1 up-regulation induces soluble vascular endothelial growth factor receptor 1, attenuating post-ischemia limb revascularization. The Journal of Biological Chemistry. 2014;289(12):8633-8644. DOI: 10.1074/jbc.M113.517219
  72. 72. Gallogly MM, Shelton MD, Qanungo S, Pai HV, Starke DW, Hoppel CL, et al. Glutaredoxin regulates apoptosis in cardiomyocytes via NFkappaB targets Bcl-2 and Bcl-xL: Implications for cardiac aging. Antioxidants & Redox Signaling. 2010;12(12):1339-1353. DOI: 10.1089/ars.2009.2791
  73. 73. Murata H, Ihara Y, Nakamura H, Yodoi J, Sumikawa K, Kondo T. Glutaredoxin exerts an antiapoptotic effect by regulating the redox state of Akt. The Journal of Biological Chemistry. 2003;278(50):50226-50233. DOI: 10.1074/jbc.M310171200
  74. 74. Pan S, Berk BC. Glutathiolation regulates tumor necrosis factor-alpha-induced caspase-3 cleavage and apoptosis: Key role for glutaredoxin in the death pathway. Circulation Research. 2007;100(2):213-219. DOI: 10.1161/01.res.0000256089.30318.20
  75. 75. Okuda M, Inoue N, Azumi H, Seno T, Sumi Y, Hirata K, et al. Expression of glutaredoxin in human coronary arteries: Its potential role in antioxidant protection against atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21(9):1483-1487. DOI: 10.1161/hq0901.095550
  76. 76. Kim J, Kim J, Kook H, Park WJ. PICOT alleviates myocardial ischemia-reperfusion injury by reducing intracellular levels of reactive oxygen species. Biochemical and Biophysical Research Communications. 2017;485(4):807-813. DOI: 10.1016/j.bbrc.2017.02.136
  77. 77. Donelson J, Wang Q , Monroe TO, Jiang X, Zhou J, Yu H, et al. Cardiac-specific ablation of glutaredoxin 3 leads to cardiac hypertrophy and heart failure. Physiological Reports. 2019;7(8):e14071. DOI: 10.14814/phy2.14071
  78. 78. Kisucka J, Chauhan AK, Patten IS, Yesilaltay A, Neumann C, Van Etten RA, et al. Peroxiredoxin1 prevents excessive endothelial activation and early atherosclerosis. Circulation Research. 2008;103(6):598-605. DOI: 10.1161/circresaha.108.174870
  79. 79. Jeong SJ, Kim S, Park JG, Jung IH, Lee MN, Jeon S, et al. Prdx1 (peroxiredoxin 1) deficiency reduces cholesterol efflux via impaired macrophage lipophagic flux. Autophagy. 2018;14(1):120-133. DOI: 10.1080/15548627.2017.1327942
  80. 80. Guo X, Yamada S, Tanimoto A, Ding Y, Wang KY, Shimajiri S, et al. Overexpression of peroxiredoxin 4 attenuates atherosclerosis in apolipoprotein E knockout mice. Antioxidants & Redox Signaling. 2012;17(10):1362-1375. DOI: 10.1089/ars.2012.4549
  81. 81. Park JG, Yoo JY, Jeong SJ, Choi JH, Lee MR, Lee MN, et al. Peroxiredoxin 2 deficiency exacerbates atherosclerosis in apolipoprotein E-deficient mice. Circulation Research. 2011;109(7):739-749. DOI: 10.1161/circresaha.111.245530
  82. 82. Choi MH, Lee IK, Kim GW, Kim BU, Han YH, Yu DY, et al. Regulation of PDGF signalling and vascular remodelling by peroxiredoxin II. Nature. 2005;435(7040):347-353. DOI: 10.1038/nature03587
  83. 83. Jang JY, Wang SB, Min JH, Chae YH, Baek JY, Yu DY, et al. Peroxiredoxin II is an antioxidant enzyme that negatively regulates collagen-stimulated platelet function. The Journal of Biological Chemistry. 2015;290(18):11432-11442. DOI: 10.1074/jbc.M115.644260
  84. 84. Li W, Febbraio M, Reddy SP, Yu DY, Yamamoto M, Silverstein RL. CD36 participates in a signaling pathway that regulates ROS formation in murine VSMCs. The Journal of Clinical Investigation. 2010;120(11):3996-4006. DOI: 10.1172/jci42823
  85. 85. Jeong SJ, Cho MJ, Ko NY, Kim S, Jung IH, Min JK, et al. Deficiency of peroxiredoxin 2 exacerbates angiotensin II-induced abdominal aortic aneurysm. Experimental & Molecular Medicine. 2020;52(9):1587-1601. DOI: 10.1038/s12276-020-00498-3
  86. 86. Tang C, Yin G, Huang C, Wang H, Gao J, Luo J, et al. Peroxiredoxin-1 ameliorates pressure overload-induced cardiac hypertrophy and fibrosis. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie. 2020;129:110357. DOI: 10.1016/j.biopha.2020.110357
  87. 87. Matsushima S, Ide T, Yamato M, Matsusaka H, Hattori F, Ikeuchi M, et al. Overexpression of mitochondrial peroxiredoxin-3 prevents left ventricular remodeling and failure after myocardial infarction in mice. Circulation. 2006;113(14):1779-1786. DOI: 10.1161/circulationaha.105.582239
  88. 88. Kumar V, Kitaeff N, Hampton MB, Cannell MB, Winterbourn CC. Reversible oxidation of mitochondrial peroxiredoxin 3 in mouse heart subjected to ischemia and reperfusion. FEBS Letters. 2009;583(6):997-1000. DOI: 10.1016/j.febslet.2009.02.018
  89. 89. Mannaa A, Hanisch FG. Redox proteomes in human physiology and disease mechanisms. Journal of Proteome Research. 2020;19(1):1-17. DOI: 10.1021/acs.jproteome.9b00586
  90. 90. Reczek CR, Chandel NS. ROS-dependent signal transduction. Current Opinion in Cell Biology. 2015;33:8-13. DOI: 10.1016/j.ceb.2014.09.010
  91. 91. Winterbourn CC, Hampton MB. Thiol chemistry and specificity in redox signaling. Free Radical Biology & Medicine. 2008;45(5):549-561. DOI: 10.1016/j.freeradbiomed.2008.05.004
  92. 92. Wojdyla K, Rogowska-Wrzesinska A. Differential alkylation-based redox proteomics—Lessons learnt. Redox Biology. 2015;6:240-252. DOI: 10.1016/j.redox.2015.08.005
  93. 93. Sheehan D, McDonagh B. The clinical potential of thiol redox proteomics. Expert Review of Proteomics. 2020;17(1):41-48. DOI: 10.1080/14789450.2020.1704260
  94. 94. Hansen RE, Winther JR. An introduction to methods for analyzing thiols and disulfides: Reactions, reagents, and practical considerations. Analytical Biochemistry. 2009;394(2):147-158. DOI: 10.1016/j.ab.2009.07.051
  95. 95. Bonzon-Kulichenko E, Camafeita E, López JA, Gómez-Serrano M, Jorge I, Calvo E, et al. Improved integrative analysis of the thiol redox proteome using filter-aided sample preparation. Journal of Proteomics. 2020;214:103624. DOI: 10.1016/j.jprot.2019.103624
  96. 96. Fu C, Hu J, Liu T, Ago T, Sadoshima J, Li H. Quantitative analysis of redox-sensitive proteome with DIGE and ICAT. Journal of Proteome Research. 2008;7(9):3789-3802. DOI: 10.1021/pr800233r
  97. 97. Martínez-Acedo P, Nuñez E, Gómez FJS, Moreno M, Ramos E, Izquierdo-Álvarez A, et al. A novel strategy for global analysis of the dynamic thiol redox proteome. Molecular & Cellular Proteomics. 2012;11(9):800-813. DOI: 10.1074/mcp.M111.016469
  98. 98. Corbacho-Alonso N, Baldán-Martín M, López JA, Rodríguez-Sánchez E, Martínez PJ, Mourino-Alvarez L, et al. Cardiovascular risk stratification based on oxidative stress for early detection of pathology. Antioxidants & Redox Signaling. 2021;35(8):602-617. DOI: 10.1089/ars.2020.8254
  99. 99. Sastre-Oliva T, Corbacho-Alonso N, Albo-Escalona D, Lopez JA, Lopez-Almodovar LF, Vázquez J, et al. The influence of coronary artery disease in the development of aortic stenosis and the importance of the albumin redox state. Antioxidants (Basel, Switzerland). 2022;11(2):317. DOI: 10.3390/antiox11020317

Written By

Laura Mourino-Alvarez, Tamara Sastre-Oliva, Nerea Corbacho-Alonso and Maria G. Barderas

Submitted: 16 May 2022 Reviewed: 14 June 2022 Published: 22 July 2022