Next Article in Journal
Nicotinamide Mononucleotide Restores NAD+ Levels to Alleviate LPS-Induced Inflammation via the TLR4/NF-κB/MAPK Signaling Pathway in Mice Granulosa Cells
Next Article in Special Issue
Myocardial DYRK1B Expression Is Increased in Patients with Impaired Cardiac Contractility and Sleep-Disordered Breathing
Previous Article in Journal
Antioxidant and Laxative Effects of Methanol Extracts of Green Pine Cones (Pinus densiflora) in Sprague-Dawley Rats with Loperamide-Induced Constipation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Deciphering Oxidative Stress in Cardiovascular Disease Progression: A Blueprint for Mechanistic Understanding and Therapeutic Innovation

1
Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
2
Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
3
Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(1), 38; https://doi.org/10.3390/antiox14010038
Submission received: 19 November 2024 / Revised: 20 December 2024 / Accepted: 27 December 2024 / Published: 31 December 2024

Abstract

:
Oxidative stress plays a pivotal role in the pathogenesis and progression of cardiovascular diseases (CVDs). This review focuses on the signaling pathways of oxidative stress during the development of CVDs, delving into the molecular regulatory networks underlying oxidative stress in various disease stages, particularly apoptosis, inflammation, fibrosis, and metabolic imbalance. By examining the dual roles of oxidative stress and the influences of sex differences on oxidative stress levels and cardiovascular disease susceptibility, this study offers a comprehensive understanding of the pathogenesis of cardiovascular diseases. The study integrates key findings from current research in three comprehensive ways. First, it outlines the major CVDs associated with oxidative stress and their respective signaling pathways, emphasizing oxidative stress’s central role in cardiovascular pathology. Second, it summarizes the cardiovascular protective effects, mechanisms of action, and animal models of various antioxidants, offering insights into future drug development. Third, it discusses the applications, advantages, limitations, and potential molecular targets of gene therapy in CVDs, providing a foundation for novel therapeutic strategies. These tables underscore the systematic and integrative nature of this study while offering a theoretical basis for precision treatment for CVDs. A major contribution of this study is the systematic review of the differential effects of oxidative stress across different stages of CVDs, in addition to the proposal of innovative, multi-level intervention strategies, which open new avenues for precision treatment of the cardiovascular system.

1. Introduction

Cardiovascular diseases (CVDs) are a significant global health and economic burden, highlighting the critical importance of maintaining cardiovascular health [1]. CVDs are among the leading causes of morbidity and mortality worldwide, primarily resulting in cardiovascular damage [2,3]. These diseases arise from a range of factors, including genetic predisposition, chronic conditions (e.g., hypertension and diabetes), unhealthy lifestyle habits (e.g., smoking and obesity), and environmental influences [4,5]. Oxidative stress is a key contributor to the pathogenesis of CVDs [6]. Research into its underlying mechanisms not only enhances our understanding of the disease but also reveals potential therapeutic targets, paving the way for more effective drugs and preventative strategies.
Oxidative stress refers to a pathological state characterized by the excessive production of reactive oxygen species (ROS) [7]. Due to their high reactivity, ROS can damage cellular components, including proteins, lipids, and DNA [8,9,10]. In cardiomyocytes, ROS can induce mitochondrial dysfunction, leading to energy metabolism disorders and myocardial injury [11]. Additionally, ROS play a role in atherosclerosis by promoting endothelial dysfunction and stimulating smooth muscle cell proliferation [12]. Targeting the oxidative stress pathway offers a promising therapeutic approach to mitigate these effects and slow CVD progression.
This review comprehensively examines the role of oxidative stress in CVDs, focusing on its heterogeneity across different disease types. It also emphasizes the importance of personalized medicine in CVD management, advocating for precision medicine strategies based on oxidative stress biomarkers as a theoretical foundation for clinical practice.

2. The Dynamic Role of Oxidative Stress in the Progression of Cardiovascular Disease

2.1. Molecular Mechanisms Underlying Oxidative Stress

Oxidative stress refers to an imbalance between the production of ROS and antioxidant defenses, leading to oxidative damage in cells and tissues (Figure 1). This imbalance is implicated in the pathogenesis of various diseases, including CVDs and neurodegenerative disorders.
ROS are pivotal in the balance between physiological signaling and pathological oxidative stress (Figure 1). The electron transport chain (ETC) in a mitochondrion serves as a primary source; electron leakage generates superoxide radicals, particularly under the conditions of impaired oxidative phosphorylation [13]. NOX enzymes are another major contributor, producing ROS as immune responses and cellular signals, but their dysregulation is linked to oxidative damage [14]. Similarly, xanthine oxidase (XO) catalyzes purine metabolism, generating superoxide and hydrogen peroxide, which are exacerbated during ischemia-reperfusion injury [15]. Endoplasmic reticulum (ER) stress further amplifies oxidative stress by disrupting protein folding and activating ROS-producing pathways [16]. Together, these systems orchestrate a delicate interplay between ROS production and cellular homeostasis, with dysregulation contributing to the pathogenesis of numerous diseases.
The ETC, which is a series of protein complexes embedded in the inner mitochondrial membrane, plays a central role in oxidative phosphorylation [17,18]. The mitochondrial respiratory chain consists of four transmembrane protein complexes that facilitate electron transfer and oxidative phosphorylation [19]. Complexes I and II introduce electrons from NADH and FADH₂ into the respiratory chain [20,21]. Complex III transfers electrons within the lipid membrane, coupling this process with proton translocation [22]. Complex IV, the terminal oxidase, transfers electrons to oxygen molecules, producing water and completing the final proton pumping process [23]. As electrons are transferred along this chain, a proton gradient is established across the inner membrane, driving ATP synthesis [24]. However, a small proportion of electrons may leak from the chain and react with molecular oxygen, generating superoxide anions—the primary ROS produced in mitochondria [19,25]. The uncoupling of oxidative phosphorylation, coupled with structural or functional abnormalities in the electron transport chain complexes, can lead to uncontrolled electron flow, increasing the risk of electron leakage and inducing oxidative damage [26,27]. Proton leak occurs in two forms: basal (associated with the mitochondrial inner membrane lipid bilayer and adenine nucleotide translocator [ANT]) and inducible (regulated by uncoupling proteins [UCPs] and ANT) [28,29]. Both constitutive and regulatory proton transporters are critical in maintaining cellular energy homeostasis. Targeting these transporters with pharmacological interventions holds promise as a novel therapeutic strategy for mitochondrial diseases.
NOX, a multi-subunit membrane-bound enzyme complex, is the primary enzyme responsible for the generation of superoxide anion (O2) through the reduction of molecular oxygen [30]. This initial step leads to the production of a variety of ROS (Figure 1). The NOX family can be broadly categorized into two groups based on structural and functional characteristics: classical NOX isoforms (Nox1–Nox4), which typically form complexes with the p22phox subunit, and dual oxidase (Duox) isoforms (Duox1 and Duox2), which require the Duoxa subunit for their activity [31]. NOX-derived ROS synergize with growth factors such as VEGF and PDGF to promote endothelial cell proliferation, migration, and tube formation through the activation of downstream signaling pathways, thereby driving angiogenesis [32,33]. Nox2 and Nox4 form heterodimers in the heart [34]. Nox2-derived ROS play a central role in the pathogenesis of angiotensin II-induced left ventricular hypertrophy and cardiac remodeling associated with pressure overload [35]. Additionally, Nox2 is involved in the cardioprotective effects of ischemic preconditioning [36]. Cardiac Nox4 expression is upregulated under various stress conditions, leading to increased ROS production. Nox5, a unique calcium-dependent NADPH oxidase, is widely expressed in the cardiovascular system [37]. Unlike its homologs, Nox5 can be activated by calcium ions without the need for additional subunits, making it a key enzyme in ROS generation in cardiovascular diseases [38]. The overexpression of Nox5 is closely associated with various cardiovascular diseases, including hypertension, aneurysms, and atherosclerosis [39]. NOX-dependent redox signaling, characterized by the generation of localized and low levels of ROS, regulates a variety of cellular processes, including proliferation, apoptosis, and migration [40].
XO is a key oxidoreductase that initially catalyzes the oxidation of hypoxanthine to xanthine and subsequently catalyzes the oxidation of xanthine to uric acid [41]. During this process, XO uses molecular oxygen as an electron acceptor, generating O2 and initiating a cascade of oxidative reactions that produce various ROS [15,42]. XO contains several electron transfer centers, including a molybdenum cofactor, FAD, and iron–sulfur clusters [42]. During catalysis, electrons are transferred from the substrate (hypoxanthine or xanthine) to these centers and, ultimately, to molecular oxygen. As a result, molecular oxygen is reduced to superoxide anion, which then initiates a cascade of reactions that generate various ROS, including hydrogen peroxide (H2O2) and hydroxyl radical (OH) [43,44].
ER stress, triggered by the accumulation of unfolded or misfolded proteins, can lead to apoptosis (Figure 1). Increased cytosolic Ca2+, released from the ER, activates kinases such as PKC and CaMKII, which phosphorylate and activate NADPH oxidases, leading to ROS generation [45,46]. The unfolded protein response (UPR) is a cellular adaptive response to ER stress that is primarily mediated by three transmembrane ER proteins: IRE1α, PERK, and ATF6 [16,47]. These sensors detect unfolded proteins in the ER lumen and initiate downstream signaling pathways. The primary mechanism underlying ROS generation during ER stress involves electron transfer and oxidative reactions during protein folding. Protein disulfide isomerase (PDI) plays a crucial role in catalyzing disulfide bond formation, and electron transfer in this process leads to the oxidation of ERO1, resulting in ROS production [48]. Nox4 interacts with PDI and directly generates superoxide anions [49,50]. Additionally, glutathione (GSH), an antioxidant, contributes to ROS production when its redox balance is disrupted [51]. The microsomal monooxygenase system, particularly cytochrome P450 2E1, also generates ROS during electron transfer [52]. Collectively, these mechanisms disrupt cellular redox homeostasis, ultimately leading to cellular damage or death.
Peroxisomes are cellular organelles responsible for various important oxidative reactions [53]. They are involved in the metabolism of fatty acids (through α-oxidation), very long chain fatty acids (VLCFAs; through β-oxidation), and purines [54]. Within peroxisomes, oxidases catalyze the oxidation of substrates, producing H2O2. To mitigate the harmful effects of hydrogen peroxide, peroxisomes contain a robust antioxidant defense system, including catalase, glutathione peroxidase, epoxide hydrolase, PMP20, and peroxidases (I, V, and VI), which collectively detoxify H2O2 [55,56]. The protective role of catalase in peroxisomes is supported by studies in human fibroblasts from patients with multiple peroxisomal enzyme deficiencies and catalase deficiency, as well as in experimental animal models, such as rats with alcoholic cardiomyopathy [57,58]. The accumulation of hydrogen peroxide in peroxisomes, resulting from either increased oxidase activity or impaired antioxidant defenses, can induce oxidative stress [59]. Therefore, understanding the interplay between peroxisomes, ROS, and oxidative stress is crucial for elucidating the cellular mechanisms underlying health and disease.

2.2. Sex-Based Differences in Redox Status and Their Implications for CVD

Young men are more susceptible to coronary heart disease, myocardial infarction, and other ischemic heart diseases due to their lower estrogen levels and higher inflammatory burden [60]. Postmenopausal women experience an increased risk of cardiovascular diseases as estrogen levels decline [61]. Female patients with cardiovascular diseases exhibit highly variable clinical presentations that are often characterized by atypical symptoms, resulting in delayed diagnosis [62]. Additionally, women have poorer outcomes in certain cardiovascular conditions, which may be linked to sex-based differences in inflammatory responses and microvascular function.
Sex differences in mitochondria represent a promising area of research in cardiovascular diseases. Mitochondrial diseases, often caused by mutations in either mitochondrial or nuclear DNA, can have a greater impact on males than females. Mitochondrial dysfunction can lead to a variety of diseases, including cardiomyopathy, and these diseases often exhibit sex-specific differences [63].
Sex hormones, particularly estrogen and testosterone, exert significant influences on oxidative stress levels [64]. Estrogen enhances the body’s antioxidant capacity by activating the Nrf2/Keap1 signaling pathway and upregulating the expression of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPx) [65]. The regulatory effects of androgens on ROS are more complex and may depend on hormone concentration.
The activation of the Nrf2 and PI3K/Akt pathways, coupled with the preservation of mitochondrial function by estrogen, enhances the antioxidant defense and promotes cell survival in the female cardiovascular system [66]. Consequently, women exhibit greater resistance to oxidative stress compared to men, who exhibit weaker signaling in these pathways [67]. Sex differences significantly influence the development and progression of cardiovascular diseases by modulating sex hormones, antioxidant systems, and redox signaling pathways.

2.3. Oxidative Stress Profile at Various Stages of CVDs

Oxidative stress levels fluctuate dynamically across different CVDs, with both the magnitude and targets of oxidative stress evolving as the diseases progress [68]. The role of oxidative stress in the developmental process of cardiovascular disease is shown in Figure 2. This dynamic process creates a vicious cycle that exacerbates disease severity (Figure 2). By elucidating the temporal changes in oxidative stress during CVD progression, we can gain deeper insights into disease pathogenesis and identify novel therapeutic targets for antioxidant interventions.

2.3.1. Early Triggering Mechanisms of Oxidative Stress in Endothelial Injury

As a pivotal regulator of cardiovascular homeostasis, the vascular endothelium is influenced by both genetic and epigenetic mechanisms [69]. Endothelial dysfunction, which is characterized by reduced nitric oxide bioavailability, increased adhesion molecule expression, and enhanced permeability, is a hallmark of atherosclerosis that initiates a cascade of events leading to plaque formation [70].
Elevated lipoprotein permeability, which is exacerbated oxidative stress, enhanced monocyte adhesion, abnormal extracellular matrix remodeling, and dysregulated coagulation/fibrinolysis, collectively characterizes endothelial dysfunction [71]. In the pathogenesis of atherosclerosis, various pathophysiological stimuli—such as pro-inflammatory cytokines, pathogen-associated molecular patterns (PAMPs), advanced glycation end products (AGEs), oxidized low-density lipoprotein (ox-LDL), and its components (e.g., lysophosphatidylcholine)—synergistically induce and aggravate endothelial dysfunction, thereby promoting the formation and progression of atherosclerotic plaques [72].
The formation of atherosclerotic lesions is closely linked to the pro-inflammatory activation of endothelial cells. Upon injury, endothelial cells lose their anti-inflammatory and anti-thrombotic properties, transitioning to a pro-inflammatory and pro-thrombotic phenotype [70]. Injured endothelial cells activate transcription factors such as NF-κB, upregulating the expression of a series of inflammation-related genes [73]. The products of these genes, including adhesion molecules (e.g., VCAM-1), chemokines (e.g., MCP-1 and fractalkine), and procoagulant factors (e.g., TF, VWF, and PAI-1), recruit monocytes, T lymphocytes, and other inflammatory cells, promoting their accumulation within the vessel wall (Figure 2) [74,75]. These inflammatory cells release additional inflammatory mediators, perpetuating a vicious cycle and exacerbating the inflammatory response within the vessel wall.
NF-κB is a pivotal transcription factor that plays a central role in the development of atherosclerosis (Figure 2) [76]. In endothelial cells, various pro-inflammatory stimuli, including IL-1, TNF, endotoxin, oxidized low-density lipoprotein, and advanced glycation end products, synergistically activate the NF-κB signaling pathway [77,78]. Additionally, alterations in hemodynamic factors, such as turbulence and low shear stress, influence NF-κB activity, further contributing to atherosclerosis development [79,80]. Endothelial cell activation can be classified into type I and type II activation [81]. Type I activation is an acute response that is typically triggered by infection or injury, leading to increased vascular permeability and leukocyte infiltration. In contrast, type II activation is a chronic response characterized by the sustained expression of inflammatory molecules in endothelial cells, and it results in a chronic inflammatory environment that promotes the formation and growth of atherosclerotic plaques. The cellular network within the vessel wall—including endothelial cells, smooth muscle cells, monocytes/macrophages, and keratinocytes—releases cytokines, growth factors, and ROS via paracrine signaling, further amplifying the inflammatory response [82]. This intricate cellular interplay drives the progression of atherosclerosis. Endothelial cell injury is the initiating event in atherosclerosis, with NF-κB activation serving as a critical step. Inflammation is a persistent core pathological feature throughout the disease process. A comprehensive understanding of these mechanisms is essential for developing more effective treatments to delay or reverse the progression of CVDs.

2.3.2. Oxidative Stress in the Formation of Atherosclerotic Lesions

Atherosclerosis is a chronic, progressive disease driven by endothelial injury, inflammation, lipid deposition, and plaque formation within the arterial intima. It is a major contributor to CVD [83], but it represents only one aspect of the multifactorial etiology underlying these conditions. After activation by ROS, MMPs can induce degradation of elastic fibers in the vascular wall and vascular dilation, ultimately leading to the formation or dissection of aortic aneurysms [84]. These mechanisms not only impair vascular function but also exacerbate atherosclerosis and its associated complications. The pathological process of atherosclerosis follows a series of key events: (1) lipoprotein oxidation induces endothelial injury, which triggers inflammation and the recruitment of monocytes; (2) these monocytes differentiate into macrophages, which then engulf oxidized low-density lipoprotein (ox-LDL), forming foam cells; (3) the accumulation of foam cells leads to the formation of fatty streaks, which may progress to the development of a fibrous cap; and (4) plaque rupture exposes the thrombogenic core, resulting in thrombosis and potential vessel occlusion [85,86,87].
Macrophages can be polarized into M1 and M2 phenotypes. M1 macrophages primarily participate in Th1 cell-mediated inflammatory responses, secreting abundant pro-inflammatory cytokines, chemokines, nitric oxide, and ROS to eliminate pathogens [88]. In contrast, M2 macrophages exhibit anti-inflammatory properties and are involved in tissue repair, angiogenesis, and other processes. They secrete anti-inflammatory cytokines such as IL-10 and TGF-β to suppress inflammation [89]. Studies have shown that foam cells respond differently to M1- and M2-polarizing stimuli. When exposed to M1-polarizing factors (e.g., LPS and IFN-γ), foam cells exhibit a weaker pro-inflammatory response compared to normal macrophages [90]. Conversely, when exposed to M2-polarizing factors (e.g., IL-4), foam cells show a similar anti-inflammatory response to normal macrophages [90].
Foam cells are pivotal components of atherosclerotic plaques that are primarily derived from macrophages but sometimes originate from vascular endothelial and smooth muscle cells [91]. These cells undergo phenotypic switching under specific conditions, contributing to plaque formation. Foam cell formation is critically dependent on lipid accumulation, particularly the uptake of ox-LDL [92]. Nox5 promotes plaque progression by inducing LDL oxidation and foam cell formation. Simultaneously, ROS generated by Nox5 activates matrix metalloproteinases (MMPs), impairing plaque stability and exacerbating the lesion [93]. Macrophages internalize ox-LDL through scavenger receptors such as SR-A1, CD36, and LOX-1 and esterify cholesterol using enzymes like ACAT1, leading to intracellular lipid accumulation and foam cell formation [94]. During atherosclerosis, macrophage cholesterol homeostasis is disrupted: lipid uptake is enhanced by the upregulation of receptors like LOX-1, while cholesterol efflux is impaired due to the downregulation of transporters such as ABCA1 and ABCG1 [95]. Pro-inflammatory cytokines, ox-LDL, lysophosphatidylcholine, and other factors synergistically promote macrophage-to-foam cell transformation, exacerbating plaque development [96]. Foam cell formation is a hallmark of atherosclerosis, driven by the dysregulation of lipid metabolism, inflammation, and phenotypic switching. A comprehensive understanding of foam cell formation mechanisms is essential for developing novel therapeutic strategies against atherosclerosis.
NLRP3 plays a pivotal role in the development of atherosclerosis [97]. As a member of the NLR family, NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) is a key component of the inflammasome [98]. The activation of NLRP3 is a multi-step process involving multiple signaling pathways. Pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) can recognize and bind to NLRP3, inducing conformational changes [99]. Simultaneously, these stimuli can activate other signaling pathways, such as the NF-κB pathway, to promote the expression of NLRP3 and pro-inflammatory cytokines like IL-1β [100]. Furthermore, alterations in intracellular environments, such as decreased potassium ion concentrations, ROS generation, and lysosomal dysfunction, can directly or indirectly activate NLRP3 [101]. Upon activation, NLRP3 recruits the apoptosis-associated speck-like protein containing CARD (ASC) and caspase-1, forming a complex that cleaves pro-IL-1β and pro-IL-18 into their active forms, triggering a robust inflammatory response [102]. The release of pro-inflammatory cytokines IL-1β and IL-18 induced by NLRP3 activation promotes the formation and development of atherosclerotic plaques [103]. Additionally, NLRP3 can polarize macrophages toward the M1 phenotype, which exhibit a pro-inflammatory phenotype, exacerbating atherosclerosis [104]. It can also promote the dedifferentiation and migration of vascular smooth muscle cells, leading to plaque instability [105]. Furthermore, NLRP3 can enhance cholesterol uptake and esterification, promoting the formation of foam cells [106].

2.3.3. Oxidative Stress and Reperfusion Injury in Acute Ischemic Events

Oxidative stress plays a pivotal role in ischemia-reperfusion injury during acute cardiovascular events such as myocardial infarction and stroke. Acute myocardial ischemia leads to a severe oxygen shortage in the myocardium. Under hypoxic conditions, myocardial cell metabolism shifts from aerobic respiration to anaerobic glycolysis, leading to reduced ATP production, lactate accumulation, and a decrease in intracellular PH [107].The acidic microenvironment further exacerbates cell injury by inhibiting mitochondrial permeability transition pore (mPTP) opening and disrupting ion homeostasis, ultimately causing cardiomyocyte death and severe cardiac dysfunction [108]. Additionally, even under low-flow ischemia or in the presence of residual oxygen, myocardial tissue can produce large amounts of ROS, further aggravating oxidative stress and accelerating myocardial injury [109].
Myocardial ischemia-reperfusion injury (MIRI) plays a pivotal role in the pathogenesis of heart disease [110]. Studies have demonstrated that ROS not only trigger a cascade of cellular events but also directly activate the NLRP3 inflammasome, playing pivotal roles in the pathogenesis of myocardial ischemia-reperfusion injury (MIRI) [111,112,113]. The activated NLRP3 inflammasome subsequently leads to the release of inflammatory cytokines, inducing a robust inflammatory response [114,115]. Moreover, the NLRP3 inflammasome plays a multifaceted role in MIRI, contributing to both cell death and endothelial dysfunction, resulting in vasoconstriction, and exacerbating cardiac injury [114].
Multiple factors contribute to the generation of ROS during myocardial ischemia-reperfusion injury, including the mitochondrial respiratory chain, xanthine oxidase, NADPH oxidase, nitric oxide synthase, and endoplasmic reticulum stress [116]. ROS generated during reperfusion oxidatively damage mitochondrial membrane proteins, phospholipids, and both mitochondrial (mtDNA) and nuclear (nDNA) genomes, thereby disrupting mitochondrial structure and function [117]. As the cell’s powerhouse, mitochondria provide energy for cardiomyocytes (HCMs), and impaired mitochondrial function directly impacts energy supply, weakening cardiac contractility and relaxation and ultimately contributing to CVDs. Mutations in mtDNA and nDNA further exacerbate mitochondrial dysfunction, resulting in excessive ROS production, decreased oxidative phosphorylation (OXPHOS) capacity, and a cascade of pathophysiological changes [118]. Mitophagy, a selective form of autophagy, serves as a quality control mechanism by degrading damaged mitochondria to maintain cellular homeostasis [119]. Impaired mitophagy leads to the accumulation of dysfunctional mitochondria, and BMAL1-mediated mitochondrial dysfunction is closely linked to dilated cardiomyopathy [120]. In myocardial ischemia-reperfusion injury, casein kinase 2α exacerbates cardiomyocyte injury by inhibiting mitophagy [121]. The induction of mitophagy can effectively alleviate mitochondrial dysfunction and lipid accumulation induced by a high-fat diet, preventing diabetic cardiomyopathy [122]. Decreased mitochondrial OXPHOS and increased ROS production are pivotal in the pathogenesis of CVDs. ROS damage mitochondrial DNA, induce apoptosis, and promote inflammation. In chronic heart failure and aneurysms, decreased OXPHOS function has been linked to reduced succinyl-CoA levels, decreased mitochondrial fusion, and loss of mitochondrial membrane potential [123]. Additionally, transcription factors like PGC-1β and microRNAs such as miR-27b-3p regulate OXPHOS, and their dysregulation is closely associated with CVDs [124].

2.3.4. Oxidative Stress Mechanisms in Cardiac Remodeling and Chronic Heart Failure

Heart failure (HF) is characterized by cardiac remodeling, which involves alterations in cardiomyocyte structure and function and remodeling of the extracellular matrix [125]. These changes result in abnormal cardiac geometry, ventricular dilation, and impaired contractile function, ultimately compromising cardiac output. Numerous studies have demonstrated elevated levels of ROS in HF patients, highlighting the significant role of oxidative stress in the pathogenesis and progression of HF [126,127]. Both rapid pacing-induced canine HF models and murine myocardial infarction (MI) models recapitulate the pathophysiological features of human HF, exhibiting remarkable similarities in cardiac structure, function, and hemodynamics [128]. Notably, studies have shown that knockout of the p47phox gene in mice attenuates post-MI cardiac remodeling and dysfunction, suggesting a crucial role for NAD(P)H oxidase in post-MI heart disease. Furthermore, other studies have provided direct evidence that angiotensin II exacerbates cardiac injury by activating NAD(P)H oxidase in vascular endothelial cells, leading to mitochondrial dysfunction [128].
ROS activate multiple signal transduction pathways, including MAPK/ERK, JNK, and p38, leading to the phosphorylation of downstream transcription factors that induce the synthesis of structural proteins in cardiomyocytes and contributing to cardiac hypertrophy [129]. Concurrently, ROS can trigger the caspase cascade, resulting in cardiomyocyte apoptosis through mitochondrial damage and endoplasmic reticulum stress. Additionally, ROS activate matrix metalloproteinases (MMPs) such as MMP-2 and MMP-9, which degrade the extracellular matrix, disrupt the myocardial microenvironment, and drive ventricular remodeling [130]. Moreover, ROS activate transcription factors like nuclear factor-κB (NF-κB), inducing the expression of inflammatory factors such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), triggering inflammatory responses, and exacerbating myocardial injury [131]. Disruption of calcium homeostasis and endothelial cell injury are also detrimental effects of ROS on the heart. These mechanisms create a vicious cycle, leading to the progressive deterioration of cardiac structure and function, ultimately promoting pathological cardiac remodeling and contributing to terminal conditions like heart failure.

2.3.5. Protective Roles of ROS in CVDs

Although excessive ROS generation can induce oxidative stress and cellular damage during ischemia-reperfusion injury (IRI), the protective role of moderate levels of ROS in IRI has been extensively studied, primarily in terms of inducing cytoprotective mechanisms and modulating signaling pathways [132].
ROS can induce myocardial preconditioning and postconditioning. Low levels of ROS generated during ischemia activate protective signaling pathways (such as PI3K/Akt, AMPK) and redox-sensitive transcription factors (such as Nrf2), enhancing antioxidant capacity and inhibiting apoptotic gene expression [133]. This “adaptive” effect reduces myocardial cell injury during subsequent ischemia-reperfusion. Postconditioning exerts its cardioprotective effects by activating multiple signaling cascades, such as the mK (ATP) and PKC pathways, and by regulating ROS levels [134]. These findings suggest that these signaling pathways are essential components of postconditioning-induced cardiovascular protection [135]. Early ROS release upon reperfusion induces endogenous protective mechanisms, such as activation of mitochondrial ATP-sensitive potassium (KATP) channels, heat shock protein (HSP) expression, and upregulation of anti-apoptotic factors, thereby attenuating IRI-induced apoptosis [136,137].
ROS generation can modulate redox-sensitive signaling pathways. Moderate levels of ROS can oxidize Keap1 protein, leading to the release and nuclear translocation of Nrf2, promoting the expression of antioxidant enzymes (such as SOD, CAT, and HO-1), enhancing cellular antioxidant capacity, and alleviating inflammation and oxidative damage in IRI [138]. ROS-mediated PI3K/Akt activation can regulate downstream eNOS generation of NO, improving microcirculation, inhibiting cardiomyocyte apoptosis, and exerting cardioprotective effects [139]. Moderate ROS can enhance mitochondrial autophagy through the AMPK pathway, maintaining mitochondrial function stability and reducing mitochondrial damage caused by IRI.
ROS generation can regulate mitochondrial function. Moderate increases in ROS levels can promote mitochondrial fusion (by upregulating fusion proteins such as OPA1), improve energy supply, and reduce oxidative stress by removing damaged mitochondria [140]. The ROS-induced opening of KATP channels can alleviate mitochondrial calcium overload, reduce membrane potential collapse, and protect mitochondrial function [141,142].
The effects of ROS exhibit a clear dose-dependent relationship. Low concentrations of ROS can serve as cellular signaling molecules, participating in various physiological processes and exerting protective effects [143]. For example, low concentrations of ROS can activate the Nrf2 pathway, inducing the expression of antioxidant enzymes and protecting cells from oxidative damage. High concentrations of ROS, however, can induce oxidative stress, leading to lipid peroxidation, protein denaturation, DNA damage, and ultimately cellular damage, inflammation, and cardiovascular diseases [144].
The protective effects of ROS in ischemia-reperfusion injury are achieved through inducing myocardial preconditioning, activating protective signaling pathways (such as Nrf2 and PI3K/Akt), regulating mitochondrial function, and maintaining redox balance. Future research should focus on precisely regulating ROS levels to maximize its protective effects, providing new strategies for the treatment of IRI.

3. The Protective Effects of Antioxidants and the Underlying Redox Signaling Mechanisms in the Heart

3.1. Redox Signaling Pathways

Redox signaling pathways are crucial for cardiovascular protection, as they regulate ROS and reactive nitrogen species (RNS) levels, modulating cellular processes to mitigate oxidative stress-induced cardiomyocyte injury. This section explores these essential pathways.

3.1.1. Nrf2/Keap1/ARE Signaling Pathway

The Nrf2/Keap1/ARE signaling pathway is a finely tuned regulatory system that orchestrates cellular responses to oxidative stress and chemical attacks [145]. Nrf2, a central transcription factor, binds to antioxidant response elements (AREs) to regulate the expression of genes involved in antioxidant defense and detoxification [146]. Keap1, a negative regulator of Nrf2, sequesters Nrf2 in the cytoplasm, inhibiting its transcriptional activity. ROS generated by Nox5 activation can upregulate the expression of antioxidant genes via the Nrf2/Keap1 pathway while simultaneously activating inflammatory responses through the NF-κB pathway, further exacerbating oxidative stress [147]. Under oxidative stress or ionizing radiation, Keap1 undergoes modifications that disrupt its interaction with Nrf2, allowing Nrf2 to translocate to the nucleus and activate target gene expression [148].
In ischemia-reperfusion injury (IRI) conditions, Nrf2 activation induces the expression of antioxidant enzymes, such as superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR), which scavenge excess ROS and mitigate oxidative damage. Additionally, Nrf2 interacts with the NF-κB signaling pathway to suppress inflammatory responses and alleviate tissue injury. Moreover, Nrf2 activates heme oxygenase-1 (HO-1) expression, promoting the degradation of heme into bilirubin, an antioxidant that provides further cellular protection [113].
In doxorubicin (DOX)-induced oxidative stress, DOX treatment upregulates Keap1 expression and inhibits Nrf2 activity through multiple mechanisms, including recruitment of TRIM21, which interferes with the separation of Nrf2 from Keap1, thereby weakening the cell’s antioxidant defense [149]. Sirtuin 1 (Sirt1) activates Nrf2 through deacetylation, enhancing its transcriptional activity. Conversely, miR-140-5p exacerbates oxidative stress by targeting Sirt2 and inhibiting Nrf2 activation. Other proteins, such as p62/Sqstm1, PKC, PI3K/Akt, and p21, regulate Nrf2 activity through various mechanisms [150,151]. Several natural compounds, including dioscin, α-linolenic acid, and sulforaphane, have been shown to enhance myocardial antioxidant capacity and alleviate DOX-induced myocardial injury by activating the Nrf2/Keap1/ARE pathway [152,153,154]. Nrf2 holds promise as a therapeutic target for DOX-induced myocardial injury, and the development of drugs that activate the Nrf2 pathway is a promising therapeutic strategy.
Type 2 diabetes mellitus (T2DM) is a major risk factor for CVD patients globally. Studies have demonstrated that individuals with diabetes are at a significantly higher risk of developing CVDs compared to those without diabetes, with this risk being positively correlated with blood glucose levels. Diabetic patients frequently exhibit cardiac damage, and even when blood glucose levels are well-controlled, the risk of cardiovascular events remains elevated relative to the general population [155]. Hinokinin, a natural compound with various pharmacological properties, has been shown to reduce cardiac oxidative stress, inflammation, and apoptosis induced by T2DM. This effect occurs through modulation of the Nrf2/Keap1/ARE pathway and inhibition of the TLR4/MyD88/NF-κB and MAPK signaling pathways, ultimately protecting the myocardium [156]. Moreover, Nrf2 activators have been found to mitigate diabetes-induced endothelial dysfunction (ED) [157]. Our in vitro study using high-glucose-induced HK-2 cells demonstrated that chitosan (COS) exerts antioxidant effects by activating the Nrf2/Keap1/ARE pathway, providing experimental evidence for its potential therapeutic value in diabetic complications [158].

3.1.2. PI3K/Akt Pathway

The PI3K/Akt pathway stands as a cornerstone of cardiomyocyte survival, orchestrating cardiovascular protection through the precise modulation of cellular growth, proliferation, and apoptosis [159]. Through the activation of the mTOR pathway, PI3K/Akt enhances protein synthesis and cellular metabolism, promoting angiogenesis and ensuring adequate myocardial oxygenation and nutrient supply. Additionally, this pathway exerts anti-inflammatory effects, reducing myocardial injury [160].
Activation of the PI3K/Akt pathway has been shown to significantly mitigate myocardial injury induced by myocardial infarction (Table 1). Both human and animal studies have demonstrated a significant upregulation of PI3K and Akt activity post-myocardial infarction. Inhibition of the PI3K/Akt pathway exacerbates cardiovascular risk and increases mortality in mice, whereas activation of this pathway by inhibiting PTEN improves cardiac function. These findings collectively underscore the therapeutic potential of targeting the PI3K/Akt pathway in myocardial infarction [161,162].
While the activation of the PI3K/Akt pathway provides protection in the early stages of acute heart failure, prolonged excessive activation can lead to myocardial remodeling and functional deterioration [163]. This pathway is involved in myocardial remodeling processes such as cardiomyocyte hypertrophy, fibrosis, and apoptosis. Chronic inflammation, a hallmark of heart failure, can be mitigated by activated protein C (APC), which inhibits the PI3K/Akt pathway [164]. Regarding angiogenesis, APC plays a dual role: while the PI3K/Akt pathway primarily promotes angiogenesis, the interaction between the two may influence angiogenesis outcomes in heart failure. Furthermore, APC can induce apoptosis in certain cells, while the PI3K/Akt pathway inhibits apoptosis. The balance between these two pathways determines cell fate in heart failure [165].

3.1.3. AMPK Pathway

AMP-activated protein kinase (AMPK), a key cellular energy sensor, plays a crucial role in maintaining cellular energy homeostasis (Table 1). In the cardiovascular system, AMPK activation exerts cardioprotective effects by promoting mitochondrial bioenergetics, enhancing fatty acid oxidation, activating antioxidant defense systems, and regulating glucose metabolism. In CVDs such as myocardial IRI and HF, AMPK activation attenuates cardiomyocyte apoptosis, inhibits inflammation, and improves cardiac function. AMPK is activated under oxidative stress conditions and exerts its cardioprotective effects by regulating downstream effectors such as peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and acetyl-CoA carboxylase (ACC).
In atherosclerosis, risk factors such as hyperglycemia and hyperlipidemia induce excessive mitochondrial ROS production, leading to endothelial cell damage [166]. AMPK exerts potent vascular protective effects by inhibiting mitochondrial ROS generation, improving mitochondrial function, attenuating inflammation, and promoting endothelial cell survival [167]. Therefore, activation of AMPK holds promise as a novel therapeutic strategy for atherosclerosis.
AMPK also plays a protective role in coronary heart disease, and its activation is considered a promising therapeutic approach [168]. Researchers are currently developing various AMPK activators, such as metformin and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), for the treatment of coronary heart disease [169]. When cellular energy levels decrease, the AMP/ATP ratio increases, activating AMPK. AMPK activation occurs primarily through several mechanisms: LKB1 phosphorylation, where LKB1, an upstream kinase of AMPK, activates AMPK by phosphorylating Thr172 on the α subunit; calcium/calmodulin-dependent protein kinase kinase β (CaMKKβ) phosphorylation, where CaMKKβ is activated upon calcium ion elevation and phosphorylates AMPK; and phosphorylation by other kinases, including TAK1 and TAK11, which can also phosphorylate AMPK [170]. AMPK is a potential therapeutic target for coronary heart disease that warrants further investigation.

3.1.4. SIRT1 Pathway

SIRT1, an NAD+-dependent deacetylase initially identified in yeast, primarily localizes to the nucleus where it regulates gene expression and influences a variety of biological processes, including cell growth, apoptosis, and metabolism [171]. In the vascular system, SIRT1 expression and activity are closely associated with vascular homeostasis [172]. It maintains vascular homeostasis by regulating multiple downstream target proteins, such as eNOS, p53, and FOXO [173,174]. SIRT1 protects vascular endothelial cells, inhibits the proliferation of vascular smooth muscle cells, and delays vascular senescence (Table 1).
In atherosclerosis, SIRT1 exhibits therapeutic potential. To investigate its impact on atherosclerosis progression, SIRT1 was overexpressed specifically in endothelial cells of ApoE−/− mice [175]. The results demonstrated that SIRT1 overexpression ameliorates endothelial dysfunction by activating the eNOS signaling pathway and suppressing endothelial inflammation, thereby attenuating atherosclerosis development. Oral administration of low-dose red wine significantly upregulates the expression of eNOS and SIRT1 in the aortas of hypercholesterolemic mice, suggesting that resveratrol may prevent atherosclerosis through the activation of the SIRT1 pathway [176].
Studies have shown that SIRT1 expression decreases with age in human vascular smooth muscle cells and is lower in atherosclerotic lesions, suggesting that SIRT1 plays an important role in maintaining vascular smooth muscle cell function [177]. Additionally, miR-34a is highly expressed in the aortas of aged mice, where it promotes senescence and inflammation by downregulating SIRT1. This suggests that SIRT1 may play a crucial role in regulating the inflammatory responses of monocytes/macrophages [178]. Donato et al. found that SIRT1 expression is decreased in the endothelial cells of the elderly and is associated with reduced NO production by eNOS and endothelial dysfunction [179]. This indicates that SIRT1 plays an important role in maintaining endothelial function. Meanwhile, Bai et al. demonstrated that SIRT1 inhibits endothelial cell senescence by deacetylating LKB1 and promoting its degradation [180]. They also found that loss of SIRT1 expression or function leads to increased nuclear accumulation of acetylated LKB1, resulting in alterations in vascular wall structure, arterial remodeling, and vascular stiffness [181]. These findings suggest that SIRT1 plays a crucial role in maintaining vascular health. Decreased SIRT1 expression is closely associated with vascular aging and atherosclerosis. SIRT1 exerts its protective effects through multiple mechanisms, including regulating eNOS activity, inhibiting endothelial cell senescence, and exerting anti-inflammatory effects [172].

3.1.5. MAPK Pathway

The MAPK pathway is a crucial intracellular signaling cascade that transmits extracellular stimuli to the nucleus, initiating a variety of cellular responses (Table 1). Typically composed of a three-kinase module—MAPKKK, MAPKK, and MAPK—the MAPK pathway regulates numerous cellular processes, including proliferation, differentiation, and apoptosis. Distinct MAPK subfamilies (ERK, JNK, and p38) target specific downstream effectors [182].
In the commonly used animal model for heart failure, transverse aortic constriction (TAC), activation of MAPK signaling pathways in cardiac tissue has been extensively studied [183]. Results consistently show that the JNK pathway is activated first, followed by the ERK and p38 MAPK pathways, suggesting a temporal hierarchy in the activation of these signaling cascades [184].
The ERK pathway plays a pivotal role in promoting cardiac hypertrophy, as evidenced by the effective suppression of hypertrophy with MEK or Raf inhibitors [185]. Grb2, an upstream regulator of the ERK pathway, is essential for ERK activation. Mice with haploinsufficiency of Grb2 exhibit resistance to pressure overload-induced cardiac hypertrophy [186]. Cardiac-specific overexpression of a dominant-negative Raf-1 or knockout of the Raf gene can also confer resistance to cardiac hypertrophy [187]. While ERK1/2 is crucial for cardiac hypertrophy, its complete deletion or reduced activity does not fully suppress hypertrophy, suggesting the involvement of other signaling pathways [188]. In contrast, the JNK and p38 pathways inhibit cardiac hypertrophy. Loss or reduced activity of JNK1/2 or p38α leads to spontaneous cardiac hypertrophy, indicating their inhibitory roles [189]. Both JNK and p38 inhibit cardiac hypertrophy by phosphorylating and inactivating the NFAT transcription factor. These findings highlight the complex interplay between different MAPK pathways in regulating cardiac hypertrophy.
MAPK signaling pathways also play a pivotal role in cardiac remodeling following myocardial infarction [190]. Among them, p38 MAPK, the most extensively studied member, is rapidly activated after myocardial infarction and promotes pathological cardiac remodeling through multiple mechanisms. For instance, p38 MAPK induces cardiomyocyte apoptosis by downregulating anti-apoptotic proteins such as Bcl-xL and Bcl-2, thereby expanding the infarct size. Additionally, it promotes collagen synthesis, leading to cardiac fibrosis, and inhibits cardiomyocyte proliferation, hindering myocardial repair and regeneration. JNK acts synergistically with p38 MAPK to promote cardiomyocyte apoptosis and fibrosis [191]. The ASK1-JNK signaling pathway plays a critical role in this process [192]. In contrast, the role of the ERK pathway in post-myocardial infarction remodeling is more complex, potentially involving both cell survival and proliferation, although the underlying mechanisms require further investigation.

3.1.6. CVDs Associated with Oxidative Stress and Their Underlying Signaling Pathways

Oxidative stress plays a pivotal role in promoting the progression of various CVDs by activating a series of signaling pathways. This table comprehensively summarizes the critical roles of oxidative stress in different CVDs, detailing the major signaling pathways involved and the specific impacts of oxidative stress on pathological processes. For example, in atherosclerosis, oxidative stress activates the NF-κB signaling pathway, leading to chronic inflammation and plaque formation. In dilated cardiomyopathy, ROS accumulation induces apoptosis and myocardial remodeling, ultimately leading to heart failure.
Table 1. Key signaling pathways and their pathological effects in oxidative stress-related CVDs.
Table 1. Key signaling pathways and their pathological effects in oxidative stress-related CVDs.
CVDsCore Signaling Pathways and Their MechanismsRole of ROS in CVDsReference
ASNF-κB
Nrf2
MAPK
PI3K/Akt
SIRT1
NF-κB drives inflammation,
Nrf2 promotes antioxidant defense,
SIRT1 regulates metabolism, and
MAPK/Akt promotes proliferation.
Increases lipid oxidation promotes plaque formation and progression.[193,194,195]
HTNRAAS
ROS
ET-1
NOX
AMPK
RAAS increases vascular tension,
NOX generates ROS,
AMPK regulates energy metabolism and improves vascular function, and
ET-1 promotes vasoconstriction.
Leads to vascular stiffening, promoting increased vascular resistance and endothelial dysfunction.[196,197]
CADNF-κB
p38 MAPK
PI3K/Akt
SIRT3
p38 MAPK regulates inflammation,
SIRT3 promotes antioxidant defense, and
NF-κB induces inflammation.
Induces endothelial cell apoptosis, promoting coronary plaque instability.[198,199]
AMIJAK/STAT
NF-κB
p38 MAPK
HIF-1α
HIF-1α activates protective genes, JAK/STAT regulates inflammation, and
p38 MAPK promotes cardiac repair.
Results in cardiomyocyte apoptosis and necrosis, leading to aggravated myocardial injury.[200,201]
DCMTGF-β
PI3K/Akt
JNK
ERK
TGF-β promotes fibrosis,
JNK/ERK induces injury, and
PI3K/Akt prevents apoptosis.
Exacerbates cardiac fibrosis, leading to ventricular remodeling and functional failure.[202,203]
HCMIGF-1
mTOR
ERK
GATA4
GATA4 induces hypertrophic genes and
IGF-1/mTOR promotes growth.
Exacerbates cardiac hypertrophy and oxidative stress, leading to cardiac dysfunction.[204]
PHET-1
TGF-β
NFAT
PI3K/Akt
mTOR
NFAT and TGF-β induce vascular smooth muscle cell proliferation and remodeling and
mTOR regulates cell proliferation.
Induces vascular smooth muscle cell proliferation and increases pulmonary arterial pressure.[205]
MIRI ROS
JAK/STAT
ERK1/2
NOX
NOX induces oxidative damage and
ERK1/2 regulates cell growth and repair.
Induces cardiomyocyte apoptosis and necrosis, leading to reperfusion injury.[206,207]
CVINOX
NF-κB
VEGF
MMP
MMPs degrade the extracellular matrix, facilitating vascular remodeling, and
VEGF promotes angiogenesis.
Increases inflammation and vascular permeability, leading to chronic vascular wall injury.[208,209]
VTEP-selectin
TGF-β
NF-κB
COX-2
COX-2 promotes inflammation and
P-selectin mediates leukocyte and platelet adhesion.
Promotes thrombus formation and exacerbates vascular occlusion.[210,211]
RHDJAK/STAT
NF-κB
IL-17
IL-17 drives chronic inflammation and induces immune responses.Induces chronic inflammation and fibrosis, impairing cardiac valve function.[212]
CAVNF-κB
mTOR
JAK/STAT
PD-1/PD-L1
PD-1/PD-L1 regulates immune suppression and mTOR regulates cell proliferation.Promotes chronic rejection and vascular remodeling, leading to allograft heart injury.[213,214]
AfibCaMKII
NF-κB
TGF-β
TGF-β induces fibrosis and atrial remodeling and CaMKII modulates myocardial electrical remodeling.Leads to atrial remodeling, increasing the incidence of atrial fibrillation.[215]
HFβ-AR
PI3K/Akt
NF-κB
Notch
Notch signaling modulates cell proliferation and apoptosis and
β-AR potentiates cardiac contractility.
Increases apoptosis and fibrosis, leading to impaired cardiac function.[216,217]
Heart
inflammation
TLR
NF-κB
MAPK
IL-1β
IL-1β mediates the inflammatory response
and TLRs mediate the recognition of pathogens and trigger immune responses.
Induces oxidative stress and immune responses, thereby exacerbating cardiac injury.[218,219]
Rheumatic carditisNF-κB
TGF-β
JAK/STAT
IL-6
IL-6 drives chronic inflammation and
JAK/STAT signaling modulates immune cell activation.
Induces chronic myocardial inflammation, thereby exacerbating cardiac injury.[220,221]
MVPTGF-β
MMP
NF-κB
SMAD
Smad signaling contributes to valvular fibrosis by promoting extracellular matrix protein synthesis.Increases valvular fibrosis, thereby impairing valve function.[222,223]
HOCMCaMKII
ROS
PI3K/Akt
ERK
ERK mediates cardiac hypertrophy and
PI3K/Akt regulates cell survival and proliferation.
Exacerbates myocardial hypertrophy and dysfunction.[224,225]
CHDWnt/β-catenin
Notch
NF-κB
SHH
Wnt and SHH signaling regulate cardiac development and
Notch regulates cell differentiation.
Causes developmental abnormalities, thereby exacerbating congenital heart disease.[226,227]
Coronary artery spasm anginaPKC
eNOS
ET-1
CaMKII
CaMKII affects myocardial contractility and
eNOS regulates nitric oxide production.
Causes coronary artery constriction, leading to ischemic events.[228]
PSVTCaMKII
PKA
RyR
SERCA
SERCA regulates calcium ion reuptake and
RyR regulates calcium ion release.
Causes arrhythmia, thereby increasing myocardial workload.[229]
CHFβ-AR
ROS
NF-κB
SIRT3
SIRT3 protects mitochondrial function and reduces oxidative stress.Promotes myocardial fibrosis and apoptosis, leading to aggravated heart failure.[230]
Left ventricular insufficiencyTGF-β
NF-κB
PI3K/Akt
IL-1β
IL-1β induces cardiac fibrosis and
TGF-β triggers structural remodeling.
Causes myocardial fibrosis.[231]
AS: Atherosclerosis; HTN: Hypertensive; CAD: Coronary heart disease; AMI: Acute myocardial infarction; DCM: Dilated cardiomyopathy; HCM: Hypertrophic cardiomyopathy; PH: Pulmonary arterial hypertension; MIRI: Myocardial ischemia-reperfusion injury; CVI: Chronic venous insufficiency; VTE: Venous thromboembolism; RHD: Rheumatic heart disease; CAV: Cardiac allograft vasculopathy; Afib: Atrial fibrillation; HF: Heart failure; MVP: Mitral valve prolapse; HOCM: Hypertrophic obstructive cardiomyopathy; CHD: Congenital heart disease; PSVT: Paroxysmal supraventricular tachycardia; and CHF: Congestive heart failure.

3.2. Antioxidants

Antioxidants have been extensively studied for their potential to mitigate oxidative damage in CVDs (Table 2). These compounds exert their effects through various mechanisms, including free radical scavenging, inhibition of lipid peroxidation, suppression of apoptosis, and modulation of redox signaling. Additionally, diverse animal models have been employed to investigate the efficacy and safety of different antioxidants in simulating human cardiovascular pathologies (Table 2).
The following table summarizes the major antioxidants currently under investigation, their applications in various CVDs, and their underlying mechanisms of action. Additionally, commonly used animal models are listed, providing a comprehensive overview of the potentials of different antioxidants in cardiovascular protection.
Table 2. Applications and Mechanisms of Antioxidants in CVDs: A Focus on Animal Models.
Table 2. Applications and Mechanisms of Antioxidants in CVDs: A Focus on Animal Models.
AntioxidantMain Mechanism of ActionCVDs AppliedAnimal ModelReference
Vitamin CAntioxidant scavenging of free radicals, reduction of lipid peroxidation, protection of endothelial function, and inhibition of inflammatory response.AS
CHD
HTN
LDLr−/− mice[232]
Vitamin EInhibits lipid peroxidation, reduces oxidative damage, and inhibits inflammation and cell proliferation.AS
MIRI
CETP transgenic rats[233]
GlutathioneIncreases antioxidant reserves, regulates redox balance, and attenuates mitochondrial damage.HTN
CHD
Myocardial fibrosis
Aldosterone-induced hypertension in C57BL/6 mice[234]
lipoic acidInhibits oxidative stress and inflammatory responses and improves mitochondrial function.DbCM
HTN
HF
RAS-activated mice[235]
CoQ10Enhances mitochondrial respiratory chain function, reduces ROS production, and improves myocardial tolerance.HF
MIRI
AS
ApoE−/− mice[236]
ResveratrolActivates SIRT1, reduces oxidative stress, and inhibits inflammation.MIRI
AS
ApoE−/− mice[237]
N-AcetylcysteineProvides GSH precursors, reduces ROS, and enhances antioxidant capacity.CHD
MIRI
MIRI in C57BL/6 Mice[238]
AstaxanthinA potent antioxidant that reduces lipid peroxidation and inhibits inflammatory responses.MIRI
HF
AS
SOD2-deficient mice[239]
QuercetinInhibits oxidative stress and lipid peroxidation and enhances anti-inflammatory capacity.HTN
AS
CHD
AT1 transgenic mice[240]
Tea polyphenolsInhibits endothelial inflammation and lipid peroxidation and protects the myocardium and blood vessels.Diabetic
AS
CMP
Leprdb/db mice[241]
Sodium thiosulfateScavenges ROS, reduces mitochondrial oxidative stress, and inhibits calcium overload and apoptosis.MIRI
HF
SIRT3 gene-deficient mice[242]
Statin drugsReduces cholesterol, inhibits ROS production, and attenuates vascular endothelial damage.AS
CHD
HTN
LDLr−/− and ApoE−/− mice[243]
Vitamin DRegulates calcium metabolism, reduces vascular smooth muscle cell proliferation, and protects the endothelium.HTN
AS
Myocardial Fibrosis
HTN-induced ApoE−/− mice[244]
Gallic acidInhibits oxidative stress and smooth muscle cell proliferation and regulates endothelial cell activity.AS
DbCM
Cholesterol-hypercholesterolemic rat model[245]
Hydrogen sulfideActs as an endogenous antioxidant molecule, regulates mitochondrial function, and inhibits apoptosis.MIRI
HF
DbCM
Diabetes-induced ZDF in rats[246]
ThioredoxinInhibits ROS generation, protects endothelial cells and mitochondria, and regulates calcium ion balance.AS
HTN
HF
Angiotensin II-induced hypertension in C57BL/6 mice[247]
AS: Aortic Stenosis; CHD: Coronary Heart Disease; HTN: Hypertension; MIRI: Myocardial Ischemia-Reperfusion Injury; HF: Heart Failure; DbCM: Diabetic Cardiomyopathy; and CMP: Cardiomyopathy.

4. Emerging Paradigms in Cardiovascular Protection

Advancements in molecular biology, gene editing, and stem cell technology have led to the development of novel cardiovascular protection strategies, bridging the gap between basic research and clinical application.

4.1. Drug Therapy

MitoTEMPO (MT) and Visomitin (SKQ1) have emerged as pioneering mitochondrial-targeted antioxidants, spearheading a new era in antioxidant therapy [248]. Functioning akin to precision surgical instruments, they directly target the cellular powerhouse, the mitochondria, to selectively eliminate deleterious ROS [248]. Derived from TEMPOL, MT, plastoquinone, and SKQ1, these compounds, despite their structural dissimilarities, share a common objective: safeguarding mitochondrial health [249]. The distinct structures of SKQ1 and MT confer upon them unique antioxidant mechanisms (Table 3). By leveraging the electron transfer capacity of plastoquinone, SKQ1 acts as an electronic “sponge”, rapidly absorbing and neutralizing harmful free radicals [250]. Conversely, MT mimics superoxide dismutase, efficiently calyzing the conversion of superoxide anions, functioning as a molecular “scavenger” [251]. The triphenylphosphonium moiety serves as their “navigation system”, precisely guiding them to the mitochondria. Research has demonstrated that both MT and SKQ1 can effectively scavenge free radicals, mitigate oxidative stress-induced cellular damage, and even improve cardiomyocyte function, offering promising therapeutic avenues for cardiovascular diseases (Table 3) [252]. In vitro experiments using H2O2- and MEN-induced oxidative damage models have revealed that both MT and SKQ1 exhibit significant cytoprotective effects, underscoring their potential as therapeutic agents in antioxidant therapy [253,254,255].
Nrf2, a pivotal transcription factor, plays an indispensable role in maintaining redox homeostasis (Table 3). Upon exposure to oxidative stress, Nrf2 is activated and rapidly translocates into the nucleus, where it binds to antioxidant response elements (AREs) to induce the expression of a battery composed of antioxidants and phase II detoxification enzymes [256]. Consequently, the cellular antioxidant capacity is enhanced, mitigating oxidative damage and protecting cells from further injury induced by oxidative stress [256]. Nrf2 activators can alleviate oxidative stress-induced damage to the cardiovascular system and prevent cardiovascular diseases [257]. By upregulating the expression of antioxidant and detoxification enzymes, such as SOD, GPX, and GST, Nrf2 activators enhance the cellular capacity to eliminate ROS and detoxify xenobiotics [258,259]. Moreover, Nrf2 activators suppress inflammatory signaling pathways, including NF-κB, thereby mitigating inflammatory responses and protecting cells from oxidative damage [260].
Small molecule scavengers hold great promise as novel therapeutic agents for cardiovascular diseases (Table 3). By directly scavenging harmful ROS, reducing inflammation, and protecting vascular endothelial cells, these compounds offer new therapeutic options for patients with cardiovascular diseases (Table 3) [261,262,263]. They have shown great potential in slowing the progression of atherosclerosis, improving cardiac function, and reducing the risk of cardiovascular events [264].
Enzyme-based therapy is a therapeutic approach that utilizes the catalytic properties of enzymes to treat diseases. Given their high specificity and efficiency as biological catalysts, enzymes hold great promise for the treatment of cardiovascular diseases (Table 3). In cardiovascular diseases, enzymes can clear oxidized low-density lipoprotein (ox-LDL), inhibit inflammatory responses, and stabilize atherosclerotic plaques [265]. They can also scavenge ROS generated by cardiomyocytes, alleviate oxidative damage, and inhibit cardiomyocyte apoptosis (Table 3). Moreover, enzymes can improve myocardial energy metabolism and enhance myocardial contractility [266]. The therapeutic efficacy of enzymes largely depends on their delivery in vivo. Current challenges in enzyme delivery include the instability of enzymes in biological fluids and the difficulty of targeting enzymes toward disease sites [267]. To address these challenges, researchers are developing various enzyme delivery systems, such as liposomes and nanoparticles [268]. Enzyme-based therapy has great potential for the treatment of cardiovascular diseases, but challenges remain. With the continuous development of nanotechnology and genetic engineering, enzyme-based therapy is expected to become a significant modality for the treatment of cardiovascular diseases in the future.
ROS metabolism inhibitors hold great promise in the treatment of cardiovascular diseases (Table 3). By inhibiting the activity of ROS-generating enzymes, enhancing antioxidant enzyme activity, and modulating redox signaling pathways, these agents effectively scavenge excessive ROS and alleviate oxidative stress [269,270]. Consequently, ROS metabolism inhibitors can delay the progression of atherosclerosis, mitigate myocardial infarction injury, and improve symptoms of heart failure, providing a novel therapeutic strategy for cardiovascular disease patients [271].
Multifunctional antioxidants are versatile compounds that offer multiple modes of action to counteract oxidative stress (Table 3). In cardiovascular diseases, these agents can directly scavenge mitochondrial-derived ROS, chelate redox-active metals, and induce antioxidant enzymes, thereby providing significant cardiovascular protection [272].
Table 3. Pharmacological Approaches Targeting Oxidative Stress and Their Characteristics.
Table 3. Pharmacological Approaches Targeting Oxidative Stress and Their Characteristics.
CategoryDrugMechanismAdvantages and InnovationReference
Mitochondria-Targeted AntioxidantsMitoTEMPOTargets mitochondria to reduce ROS generation and improve mitochondrial function.Targeting mitochondria with high specificity, directly acting on major ROS production sites, and reducing side effects.[273]
SkQ1Penetrates the mitochondrial membrane to reduce oxidative damage and delay cell apoptosis.Unique mitochondrial penetration mechanism for effective myocardial cell protection.[274]
Nrf2 ActivatorsBardoxolone methylActivates the Nrf2/ARE pathway to enhance antioxidant enzyme (e.g., HO-1) expression.Enhances endogenous antioxidant capacity via transcriptional regulation, providing stable long-term effects.[275]
Small Molecule ScavengersEdaravone derivativesScavenges free radicals and reduces acute ROS levels.Optimized molecular structure for higher efficiency and rapid action.
TempolMimics superoxide dismutase (SOD) to inhibit superoxide production.Simple synthetic pathway, providing an economical antioxidant treatment option.
Enzyme-Based TherapiesPEG-SOD/PEG-CatalaseModifies enzymes to extend circulation time and improve ROS scavenging capacity.Enhanced stability and bioavailability, reducing the need for frequent administration.
ROS Metabolism InhibitorsGKT137831Inhibits NOX to reduce ROS production.High-specificity NOX inhibition with minimal side effects.
p66Shc InhibitorTargets upstream regulators of mitochondrial ROS production, delaying cardiovascular aging.Innovative target directly addressing core mitochondrial oxidative stress mechanisms.
Multifunctional Antioxidant MoleculesRTA 408Simultaneously scavenges ROS and inhibits inflammation.Integrated mechanism combining anti-inflammatory and antioxidant functions, broadening indications.

4.2. Gene Therapy

Gene therapy, which has shown significant success in oncology and genetic disorders, is now being investigated for cardiovascular applications (Table 4). By modifying gene expression, this approach provides new therapeutic possibilities for conditions such as peripheral arterial disease and myocardial ischemia (Table 4) [276].
Table 4. Gene therapy for CVDs.
Table 4. Gene therapy for CVDs.
TypologyAdvantagesDisadvantagesRelated ResearchReference
Gene editingHigh-precision targeted gene editing enabling permanent repair and reducing recurrence risk in monogenic diseases.The potential risks of this technology include off-target mutations, immune responses, and challenges in precise delivery, which are coupled with high technical complexity and cost.CRISPR-mediated gene editing ameliorates cardiac hypertrophy and improves cardiac function in a murine model of MYBPC3-related cardiomyopathy.[277]
Gene replacement therapyThe precise targeting and repair of defective genes, leading to the restoration of normal cellular physiology, highlighting its broad clinical potential.The limitations of this technology, including low insertion efficiency, off-target effects, and immunogenicity, necessitate further investigations into its long-term safety profile.DMD gene replacement significantly improves cardiac function in a mouse model of Duchenne muscular dystrophy.[278]
RNA interferenceHighly specific targeting of specific gene expression to inhibit pathological effects of disease-causing genes; applicable to reversible regulation without affecting gene ontology; and short duration of action to modulate efficacy and safety.Shorter expression time, requiring repeated administration; lower stability in vivo, difficult to resist enzymatic breakdown; complex drug delivery and dose regulation, frequent dosing cycles; and may trigger cytotoxic and immune responses.RNAi inhibition of PCSK9 significantly reduces LDL-C levels and inhibits the course of atherosclerosis in ApoE−/− mice.[279]
Gene enhancement therapyCan upregulate protective gene expression and enhance antioxidant, anti-inflammatory and metabolic regulation functions; suitable for chronic diseases lacking protective mechanisms; and can delay lesions by modulating multiple protective pathways.Higher dosage requirements, overexpression may trigger toxic effects; dependence on the level of gene expression makes it difficult to control the side effects of gene overexpression; the immune system may reject exogenous genes; and long-term monitoring and observation of safety is required.SIRT1 overexpression protects against myocardial ischemia-reperfusion injury.[280]
AAV-mediated gene deliveryPersistent gene expression, which facilitates long-term treatment; low immunogenicity, which reduces the risk of immune rejection; cardiac-specific delivery potential, which enhances therapeutic efficacy; and suitability for long-acting treatments for chronic diseases and genetic defects.The limited capacity of AAV vectors restricts large-scale gene delivery; it is difficult to completely avoid immune recognition, so we need to be vigilant about potential immune reactions; the uneven integration of vectors may trigger gene insertion mutations; and the risk of long-term expression needs to be adequately verified.AAV delivery of SERCA2a restores calcium homeostasis, reduces myocardial fibrosis, and restores cardiac function in mouse and porcine models of heart failure.[281]
Genetic vaccineStimulates immune system regulation and reduces arterial inflammation and lipid accumulation; intervenes well in the early stages of disease; is easy to administer multiple times at relatively low cost; and can be personalised to improve vaccine specificity.The immune response is difficult to control accurately and may trigger an autoimmune response; there are individual differences in vaccine tolerance and durability; the vaccine development and evaluation cycle is long; and some of the vaccine components may cause toxicity or side effects.Gene vaccine targeting; CD68 and LDLR reduce arterial plaque formation and enhance anti-inflammation and lipid metabolism in a mouse model of atherosclerosis.[282]

4.3. Cellular and Regenerative Medicine

4.3.1. Cardiac Cell Therapy

Cardiac cell therapy is an emerging therapeutic strategy for heart diseases that involves transplanting healthy cells to repair damaged cardiac tissue (Table 4). These cells, which are derived from adult tissues such as bone marrow, skeletal muscle, the heart itself, or pluripotent stem cells, can function in two ways: (1) directly replacing damaged cardiac tissue to restore heart function and (2) indirectly promoting cardiac repair by secreting molecules and microvesicles that activate immune regulation and cardiac regenerative mechanisms [283]. Current regenerative approaches for CVDs primarily involve mesenchymal stem cells (MSCs), cancer stem cells (CSCs), induced pluripotent stem cells (iPSCs), and gene therapy [284]. MSCs, known for their safety and accessibility, are the most commonly employed [285]. However, their therapeutic effects are primarily mediated through paracrine mechanisms. While CSCs and iPSCs hold potential, they face challenges related to safety and technical feasibility.
Stem cell therapy holds immense promise for addressing oxidative stress in cardiovascular diseases through multifaceted mechanisms [286]. These include the secretion of antioxidative enzymes like superoxide dismutase (SOD) and glutathione peroxidase (GPx), effectively mitigating oxidative damage [287]. Furthermore, the release of vascular endothelial growth factor (VEGF) enhances angiogenesis, improving myocardial oxygenation and metabolism. Stem cells also modulate immune responses to alleviate inflammation while promoting myocardial regeneration via differentiation and paracrine signaling. Recent advancements in stem cell-derived exosome therapy offer targeted delivery of antioxidative molecules, reducing systemic side effects [288]. Integration with gene editing and nanotechnology further optimizes therapeutic efficacy, paving the way for transformative clinical applications.

4.3.2. Exocrine Therapy

Exosome therapy is an innovative approach for treating CVDs that uses extracellular vesicles, known as exosomes, as therapeutic carriers [289]. Exosomes have gained recognition as versatile agents in cardiovascular health (Table 4). Studies have demonstrated that exosomes derived from various sources, including healthy individuals and cardiac surgery patients, can promote angiogenesis and protect cardiomyocytes [290]. Furthermore, animal models have shown that exosomes are actively released by cardiomyocytes and endothelial cells in response to ischemic injury, indicating their role in tissue repair and remodeling [291]. These findings highlight the potential of exosomes as promising therapeutic agents for CVDs.
Exosomal molecular markers, such as proteins, microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circulating DNA, provide valuable support for the early diagnosis of CVDs. Protein markers, including cardiac troponins (cTnI and cTnT), creatine kinase-MB (CK-MB), and inflammatory markers like tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), can indicate early myocardial injury and inflammation [292]. MicroRNA markers such as miR-21, miR-126, miR-1, and miR-133a show early signs under conditions such as myocardial hypertrophy, endothelial cell repair, and acute myocardial infarction [293]. Long non-coding RNA (lncRNA) markers like LIPCAR, H19, ANRIL, and KCNQ1OT1 are linked to myocardial remodeling, fibrosis, and plaque formation, suggesting long-term risks [294]. Additionally, increases in circulating DNA, including mitochondrial DNA (mtDNA) and cell-free DNA (cfDNA), can reflect cellular damage in acute events [289]. The combined detection of multiple molecular markers holds significant clinical value for the early diagnosis of CVDs.
Exosome therapy represents a promising emerging strategy for mitigating oxidative stress in cardiovascular diseases [111]. It functions primarily by delivering antioxidative enzymes, regulatory miRNAs (e.g., miR-126 and miR-210), and proteins to reduce ROS accumulation [295]. Additionally, it modulates inflammatory pathways, breaking the vicious cycle between inflammation and oxidative stress while promoting angiogenesis and myocardial regeneration through the release of growth factors like VEGF and HGF [296]. By improving mitochondrial function and optimizing myocardial energy metabolism, exosome therapy demonstrates high specificity and minimal side effects. Integrated with gene editing and nanotechnology, it holds transformative potential for cardiovascular precision medicine.

4.4. Inflammation Regulation and Immunotherapy

IL-1β, a pro-inflammatory cytokine, plays a crucial role in the development of CVDs [297]. By activating signaling pathways such as NF-κB, IL-1β promotes inflammation and contributes to the formation and rupture of atherosclerotic plaques (Table 4). Canakinumab, a selective IL-1β receptor antagonist, has been shown to effectively inhibit inflammation and stabilize plaques, thereby reducing the risk of cardiovascular events [298]. Large-scale clinical trials have provided compelling evidence supporting the therapeutic efficacy of canakinumab in high-risk patients [299].
Anti-PCSK9 monoclonal antibodies have demonstrated significant potential in the treatment of CVDs [3]. By specifically binding to and inhibiting the PCSK9 protein, these antibodies upregulate the expression of LDL receptors (LDLR), leading to increased LDL clearance and reduced plasma LDL cholesterol (LDL-C) levels [300]. Large-scale randomized controlled trials have consistently shown that anti-PCSK9 antibody treatment significantly reduces LDL-C levels and the risk of major adverse cardiovascular events in high-risk patients [301,302]. These findings establish anti-PCSK9 antibodies as a valuable therapeutic option for managing CVDs.

4.5. Metabolic Regulator

Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) have emerged as a novel class of anti-diabetic drugs with effects extending beyond glycemic control [303]. By mimicking the actions of endogenous glucagon-like peptide-1, GLP-1 RAs exert multifaceted effects on glucose and lipid metabolism. On the one hand, GLP-1 RAs promote pancreatic β-cell proliferation and function, enhance insulin secretion, and slow gastric emptying, leading to increased satiety and effective glycemic control [304]. On the other hand, GLP-1 RAs improve endothelial function, inhibit inflammation, and stabilize atherosclerotic plaques, thereby providing significant cardiovascular protection (Table 4). Compared with traditional anti-diabetic drugs, GLP-1 RAs have been shown to more effectively reduce the incidence of cardiovascular events, including nonfatal myocardial infarction, nonfatal stroke, and cardiovascular death, in patients with type 2 diabetes [305]. These findings suggest that GLP-1 RAs are not only effective anti-diabetic agents but also promising cardiovascular protective agents with broad clinical applications.
Sodium-glucose cotransporter 2 (SGLT2) inhibitors represent another class of novel anti-diabetic agents that have demonstrated remarkable efficacy in improving outcomes in patients with CVD [306]. By inhibiting the sodium-glucose cotransporter 2 in the proximal renal tubule, SGLT2 inhibitors effectively lower blood glucose levels and body weight, increase urinary sodium excretion, and reduce cardiac workload [307]. Additionally, SGLT2 inhibitors exert multiple beneficial effects, including improved myocardial energy metabolism, reduced myocardial hypertrophy, and attenuated inflammatory responses [308].

5. Outlook and Future Perspective

The evolving understanding of oxidative stress in CVD has significantly influenced current therapeutic strategies. Despite advances in identifying redox signaling pathways and their contributions to disease progression, challenges persist in translating these insights into effective, targeted therapies. Future research must continue to elucidate the complex interplay between oxidative stress and cardiovascular pathophysiology, particularly its stage-specific roles—from early endothelial injury to advanced cardiac remodeling. Characterizing these processes at various disease stages may help identify novel, stage-specific targets to enhance therapeutic precision.
Antioxidant therapies have shown promising potential, yet their clinical efficacy remains inconsistent, partly due to the nonspecific nature of many antioxidants and the complexities of redox signaling in diverse cellular contexts. To address these challenges, future studies should focus on developing selective antioxidants that target specific signaling pathways, such as Nrf2/Keap1/ARE, SIRT1, and PI3K/Akt, while avoiding interference with essential physiological redox processes. Further exploration of cross-talk between these pathways may reveal synergies that could be leveraged in multi-target therapies for more robust cardiovascular protection.
Emerging strategies, including gene therapy, cellular and regenerative approaches, and inflammation regulation, offer a promising future for the treatment of CVDs driven by oxidative stress. Gene therapy holds particular promise in addressing genetic predispositions and modifying redox-sensitive genes to enhance antioxidant capacity. Meanwhile, cell-based therapies and regenerative medicine may facilitate tissue repair and functional recovery after injury. Additionally, immunomodulatory approaches that mitigate inflammation could complement antioxidant strategies, offering dual benefits in redox and immune regulation.
Ultimately, integrating metabolic regulation with oxidative stress management represents a promising frontier. Modulating metabolic pathways, including those involving AMPK and mitochondrial dynamics, may reduce oxidative stress and enhance cellular resilience, offering a holistic approach to CVD management. For successful clinical translation, future investigations must prioritize refining these novel interventions within robust preclinical and clinical frameworks, emphasizing safety, efficacy, and precision targeting. Such multifaceted approaches are likely to pave the way for more effective and personalized cardiovascular therapies, ultimately reducing the global burden of CVD.

Funding

This study was supported by the National Natural Science Foundation of China (grant no. 82300526) and the Natural Science Foundation of Hubei province (grant no. 2022CFB648).

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Tomii, D.; Pilgrim, T.; Borger, M.A.; De Backer, O.; Lanz, J.; Reineke, D.; Siepe, M.; Windecker, S. Aortic Stenosis and Coronary Artery Disease: Decision-Making Between Surgical and Transcatheter Management. Circulation 2024, 150, 2046–2069. [Google Scholar] [CrossRef] [PubMed]
  2. Zheng, W.C.; Chan, W.; Dart, A.; Shaw, J.A. Novel therapeutic targets and emerging treatments for atherosclerotic cardiovascular disease. Eur. Heart J. Cardiovasc. Pharmacother. 2024, 10, 53–67. [Google Scholar] [CrossRef] [PubMed]
  3. Hummelgaard, S.; Vilstrup, J.P.; Gustafsen, C.; Glerup, S.; Weyer, K. Targeting PCSK9 to tackle cardiovascular disease. Pharmacol. Ther. 2023, 249, 108480. [Google Scholar] [CrossRef] [PubMed]
  4. Rajendran, A.; Minhas, A.S.; Kazzi, B.; Varma, B.; Choi, E.; Thakkar, A.; Michos, E.D. Sex-specific differences in cardiovascular risk factors and implications for cardiovascular disease prevention in women. Atherosclerosis 2023, 384, 117269. [Google Scholar] [CrossRef]
  5. Ariza Corbo, M.J.; Muñiz-Grijalvo, O.; Blanco Echevarría, A.; Díaz-Díaz, J.L. Genetic basis of hypertriglyceridemia. Clin. Investig. Arterioscler. 2024, 36 (Suppl. 2), S3–S12. [Google Scholar] [CrossRef] [PubMed]
  6. Steven, S.; Frenis, K.; Oelze, M.; Kalinovic, S.; Kuntic, M.; Bayo Jimenez, M.T.; Vujacic-Mirski, K.; Helmstädter, J.; Kröller-Schön, S.; Münzel, T.; et al. Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease. Oxidative Med. Cell. Longev. 2019, 2019, 7092151. [Google Scholar] [CrossRef] [PubMed]
  7. Spinelli, S.; Marino, A.; Morabito, R.; Remigante, A. Interplay Between Metabolic Pathways and Increased Oxidative Stress in Human Red Blood Cells. Cells 2024, 13, 2026. [Google Scholar] [CrossRef] [PubMed]
  8. Shahidi, F.; Zhong, Y. Lipid oxidation and improving the oxidative stability. Chem. Soc. Rev. 2010, 39, 4067–4079. [Google Scholar] [CrossRef] [PubMed]
  9. Hauck, A.K.; Huang, Y.; Hertzel, A.V.; Bernlohr, D.A. Adipose oxidative stress and protein carbonylation. J. Biol. Chem. 2019, 294, 1083–1088. [Google Scholar] [CrossRef] [PubMed]
  10. Dhalla, N.S.; Elimban, V.; Bartekova, M.; Adameova, A. Involvement of Oxidative Stress in the Development of Subcellular Defects and Heart Disease. Biomedicines 2022, 10, 393. [Google Scholar] [CrossRef]
  11. Evans, C.E.; Cober, N.D.; Dai, Z.; Stewart, D.J.; Zhao, Y.Y. Endothelial cells in the pathogenesis of pulmonary arterial hypertension. Eur. Respir. J. 2021, 58, 2003957. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, X.; Xu, X.D.; Ma, M.Q.; Liang, Y.; Cai, Y.B.; Zhu, Z.X.; Xu, T.; Zhu, L.; Ren, K. The mechanisms of ferroptosis and its role in atherosclerosis. Biomed. Pharmacother. 2024, 171, 116112. [Google Scholar] [CrossRef] [PubMed]
  13. Nolfi-Donegan, D.; Braganza, A.; Shiva, S. Mitochondrial electron transport chain: Oxidative phosphorylation, oxidant production, and methods of measurement. Redox Biol. 2020, 37, 101674. [Google Scholar] [CrossRef]
  14. Pecchillo Cimmino, T.; Ammendola, R.; Cattaneo, F.; Esposito, G. NOX Dependent ROS Generation and Cell Metabolism. Int. J. Mol. Sci. 2023, 24, 2086. [Google Scholar] [CrossRef]
  15. Bortolotti, M.; Polito, L.; Battelli, M.G.; Bolognesi, A. Xanthine oxidoreductase: One enzyme for multiple physiological tasks. Redox Biol. 2021, 41, 101882. [Google Scholar] [CrossRef] [PubMed]
  16. Guo, Z.; Chi, R.; Peng, Y.; Sun, K.; Liu, H.; Guo, F.; Guo, J. The Role and Interactive Mechanism of Endoplasmic Reticulum Stress and Ferroptosis in Musculoskeletal Disorders. Biomolecules 2024, 14, 1369. [Google Scholar] [CrossRef] [PubMed]
  17. Zhao, R.Z.; Jiang, S.; Zhang, L.; Yu, Z.B. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int. J. Mol. Med. 2019, 44, 3–15. [Google Scholar] [CrossRef]
  18. Guo, R.; Zong, S.; Wu, M.; Gu, J.; Yang, M. Architecture of Human Mitochondrial Respiratory Megacomplex I2III2IV2. Cell 2017, 170, 1247–1257.e12. [Google Scholar] [CrossRef]
  19. Panov, A.V.; Mayorov, V.I.; Dikalov, S.I. Role of Fatty Acids β-Oxidation in the Metabolic Interactions Between Organs. Int. J. Mol. Sci. 2024, 25, 12740. [Google Scholar] [CrossRef] [PubMed]
  20. Hunte, C.; Zickermann, V.; Brandt, U. Functional modules and structural basis of conformational coupling in mitochondrial complex I. Science 2010, 329, 448–451. [Google Scholar] [CrossRef]
  21. Esteban-Amo, M.J.; Jiménez-Cuadrado, P.; Serrano-Lorenzo, P.; de la Fuente, M.; Simarro, M. Succinate Dehydrogenase and Human Disease: Novel Insights into a Well-Known Enzyme. Biomedicines 2024, 12, 2050. [Google Scholar] [CrossRef]
  22. Čunátová, K.; Fernández-Vizarra, E. Pathological variants in nuclear genes causing mitochondrial complex III deficiency: An update. J. Inherit. Metab. Dis. 2024, 47, 1278–1291. [Google Scholar] [CrossRef] [PubMed]
  23. Kadenbach, B.; Hüttemann, M. The subunit composition and function of mammalian cytochrome c oxidase. Mitochondrion 2015, 24, 64–76. [Google Scholar] [CrossRef] [PubMed]
  24. Zotta, A.; O’Neill, L.A.J.; Yin, M. Unlocking potential: The role of the electron transport chain in immunometabolism. Trends Immunol. 2024, 45, 259–273. [Google Scholar] [CrossRef] [PubMed]
  25. Pham, L.; Arroum, T.; Wan, J.; Pavelich, L.; Bell, J.; Morse, P.T.; Lee, I.; Grossman, L.I.; Sanderson, T.H.; Malek, M.H.; et al. Regulation of mitochondrial oxidative phosphorylation through tight control of cytochrome c oxidase in health and disease—Implications for ischemia/reperfusion injury, inflammatory diseases, diabetes, and cancer. Redox Biol. 2024, 78, 103426. [Google Scholar] [CrossRef]
  26. Le Bras, M.; Clément, M.V.; Pervaiz, S.; Brenner, C. Reactive oxygen species and the mitochondrial signaling pathway of cell death. Histol. Histopathol. 2005, 20, 205–219. [Google Scholar] [CrossRef]
  27. Pelicano, H.; Carney, D.; Huang, P. ROS stress in cancer cells and therapeutic implications. Drug Resist. Updates 2004, 7, 97–110. [Google Scholar] [CrossRef]
  28. Brand, M.D.; Pakay, J.L.; Ocloo, A.; Kokoszka, J.; Wallace, D.C.; Brookes, P.S.; Cornwall, E.J. The basal proton conductance of mitochondria depends on adenine nucleotide translocase content. Biochem. J. 2005, 392, 353–362. [Google Scholar] [CrossRef]
  29. Roussel, D.; Harding, M.; Runswick, M.J.; Walker, J.E.; Brand, M.D. Does any yeast mitochondrial carrier have a native uncoupling protein function? J. Bioenerg. Biomembr. 2002, 34, 165–176. [Google Scholar] [CrossRef]
  30. O’Reilly, A.; Zhao, W.; Wickström, S.; Arnér, E.S.J.; Kiessling, R. Reactive oxygen species: Janus-faced molecules in the era of modern cancer therapy. J. ImmunoTher. Cancer 2024, 12, e009409. [Google Scholar] [CrossRef]
  31. Gan, Z.; van der Stelt, I.; Li, W.; Hu, L.; Song, J.; Grefte, S.; van de Westerlo, E.; Zhang, D.; van Schothorst, E.M.; Claahsen-van der Grinten, H.L.; et al. Mitochondrial Nicotinamide Nucleotide Transhydrogenase: Role in Energy Metabolism, Redox Homeostasis, and Cancer. Antioxid. Redox Signal. 2024, 41, 927–956. [Google Scholar] [CrossRef]
  32. Kamal, R.; Awasthi, A.; Pundir, M.; Thakur, S. Healing the diabetic wound: Unlocking the secrets of genes and pathways. Eur. J. Pharmacol. 2024, 975, 176645. [Google Scholar] [CrossRef]
  33. Akhigbe, R.; Ajayi, A. The impact of reactive oxygen species in the development of cardiometabolic disorders: A review. Lipids Health Dis. 2021, 20, 23. [Google Scholar] [CrossRef]
  34. Silveira, T.H.R.; Silva, F.H.; Hill, W.G.; Antunes, E.; de Oliveira, M.G. Targeting NADPH Oxidase as an Approach for Diabetic Bladder Dysfunction. Antioxidants 2024, 13, 1155. [Google Scholar] [CrossRef]
  35. Yan, H.; Yin, Y.; Zhou, Y.; Li, Z.; Li, Y.; Ren, L.; Wen, J.; Wang, W. Regulation of cardiovascular diseases by histone deacetylases and NADPH oxidases. Redox Biol. 2024, 77, 103379. [Google Scholar] [CrossRef] [PubMed]
  36. Yuan, Q.; Tang, B.; Zhang, C. Signaling pathways of chronic kidney diseases, implications for therapeutics. Signal Transduct. Target. Ther. 2022, 7, 182. [Google Scholar] [CrossRef]
  37. García, J.G.; Ansorena, E.; Izal, I.; Zalba, G.; de Miguel, C.; Milagro, F.I. Structure, regulation, and physiological functions of NADPH oxidase 5 (NOX5). J. Physiol. Biochem. 2023, 79, 383–395. [Google Scholar] [CrossRef]
  38. Sylvester, A.L.; Zhang, D.X.; Ran, S.; Zinkevich, N.S. Inhibiting NADPH Oxidases to Target Vascular and Other Pathologies: An Update on Recent Experimental and Clinical Studies. Biomolecules 2022, 12, 823. [Google Scholar] [CrossRef]
  39. Vermot, A.; Petit-Härtlein, I.; Smith, S.M.E.; Fieschi, F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants 2021, 10, 890. [Google Scholar] [CrossRef] [PubMed]
  40. Ramos-Mondragón, R.; Lozhkin, A.; Vendrov, A.E.; Runge, M.S.; Isom, L.L.; Madamanchi, N.R. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants 2023, 12, 1833. [Google Scholar] [CrossRef]
  41. Furuhashi, M. New insights into purine metabolism in metabolic diseases: Role of xanthine oxidoreductase activity. Am. J. Physiol. Endocrinol. Metab. 2020, 319, E827–E834. [Google Scholar] [CrossRef]
  42. Mokhosoev, I.M.; Astakhov, D.V.; Terentiev, A.A.; Moldogazieva, N.T. Human Cytochrome P450 Cancer-Related Metabolic Activities and Gene Polymorphisms: A Review. Cells 2024, 13, 1958. [Google Scholar] [CrossRef]
  43. Knaus, U.G. Oxidants in Physiological Processes. Handb. Exp. Pharmacol. 2021, 264, 27–47. [Google Scholar] [CrossRef] [PubMed]
  44. Ullah, Z.; Yue, P.; Mao, G.; Zhang, M.; Liu, P.; Wu, X.; Zhao, T.; Yang, L. A comprehensive review on recent xanthine oxidase inhibitors of dietary based bioactive substances for the treatment of hyperuricemia and gout: Molecular mechanisms and perspective. Int. J. Biol. Macromol. 2024, 278, 134832. [Google Scholar] [CrossRef]
  45. Engin, A. Protein Kinases in Obesity, and the Kinase-Targeted Therapy. Adv. Exp. Med. Biol. 2024, 1460, 199–229. [Google Scholar] [CrossRef]
  46. Benítez-King, G.; Argueta, J.; Miranda-Riestra, A.; Muñoz-Delgado, J.; Estrada-Reyes, R. Interaction of the Melatonin/Ca2+-CaM Complex with Calmodulin Kinase II: Physiological Importance. Mol. Pharmacol. 2024, 106, 3–12. [Google Scholar] [CrossRef]
  47. Mirzavi, F.; Rajabian, A.; Hosseini, H. The potential protective role of carotenoids from saffron: A focus on endoplasmic reticulum stress-related organ damage. Food Sci. Nutr. 2024, 12, 6108–6122. [Google Scholar] [CrossRef]
  48. Engin, A. Nonalcoholic Fatty Liver Disease and Staging of Hepatic Fibrosis. Adv. Exp. Med. Biol. 2024, 1460, 539–574. [Google Scholar] [CrossRef]
  49. Laurindo, F.R.; Pescatore, L.A.; Fernandes Dde, C. Protein disulfide isomerase in redox cell signaling and homeostasis. Free Radic. Biol. Med. 2012, 52, 1954–1969. [Google Scholar] [CrossRef] [PubMed]
  50. Meneses-Valdés, R.; Gallero, S.; Henríquez-Olguín, C.; Jensen, T.E. Exploring NADPH oxidases 2 and 4 in cardiac and skeletal muscle adaptations—A cross-tissue comparison. Free Radic. Biol. Med. 2024, 223, 296–305. [Google Scholar] [CrossRef]
  51. Nkhumeleni, Z.; Phoswa, W.N.; Mokgalaboni, K. Purslane Ameliorates Inflammation and Oxidative Stress in Diabetes Mellitus: A Systematic Review. Int. J. Mol. Sci. 2024, 25, 12276. [Google Scholar] [CrossRef] [PubMed]
  52. Massart, J.; Begriche, K.; Hartman, J.H.; Fromenty, B. Role of Mitochondrial Cytochrome P450 2E1 in Healthy and Diseased Liver. Cells 2022, 11, 288. [Google Scholar] [CrossRef] [PubMed]
  53. Dai, C.; Das Gupta, S.; Wang, Z.; Jiang, H.; Velkov, T.; Shen, J. T-2 toxin and its cardiotoxicity: New insights on the molecular mechanisms and therapeutic implications. Food Chem. Toxicol. 2022, 167, 113262. [Google Scholar] [CrossRef] [PubMed]
  54. Antelo-Cea, D.A.; Martínez-Rojas, L.; Cabrerizo-Ibáñez, I.; Roudi Rashtabady, A.; Hernández-Alvarez, M.I. Regulation of Mitochondrial and Peroxisomal Metabolism in Female Obesity and Type 2 Diabetes. Int. J. Mol. Sci. 2024, 25, 11237. [Google Scholar] [CrossRef]
  55. Lan, R.; Zhang, M.J.; Liu, K.; Meng, F.F.; Xu, X.H.; Wang, C.C.; Zhang, M.Q.; Yan, Y.; Kou, J.J.; Zhao, L.L.; et al. Cytochrome P450-derived Epoxyeicosatrienoic Acid, the Regulation of Cardiovascular-related Diseases, and the Implication for Pulmonary Hypertension. Cardiovasc. Drugs Ther. 2024, 38, 1–17. [Google Scholar] [CrossRef]
  56. Blagov, A.V.; Summerhill, V.I.; Sukhorukov, V.N.; Zhigmitova, E.B.; Postnov, A.Y.; Orekhov, A.N. Potential use of antioxidants for the treatment of chronic inflammatory diseases. Front. Pharmacol. 2024, 15, 1378335. [Google Scholar] [CrossRef]
  57. Lu, Y.; George, J. Interaction between fatty acid oxidation and ethanol metabolism in liver. Am. J. Physiol. Gastrointest. Liver Physiol. 2024, 326, G483–G494. [Google Scholar] [CrossRef] [PubMed]
  58. Baker, A.; Lin, C.C.; Lett, C.; Karpinska, B.; Wright, M.H.; Foyer, C.H. Catalase: A critical node in the regulation of cell fate. Free Radic. Biol. Med. 2023, 199, 56–66. [Google Scholar] [CrossRef] [PubMed]
  59. Rius-Pérez, S. p53 at the crossroad between mitochondrial reactive oxygen species and necroptosis. Free Radic. Biol. Med. 2023, 207, 183–193. [Google Scholar] [CrossRef]
  60. Di Florio, D.N.; Sin, J.; Coronado, M.J.; Atwal, P.S.; Fairweather, D. Sex differences in inflammation, redox biology, mitochondria and autoimmunity. Redox Biol. 2020, 31, 101482. [Google Scholar] [CrossRef] [PubMed]
  61. Olié, V.; Grave, C.; Helft, G.; Nguyen-Thanh, V.; Andler, R.; Quatremere, G.; Pasquereau, A.; Lahaie, E.; Lailler, G.; Verdot, C.; et al. Epidemiology of cardiovascular risk factors: Behavioural risk factors. Arch. Cardiovasc. Dis. 2024, 117, 770–784. [Google Scholar] [CrossRef]
  62. Lailler, G.; Gabet, A.; Grave, C.; Boudet-Berquier, J.; El Rafei, R.; Regnault, N.; Acar, P.; Thomas-Chabaneix, J.; Tuppin, P.; Béjot, Y.; et al. Cardiovascular hospitalizations and deaths in adults, children and pregnant women. Arch. Cardiovasc. Dis. 2024, 117, 751–760. [Google Scholar] [CrossRef]
  63. Vona, R.; Ascione, B.; Malorni, W.; Straface, E. Mitochondria and Sex-Specific Cardiac Function. Adv. Exp. Med. Biol. 2018, 1065, 241–256. [Google Scholar] [CrossRef]
  64. Robert, J. Sex differences in vascular endothelial cells. Atherosclerosis 2023, 384, 117278. [Google Scholar] [CrossRef] [PubMed]
  65. Lv, Q.; Xu, W.; Yang, F.; Li, J.; Wei, W.; Chen, X.; Liu, Y.; Zhang, Z. Protective and Detoxifying Effects of Resveratrol on Zearalenone-Mediated Toxicity: A Review. Int. J. Mol. Sci. 2024, 25, 11003. [Google Scholar] [CrossRef] [PubMed]
  66. Krzyżewska, A.; Kurakula, K. Sex Dimorphism in Pulmonary Arterial Hypertension Associated with Autoimmune Diseases. Arterioscler. Thromb. Vasc. Biol. 2024, 44, 2169–2190. [Google Scholar] [CrossRef] [PubMed]
  67. Luo, Y.; Safabakhsh, S.; Palumbo, A.; Fiset, C.; Shen, C.; Parker, J.; Foster, L.J.; Laksman, Z. Sex-Based Mechanisms of Cardiac Development and Function: Applications for Induced-Pluripotent Stem Cell Derived-Cardiomyocytes. Int. J. Mol. Sci. 2024, 25, 5964. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, W.; Kang, P.M. Oxidative Stress and Antioxidant Treatments in Cardiovascular Diseases. Antioxidants 2020, 9, 1292. [Google Scholar] [CrossRef] [PubMed]
  69. Padmanaban, A.M.; Ganesan, K.; Ramkumar, K.M. A Co-Culture System for Studying Cellular Interactions in Vascular Disease. Bioengineering 2024, 11, 1090. [Google Scholar] [CrossRef] [PubMed]
  70. Yu, F.; Chen, J.; Zhang, X.; Ma, Z.; Wang, J.; Wu, Q. Role of Neutrophil Extracellular Traps in Hypertension and Their Impact on Target Organs. J. Clin. Hypertens. 2024, 26, 1323–1534. [Google Scholar] [CrossRef]
  71. Manoj, H.; Gomes, S.M.; Thimmappa, P.Y.; Nagareddy, P.R.; Jamora, C.; Joshi, M.B. Cytokine signalling in formation of neutrophil extracellular traps: Implications for health and diseases. Cytokine Growth Factor Rev. 2024; in press. [Google Scholar] [CrossRef]
  72. Latif, F.; Mubbashir, A.; Khan, M.S.; Shaikh, Z.; Memon, A.; Alvares, J.; Azhar, A.; Jain, H.; Ahmed, R.; Kanagala, S.G. Trimethylamine N-oxide in cardiovascular disease: Pathophysiology and the potential role of statins. Life Sci. 2024, 361, 123304. [Google Scholar] [CrossRef]
  73. Wang, L.; Cao, Y.; Gorshkov, B.; Zhou, Y.; Yang, Q.; Xu, J.; Ma, Q.; Zhang, X.; Wang, J.; Mao, X.; et al. Ablation of endothelial Pfkfb3 protects mice from acute lung injury in LPS-induced endotoxemia. Pharmacol. Res. 2019, 146, 104292. [Google Scholar] [CrossRef] [PubMed]
  74. Xu, S.; Ilyas, I.; Little, P.J.; Li, H.; Kamato, D.; Zheng, X.; Luo, S.; Li, Z.; Liu, P.; Han, J.; et al. Endothelial Dysfunction in Atherosclerotic Cardiovascular Diseases and Beyond: From Mechanism to Pharmacotherapies. Pharmacol. Rev. 2021, 73, 924–967. [Google Scholar] [CrossRef] [PubMed]
  75. Alvandi, Z.; Bischoff, J. Endothelial-Mesenchymal Transition in Cardiovascular Disease. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 2357–2369. [Google Scholar] [CrossRef]
  76. Eity, T.A.; Bhuia, M.S.; Chowdhury, R.; Ahmmed, S.; Salehin, S.; Akter, R.; Islam, M.T. Therapeutic Efficacy of Quercetin and Its Nanoformulation Both the Mono- or Combination Therapies in the Management of Cancer: An Update with Molecular Mechanisms. J. Trop. Med. 2024, 2024, 5594462. [Google Scholar] [CrossRef] [PubMed]
  77. Ren, J.; Liu, K.; Wu, B.; Lu, X.; Sun, L.; Privratsky, J.R.; Xing, C.; Robson, M.J.; Mao, H.; Blakely, R.D.; et al. Divergent Actions of Renal Tubular and Endothelial Type 1 IL-1 Receptor Signaling in Toxin-Induced AKI. J. Am. Soc. Nephrol. 2023, 34, 1629–1646. [Google Scholar] [CrossRef]
  78. Ge, Y.L.; Li, P.J.; Bu, Y.R.; Zhang, B.; Xu, J.; He, S.Y.; Cao, Q.L.; Bai, Y.G.; Ma, J.; Zhang, L.; et al. TNF-α and RPLP0 drive the apoptosis of endothelial cells and increase susceptibility to high-altitude pulmonary edema. Apoptosis 2024, 29, 1600–1618. [Google Scholar] [CrossRef]
  79. Palikuqi, B.; Nguyen, D.T.; Li, G.; Schreiner, R.; Pellegata, A.F.; Liu, Y.; Redmond, D.; Geng, F.; Lin, Y.; Gómez-Salinero, J.M.; et al. Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis. Nature 2020, 585, 426–432. [Google Scholar] [CrossRef]
  80. Chiu, J.J.; Chien, S. Effects of disturbed flow on vascular endothelium: Pathophysiological basis and clinical perspectives. Physiol. Rev. 2011, 91, 327–387. [Google Scholar] [CrossRef]
  81. Tokarz-Deptuła, B.; Baraniecki, Ł.; Palma, J.; Stosik, M.; Deptuła, W. Characterization of Platelet Receptors and Their Involvement in Immune Activation of These Cells. Int. J. Mol. Sci. 2024, 25, 12611. [Google Scholar] [CrossRef]
  82. Steinberg, D. The LDL modification hypothesis of atherogenesis: An update. J. Lipid Res. 2009, 50, S376–S381. [Google Scholar] [CrossRef]
  83. Rajpoot, A.; Aggarwal, T.; Sharma, V. Unraveling the enigma of cardiac damage caused by lead: Understanding the intricate relationship between oxidative stress and other multifactorial mechanisms. Toxicology 2024, 509, 153984. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Q.; Liu, J.; Duan, H.; Li, R.; Peng, W.; Wu, C. Activation of Nrf2/HO-1 signaling: An important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. J. Adv. Res. 2021, 34, 43–63. [Google Scholar] [CrossRef]
  85. Jacob, R.; Khan, M. Cardiac Biomarkers: What Is and What Can Be. Indian J. Cardiovasc. Dis. Women WINCARS 2018, 3, 240–244. [Google Scholar] [CrossRef] [PubMed]
  86. Kirichenko, T.V.; Sobenin, I.A.; Nikolic, D.; Rizzo, M.; Orekhov, A.N. Anti-cytokine therapy for prevention of atherosclerosis. Phytomedicine 2016, 23, 1198–1210. [Google Scholar] [CrossRef] [PubMed]
  87. Ouweneel, A.B.; Van Eck, M. Lipoproteins as modulators of atherothrombosis: From endothelial function to primary and secondary coagulation. Vascul. Pharmacol. 2016, 82, 1–10. [Google Scholar] [CrossRef]
  88. Engin, A. Endothelial Dysfunction in Obesity and Therapeutic Targets. Adv. Exp. Med. Biol. 2024, 1460, 489–538. [Google Scholar] [CrossRef] [PubMed]
  89. Kardassis, D.; Vindis, C.; Stancu, C.S.; Toma, L.; Gafencu, A.V.; Georgescu, A.; Alexandru-Moise, N.; Molica, F.; Kwak, B.R.; Burlacu, A.; et al. Unravelling molecular mechanisms in atherosclerosis using cellular models and omics technologies. Vascul. Pharmacol. 2024, 158, 107452. [Google Scholar] [CrossRef]
  90. Yang, B.; Hang, S.; Xu, S.; Gao, Y.; Yu, W.; Zang, G.; Zhang, L.; Wang, Z. Macrophage polarisation and inflammatory mechanisms in atherosclerosis: Implications for prevention and treatment. Heliyon 2024, 10, e32073. [Google Scholar] [CrossRef]
  91. Song, J.; Cao, C.; Wang, Z.; Li, H.; Yang, L.; Kang, J.; Meng, H.; Li, L.; Liu, J. Mechanistic insights into the regression of atherosclerotic plaques. Front. Physiol. 2024, 15, 1473709. [Google Scholar] [CrossRef]
  92. Saki, N.; Haybar, H.; Maniati, M.; Davari, N.; Javan, M.; Moghimian-Boroujeni, B. Modification macrophage to foam cells in atherosclerosis disease: Some factors stimulate or inhibit this process. J. Diabetes Metab. Disord. 2024, 23, 1687–1697. [Google Scholar] [CrossRef] [PubMed]
  93. Brown, O.I.; Bridge, K.I.; Kearney, M.T. Nicotinamide Adenine Dinucleotide Phosphate Oxidases in Glucose Homeostasis and Diabetes-Related Endothelial Cell Dysfunction. Cells 2021, 10, 2315. [Google Scholar] [CrossRef]
  94. Yu, X.H.; Fu, Y.C.; Zhang, D.W.; Yin, K.; Tang, C.K. Foam cells in atherosclerosis. Clin. Chim. Acta 2013, 424, 245–252. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, W.; Wu, X.; Hu, J.; Liu, X.; Guo, Z.; Wu, J.; Shao, Y.; Hao, M.; Zhang, S.; Hu, W.; et al. The translational potential of miR-26 in atherosclerosis and development of agents for its target genes ACC1/2, COL1A1, CPT1A, FBP1, DGAT2, and SMAD7. Cardiovasc. Diabetol. 2024, 23, 21. [Google Scholar] [CrossRef] [PubMed]
  96. Ma, J.; Zhang, H.; Chen, Y.; Liu, X.; Tian, J.; Shen, W. The Role of Macrophage Iron Overload and Ferroptosis in Atherosclerosis. Biomolecules 2022, 12, 1702. [Google Scholar] [CrossRef]
  97. Obare, L.M.; Temu, T.; Mallal, S.A.; Wanjalla, C.N. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ. Res. 2024, 134, 1515–1545. [Google Scholar] [CrossRef] [PubMed]
  98. Davidson, S.M.; Adameová, A.; Barile, L.; Cabrera-Fuentes, H.A.; Lazou, A.; Pagliaro, P.; Stensløkken, K.O.; Garcia-Dorado, D. Mitochondrial and mitochondrial-independent pathways of myocardial cell death during ischaemia and reperfusion injury. J. Cell. Mol. Med. 2020, 24, 3795–3806. [Google Scholar] [CrossRef] [PubMed]
  99. Pandey, A.; Li, Z.; Gautam, M.; Ghosh, A.; Man, S.M. Molecular mechanisms of emerging inflammasome complexes and their activation and signaling in inflammation and pyroptosis. Immunol. Rev. 2024, 38, 1–18. [Google Scholar] [CrossRef]
  100. Jiang, Y.; Xing, W.; Li, Z.; Zhao, D.; Xiu, B.; Xi, Y.; Bai, S.; Li, X.; Zhang, Z.; Zhang, W.; et al. The calcium-sensing receptor alleviates endothelial inflammation in atherosclerosis through regulation of integrin β1-NLRP3 inflammasome. FEBS J. 2024, 291, 1–18. [Google Scholar] [CrossRef]
  101. Minutoli, L.; Puzzolo, D.; Rinaldi, M.; Irrera, N.; Marini, H.; Arcoraci, V.; Bitto, A.; Crea, G.; Pisani, A.; Squadrito, F.; et al. ROS-Mediated NLRP3 Inflammasome Activation in Brain, Heart, Kidney, and Testis Ischemia/Reperfusion Injury. Oxidative Med. Cell. Longev. 2016, 2016, 2183026. [Google Scholar] [CrossRef] [PubMed]
  102. Hao, W.; Feng, C. Research progress on pyroptosis and its effect on the central nervous system. Neurobiol. Dis. 2023, 188, 106333. [Google Scholar] [CrossRef]
  103. Gupta, S.; Cassel, S.L.; Sutterwala, F.S.; Dagvadorj, J. Regulation of the NLRP3 inflammasome by autophagy and mitophagy. Immunol. Rev. 2024. [Google Scholar] [CrossRef] [PubMed]
  104. Gao, Y.; Xu, X.; Zhang, X. Targeting different phenotypes of macrophages: A potential strategy for natural products to treat inflammatory bone and joint diseases. Phytomedicine 2023, 118, 154952. [Google Scholar] [CrossRef] [PubMed]
  105. Wortmann, M.; Peters, A.S.; Erhart, P.; Körfer, D.; Böckler, D.; Dihlmann, S. Inflammasomes in the Pathophysiology of Aortic Disease. Cells 2021, 10, 2433. [Google Scholar] [CrossRef] [PubMed]
  106. Bäck, M.; Yurdagul, A., Jr.; Tabas, I.; Öörni, K.; Kovanen, P.T. Inflammation and its resolution in atherosclerosis: Mediators and therapeutic opportunities. Nat. Rev. Cardiol. 2019, 16, 389–406. [Google Scholar] [CrossRef]
  107. Prouse, T.; Majumder, S.; Majumder, R. Functions of TAM Receptors and Ligands Protein S and Gas6 in Atherosclerosis and Cardiovascular Disease. Int. J. Mol. Sci. 2024, 25, 12736. [Google Scholar] [CrossRef]
  108. Zhou, D.; Yang, Y.; Han, R.; He, J.; Liu, D.; Xia, W.; Cai, Y.; Perek, B.; Xia, Z. Ferroptosis and its Potential Determinant Role in Myocardial Susceptibility to Ischemia/Reperfusion Injury in Diabetes. Rev. Cardiovasc. Med. 2024, 25, 360. [Google Scholar] [CrossRef] [PubMed]
  109. Jin, B.; Zhang, Z.; Zhang, Y.; Yang, M.; Wang, C.; Xu, J.; Zhu, Y.; Mi, Y.; Jiang, J.; Sun, Z. Ferroptosis and myocardial ischemia-reperfusion: Mechanistic insights and new therapeutic perspectives. Front. Pharmacol. 2024, 15, 1482986. [Google Scholar] [CrossRef]
  110. Carden, D.L.; Granger, D.N. Pathophysiology of ischaemia-reperfusion injury. J. Pathol. 2000, 190, 255–266. [Google Scholar] [CrossRef]
  111. Chen, M.; Wang, R.; Liao, L.; Li, Y.; Sun, X.; Wu, H.; Lan, Q.; Deng, Z.; Liu, P.; Xu, T.; et al. DanShen Decoction targets miR-93-5p to provide protection against MI/RI by regulating the TXNIP/NLRP3/Caspase-1 signaling pathway. Phytomedicine 2024, 135, 156225. [Google Scholar] [CrossRef] [PubMed]
  112. Wei, X.H.; Chen, J.; Wu, X.F.; Zhang, Q.; Xia, G.Y.; Chu, X.Y.; Xia, H.; Lin, S.; Shang, H.C. Salvianolic acid B alleviated myocardial ischemia-reperfusion injury via modulating SIRT3-mediated crosstalk between mitochondrial ROS and NLRP3. Phytomedicine 2024, 136, 156260. [Google Scholar] [CrossRef] [PubMed]
  113. Li, H.; Yang, D.H.; Zhang, Y.; Zheng, F.; Gao, F.; Sun, J.; Shi, G. Geniposide suppresses NLRP3 inflammasome-mediated pyroptosis via the AMPK signaling pathway to mitigate myocardial ischemia/reperfusion injury. Chin. Med. 2022, 17, 73. [Google Scholar] [CrossRef]
  114. Pagliaro, P.; Penna, C. Inhibitors of NLRP3 Inflammasome in Ischemic Heart Disease: Focus on Functional and Redox Aspects. Antioxidants 2023, 12, 1396. [Google Scholar] [CrossRef] [PubMed]
  115. Alloatti, G.; Penna, C.; Comità, S.; Tullio, F.; Aragno, M.; Biasi, F.; Pagliaro, P. Aging, sex and NLRP3 inflammasome in cardiac ischaemic disease. Vascul. Pharmacol. 2022, 145, 107001. [Google Scholar] [CrossRef]
  116. Bugger, H.; Pfeil, K. Mitochondrial ROS in myocardial ischemia reperfusion and remodeling. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165768. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, C.; Zhang, Z.; Zhang, W.; Liu, X. Mitochondrial dysfunction and mitochondrial therapies in heart failure. Pharmacol. Res. 2022, 175, 106038. [Google Scholar] [CrossRef] [PubMed]
  118. Atici, A.E.; Crother, T.R.; Noval Rivas, M. Mitochondrial quality control in health and cardiovascular diseases. Front. Cell Dev. Biol. 2023, 11, 1290046. [Google Scholar] [CrossRef]
  119. Chen, T.H.; Wang, H.C.; Chang, C.J.; Lee, S.Y. Mitochondrial Glutathione in Cellular Redox Homeostasis and Disease Manifestation. Int. J. Mol. Sci. 2024, 25, 1314. [Google Scholar] [CrossRef]
  120. Peng, Z.; Liang, Y.; Liu, X.; Shao, J.; Hu, N.; Zhang, X. New insights into the mechanisms of diabetic kidney disease: Role of circadian rhythm and Bmal1. Biomed. Pharmacother. 2023, 166, 115422. [Google Scholar] [CrossRef]
  121. Fu, L.; Zhang, L. Physiological functions of CKIP-1: From molecular mechanisms to therapy implications. Ageing Res. Rev. 2019, 53, 100908. [Google Scholar] [CrossRef]
  122. Oduro, P.K.; Zheng, X.; Wei, J.; Yang, Y.; Wang, Y.; Zhang, H.; Liu, E.; Gao, X.; Du, M.; Wang, Q. The cGAS-STING signaling in cardiovascular and metabolic diseases: Future novel target option for pharmacotherapy. Acta Pharm. Sin. B 2022, 12, 50–75. [Google Scholar] [CrossRef]
  123. Takada, S.; Maekawa, S.; Furihata, T.; Kakutani, N.; Setoyama, D.; Ueda, K.; Nambu, H.; Hagiwara, H.; Handa, H.; Fumoto, Y.; et al. Succinyl-CoA-based energy metabolism dysfunction in chronic heart failure. Proc. Natl. Acad. Sci. USA 2022, 119, e2203628119. [Google Scholar] [CrossRef] [PubMed]
  124. Li, G.; Shao, Y.; Guo, H.C.; Zhi, Y.; Qiao, B.; Ma, K.; Du, J.; Lai, Y.Q.; Li, Y. MicroRNA-27b-3p down-regulates FGF1 and aggravates pathological cardiac remodelling. Cardiovasc. Res. 2022, 118, 2139–2151. [Google Scholar] [CrossRef]
  125. Mongirdienė, A.; Skrodenis, L.; Varoneckaitė, L.; Mierkytė, G.; Gerulis, J. Reactive Oxygen Species Induced Pathways in Heart Failure Pathogenesis and Potential Therapeutic Strategies. Biomedicines 2022, 10, 602. [Google Scholar] [CrossRef] [PubMed]
  126. Noctor, G.; Foyer, C.H. Intracellular Redox Compartmentation and ROS-Related Communication in Regulation and Signaling. Plant Physiol. 2016, 171, 1581–1592. [Google Scholar] [CrossRef] [PubMed]
  127. Cortassa, S.; Juhaszova, M.; Aon, M.A.; Zorov, D.B.; Sollott, S.J. Mitochondrial Ca2+, redox environment and ROS emission in heart failure: Two sides of the same coin? J. Mol. Cell. Cardiol. 2021, 151, 113–125. [Google Scholar] [CrossRef] [PubMed]
  128. Tsutsui, H.; Kinugawa, S.; Matsushima, S. Oxidative stress and heart failure. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H2181–H2190. [Google Scholar] [CrossRef]
  129. Bej, E.; Cesare, P.; d’Angelo, M.; Volpe, A.R.; Castelli, V. Neuronal Cell Rearrangement During Aging: Antioxidant Compounds as a Potential Therapeutic Approach. Cells 2024, 13, 1945. [Google Scholar] [CrossRef] [PubMed]
  130. Prasad, S.; Kulshreshtha, A.; Lall, R.; Gupta, S.C. Inflammation and ROS in arthritis: Management by Ayurvedic medicinal plants. Food Funct. 2021, 12, 8227–8247. [Google Scholar] [CrossRef]
  131. Li, D.; Rui, Y.X.; Guo, S.D.; Luan, F.; Liu, R.; Zeng, N. Ferulic acid: A review of its pharmacology, pharmacokinetics and derivatives. Life Sci. 2021, 284, 119921. [Google Scholar] [CrossRef] [PubMed]
  132. Downey, J.M.; Cohen, M.V. A really radical observation—A comment on Penna et al. in Basic Res Cardiol (2006) 101:180–189. Basic. Res. Cardiol. 2006, 101, 190–191. [Google Scholar] [CrossRef] [PubMed]
  133. Khan, M.Z.; Chen, W.; Liu, X.; Kou, X.; Khan, A.; Khan, R.U.; Zahoor, M.; Wang, C. An Overview of Bioactive Compounds’ Role in Modulating the Nrf2/Keap1/NF-κB Pathway to Alleviate Lipopolysaccharide-Induced Endometritis. Int. J. Mol. Sci. 2024, 25, 10319. [Google Scholar] [CrossRef]
  134. Penna, C.; Rastaldo, R.; Mancardi, D.; Raimondo, S.; Cappello, S.; Gattullo, D.; Losano, G.; Pagliaro, P. Post-conditioning induced cardioprotection requires signaling through a redox-sensitive mechanism, mitochondrial ATP-sensitive K+ channel and protein kinase C activation. Basic. Res. Cardiol. 2006, 101, 180–189. [Google Scholar] [CrossRef]
  135. Tsutsumi, Y.M.; Yokoyama, T.; Horikawa, Y.; Roth, D.M.; Patel, H.H. Reactive oxygen species trigger ischemic and pharmacological postconditioning: In vivo and in vitro characterization. Life Sci. 2007, 81, 1223–1227. [Google Scholar] [CrossRef]
  136. Voronkov, N.S.; Popov, S.V.; Naryzhnaya, N.V.; Prasad, N.R.; Petrov, I.M.; Kolpakov, V.V.; Tomilova, E.A.; Sapozhenkova, E.V.; Maslov, L.N. Effect of Cold Adaptation on the State of Cardiovascular System and Cardiac Tolerance to Ischemia/Reperfusion Injury. Iran. Biomed. J. 2024, 28, 59–70. [Google Scholar] [CrossRef]
  137. Xu, M.; Wu, G.; You, Q.; Chen, X. The Landscape of Smart Biomaterial-Based Hydrogen Therapy. Adv. Sci. 2024, 11, e2401310. [Google Scholar] [CrossRef]
  138. Di Pietro, M.; Filardo, S.; Falasca, F.; Turriziani, O.; Sessa, R. Infectious Agents in Atherosclerotic Cardiovascular Diseases through Oxidative Stress. Int. J. Mol. Sci. 2017, 18, 2459. [Google Scholar] [CrossRef]
  139. Ju, S.; Singh, M.K.; Han, S.; Ranbhise, J.; Ha, J.; Choe, W.; Yoon, K.S.; Yeo, S.G.; Kim, S.S.; Kang, I. Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species. Int. J. Mol. Sci. 2024, 25, 12387. [Google Scholar] [CrossRef] [PubMed]
  140. Procaccio, V.; Bris, C.; Chao de la Barca, J.M.; Oca, F.; Chevrollier, A.; Amati-Bonneau, P.; Bonneau, D.; Reynier, P. Perspectives of drug-based neuroprotection targeting mitochondria. Rev. Neurol. 2014, 170, 390–400. [Google Scholar] [CrossRef]
  141. Sommers, O.; Tomsine, R.A.; Khacho, M. Mitochondrial Dynamics Drive Muscle Stem Cell Progression from Quiescence to Myogenic Differentiation. Cells 2024, 13, 1773. [Google Scholar] [CrossRef]
  142. Maqoud, F.; Scala, R.; Hoxha, M.; Zappacosta, B.; Tricarico, D. ATP-sensitive Potassium Channel Subunits in Neuroinflammation: Novel Drug Targets in Neurodegenerative Disorders. CNS Neurol. Disord. Drug Targets 2022, 21, 130–149. [Google Scholar] [CrossRef]
  143. Macrì, R.; Musolino, V.; Gliozzi, M.; Carresi, C.; Maiuolo, J.; Nucera, S.; Scicchitano, M.; Bosco, F.; Scarano, F.; Ruga, S.; et al. Ferula L. Plant Extracts and Dose-Dependent Activity of Natural Sesquiterpene Ferutinin: From Antioxidant Potential to Cytotoxic Effects. Molecules 2020, 25, 5768. [Google Scholar] [CrossRef]
  144. Wang, L.; Apel, K. Dose-dependent effects of 1O2 in chloroplasts are determined by its timing and localization of production. J. Exp. Bot. 2019, 70, 29–40. [Google Scholar] [CrossRef] [PubMed]
  145. Kopacz, A.; Kloska, D.; Forman, H.J.; Jozkowicz, A.; Grochot-Przeczek, A. Beyond repression of Nrf2: An update on Keap1. Free Radic. Biol. Med. 2020, 157, 63–74. [Google Scholar] [CrossRef] [PubMed]
  146. Tanase, D.M.; Gosav, E.M.; Anton, M.I.; Floria, M.; Seritean Isac, P.N.; Hurjui, L.L.; Tarniceriu, C.C.; Costea, C.F.; Ciocoiu, M.; Rezus, C. Oxidative Stress and NRF2/KEAP1/ARE Pathway in Diabetic Kidney Disease (DKD): New Perspectives. Biomolecules 2022, 12, 1227. [Google Scholar] [CrossRef] [PubMed]
  147. Shi, S.; Chen, Y.; Luo, Z.; Nie, G.; Dai, Y. Role of oxidative stress and inflammation-related signaling pathways in doxorubicin-induced cardiomyopathy. Cell Commun. Signal. 2023, 21, 61. [Google Scholar] [CrossRef]
  148. Chauhan, W.; Zennadi, R. Keap1-Nrf2 Heterodimer: A Therapeutic Target to Ameliorate Sickle Cell Disease. Antioxidants 2023, 12, 740. [Google Scholar] [CrossRef]
  149. Wu, L.; Wang, L.; Du, Y.; Zhang, Y.; Ren, J. Mitochondrial quality control mechanisms as therapeutic targets in doxorubicin-induced cardiotoxicity. Trends Pharmacol. Sci. 2023, 44, 34–49. [Google Scholar] [CrossRef] [PubMed]
  150. Singh, V.; Ubaid, S. Role of Silent Information Regulator 1 (SIRT1) in Regulating Oxidative Stress and Inflammation. Inflammation 2020, 43, 1589–1598. [Google Scholar] [CrossRef] [PubMed]
  151. Zhao, L.; Qi, Y.; Xu, L.; Tao, X.; Han, X.; Yin, L.; Peng, J. MicroRNA-140-5p aggravates doxorubicin-induced cardiotoxicity by promoting myocardial oxidative stress via targeting Nrf2 and Sirt2. Redox Biol. 2018, 15, 284–296. [Google Scholar] [CrossRef] [PubMed]
  152. Zhao, L.; Tao, X.; Qi, Y.; Xu, L.; Yin, L.; Peng, J. Protective effect of dioscin against doxorubicin-induced cardiotoxicity via adjusting microRNA-140-5p-mediated myocardial oxidative stress. Redox Biol. 2018, 16, 189–198. [Google Scholar] [CrossRef] [PubMed]
  153. Bertoni, C.; Abodi, M.; D’Oria, V.; Milani, G.P.; Agostoni, C.; Mazzocchi, A. Alpha-Linolenic Acid and Cardiovascular Events: A Narrative Review. Int. J. Mol. Sci. 2023, 24, 14319. [Google Scholar] [CrossRef] [PubMed]
  154. Uddin, M.S.; Mamun, A.A.; Jakaria, M.; Thangapandiyan, S.; Ahmad, J.; Rahman, M.A.; Mathew, B.; Abdel-Daim, M.M.; Aleya, L. Emerging promise of sulforaphane-mediated Nrf2 signaling cascade against neurological disorders. Sci. Total Environ. 2020, 707, 135624. [Google Scholar] [CrossRef] [PubMed]
  155. Davis, T.M.; Coleman, R.L.; Holman, R.R. Prognostic significance of silent myocardial infarction in newly diagnosed type 2 diabetes mellitus: United Kingdom Prospective Diabetes Study (UKPDS) 79. Circulation 2013, 127, 980–987. [Google Scholar] [CrossRef] [PubMed]
  156. Cenci, A.; Macchia, I.; La Sorsa, V.; Sbarigia, C.; Di Donna, V.; Pietraforte, D. Mechanisms of Action of Ozone Therapy in Emerging Viral Diseases: Immunomodulatory Effects and Therapeutic Advantages with Reference to SARS-CoV-2. Front. Microbiol. 2022, 13, 871645. [Google Scholar] [CrossRef] [PubMed]
  157. Karan, A.; Bhakkiyalakshmi, E.; Jayasuriya, R.; Sarada, D.V.L.; Ramkumar, K.M. The pivotal role of nuclear factor erythroid 2-related factor 2 in diabetes-induced endothelial dysfunction. Pharmacol. Res. 2020, 153, 104601. [Google Scholar] [CrossRef] [PubMed]
  158. Liu, Y.; Zhang, R.; Zou, J.; Yin, H.; Zhao, M.; Zhao, L. The impact of chitooligosaccharides with a certain degree of polymerization on diabetic nephropathic mice and high glucose-damaged HK-2 cells. Food Sci. Nutr. 2024, 12, 4173–4184. [Google Scholar] [CrossRef]
  159. Nie, C.; He, T.; Zhang, W.; Zhang, G.; Ma, X. Branched Chain Amino Acids: Beyond Nutrition Metabolism. Int. J. Mol. Sci. 2018, 19, 954. [Google Scholar] [CrossRef]
  160. Ramasubbu, K.; Devi Rajeswari, V. Impairment of insulin signaling pathway PI3K/Akt/mTOR and insulin resistance induced AGEs on diabetes mellitus and neurodegenerative diseases: A perspective review. Mol. Cell. Biochem. 2023, 478, 1307–1324. [Google Scholar] [CrossRef] [PubMed]
  161. Meng, H.; Zhang, Y.; An, S.T.; Chen, Y. Annexin A3 gene silencing promotes myocardial cell repair through activation of the PI3K/Akt signaling pathway in rats with acute myocardial infarction. J. Cell. Physiol. 2019, 234, 10535–10546. [Google Scholar] [CrossRef]
  162. Chen, X.; Zhabyeyev, P.; Azad, A.K.; Vanhaesebroeck, B.; Grueter, C.E.; Murray, A.G.; Kassiri, Z.; Oudit, G.Y. Pharmacological and cell-specific genetic PI3Kα inhibition worsens cardiac remodeling after myocardial infarction. J. Mol. Cell. Cardiol. 2021, 157, 17–30. [Google Scholar] [CrossRef]
  163. Qin, W.; Cao, L.; Massey, I.Y. Role of PI3K/Akt signaling pathway in cardiac fibrosis. Mol. Cell. Biochem. 2021, 476, 4045–4059. [Google Scholar] [CrossRef] [PubMed]
  164. Puthusseri, B.; Marudamuthu, A.; Tiwari, N.; Fu, J.; Idell, S.; Shetty, S. Regulation of p53-mediated changes in the uPA-fibrinolytic system and in lung injury by loss of surfactant protein C expression in alveolar epithelial cells. Am. J. Physiol. Lung Cell Mol. Physiol. 2017, 312, L783–L796. [Google Scholar] [CrossRef] [PubMed]
  165. Cameselle-García, S.; Abdulkader-Nallib, I.; Sánchez-Ares, M.; Cameselle-Teijeiro, J.M. Cribriform morular thyroid carcinoma: Clinicopathological and molecular basis for both a preventive and therapeutic approach for a rare tumor (Review). Oncol. Rep. 2024, 52, 119. [Google Scholar] [CrossRef] [PubMed]
  166. Alfadda, A.A.; Sallam, R.M. Reactive oxygen species in health and disease. J. Biomed. Biotechnol. 2012, 2012, 936486. [Google Scholar] [CrossRef] [PubMed]
  167. Kobayashi, H.; Imanaka, S. Mitochondrial DNA Damage and Its Repair Mechanisms in Aging Oocytes. Int. J. Mol. Sci. 2024, 25, 13144. [Google Scholar] [CrossRef] [PubMed]
  168. Ouyang, C.; You, J.; Xie, Z. The interplay between autophagy and apoptosis in the diabetic heart. J. Mol. Cell. Cardiol. 2014, 71, 71–80. [Google Scholar] [CrossRef]
  169. Penugurti, V.; Manne, R.K.; Bai, L.; Kant, R.; Lin, H.K. AMPK: The energy sensor at the crossroads of aging and cancer. Semin. Cancer Biol. 2024, 106–107, 15–27. [Google Scholar] [CrossRef]
  170. Abdul Khaliq, H.; Alhouayek, M.; Quetin-Leclercq, J.; Muccioli, G.G. 5’AMP-activated protein kinase: An emerging target of phytochemicals to treat chronic inflammatory diseases. Crit. Rev. Food Sci. Nutr. 2024, 64, 4763–4788. [Google Scholar] [CrossRef]
  171. Zhou, R.; Barnes, K.; Gibson, S.; Fillmore, N. Dual-edged role of SIRT1 in energy metabolism and cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 2024, 327, H1162–H1173. [Google Scholar] [CrossRef]
  172. Kitada, M.; Ogura, Y.; Koya, D. The protective role of Sirt1 in vascular tissue: Its relationship to vascular aging and atherosclerosis. Aging 2016, 8, 2290–2307. [Google Scholar] [CrossRef] [PubMed]
  173. Campagna, R.; Mazzanti, L.; Pompei, V.; Alia, S.; Vignini, A.; Emanuelli, M. The Multifaceted Role of Endothelial Sirt1 in Vascular Aging: An Update. Cells 2024, 13, 1469. [Google Scholar] [CrossRef] [PubMed]
  174. Sazdova, I.; Hadzi-Petrushev, N.; Keremidarska-Markova, M.; Stojchevski, R.; Sopi, R.; Shileiko, S.; Mitrokhin, V.; Gagov, H.; Avtanski, D.; Lubomirov, L.T.; et al. SIRT-associated attenuation of cellular senescence in vascular wall. Mech. Ageing Dev. 2024, 220, 111943. [Google Scholar] [CrossRef]
  175. Zhang, Q.J.; Wang, Z.; Chen, H.Z.; Zhou, S.; Zheng, W.; Liu, G.; Wei, Y.S.; Cai, H.; Liu, D.P.; Liang, C.C. Endothelium-specific overexpression of class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein E-deficient mice. Cardiovasc. Res. 2008, 80, 191–199. [Google Scholar] [CrossRef] [PubMed]
  176. Napoli, C.; Balestrieri, M.L.; Sica, V.; Lerman, L.O.; Crimi, E.; De Rosa, G.; Schiano, C.; Servillo, L.; D’Armiento, F.P. Beneficial effects of low doses of red wine consumption on perturbed shear stress-induced atherogenesis. Heart Vessel. 2008, 23, 124–133. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, F.; Chen, H.Z. Histone Deacetylase SIRT1, Smooth Muscle Cell Function, and Vascular Diseases. Front. Pharmacol. 2020, 11, 537519. [Google Scholar] [CrossRef]
  178. Law, M.; Wang, P.C.; Zhou, Z.Y.; Wang, Y. From Microcirculation to Aging-Related Diseases: A Focus on Endothelial SIRT1. Pharmaceuticals 2024, 17, 1495. [Google Scholar] [CrossRef]
  179. Donato, A.J.; Magerko, K.A.; Lawson, B.R.; Durrant, J.R.; Lesniewski, L.A.; Seals, D.R. SIRT-1 and vascular endothelial dysfunction with ageing in mice and humans. J. Physiol. 2011, 589, 4545–4554. [Google Scholar] [CrossRef] [PubMed]
  180. Goyal, A.; Kumari, A.; Verma, A.; Chaudhary, V.; Agrawal, V.; Yadav, H.N. Silent Information Regulator 1/Peroxisome Proliferator-Activated Receptor-γ Coactivator-1α Axis: A Promising Target for Parkinson’s and Alzheimer’s Disease Therapies. J. Biochem. Mol. Toxicol. 2024, 38, e70078. [Google Scholar] [CrossRef] [PubMed]
  181. Kauppinen, A.; Suuronen, T.; Ojala, J.; Kaarniranta, K.; Salminen, A. Antagonistic crosstalk between NF-κB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell. Signal. 2013, 25, 1939–1948. [Google Scholar] [CrossRef]
  182. Xue, H.Z.; Chen, Y.; Wang, S.D.; Yang, Y.M.; Cai, L.Q.; Zhao, J.X.; Huang, W.J.; Xiao, Y.H. Radix Astragali and Its Representative Extracts for Diabetic Nephropathy: Efficacy and Molecular Mechanism. J. Diabetes Res. 2024, 2024, 5216113. [Google Scholar] [CrossRef] [PubMed]
  183. Xian, Y.; Ye, J.; Tang, Y.; Zhang, N.; Peng, C.; Huang, W.; He, G. Deubiquitinases as novel therapeutic targets for diseases. MedComm 2024, 5, e70036. [Google Scholar] [CrossRef] [PubMed]
  184. Cazzola, M.; Calzetta, L.; Rogliani, P.; Matera, M.G. Emerging Anti-Inflammatory COPD Treatments: Potential Cardiovascular Impacts. Int. J. Chronic Obstr. Pulm. Dis. 2024, 19, 2481–2495. [Google Scholar] [CrossRef]
  185. Khanahmadi, M.; Ebrahimi Fard, M.; Baghani, M.; Shayan, M.; Baghani, M. Exploring STK3 in melanoma: A systematic review of signaling networks and therapeutic opportunities. Mol. Biol. Rep. 2024, 52, 8. [Google Scholar] [CrossRef] [PubMed]
  186. Puranik, N.; Jung, H.; Song, M. SPROUTY2, a Negative Feedback Regulator of Receptor Tyrosine Kinase Signaling, Associated with Neurodevelopmental Disorders: Current Knowledge and Future Perspectives. Int. J. Mol. Sci. 2024, 25, 11043. [Google Scholar] [CrossRef]
  187. Ramli, F.F.; Ali, A.; Ibrahim, N. Molecular-Signaling Pathways of Ginsenosides Rb in Myocardial Ischemia-Reperfusion Injury: A Mini Review. Int. J. Med. Sci. 2022, 19, 65–73. [Google Scholar] [CrossRef]
  188. Spirrison, A.N.; Lannigan, D.A. RSK1 and RSK2 as therapeutic targets: An up-to-date snapshot of emerging data. Expert Opin. Ther. Targets 2024, 27, 1–13. [Google Scholar] [CrossRef] [PubMed]
  189. Liang, Q.; Bueno, O.F.; Wilkins, B.J.; Kuan, C.Y.; Xia, Y.; Molkentin, J.D. c-Jun N-terminal kinases (JNK) antagonize cardiac growth through cross-talk with calcineurin-NFAT signaling. EMBO J. 2003, 22, 5079–5089. [Google Scholar] [CrossRef] [PubMed]
  190. Attachaipanich, T.; Chattipakorn, S.C.; Chattipakorn, N. Cardiovascular toxicities by calcineurin inhibitors: Cellular mechanisms behind clinical manifestations. Acta Physiol. 2024, 240, e14199. [Google Scholar] [CrossRef] [PubMed]
  191. Lau, J.M.; Jin, X.; Ren, J.; Avery, J.; DeBosch, B.J.; Treskov, I.; Lupu, T.S.; Kovacs, A.; Weinheimer, C.; Muslin, A.J. The 14-3-3τ phosphoserine-binding protein is required for cardiomyocyte survival. Mol. Cell. Biol. 2007, 27, 1455–1466. [Google Scholar] [CrossRef] [PubMed]
  192. Muslin, A.J. MAPK signalling in cardiovascular health and disease: Molecular mechanisms and therapeutic targets. Clin. Sci. 2008, 115, 203–218. [Google Scholar] [CrossRef]
  193. Feng, X.; Du, M.; Li, S.; Zhang, Y.; Ding, J.; Wang, J.; Wang, Y.; Liu, P. Hydroxysafflor yellow A regulates lymphangiogenesis and inflammation via the inhibition of PI3K on regulating AKT/mTOR and NF-κB pathway in macrophages to reduce atherosclerosis in ApoE-/- mice. Phytomedicine 2023, 112, 154684. [Google Scholar] [CrossRef] [PubMed]
  194. Fang, S.; Wan, X.; Zou, X.; Sun, S.; Hao, X.; Liang, C.; Zhang, Z.; Zhang, F.; Sun, B.; Li, H.; et al. Arsenic trioxide induces macrophage autophagy and atheroprotection by regulating ROS-dependent TFEB nuclear translocation and AKT/mTOR pathway. Cell Death Dis. 2021, 12, 88. [Google Scholar] [CrossRef]
  195. D’Onofrio, N.; Servillo, L.; Balestrieri, M.L. SIRT1 and SIRT6 Signaling Pathways in Cardiovascular Disease Protection. Antioxid. Redox Signal. 2018, 28, 711–732. [Google Scholar] [CrossRef] [PubMed]
  196. Te Riet, L.; van Esch, J.H.; Roks, A.J.; van den Meiracker, A.H.; Danser, A.H. Hypertension: Renin-angiotensin-aldosterone system alterations. Circ. Res. 2015, 116, 960–975. [Google Scholar] [CrossRef] [PubMed]
  197. Motte, S.; McEntee, K.; Naeije, R. Endothelin receptor antagonists. Pharmacol. Ther. 2006, 110, 386–414. [Google Scholar] [CrossRef]
  198. Valanti, E.K.; Dalakoura-Karagkouni, K.; Fotakis, P.; Vafiadaki, E.; Mantzoros, C.S.; Chroni, A.; Zannis, V.; Kardassis, D.; Sanoudou, D. Reconstituted HDL-apoE3 promotes endothelial cell migration through ID1 and its downstream kinases ERK1/2, AKT and p38 MAPK. Metabolism 2022, 127, 154954. [Google Scholar] [CrossRef] [PubMed]
  199. Zheng, Y.; Li, Y.; Ran, X.; Wang, D.; Zheng, X.; Zhang, M.; Yu, B.; Sun, Y.; Wu, J. Mettl14 mediates the inflammatory response of macrophages in atherosclerosis through the NF-κB/IL-6 signaling pathway. Cell. Mol. Life Sci. 2022, 79, 311. [Google Scholar] [CrossRef]
  200. Li, X.; Liu, R.; Liu, W.; Liu, X.; Fan, Z.; Cui, J.; Wu, Y.; Yin, H.; Lin, Q. Panax quinquefolium L. and Salvia miltiorrhiza Bunge. Enhances Angiogenesis by Regulating the miR-155-5p/HIF-1α/VEGF Axis in Acute Myocardial Infarction. Drug Des. Dev. Ther. 2023, 17, 3249–3267. [Google Scholar] [CrossRef] [PubMed]
  201. Negoro, S.; Kunisada, K.; Tone, E.; Funamoto, M.; Oh, H.; Kishimoto, T.; Yamauchi-Takihara, K. Activation of JAK/STAT pathway transduces cytoprotective signal in rat acute myocardial infarction. Cardiovasc. Res. 2000, 47, 797–805. [Google Scholar] [CrossRef] [PubMed]
  202. Yin, H.; Guo, X.; Chen, Y.; Zeng, Y.; Mo, X.; Hong, S.; He, H.; Li, J.; Steinmetz, R.; Liu, Q. TAB2 deficiency induces dilated cardiomyopathy by promoting RIPK1-dependent apoptosis and necroptosis. J. Clin. Investig. 2022, 132, e152297. [Google Scholar] [CrossRef] [PubMed]
  203. Shahzadi, S.K.; Marzook, H.; Qaisar, R.; Ahmad, F. Nicotinamide riboside kinase-2 inhibits JNK pathway and limits dilated cardiomyopathy in mice with chronic pressure overload. Clin. Sci. 2022, 136, 181–196. [Google Scholar] [CrossRef]
  204. Khalilimeybodi, A.; Saucerman, J.J.; Rangamani, P. Modeling cardiomyocyte signaling and metabolism predicts genotype-to-phenotype mechanisms in hypertrophic cardiomyopathy. Comput. Biol. Med. 2024, 175, 108499. [Google Scholar] [CrossRef] [PubMed]
  205. El Chami, H.; Hassoun, P.M. Inflammatory mechanisms in the pathogenesis of pulmonary arterial hypertension. Compr. Physiol. 2011, 1, 1929–1941. [Google Scholar] [CrossRef] [PubMed]
  206. Yang, Y.; Shao, M.; Yao, J.; Yang, S.; Cheng, W.; Ma, L.; Li, W.; Cao, J.; Zhang, Y.; Hu, Y.; et al. Neocryptotanshinone protects against myocardial ischemia-reperfusion injury by promoting autolysosome degradation of protein aggregates via the ERK1/2-Nrf2-LAMP2 pathway. Phytomedicine 2023, 110, 154625. [Google Scholar] [CrossRef]
  207. Xu, M.; Li, X.; Song, L. Baicalin regulates macrophages polarization and alleviates myocardial ischaemia/reperfusion injury via inhibiting JAK/STAT pathway. Pharm. Biol. 2020, 58, 655–663. [Google Scholar] [CrossRef]
  208. Slonková, V.; Slonková, V., Jr.; Vašků, A.; Vašků, V. Genetic predisposition for chronic venous insufficiency in several genes for matrix metalloproteinases (MMP-2, MMP-9, MMP-12) and their inhibitor TIMP-2. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 1746–1752. [Google Scholar] [CrossRef]
  209. Bharath, V.; Kahn, S.R.; Lazo-Langner, A. Genetic polymorphisms of vein wall remodeling in chronic venous disease: A narrative and systematic review. Blood 2014, 124, 1242–1250. [Google Scholar] [CrossRef]
  210. Swamy, S.; Ueland, T.; Hansen, J.B.; Snir, O.; Brækkan, S.K. Plasma levels of P-selectin and future risk of incident venous thromboembolism. J. Thromb. Haemost. 2023, 21, 2451–2460. [Google Scholar] [CrossRef] [PubMed]
  211. Patrono, C. The Multifaceted Clinical Readouts of Platelet Inhibition by Low-Dose Aspirin. J. Am. Coll. Cardiol. 2015, 66, 74–85. [Google Scholar] [CrossRef]
  212. Ji, E.; Lee, S. Antibody-Based Therapeutics for Atherosclerosis and Cardiovascular Diseases. Int. J. Mol. Sci. 2021, 22, 5770. [Google Scholar] [CrossRef] [PubMed]
  213. Gremese, E.; Alivernini, S.; Ferraccioli, E.S.; Ferraccioli, G. Checkpoint inhibitors (CPI) and autoimmune chronic inflammatory diseases (ACIDs): Tolerance and loss of tolerance in the occurrence of immuno-rheumatologic manifestations. Clin. Immunol. 2020, 214, 108395. [Google Scholar] [CrossRef]
  214. Sperry, B.W.; Zein, R.E.; Fendler, T.J.; Sauer, A.J.; Khumri, T.M.; Magalski, A.; Austin, B.A.; Safley, D.; Kao, A.C. Stabilization of Rapidly Progressive Cardiac Allograft Vasculopathy Using mTOR Inhibition After Heart Transplantation. J. Card. Fail. 2024, 30, 613–617. [Google Scholar] [CrossRef] [PubMed]
  215. Nattel, S.; Heijman, J.; Zhou, L.; Dobrev, D. Molecular Basis of Atrial Fibrillation Pathophysiology and Therapy: A Translational Perspective. Circ. Res. 2020, 127, 51–72. [Google Scholar] [CrossRef]
  216. Trac, D.; Maxwell, J.T.; Brown, M.E.; Xu, C.; Davis, M.E. Aggregation of Child Cardiac Progenitor Cells Into Spheres Activates Notch Signaling and Improves Treatment of Right Ventricular Heart Failure. Circ. Res. 2019, 124, 526–538. [Google Scholar] [CrossRef]
  217. Qian, J.F.; Liang, S.Q.; Wang, Q.Y.; Xu, J.C.; Luo, W.; Huang, W.J.; Wu, G.J.; Liang, G. Isoproterenol induces MD2 activation by β-AR-cAMP-PKA-ROS signalling axis in cardiomyocytes and macrophages drives inflammatory heart failure. Acta Pharmacol. Sin. 2024, 45, 531–544. [Google Scholar] [CrossRef] [PubMed]
  218. Higashikuni, Y.; Liu, W.; Numata, G.; Tanaka, K.; Fukuda, D.; Tanaka, Y.; Hirata, Y.; Imamura, T.; Takimoto, E.; Komuro, I.; et al. NLRP3 Inflammasome Activation Through Heart-Brain Interaction Initiates Cardiac Inflammation and Hypertrophy During Pressure Overload. Circulation 2023, 147, 338–355. [Google Scholar] [CrossRef] [PubMed]
  219. Oka, T.; Hikoso, S.; Yamaguchi, O.; Taneike, M.; Takeda, T.; Tamai, T.; Oyabu, J.; Murakawa, T.; Nakayama, H.; Nishida, K.; et al. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature 2012, 485, 251–255. [Google Scholar] [CrossRef] [PubMed]
  220. Aletaha, D.; Kerschbaumer, A.; Kastrati, K.; Dejaco, C.; Dougados, M.; McInnes, I.B.; Sattar, N.; Stamm, T.A.; Takeuchi, T.; Trauner, M.; et al. Consensus statement on blocking interleukin-6 receptor and interleukin-6 in inflammatory conditions: An update. Ann. Rheum. Dis. 2023, 82, 773–787. [Google Scholar] [CrossRef]
  221. Kumar, R.K.; Antunes, M.J.; Beaton, A.; Mirabel, M.; Nkomo, V.T.; Okello, E.; Regmi, P.R.; Reményi, B.; Sliwa-Hähnle, K.; Zühlke, L.J.; et al. Contemporary Diagnosis and Management of Rheumatic Heart Disease: Implications for Closing the Gap: A Scientific Statement from the American Heart Association. Circulation 2020, 142, e337–e357. [Google Scholar] [CrossRef]
  222. Kruithof, B.P.T.; Paardekooper, L.; Hiemstra, Y.L.; Goumans, M.J.; Palmen, M.; Delgado, V.; Klautz, R.J.M.; Ajmone Marsan, N. Stress-induced remodelling of the mitral valve: A model for leaflet thickening and superimposed tissue formation in mitral valve disease. Cardiovasc. Res. 2020, 116, 931–943. [Google Scholar] [CrossRef] [PubMed]
  223. Hagler, M.A.; Hadley, T.M.; Zhang, H.; Mehra, K.; Roos, C.M.; Schaff, H.V.; Suri, R.M.; Miller, J.D. TGF-β signalling and reactive oxygen species drive fibrosis and matrix remodelling in myxomatous mitral valves. Cardiovasc. Res. 2013, 99, 175–184. [Google Scholar] [CrossRef] [PubMed]
  224. Wang, L.; Li, L.; Zhao, D.; Yuan, H.; Zhang, H.; Chen, J.; Pang, D.; Lu, Y.; Ouyang, H. MYH7 R453C induced cardiac remodelling via activating TGF-β/Smad2/3, ERK1/2 and Nox4/ROS/NF-κB signalling pathways. Open Biol. 2024, 14, 230427. [Google Scholar] [CrossRef]
  225. Robinson, P.; Liu, X.; Sparrow, A.; Patel, S.; Zhang, Y.H.; Casadei, B.; Watkins, H.; Redwood, C. Hypertrophic cardiomyopathy mutations increase myofilament Ca(2+) buffering, alter intracellular Ca2+ handling, and stimulate Ca2+-dependent signaling. J. Biol. Chem. 2018, 293, 10487–10499. [Google Scholar] [CrossRef] [PubMed]
  226. Moncla, L.M.; Briend, M.; Bossé, Y.; Mathieu, P. Calcific aortic valve disease: Mechanisms, prevention and treatment. Nat. Rev. Cardiol. 2023, 20, 546–559. [Google Scholar] [CrossRef]
  227. Miao, Y.; Tian, L.; Martin, M.; Paige, S.L.; Galdos, F.X.; Li, J.; Klein, A.; Zhang, H.; Ma, N.; Wei, Y.; et al. Intrinsic Endocardial Defects Contribute to Hypoplastic Left Heart Syndrome. Cell Stem Cell 2020, 27, 574–589.e8. [Google Scholar] [CrossRef]
  228. Yoshimura, T.; Hisatomi, A.; Kajihara, S.; Yasutake, T.; Ogawa, Y.; Mizuta, T.; Ozaki, I.; Utsunomiyai, T.; Yamamoto, K. The relationship between insulin resistance and polymorphisms of the endothelial nitric oxide synthase gene in patients with coronary artery disease. J. Atheroscler. Thromb. 2003, 10, 43–47. [Google Scholar] [CrossRef] [PubMed]
  229. Motloch, L.J.; Cacheux, M.; Ishikawa, K.; Xie, C.; Hu, J.; Aguero, J.; Fish, K.M.; Hajjar, R.J.; Akar, F.G. Primary Effect of SERCA 2a Gene Transfer on Conduction Reserve in Chronic Myocardial Infarction. J. Am. Heart Assoc. 2018, 7, e009598. [Google Scholar] [CrossRef]
  230. Hu, J.; Liu, T.; Fu, F.; Cui, Z.; Lai, Q.; Zhang, Y.; Yu, B.; Liu, F.; Kou, J.; Li, F. Omentin1 ameliorates myocardial ischemia-induced heart failure via SIRT3/FOXO3a-dependent mitochondrial dynamical homeostasis and mitophagy. J. Transl. Med. 2022, 20, 447. [Google Scholar] [CrossRef]
  231. Kundu, S.; Gairola, S.; Verma, S.; Mugale, M.N.; Sahu, B.D. Chronic kidney disease activates the HDAC6-inflammatory axis in the heart and contributes to myocardial remodeling in mice: Inhibition of HDAC6 alleviates chronic kidney disease-induced myocardial remodeling. Basic Res. Cardiol. 2024, 119, 831–852. [Google Scholar] [CrossRef] [PubMed]
  232. Yuan, S.; Zheng, J.S.; Mason, A.M.; Burgess, S.; Larsson, S.C. Genetically predicted circulating vitamin C in relation to cardiovascular disease. Eur. J. Prev. Cardiol. 2022, 28, 1829–1837. [Google Scholar] [CrossRef] [PubMed]
  233. Violi, F.; Nocella, C.; Loffredo, L.; Carnevale, R.; Pignatelli, P. Interventional study with vitamin E in cardiovascular disease and meta-analysis. Free Radic. Biol. Med. 2022, 178, 26–41. [Google Scholar] [CrossRef] [PubMed]
  234. Ghrayeb, A.; Finney, A.C.; Agranovich, B.; Peled, D.; Anand, S.K.; McKinney, M.P.; Sarji, M.; Yang, D.; Weissman, N.; Drucker, S.; et al. Serine synthesis via reversed SHMT2 activity drives glycine depletion and acetaminophen hepatotoxicity in MASLD. Cell Metab. 2024, 36, 116–129.e7. [Google Scholar] [CrossRef]
  235. Wang, Y.; Zheng, Y.; Qi, B.; Liu, Y.; Cheng, X.; Feng, J.; Gao, W.; Li, T. α-Lipoic acid alleviates myocardial injury and induces M2b macrophage polarization after myocardial infarction via HMGB1/NF-kB signaling pathway. Int. Immunopharmacol. 2023, 121, 110435. [Google Scholar] [CrossRef] [PubMed]
  236. Gutierrez-Mariscal, F.M.; de la Cruz-Ares, S.; Torres-Peña, J.D.; Alcalá-Diaz, J.F.; Yubero-Serrano, E.M.; López-Miranda, J. Coenzyme Q(10) and Cardiovascular Diseases. Antioxidants 2021, 10, 906. [Google Scholar] [CrossRef]
  237. Dyck, G.J.B.; Raj, P.; Zieroth, S.; Dyck, J.R.B.; Ezekowitz, J.A. The Effects of Resveratrol in Patients with Cardiovascular Disease and Heart Failure: A Narrative Review. Int. J. Mol. Sci. 2019, 20, 904. [Google Scholar] [CrossRef]
  238. Martinez de Lizarrondo, S.; Gakuba, C.; Herbig, B.A.; Repessé, Y.; Ali, C.; Denis, C.V.; Lenting, P.J.; Touzé, E.; Diamond, S.L.; Vivien, D.; et al. Potent Thrombolytic Effect of N-Acetylcysteine on Arterial Thrombi. Circulation 2017, 136, 646–660. [Google Scholar] [CrossRef]
  239. Ciaraldi, T.P.; Boeder, S.C.; Mudaliar, S.R.; Giovannetti, E.R.; Henry, R.R.; Pettus, J.H. Astaxanthin, a natural antioxidant, lowers cholesterol and markers of cardiovascular risk in individuals with prediabetes and dyslipidaemia. Diabetes Obes. Metab. 2023, 25, 1985–1994. [Google Scholar] [CrossRef]
  240. Ferenczyova, K.; Kalocayova, B.; Bartekova, M. Potential Implications of Quercetin and its Derivatives in Cardioprotection. Int. J. Mol. Sci. 2020, 21, 1585. [Google Scholar] [CrossRef] [PubMed]
  241. Guo, J.; Li, K.; Lin, Y.; Liu, Y. Protective effects and molecular mechanisms of tea polyphenols on cardiovascular diseases. Front. Nutr. 2023, 10, 1202378. [Google Scholar] [CrossRef] [PubMed]
  242. Zhang, M.Y.; Dugbartey, G.J.; Juriasingani, S.; Sener, A. Hydrogen Sulfide Metabolite, Sodium Thiosulfate: Clinical Applications and Underlying Molecular Mechanisms. Int. J. Mol. Sci. 2021, 22, 6452. [Google Scholar] [CrossRef]
  243. Ballantyne, C.M.; Laufs, U.; Ray, K.K.; Leiter, L.A.; Bays, H.E.; Goldberg, A.C.; Stroes, E.S.; MacDougall, D.; Zhao, X.; Catapano, A.L. Bempedoic acid plus ezetimibe fixed-dose combination in patients with hypercholesterolemia and high CVD risk treated with maximally tolerated statin therapy. Eur. J. Prev. Cardiol. 2020, 27, 593–603. [Google Scholar] [CrossRef] [PubMed]
  244. de la Guía-Galipienso, F.; Martínez-Ferran, M.; Vallecillo, N.; Lavie, C.J.; Sanchis-Gomar, F.; Pareja-Galeano, H. Vitamin D and cardiovascular health. Clin. Nutr. 2021, 40, 2946–2957. [Google Scholar] [CrossRef] [PubMed]
  245. Clark, M.; Centner, A.M.; Ukhanov, V.; Nagpal, R.; Salazar, G. Gallic acid ameliorates atherosclerosis and vascular senescence and remodels the microbiome in a sex-dependent manner in ApoE(-/-) mice. J. Nutr. Biochem. 2022, 110, 109132. [Google Scholar] [CrossRef] [PubMed]
  246. Ahmed, H.H.; Taha, F.M.; Omar, H.S.; Elwi, H.M.; Abdelnasser, M. Hydrogen sulfide modulates SIRT1 and suppresses oxidative stress in diabetic nephropathy. Mol. Cell. Biochem. 2019, 457, 1–9. [Google Scholar] [CrossRef]
  247. El Hadri, K.; Smith, R.; Duplus, E.; El Amri, C. Inflammation, Oxidative Stress, Senescence in Atherosclerosis: Thioredoxine-1 as an Emerging Therapeutic Target. Int. J. Mol. Sci. 2021, 23, 77. [Google Scholar] [CrossRef]
  248. Liu, B.; Chen, L.; Gao, M.; Dai, M.; Zheng, Y.; Qu, L.; Zhang, J.; Gong, G. A comparative study of the efficiency of mitochondria-targeted antioxidants MitoTEMPO and SKQ1 under oxidative stress. Free Radic. Biol. Med. 2024, 224, 117–129. [Google Scholar] [CrossRef]
  249. Chang, B.; Su, Y.; Li, T.; Zheng, Y.; Yang, R.; Lu, H.; Wang, H.; Ding, Y. Mito-TEMPO Ameliorates Sodium Palmitate Induced Ferroptosis in MIN6 Cells through PINK1/Parkin-Mediated Mitophagy. Biomed. Environ. Sci. 2024, 37, 1128–1141. [Google Scholar] [CrossRef]
  250. Zinovkina, L.A.; Makievskaya, C.I.; Galkin, I.I.; Zinovkin, R.A. Mitochondria-targeted Uncouplers Decrease Inflammatory Reactions in Endothelial Cells by Enhancing Methylation of the ICAM1 Gene Promoter. Curr. Mol. Pharmacol. 2024, 17, e150823219723. [Google Scholar] [CrossRef]
  251. Liu, S.Q.; Xie, S.Y.; Zhang, T.; Zhang, H.; Chen, M.Y.; Xing, Y.; Zhao, N.; Li, L.; Chen, S.; Wang, S.S.; et al. Impeding Nucleotide-Binding Oligomerization Domain-Like Receptor 3 Inflammasome Ameliorates Cardiac Remodeling and Dysfunction in Obesity-Associated Cardiomyopathy. J. Am. Heart Assoc. 2024, 13, e035234. [Google Scholar] [CrossRef] [PubMed]
  252. Song, J.; Sheng, J.; Lei, J.; Gan, W.; Yang, Y. Mitochondrial Targeted Antioxidant SKQ1 Ameliorates Acute Kidney Injury by Inhibiting Ferroptosis. Oxidative Med. Cell. Longev. 2022, 2022, 2223957. [Google Scholar] [CrossRef]
  253. Jabůrek, M.; Klöppel, E.; Průchová, P.; Mozheitova, O.; Tauber, J.; Engstová, H.; Ježek, P. Mitochondria to plasma membrane redox signaling is essential for fatty acid β-oxidation-driven insulin secretion. Redox Biol. 2024, 75, 103283. [Google Scholar] [CrossRef]
  254. Park, C.; Cha, H.J.; Kim, M.Y.; Bang, E.; Moon, S.K.; Yun, S.J.; Kim, W.J.; Noh, J.S.; Kim, G.Y.; Cho, S.; et al. Phloroglucinol Attenuates DNA Damage and Apoptosis Induced by Oxidative Stress in Human Retinal Pigment Epithelium ARPE-19 Cells by Blocking the Production of Mitochondrial ROS. Antioxidants 2022, 11, 2353. [Google Scholar] [CrossRef] [PubMed]
  255. Zheng, J.; Zhao, L.; Liu, Y.; Chen, M.; Guo, X.; Wang, J. N-acetylcysteine, a small molecule scavenger of reactive oxygen species, alleviates cardiomyocyte damage by regulating OPA1-mediated mitochondrial quality control and apoptosis in response to oxidative stress. J. Thorac. Dis. 2024, 16, 5323–5336. [Google Scholar] [CrossRef]
  256. Quiles, J.M.; Pepin, M.E.; Sunny, S.; Shelar, S.B.; Challa, A.K.; Dalley, B.; Hoidal, J.R.; Pogwizd, S.M.; Wende, A.R.; Rajasekaran, N.S. Identification of Nrf2-responsive microRNA networks as putative mediators of myocardial reductive stress. Sci. Rep. 2021, 11, 11977. [Google Scholar] [CrossRef] [PubMed]
  257. Wang, L.; Yang, W.; Song, Y. Impairing Ferroptosis Through the PI3K/Akt/Nrf2 Pathway: The Way for Nerve Growth Factor to Mitigate Hypoxia-induced Cardiomyocyte Damage. Cell Biochem. Biophys. 2024, 1–11. [Google Scholar] [CrossRef]
  258. Dong, S.; Zhang, C.; Wang, Y.; Liu, S.; Yang, J.; Li, L.; Ma, Y.; Liu, J. The protective effect of rutin on sciatic nerve injury in acrylamide-exposed rats and its mechanisms. Food Chem. Toxicol. 2024, 195, 115106. [Google Scholar] [CrossRef] [PubMed]
  259. Asle-Rousta, M.; Peirovy, Y. Neuroprotective Effects of Thymol and p-Cymene in Immobilized Male rats through Alterations in Molecular, Biochemical, Histological, and Behavioral Parameters. Neurochem. Res. 2024, 50, 5. [Google Scholar] [CrossRef]
  260. Zheng, X.B.; Wang, C.; Zhang, M.; Yao, B.Q.; Wu, H.Y.; Hou, S.X. Exogenous H2S targeting PI3K/AKT/mTOR pathway alleviates chronic intermittent hypoxia-induced myocardial damage through inhibiting oxidative stress and enhancing autophagy. Sleep Breath. 2024, 29, 43. [Google Scholar] [CrossRef]
  261. Sahasrabudhe, S.A.; Terluk, M.R.; Kartha, R.V. N-acetylcysteine Pharmacology and Applications in Rare Diseases-Repurposing an Old Antioxidant. Antioxidants 2023, 12, 1316. [Google Scholar] [CrossRef]
  262. Song, G.; Wang, J.; Liu, J.; Ruan, Y. Dimethyl fumarate ameliorates erectile dysfunction in bilateral cavernous nerve injury rats by inhibiting oxidative stress and NLRP3 inflammasome-mediated pyroptosis of nerve via activation of Nrf2/HO-1 signaling pathway. Redox Biol. 2023, 68, 102938. [Google Scholar] [CrossRef] [PubMed]
  263. Carlström, M.; Weitzberg, E.; Lundberg, J.O. Nitric Oxide Signaling and Regulation in the Cardiovascular System: Recent Advances. Pharmacol. Rev. 2024, 76, 1038–1062. [Google Scholar] [CrossRef]
  264. Tomar, A.; Sahoo, S.; Aathi, M.; Kuila, S.; Khan, M.A.; Ravi, G.R.R.; Jeyaraman, J.; Mehta, J.L.; Varughese, K.I.; Arockiasamy, A. Exploring the druggability of oxidized low-density lipoprotein (ox-LDL) receptor, LOX-1, a proatherogenic drug target involved in atherosclerosis. Biochem. Biophys. Res. Commun. 2022, 623, 59–65. [Google Scholar] [CrossRef] [PubMed]
  265. Singh, P.; Sun, J.; Cavalera, M.; Al-Sharify, D.; Matthes, F.; Barghouth, M.; Tengryd, C.; Dunér, P.; Persson, A.; Sundius, L.; et al. Dysregulation of MMP2-dependent TGF-ß2 activation impairs fibrous cap formation in type 2 diabetes-associated atherosclerosis. Nat. Commun. 2024, 15, 10464. [Google Scholar] [CrossRef]
  266. Zeng, L.; Zhang, X.; Huang, Z.; Song, S.; Li, M.; Wang, T.; Sun, A.; Ge, J. Ubiquitin proteasome system in cardiac fibrosis. J. Adv. Res. 2024; in press. [Google Scholar] [CrossRef] [PubMed]
  267. Haase, V.H.; Tanaka, T.; Koury, M.J. Hypoxia-inducible factor activators: A novel class of oral drugs for the treatment of anemia of chronic kidney disease. Hematol. Am. Soc. Hematol. Educ. Program. 2024, 2024, 409–418. [Google Scholar] [CrossRef] [PubMed]
  268. Ye, Q.; Zhang, M.; Li, S.; Liu, W.; Xu, C.; Li, Y.; Xie, R. Controlled Stimulus-Responsive Delivery Systems for Osteoarthritis Treatment. Int. J. Mol. Sci. 2024, 25, 11799. [Google Scholar] [CrossRef] [PubMed]
  269. Zhou, Q.; Zhang, Y.; Shi, W.; Lu, L.; Wei, J.; Wang, J.; Zhang, H.; Pu, Y.; Yin, L. Angiotensin II Induces Vascular Endothelial Dysfunction by Promoting Lipid Peroxidation-Mediated Ferroptosis via CD36. Biomolecules 2024, 14, 1456. [Google Scholar] [CrossRef] [PubMed]
  270. Tian, M.; Huang, X.; Li, M.; Lou, P.; Ma, H.; Jiang, X.; Zhou, Y.; Liu, Y. Ferroptosis in diabetic cardiomyopathy: From its mechanisms to therapeutic strategies. Front. Endocrinol. 2024, 15, 1421838. [Google Scholar] [CrossRef] [PubMed]
  271. Alsereidi, F.R.; Khashim, Z.; Marzook, H.; Al-Rawi, A.M.; Salomon, T.; Almansoori, M.K.; Madkour, M.M.; Hamam, A.M.; Ramadan, M.M.; Peterson, Q.P.; et al. Dapagliflozin mitigates cellular stress and inflammation through PI3K/AKT pathway modulation in cardiomyocytes, aortic endothelial cells, and stem cell-derived β cells. Cardiovasc. Diabetol. 2024, 23, 388. [Google Scholar] [CrossRef] [PubMed]
  272. Drakontaeidi, A.; Papanotas, I.; Pontiki, E. Multitarget Pharmacology of Sulfur-Nitrogen Heterocycles: Anticancer and Antioxidant Perspectives. Antioxidants 2024, 13, 898. [Google Scholar] [CrossRef] [PubMed]
  273. Wu, P.; Barros-Becker, F.; Ogelman, R.; Camci, E.D.; Linbo, T.H.; Simon, J.A.; Rubel, E.W.; Raible, D.W. Multiple mechanisms of aminoglycoside ototoxicity are distinguished by subcellular localization of action. Front. Neurol. 2024, 15, 1480435. [Google Scholar] [CrossRef]
  274. Lyamzaev, K.G.; Huan, H.; Panteleeva, A.A.; Simonyan, R.A.; Avetisyan, A.V.; Chernyak, B.V. Exogenous Iron Induces Mitochondrial Lipid Peroxidation, Lipofuscin Accumulation, and Ferroptosis in H9c2 Cardiomyocytes. Biomolecules 2024, 14, 730. [Google Scholar] [CrossRef] [PubMed]
  275. Zhang, W.; Shi, C.; Yao, Z.; Qian, S. Bardoxolone methyl attenuates doxorubicin-induced cardiotoxicity by inhibiting the TXNIP-NLRP3 pathway through Nrf2 activation. Environ. Toxicol. 2024, 39, 1936–1950. [Google Scholar] [CrossRef] [PubMed]
  276. Shimamura, M.; Nakagami, H.; Sanada, F.; Morishita, R. Progress of Gene Therapy in Cardiovascular Disease. Hypertension 2020, 76, 1038–1044. [Google Scholar] [CrossRef] [PubMed]
  277. Nie, J.; Han, Y.; Jin, Z.; Hang, W.; Shu, H.; Wen, Z.; Ni, L.; Wang, D.W. Homology-directed repair of an MYBPC3 gene mutation in a rat model of hypertrophic cardiomyopathy. Gene Ther. 2023, 30, 520–527. [Google Scholar] [CrossRef]
  278. Argiro, A.; Bui, Q.; Hong, K.N.; Ammirati, E.; Olivotto, I.; Adler, E. Applications of Gene Therapy in Cardiomyopathies. JACC Heart Fail. 2024, 12, 248–260. [Google Scholar] [CrossRef] [PubMed]
  279. Swerdlow, D.I.; Rider, D.A.; Yavari, A.; Wikström Lindholm, M.; Campion, G.V.; Nissen, S.E. Treatment and prevention of lipoprotein(a)-mediated cardiovascular disease: The emerging potential of RNA interference therapeutics. Cardiovasc. Res. 2022, 118, 1218–1231. [Google Scholar] [CrossRef] [PubMed]
  280. Nemet, I.; Saha, P.P.; Gupta, N.; Zhu, W.; Romano, K.A.; Skye, S.M.; Cajka, T.; Mohan, M.L.; Li, L.; Wu, Y.; et al. A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors. Cell 2020, 180, 862–877.e22. [Google Scholar] [CrossRef]
  281. Davis, J.R.; Banskota, S.; Levy, J.M.; Newby, G.A.; Wang, X.; Anzalone, A.V.; Nelson, A.T.; Chen, P.J.; Hennes, A.D.; An, M.; et al. Efficient prime editing in mouse brain, liver and heart with dual AAVs. Nat. Biotechnol. 2024, 42, 253–264. [Google Scholar] [CrossRef] [PubMed]
  282. Dzau, V.J.; Hodgkinson, C.P. RNA Therapeutics for the Cardiovascular System. Circulation 2024, 149, 707–716. [Google Scholar] [CrossRef]
  283. Menasché, P.; Vanneaux, V.; Fabreguettes, J.R.; Bel, A.; Tosca, L.; Garcia, S.; Bellamy, V.; Farouz, Y.; Pouly, J.; Damour, O.; et al. Towards a clinical use of human embryonic stem cell-derived cardiac progenitors: A translational experience. Eur. Heart J. 2015, 36, 743–750. [Google Scholar] [CrossRef]
  284. Bagno, L.; Hatzistergos, K.E.; Balkan, W.; Hare, J.M. Mesenchymal Stem Cell-Based Therapy for Cardiovascular Disease: Progress and Challenges. Mol. Ther. 2018, 26, 1610–1623. [Google Scholar] [CrossRef] [PubMed]
  285. Wang, Y.; Xu, F.; Ma, J.; Shi, J.; Chen, S.; Liu, Z.; Liu, J. Effect of stem cell transplantation on patients with ischemic heart failure: A systematic review and meta-analysis of randomized controlled trials. Stem Cell Res. Ther. 2019, 10, 125. [Google Scholar] [CrossRef] [PubMed]
  286. Han, W.; Xiong, W.; Sun, W.; Liu, W.; Zhang, Y.; Li, C.; Gu, N.; Shen, Y.; Qiu, Z.; Li, C. YTHDC1 Mitigates Apoptosis in Bone Marrow Mesenchymal Stem Cells by Inhibiting NfƙBiα and Augmenting Cardiac Function Following Myocardial Infarction. Cell Transplant. 2024, 33, 9636897241290910. [Google Scholar] [CrossRef]
  287. Mu, L.; Dong, R.; Li, C.; Chen, J.; Huang, Y.; Li, T.; Guo, B. ROS responsive conductive microspheres loaded with salvianolic acid B as adipose derived stem cell carriers for acute myocardial infarction treatment. Biomaterials 2025, 314, 122849. [Google Scholar] [CrossRef]
  288. Javaid, A.; Sharma, K.K.; Ka, A.; Verma, A.; Mudavath, S.L. Therapeutic Potential of Molsidomine-Loaded Liquid Crystal Nanoparticles for the Treatment and Management of Niacin-Induced Varicose Veins: In Vitro and In Vivo Studies. ACS Appl. Bio Mater. 2024, 7, 7306–7323. [Google Scholar] [CrossRef]
  289. Sahoo, S.; Adamiak, M.; Mathiyalagan, P.; Kenneweg, F.; Kafert-Kasting, S.; Thum, T. Therapeutic and Diagnostic Translation of Extracellular Vesicles in Cardiovascular Diseases: Roadmap to the Clinic. Circulation 2021, 143, 1426–1449. [Google Scholar] [CrossRef] [PubMed]
  290. Vicencio, J.M.; Yellon, D.M.; Sivaraman, V.; Das, D.; Boi-Doku, C.; Arjun, S.; Zheng, Y.; Riquelme, J.A.; Kearney, J.; Sharma, V.; et al. Plasma exosomes protect the myocardium from ischemia-reperfusion injury. J. Am. Coll. Cardiol. 2015, 65, 1525–1536. [Google Scholar] [CrossRef] [PubMed]
  291. Loyer, X.; Zlatanova, I.; Devue, C.; Yin, M.; Howangyin, K.Y.; Klaihmon, P.; Guerin, C.L.; Kheloufi, M.; Vilar, J.; Zannis, K.; et al. Intra-Cardiac Release of Extracellular Vesicles Shapes Inflammation Following Myocardial Infarction. Circ. Res. 2018, 123, 100–106. [Google Scholar] [CrossRef] [PubMed]
  292. Skeik, N.; Patel, D.C. A review of troponins in ischemic heart disease and other conditions. Int. J. Angiol. 2007, 16, 53–58. [Google Scholar] [CrossRef]
  293. Li, B.; Cao, Y.; Sun, M.; Feng, H. Expression, regulation, and function of exosome-derived miRNAs in cancer progression and therapy. FASEB J. 2021, 35, e21916. [Google Scholar] [CrossRef] [PubMed]
  294. Hashemi, M.; Moosavi, M.S.; Abed, H.M.; Dehghani, M.; Aalipour, M.; Heydari, E.A.; Behroozaghdam, M.; Entezari, M.; Salimimoghadam, S.; Gunduz, E.S.; et al. Long non-coding RNA (lncRNA) H19 in human cancer: From proliferation and metastasis to therapy. Pharmacol. Res. 2022, 184, 106418. [Google Scholar] [CrossRef] [PubMed]
  295. Lee, T.L.; Shen, W.C.; Chen, Y.C.; Lai, T.C.; Lin, S.R.; Lin, S.W.; Yu, I.S.; Yeh, Y.H.; Li, T.K.; Lee, I.T.; et al. Mir221- and Mir222-enriched adsc-exosomes mitigate PM exposure-exacerbated cardiac ischemia-reperfusion injury through the modulation of the BNIP3-MAP1LC3B-BBC3/PUMA pathway. Autophagy 2024, 20, 1–20. [Google Scholar] [CrossRef] [PubMed]
  296. Liu, J.; Wang, Z.; Lin, A.; Zhang, N. Exosomes from Hypoxic Pretreatment ADSCs Ameliorate Cardiac Damage Post-MI via Activated circ-Stt3b/miR-15a-5p/GPX4 Signaling and Decreased Ferroptosis. Cardiovasc. Toxicol. 2024, 24, 1215–1225. [Google Scholar] [CrossRef] [PubMed]
  297. Elhage, R.; Maret, A.; Pieraggi, M.T.; Thiers, J.C.; Arnal, J.F.; Bayard, F. Differential effects of interleukin-1 receptor antagonist and tumor necrosis factor binding protein on fatty-streak formation in apolipoprotein E-deficient mice. Circulation 1998, 97, 242–244. [Google Scholar] [CrossRef] [PubMed]
  298. Ridker, P.M.; Rane, M. Interleukin-6 Signaling and Anti-Interleukin-6 Therapeutics in Cardiovascular Disease. Circ. Res. 2021, 128, 1728–1746. [Google Scholar] [CrossRef] [PubMed]
  299. Grebe, A.; Hoss, F.; Latz, E. NLRP3 Inflammasome and the IL-1 Pathway in Atherosclerosis. Circ. Res. 2018, 122, 1722–1740. [Google Scholar] [CrossRef]
  300. Ding, Z.; Pothineni, N.V.K.; Goel, A.; Lüscher, T.F.; Mehta, J.L. PCSK9 and inflammation: Role of shear stress, pro-inflammatory cytokines, and LOX-1. Cardiovasc. Res. 2020, 116, 908–915. [Google Scholar] [CrossRef]
  301. Newman, C.B.; Tobert, J.A. Targeting PCSK9 with Antibodies and Gene Silencing to Reduce LDL Cholesterol. J. Clin. Endocrinol. Metab. 2023, 108, 784–790. [Google Scholar] [CrossRef]
  302. Dutka, M.; Zimmer, K.; Ćwiertnia, M.; Ilczak, T.; Bobiński, R. The role of PCSK9 in heart failure and other cardiovascular diseases-mechanisms of action beyond its effect on LDL cholesterol. Heart Fail. Rev. 2024, 29, 917–937. [Google Scholar] [CrossRef] [PubMed]
  303. Neuen, B.L.; Heerspink, H.J.L.; Vart, P.; Claggett, B.L.; Fletcher, R.A.; Arnott, C.; de Oliveira Costa, J.; Falster, M.O.; Pearson, S.A.; Mahaffey, K.W.; et al. Estimated Lifetime Cardiovascular, Kidney, and Mortality Benefits of Combination Treatment with SGLT2 Inhibitors, GLP-1 Receptor Agonists, and Nonsteroidal MRA Compared with Conventional Care in Patients with Type 2 Diabetes and Albuminuria. Circulation 2024, 149, 450–462. [Google Scholar] [CrossRef]
  304. Nauck, M.A.; D’Alessio, D.A. Tirzepatide, a dual GIP/GLP-1 receptor co-agonist for the treatment of type 2 diabetes with unmatched effectiveness regrading glycaemic control and body weight reduction. Cardiovasc. Diabetol. 2022, 21, 169. [Google Scholar] [CrossRef]
  305. Marx, N.; Husain, M.; Lehrke, M.; Verma, S.; Sattar, N. GLP-1 Receptor Agonists for the Reduction of Atherosclerotic Cardiovascular Risk in Patients with Type 2 Diabetes. Circulation 2022, 146, 1882–1894. [Google Scholar] [CrossRef]
  306. Vallon, V.; Verma, S. Effects of SGLT2 Inhibitors on Kidney and Cardiovascular Function. Annu. Rev. Physiol. 2021, 83, 503–528. [Google Scholar] [CrossRef] [PubMed]
  307. Brown, E.; Heerspink, H.J.L.; Cuthbertson, D.J.; Wilding, J.P.H. SGLT2 inhibitors and GLP-1 receptor agonists: Established and emerging indications. Lancet 2021, 398, 262–276. [Google Scholar] [CrossRef] [PubMed]
  308. Trepiccione, F.; Iervolino, A.; D’Acierno, M.; Siccardi, S.; Costanzo, V.; Sardella, D.; De La Motte, L.R.; D’Apolito, L.; Miele, A.; Perna, A.F.; et al. The SGLT2 inhibitor dapagliflozin improves kidney function in glycogen storage disease XI. Sci. Transl. Med. 2023, 15, eabn4214. [Google Scholar] [CrossRef]
Figure 1. Generation and roles of ROS. ROS are highly reactive molecules containing oxygen produced during cellular metabolism. Excessive ROS production can lead to oxidative stress, resulting in cellular damage. As depicted in the figure, intracellular ROS primarily originate from the electron transport chain (ETC), NADPH oxidase (NOX), xanthine oxidase (XO), cyclooxygenase (COX), nitric oxide synthase (NOS), and cytochrome P450 (CYP450). ROS can oxidize biomolecules, including lipids, proteins, and DNA, impairing cellular structure and function, triggering inflammatory responses, and promoting apoptosis, ultimately contributing to the pathogenesis of CVDs.
Figure 1. Generation and roles of ROS. ROS are highly reactive molecules containing oxygen produced during cellular metabolism. Excessive ROS production can lead to oxidative stress, resulting in cellular damage. As depicted in the figure, intracellular ROS primarily originate from the electron transport chain (ETC), NADPH oxidase (NOX), xanthine oxidase (XO), cyclooxygenase (COX), nitric oxide synthase (NOS), and cytochrome P450 (CYP450). ROS can oxidize biomolecules, including lipids, proteins, and DNA, impairing cellular structure and function, triggering inflammatory responses, and promoting apoptosis, ultimately contributing to the pathogenesis of CVDs.
Antioxidants 14 00038 g001
Figure 2. Pathophysiological processes in CVDs. This figure provides a visual representation of the characteristic features of oxidative stress at different stages of cardiovascular diseases and their potential underlying causes, demonstrating the close relationship between oxidative stress and disease progression.
Figure 2. Pathophysiological processes in CVDs. This figure provides a visual representation of the characteristic features of oxidative stress at different stages of cardiovascular diseases and their potential underlying causes, demonstrating the close relationship between oxidative stress and disease progression.
Antioxidants 14 00038 g002
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

Zhang, Z.; Guo, J. Deciphering Oxidative Stress in Cardiovascular Disease Progression: A Blueprint for Mechanistic Understanding and Therapeutic Innovation. Antioxidants 2025, 14, 38. https://doi.org/10.3390/antiox14010038

AMA Style

Zhang Z, Guo J. Deciphering Oxidative Stress in Cardiovascular Disease Progression: A Blueprint for Mechanistic Understanding and Therapeutic Innovation. Antioxidants. 2025; 14(1):38. https://doi.org/10.3390/antiox14010038

Chicago/Turabian Style

Zhang, Zhaoshan, and Jiawei Guo. 2025. "Deciphering Oxidative Stress in Cardiovascular Disease Progression: A Blueprint for Mechanistic Understanding and Therapeutic Innovation" Antioxidants 14, no. 1: 38. https://doi.org/10.3390/antiox14010038

APA Style

Zhang, Z., & Guo, J. (2025). Deciphering Oxidative Stress in Cardiovascular Disease Progression: A Blueprint for Mechanistic Understanding and Therapeutic Innovation. Antioxidants, 14(1), 38. https://doi.org/10.3390/antiox14010038

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