Next Article in Journal
Pulmonary Function Among COVID-19 Patients in Home Isolation Program
Previous Article in Journal
Emergency Laparoscopic Cholecystectomy Pathway Reduces Elective Waiting Times and Preoperative Admissions: A Prospective Propensity-Matched Cohort Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiological Mechanisms of Diabetes-Induced Macrovascular and Microvascular Complications: The Role of Oxidative Stress

Department of Physiology, Wayne State University, Detroit, MI 48202, USA
Med. Sci. 2025, 13(3), 87; https://doi.org/10.3390/medsci13030087
Submission received: 11 May 2025 / Revised: 29 June 2025 / Accepted: 1 July 2025 / Published: 2 July 2025

Abstract

Diabetic vascular diseases have emerged as a significant concern in medical research due to their considerable impact on human health. The challenge lies in the insufficient understanding of the intricate pathophysiological mechanisms associated with different forms of diabetic vascular diseases, which hampers our ability to identify effective treatment targets. Addressing this knowledge gap is essential for developing successful interventions. Unraveling the molecular pathways through which diabetes leads to microvascular and macrovascular complications in vital organs such as the heart, brain, kidneys, retina, and extremities is crucial. Notably, oxidative stress resulting from hyperglycemia is the key factor in initiating these complications. This review aims to elucidate the specific molecular mechanisms by which oxidative stress drives microvascular and macrovascular diseases and to highlight promising therapeutic advancements that offer hope for effective treatment solutions.

1. Introduction

Diabetes mellitus (DM) is a chronic endocrine and metabolic condition characterized by either insulin deficiency, insulin resistance, or a combination of both [1]. This disorder leads to elevated blood sugar levels, known as hyperglycemia, and can result in various diabetic vascular diseases over time. DM and its complications are rising at an alarming rate and have become a global threat to human lives, which requires urgent attention. In 2019, the global prevalence of diabetes was estimated at 9.3%, affecting 463 million people. This figure is projected to increase to 10.2% (578 million) by 2030 and 10.9% (700 million) by 2045 [2]. The increased prevalence of diabetes is mainly due to several factors, including an aging population, socio-economic growth, unhealthy dietary habits, and a sedentary lifestyle [3]. More than 90% of diabetes cases represent type 2 DM (T2DM) [4]. Diabetic complications affect multiple organ systems, including the heart, brain, kidneys, eyes, and peripheral blood vessels [5]. Diabetes poses a significant health burden primarily because of its vascular complications. These complications can be categorized into two groups (1) macrovascular complications, including coronary artery disease (CAD), cerebrovascular disease, and peripheral artery disease, and (2) microvascular complications, including diabetic kidney disease (DKD), diabetic retinopathy (DR), diabetic neuropathy, and cardiomyopathy [6].
The coexistence of multiple vascular complications results in a considerably worse prognosis [7]. Recent research into the complexities of diabetic complications has significantly enhanced our understanding of the disease’s underlying mechanisms. Yet, the growing fragmentation of medical specialties often leads to a narrow focus on isolated problems, risking a comprehensive view of the issue. It is crucial to adopt a holistic approach that investigates the interconnected nature of diabetic complications across various systems and vascular conditions. By doing so, we can better address the challenges posed by diabetes and improve outcomes for millions affected by this devastating disease.
Oxidative stress plays a significant role in diabetes-induced pathophysiology of vascular complications. In recent years, this condition has received considerable attention due to its profound implications for human health, especially its connection to diabetes.
Elevated reactive oxygen species (ROS) are the hallmark of oxidative stress in DM. At physiological concentrations, ROS serve as important second messengers that transmit intracellular signals involved in various biological processes. However, when ROS production becomes excessive and exceeds the capacity of antioxidant defense systems or when antioxidant enzymes are impaired, oxidative stress occurs [8,9]. ROS, which includes free radicals and other reactive molecules, are natural byproducts of normal cellular metabolism. In diabetes, chronic high blood sugar levels and mitochondrial dysfunction lead to increased production of ROS, further worsening oxidative stress. The excessive accumulation of ROS can damage various cellular components, including proteins, lipids, and DNA, leading to cellular dysfunction and disruption of normal physiological processes [10]. This damage may trigger inflammation and impair the function of essential cellular structures, ultimately contributing to the onset and progression of several diseases, including cardiovascular disease (CVD). Although oxidative stress is a secondary incidence of hyperglycemia in diabetes, it exacerbates the progression of diabetes and its complications [11,12,13]. Oxidative stress has been shown to impair two major mechanisms that fail during diabetes: insulin production by the pancreatic β-cells and tissue-specific insulin signaling [14]. Oxidative stress in diabetes plays a dual role. It not only contributes to the onset of diabetes but also deteriorates the condition and its associated complications. Experimental evidence highlights the involvement of ROS in the impaired function of β-cells, which is caused by autoimmune reactions, cytokines, and inflammatory proteins in type 1 diabetes [15]. Additionally, hyperglycemia has been observed to promote oxidative stress through the generation of free radicals and the suppression of antioxidant defense systems [16]. In chronic hyperglycemia, the production of ROS is sustained, leading to a significant reduction in antioxidant enzymes and non-enzymatic antioxidants across various tissues [17]. This exacerbates oxidative stress, which explains why individuals with diabetes tend to have higher levels of oxidative stress compared to healthy individuals. This review discussed the sources, and the molecular mechanisms involved in the initiation of oxidative stress in diabetes, as well as the role of oxidative stress in major macro and microvascular complications in diabetes, including atherosclerosis, DR and DKD.

2. Sources of ROS in DM

2.1. Mitochondria

Superoxide anion is mainly produced in the mitochondria, particularly at complexes I and III of the electron transport chain (ETC) [18]. This production occurs when electrons leak from the ETC, resulting in the partial reduction of molecular oxygen into superoxide instead of water [19]. Mitochondrial ROS (mtROS) can also disrupt the function of ETC complexes by oxidizing their iron-sulfur centers, which further elevates ROS production [20,21]. At physiological levels, ROS serve critical roles as signaling molecules that regulate important processes, including cell growth, differentiation, senescence, apoptosis, and autophagy [8,22]. However, when present at pathological levels, ROS can cause cellular dysfunction, cell death, and organ failure [23]. Numerous studies have highlighted the strong link between ROS generation in mitochondria, endothelial dysfunction, and heightened cardiovascular risk [24,25]. The production of ROS promotes the accumulation of leukocytes in vessel walls through processes like rolling, adhesion, and transmigration across the endothelial barrier [26]. In diabetic conditions, blood polymorphonuclear leukocytes (PMNs) demonstrate increased mtROS production [27], reduced oxygen consumption [28], and decreased rolling velocity [29], which correlates with an enhanced interaction with endothelial cells. This interaction is one of the early steps leading to endothelial damage [30]. Excessive mtROS production from the ETC is linked to the hyperglycemic damage seen in diabetes [16,31]. In contrast, inhibiting complex II in bovine aortic endothelial cells (BAECs) has been shown to block glucose-induced activation of protein kinase C (PKC), activation of NFĸB, and formation of AGEs [32]. While the ETC complexes are the primary sources of mtROS, other proteins can also induce mitochondrial oxidative stress, including p66Shc, a 66 kDa adaptor protein that belongs to the ShcA protein family [33]. It transmits pro-apoptotic signals by generating mtROS and can contribute to the regulation of lifespan in mammals [34]. A portion of p66Shc is located in the mitochondrial intermembrane space [35], where it acts as a redox enzyme, generating ROS through the oxidation of cytochrome c [36]. This process also results in the partial reduction of molecular oxygen to superoxide. Overexpression of p66Shc leads to endothelial dysfunction, characterized by increased ROS production, elevated levels of E-selectin, and enhanced leukocyte transmigration across the human umbilical vein endothelial cell (HUVEC) monolayer [37]. Additionally, elevated p66Shc levels have been implicated in circulating leukocytes from diabetic patients, as well as in the aorta and renal cortex of diabetes models [38,39]. The p66Shc protein is also implicated in β-cells dysfunction and insulin resistance caused by saturated fatty acids and excessive body fat [40]. Silencing p66Shc gene has been shown to reduce ROS production, enhanced endothelium-dependent vasorelaxation, and diminished apoptosis by restricting cytochrome c release, caspase 3 activity, and poly (ADP-ribose) polymerase cleavage [41]. In contrast, knocking down p66Shc in endothelial cells from diabetic mice reduces ROS production, prevents the formation of the AGE precursor methylglyoxal [42], and protects against ROS-mediated atherosclerosis, diabetes-induced endothelial dysfunction, and glomerulopathy [39]. Therefore, targeting p66Shc may be a promising approach to reduce mtROS production and subsequent EC dysfunction.

2.2. NADPH Oxidase

The NADPH oxidases (NOX), consisting of seven membrane-bound enzyme complexes, reduce molecular oxygen to superoxide using NADPH as the electron donor [43,44]. Their primary role is to generate ROS, contributing to the ROS burst and bacterial elimination in phagocytes [32,45,46]. Among the isoforms, NOX4 is most prevalent in endothelial cells compared to NOX1, NOX2, and NOX5 [47]. In non-phagocytic cells under normal physiological conditions, moderate levels of ROS are produced by the catalysis of NOX enzymes, essential for typical redox signaling [48]. Specifically, NOX2 and NOX4 promote endothelial cell proliferation and survival by ROS-mediated activation of p38 MAPK and Akt pathways [49,50]. The significant decrease in ROS-mediated tube formation and wound healing in human microvascular endothelial cells (HMECs) after siRNA-mediated NOX4 knockdown further underscores its crucial role in angiogenesis and endothelial cell proliferation [51]. Conversely, in pathological states, NOX4 might lead to a pro-thrombogenic endothelial phenotype. A study demonstrated that siRNA-mediated NOX4 knockdown reduced the endotoxin-induced expression of intercellular adhesion molecule-1 (ICAM-1), interleukin-8 (IL-8), and monocyte chemoattractant protein-1 (MCP-1) in human aortic endothelial cells (HAECs) [52]. Moreover, elevated NOX4 levels and subsequent augmentation of oxidative stress are associated with idiopathic pulmonary fibrosis [53,54] and the progression of diabetic nephropathy in both humans and animal models [55]. Nonetheless, NOX4 is implicated to have protective effects on the vasculature by preventing endothelial dysfunction in ischemic or inflammatory damage [56]. Furthermore, overexpressed endothelial NOX4 has been shown to improve vasodilation and lower blood pressure in transgenic mice through the generation of H2O2 [46,57], whereas NOX4 knockout mice displayed worsened angiotensin II-induced inflammation, endothelial dysfunction, and vascular remodeling [56]. Like NOX2 and NOX4 isoforms, NOX1 is also expressed in endothelial cells, but at a lower level [58]. Endothelial NOX1 is implicated in ROS generation under pathological conditions as evident by increased NOX1 expression in endothelial cells subjected to oscillatory shear stress [59]. Furthermore, siRNA-mediated NOX activator protein 1 knockdown has been shown to decrease ROS generation in HUVECs exposed to oxidized low-density lipoproteins (Ox-LDL) [60]. Unlike the other NOX isoforms, the activity of NOX5 is regulated by intracellular calcium ion (Ca2+) levels due to its four Ca2+ binding sites in the N-terminal calmodulin-like domain with elevated NOX5 expression is implicated in CAD patient arteries [61]. Studies in animals showed that the NOX1, NOX2, and NOX5 isoforms augment endothelial dysfunction, inflammation, and apoptosis in animal models of induced hypertension [62,63], diabetes [64], or atherosclerosis [62].

2.3. Uncoupled Endothelial Nitric Oxide Synthase

Nitric oxide synthases (NOS) are a group of enzymes responsible for producing NO and citrulline from oxygen and L-arginine. This process involves the transfer of electrons from NADPH, which is attached to the C-terminal reductase domain, to the heme iron and the cofactor tetrahydrobiopterin (BH4) found in the N-terminal oxygenase domain. This transfer is essential for the reduction and incorporation of O2 into L-arginine, resulting in the formation of NO and citrulline [32,65]. NO is generated by three distinct isoforms of NOS, including neuronal NOS (nNOS), inducible NOS (iNOS), and endothelial NOS (eNOS), the latter being the most prevalent in the endothelium [66]. NO generated by eNOS plays a crucial role in maintaining vascular homeostasis [67]. However, in unhealthy physiological conditions, including diabetes or other metabolic abnormalities, a scarcity of substrates or cofactors for eNOS may produce superoxide instead of NO, a phenomenon referred to as “uncoupling” [68]. Additionally, NO can react with superoxide to form peroxynitrite, a strong oxidant that leads to protein nitration, mitochondrial dysfunction, and damage or death of endothelial cells [69,70,71,72]. A decrease in BH4 bioavailability appears to be the primary factor driving eNOS uncoupling linked to vascular injury [32,46,66,67]. In ROS-induced endothelial dysfunction, BH4 is converted to BH2, which cannot serve as a cofactor for eNOS, contributing to the uncoupling [73]. Studies have shown that knocking out p47phox, a subunit of NOX, as well as the inhibition of another subunit, Rac1, have been shown to prevent both eNOS uncoupling and BH4 oxidation, highlighting the critical function of NOX in decreasing BH4 availability and facilitating eNOS uncoupling. Additionally, studies in humans demonstrated that endothelial function is significantly improved in patients with diabetes [74], hypertension [75], or hypercholesterolemia [76] after BH4 supplementation. An endogenous competitive inhibitor, asymmetric dimethylarginine (ADMA), is implicated in NOS uncoupling, as evident in pulmonary arterial endothelial cells, where it promotes eNOS uncoupling [77]. It has been reported that in patients with advanced atherosclerosis, eNOS uncoupling and O2· production within the vascular endothelium is positively correlated with Serum ADMA levels [78]. Multiple in vitro studies showed that when endothelial cells are exposed to various stimuli mimicking metabolic changes leading to endothelial dysfunction, such as native LDL and Ox-LDL [79], angiostatin [80], homocysteine [81], and high glucose [82,83], they become increasingly vulnerable to eNOS uncoupling. eNOS uncoupling is implicated in promoting the mobilization and function of endothelial progenitor cells (EPCs). For instance, ROS levels significantly augmented in EPCs obtained from diabetic patients compared to control subjects, whereas the inhibition of eNOS attenuated this ROS elevation. Additionally, high glucose treatment significantly decreased BH4 levels and altered migration capability in the cultured EPCs, which were restored by eNOS inhibition and the administration of exogenous BH4 [84].

2.4. Xanthine Oxidase

Xanthine oxidase (XO) belongs to the xanthine oxidoreductase (XOR) family, which play a vital role in purine degradation by catalyzing the conversion of hypoxanthine to xanthine and later xanthine to uric acid. This enzyme operates as a homodimer, around 300 kDa in size, with each monomer containing a molybdopterin (Mo-Pt) cofactor, two iron-sulfur (Fe-S) centers, and a flavin adenine dinucleotide (FAD) domain [85,86]. Typically, expressed in its dehydrogenase form (XDH), XOR can switch to its oxidase form (XO) under inflammatory conditions due to the oxidation of cysteine residues 535 and 992 or through proteolytic conversion [85,87]. In the process of converting xanthine into uric acid, XDH facilitates the reduction of NAD+ to NADH, whereas the XO form reduces molecular oxygen to superoxide (O2·) and H2O2. XO-induced generation of ROS is the primary contributor to oxidative stress in ischemia/reperfusion injuries. Pharmacological Inhibition of XO with allopurinol is shown to decrease vascular remodeling and oxidative stress in a rodent model of hypoxia-induced pulmonary hypertension [88]. Additionally, XO is associated with elevated ROS levels and vascular damage in diabetes, as elevated XO levels are detected in the plasma of type 1 [89] and type 2 [90] diabetic patients. Furthermore, allopurinol administration reduced plasma lipoperoxides in those with type 1 diabetes and attenuated increased superoxide levels in the aortic rings of diabetic rabbits [89]. Moreover, a connection between elevated XO levels and atherosclerosis was established. Bovine aortic endothelial cells (BAECs) subjected to oscillatory shear stress exhibited increased superoxide production and XO activity, which was suppressed by the XO inhibitor oxypurinol [91]. Notably, increased oxidative stress-induced activation of NOX leads to XO production [92]. Apocynin-mediated NOX inhibition blocked superoxide generation and the conversion of XDH to XO in BAECs under shear stress. Additionally, BAECs treated with AngII showed higher XO levels, an effect negated by NOX inhibition. Importantly, under certain pathological circumstances, including liver and intestinal diseases [93] and hypoxia [94] XDH is released into the bloodstream and transformed into the XO form, which binds to sulphated glycosaminoglycans (GAGs) expressed on endothelial cell surfaces, thereby further promoting ROS generation and endothelial dysfunction [95]. It is noteworthy that inhibiting circulating XO activity enhanced endothelial function in patients suffering from CAD [92], chronic heart failure [96], as well as type 1 DM [89] and T2DM [97] diabetes.

2.5. Nutrient Excess

In cells, overproduction of ROS often occurs when there is an excessive nutrient supply, such as high glucose or elevated free fatty acids, combined with low energy demand. This state leads to elevated intracellular ATP levels, which inhibit ADP availability and slow electron flow along the ETC in mitochondria. When the ETC is slowed, the electron carriers, particularly at Complexes I and III, remain in a reduced state for longer, increasing the likelihood of electrons leaking directly to molecular oxygen, thereby forming superoxide anion (O2•) as a primary ROS. In hyperglycemia, increased glycolytic flux leads to an abundance of pyruvate entering the tricarboxylic acid (TCA) cycle, generating excess NADH and FADH2, which further supply electrons to the ETC and exacerbate the reduced state of its components. Similarly, elevated free fatty acids in obesity undergo β-oxidation, producing additional NADH and FADH2, contributing to the electron overload in mitochondria. The resulting mitochondrial ROS production can damage mitochondrial DNA, proteins, and lipids, further impairing ETC function and creating a vicious cycle of ROS generation. In endothelial cells, this oxidative environment reduces NO bioavailability by reacting with NO to form peroxynitrite, leading to endothelial dysfunction, vasoconstriction, and inflammation. These processes collectively link excess nutrient-induced metabolic conditions, such as hyperglycemia and obesity, to mitochondrial ROS overproduction and subsequent vascular dysfunction, which are key contributors to the development of diabetic vascular complications [98]. Oxidative stress often arises early in response to rapid or sustained hyperglycemia. Several cellular changes occur in response to oxidative stress from hyperglycemia, including: (i) generation of peroxynitrites; (ii) increased levels of ADMA, a competitive eNOS inhibitor; (iii) reduced insulin-induced generation of NO [99]; (iv) Augmented diacylglycerol (DAG) synthesis followed by PKC activation, which are linked to NOX activation, reduced insulin-stimulated NO production [100], upregulated expression of adhesion molecules, increased endothelin-1 (ET-1) release [101], and p66Shc activation [102,103]; (v) decreased activity and/or expression of antioxidant enzymes [104]; (vi) formation of AGEs [105]; and (vii) activation of protein phosphatase 2A (PP2A) [106]. AGEs possess pro-inflammatory characteristics created when reducing sugars react with amino groups in proteins, lipids, and nucleic acids through a non-enzymatic Maillard reaction. AGEs can be generated through other mechanisms, including glucose oxidation, lipid peroxidation, or the polyol pathway [107,108,109], accumulating in blood vessels and contributing to the pathophysiology of both microvascular and macrovascular complications associated with diabetes. For instance, patients with Type 2 diabetes exhibit elevated levels of compounds like glyoxal, methylglyoxal, and 3-deoxyglucosone [110]. AGEs can exacerbate diabetic complications by impairing cellular structures, binding specific basement membrane molecules in the extracellular matrix, and interacting with receptors for AGEs (RAGEs) on cell surfaces, relaying stress signals [111]. In endothelial cells, the interaction between AGEs and RAGEs boosts ROS production and increases adhesion molecules, including vascular cell adhesion molecule-1 (VCAM-1), ICAM-1, and E-selectin [111,112], resulting in NO bioavailability due to reduced eNOS activity, while promoting cell apoptosis [113,114,115]. A study demonstrated that cultured endothelial cells exposed to AGE methylglyoxal (MGO) significantly reduced the Bcl2/Bax ratio, leading to increased apoptosis. This was associated with an augmented production of ROS by NOX4, diminished eNOS activity, mitochondrial membrane potential, and nuclear translocation of NFκB, all of which could be reduced by pretreating endothelial cells with phosphocreatine [115]. Another potential mechanism of high glucose-induced ROS production is the increased expression and activation of PP2A, a serine/threonine phosphatase that dephosphorylates various substrates involved in cellular signaling, including p66Shc. PP2A catalyzes the dephosphorylation of p66Shc at Ser36 and promotes its translocation into mitochondria to induce oxidative stress [103]. A study showed that high glucose-treated HUVECs significantly augmented intracellular calcium levels, enhanced the phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and cAMP response element-binding protein (CREB), ultimately upregulated both PP2A expression and activity, as well as ICAM-1 expression. However, blocking the high glucose-induced expression of PP2A prevented O2· accumulation, enhanced NO production, and inhibited ICAM-1 expression, thereby exacerbating endothelial dysfunction [116]. Likewise, increased lipid levels contribute to endothelial cell dysfunction and death due to increased oxidative stress. For instance, increased levels of palmitate, the primary saturated fatty acid in human plasma [117,118], significantly disrupt insulin signaling in endothelial cells. Specifically, treating bovine aortic endothelial cells (BAECs) with palmitate significantly diminished insulin-mediated tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1) and the subsequent serine phosphorylation of Akt and eNOS, along with decreased NO production, due to increased inhibitor of NFκB kinase beta (IKKβ) activity and subsequent phosphorylation of IRS-1 that inhibited the signaling pathway [118]. Another study reported that palmitate-treated HMECs augmented NOX4-induced ROS production, NFκB activation, and upregulation of IL-6 and ICAM-1 expression [119]. Furthermore, chronic exposure to palmitate resulted in apoptosis of endothelial and progenitor cells through the activation of stress-activated protein kinases JNK and p38 MAPK [120,121,122,123]. Collectively, these findings suggest that increased plasma levels of saturated fatty acids can induce endothelial dysfunction and apoptosis while impairing endothelial repair by damaging endothelial progenitor cells. Importantly, elevated levels of saturated fatty acids promote the formation and accumulation of detrimental lipid metabolites like ceramide [124], which is known to induce superoxide production and peroxynitrite formation through NOX in bovine coronary arterial endothelial cells [125], while declining bioactive NO levels in human endothelial cells [126]. In contrast, inhibiting ceramide synthesis in fat-fed streptozotocin (STZ)-treated rats has been shown to improve endothelial dysfunction by enhancing phosphorylation and NO release via the PI3K/Akt/eNOS pathway.

2.6. Peroxiredoxins

Peroxiredoxins (Prx) are thiol-specific enzymes that convert H2O2 into water by utilizing active cysteine residues. In mammals, six isoforms of peroxiredoxins (Prx1–6) have been characterized so far. Isoforms Prx1–5 require two cysteine residues for function, whereas Prx6 requires only one [127]. Multiple studies revealed the essential role of Prx as ROS scavengers, particularly in protecting lung tissues from ROS-induced cytotoxicity [128,129,130]. Silencing Prx2 with siRNA resulted in the inactivation of the vascular endothelial growth factor receptor-2 (VEGFR2) due to H2O2-induced oxidation of a crucial cysteine residue and subsequently reduced chemotactic mobility, proliferation, and VEGF-induced tube formation of HAECs. Interestingly, VEGF-induced VEGFR2 activity was not altered upon knockdown of Prx1 and Gpx1, indicating that Prx2 is the specific cytosolic antioxidant enzyme responsible for regulating basal H2O2 levels in endothelial cells [131]. Prx6 KO mice exhibited dysfunctional and apoptotic endothelial cells around skin wounds, which were correlated with damage to blood vessels and hemorrhages. Moreover, the siRNA-mediated knockdown of Prx6 significantly decreased the survival of endothelial cells subjected to H2O2 exposure [132]. Prx1 isoform plays an important role in the antioxidative and anti-inflammatory effects in BAECs upon laminar shear stress [133].

2.7. Thioredoxin

Thioredoxins (Trxs) are a group of 12 kDa oxidoreductases found in the cytosol and mitochondria. They are essential for reducing disulfides and sulfenic acids that develop from oxidative stress in proteins, both within and between cells. For instance, Trx possesses cysteine residues in Prx in a reduced state, preserving the enzyme’s catalytic activity [32]. Beyond the regulation of redox conditions of various targets, Trx aids the survival of endothelial cells. In these cells, H2O2 has been reported to regulate Trx protein levels. For instance, H2O2 at low concentrations elevates Trx levels and shields against apoptosis of endothelial cells, whereas higher concentrations augment apoptosis through Trx degradation via a cathepsin-D dependent mechanism [134]. Trx also affects endothelial function by binding to specific proteins and transcription factors. Laminar shear stress is implicated to augment the activity of Trx due to the decreased expression of thioredoxin binding protein (TXNIP), facilitating Trx’s binding with apoptosis signal-related kinase-1 (ASK1) following the inhibition of ASK1-mediated activation of JNK and p38 MAPK in response to TNF [135]. Additionally, in HUVECs, the prevention of apoptosis from A low dose of H2O2 induces the translocation of Trx into the nucleus, where it binds to several transcription factors, improving their interaction with antioxidant responsive elements (AREs) and promoting the expression of glutathione S-transferase P1, which is crucial for preventing apoptosis [136]. Beyond Trx, other proteins can also contribute to the buffering of ROS. For example, Paraoxonase 2 (PON2) binds to coenzyme Q10 within respiratory complex III, decreasing ROS production at that site [137]. PON2 KO mice have shown diminished activity in complexes I and III, along with lower oxygen consumption and ATP production. Additionally, these mice display a more severe atherogenic phenotype than controls when fed a high-fat, high-cholesterol diet [138].

2.8. Uncoupling Proteins

Uncoupling proteins (UCPs), which consist of five mitochondrial protein carriers, transport protons from the intermembrane space to the matrix, reducing the mitochondrial membrane potential followed by uncoupling ATP synthesis. Given the necessity of high levels of mitochondrial membrane potential for ROS production, uncoupling proteins may serve as a form of antioxidant defense. Indeed, overexpressing UCP1 has been shown to diminish ROS production and apoptosis in endothelial cells triggered by high glucose levels [32,139], while UCP2 knockdown augmented mitochondrial membrane potential and superoxide anion levels in murine endothelial cells [140]. Hyperglycemia-induced upregulation of UCP2 is implicated in endothelial cells [141]. Animal studies showed that UCP2 upregulation enhances endothelium-dependent relaxation in aortic rings from diabetic mice [142], as well as prevents endothelial cell apoptosis resulting from elevated free fatty acids and ROS [143].

2.9. Superoxide Dismutases

Superoxide dismutases (SODs) are a group of enzymes that facilitate the transformation of superoxide anion into H2O2. There are three distinct isoforms of SOD located in various subcellular compartments: a cytosolic copper-zinc SOD (CuZnSOD), a manganese SOD (MnSOD) predominantly found in mitochondria, and an extracellular CuZnSOD (SOD3) that binds to cell surface heparin sulfate proteoglycans [144,145]. Inhibition of SOD1 in endothelial cells augmented superoxide production and diminished the phosphorylation of extracellular signal–regulated kinases (ERK) 1/2 mediated by fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor (VEGF), thereby decreasing angiogenesis [146]. Beyond its traditional role in scavenging ROS, H2O2 produced by SOD1 has been identified as an endothelium-dependent hyperpolarization factor in vivo [147]. SOD2, an inducible enzyme featuring manganese at its active site, catalyzes superoxide generation in the mitochondrial matrix [148]. SOD2-mediated generation of H2O2 has been shown to promote endothelial cell sprouting and angiogenesis [149]. Moreover, SOD2 expression levels are significantly higher in human endothelial progenitor cells relative to differentiated endothelial cells, making them more resilient to oxidative stress [150]. Elevated superoxide levels and impaired relaxation of aortas in response to acetylcholine have been observed in experimental atherosclerosis models with SOD2 deficiency [151]. Furthermore, decreased SOD2 levels were detected in the pulmonary artery endothelial cells of fetal lambs with chronic pulmonary hypertension [152]. DM-induced ROS production also impairs SOD2 expression and activity, thereby exacerbating ROS-mediated endothelial dysfunction. Notably, a positive correlation was found between decreased SOD2 levels and impaired wound healing and angiogenesis in endothelial progenitor cells isolated from diabetic mice [153]. SOD3 serves as the predominant SOD in the vascular extracellular space [154]. Studies demonstrated that decreased SOD3 levels are linked to elevated ROS levels in the aortas of older rats [155], while upregulated expression of SOD3 enhanced endothelial function in hypertensive [156] and heart failure rat models [157].

2.10. Catalase and Peroxidases

Antioxidant enzymes, catalase and peroxidases, catalyze the conversion of H2O2 into molecular oxygen and water. Catalase is a 240-kDa homotetrameric heme-containing peroxisomal protein, predominantly expressed in the liver, lungs, and kidneys [158]. Although catalase’s role in the endothelium is not well-known as its activity is not well studied in normal physiological conditions, in endothelial cells, it plays an active role in the adaptive response of cells against oxidative stress and may be modulated by oxidative factors like oxLDL [159]. Glutathione peroxidase (GPx) is an 85-kDa selenium-dependent protein, exerts its catalytic activities using monomeric glutathione as an electron donor. The oxidized form of glutathione is then reverted to its reduced state by the action of glutathione reductase. Four types of glutathione peroxidases have been identified so far in mammals, with isoform 1 (GPx-1) being the most abundantly expressed [160]. In the endothelial cells, GPx-1 is present in both the mitochondria and cytoplasm [161]. Upregulation of GPx-1 expression is implicated in protecting cultured human primary pulmonary artery endothelial cells from H2O2-induced cytotoxicity [162]. In contrast, the knockdown of GPx-1 enhances leukocyte adhesion to endothelial cells and promotes a pro-inflammatory phenotype in aging [163]. Furthermore, the mesenteric artery of GPx-1 KO mice demonstrated impaired vasodilation due to reduced NO bioavailability and elevated oxidative stress [160]. MicroRNAs play an important role in regulating genes involved in the antioxidant response to elevated glucose levels; for instance, a study revealed that upregulation of miRNA-185 coincided with the suppression of GPx-1 levels in HUVECs exposed to fluctuating high glucose [164]. Possible sources of ROS are illustrated in Figure 1.

3. Mechanism of Oxidative Stress in DM

In diabetes, oxidative stress results from a complex interplay of multiple factors, such as the buildup of glycolysis intermediates, formation of advanced glycation end products (AGEs), activation of PKC, polyol, and hexosamine pathways [17] (Table 1).

3.1. Buildup of Glycolysis Intermediates

Under typical physiological conditions, cellular processes, including glucose oxidation, lead to the mitochondrial production of superoxide anion radicals. This production occurs at a level manageable by the body’s antioxidant defenses [18,176]. However, in hyperglycemic conditions, the overproduction of superoxide anion radicals can overwhelm these antioxidant systems, resulting in oxidative stress that damages nuclear DNA and other biomolecules [177]. In response to DNA damage, the DNA repair enzyme poly-ADP-ribose polymerase-1 (PARP-1) becomes activated [178]. This enzyme inhibits GAPDH, causing levels of Glyceraldehyde 3-phosphate (G3P) and other glycolytic intermediates such as fructose-6-phosphate and glucose-6-phosphate, as well as glucose, to rise [179]. The accumulation of these molecules triggers additional pro-oxidative pathways like the AGE and PKC pathways from increased G3P, and the hexosamine and polyol pathways due to elevated F-6-P and glucose levels, respectively [17]. Additionally, the buildup of G3P can lead to its autooxidation, which produces hydrogen peroxide (H2O2) and exacerbates oxidative stress. Likewise, the accumulation of glucose can result in its autooxidation, yielding glyoxal, a precursor to AGEs, which further contributes to cellular oxidative stress [10].

3.2. AGEs/RAGE Pathway

AGEs are highly reactive and irreversible end products resulting from non-enzymatic reactions involving glucuronyl groups and free amino groups, including those found in lipids and proteins [180]. Prolonged hyperglycemia significantly accelerates the production of AGEs, while excessive AGEs directly enhance the production of ROS. Importantly, the generated ROS reciprocally stimulates further AGE production, thereby exacerbating oxidative stress damages to different cells in different organs. A substantial body of research indicates that the receptor for AGEs (RAGE) is increasingly expressed in vascular endothelial cells, immune cells, monocytes/macrophages, neurons, cardiomyocytes, adipocytes, glomerular epithelial cells, podocytes and alveolar epithelial cells following hyperglycemic stimulation [167]. In endothelial cells, the interaction of RAGE with AGEs activates NOX, resulting in increased ROS production, which disturbs molecular conformation and alters enzyme activity, thereby inducing oxidative stress responses. Consequently, the activated oxidative stress augments the activation of different downstream signaling pathways, including NFκB, TNF, JNK, and p38-MAPK, leading to an increased release of adhesion molecules, vascular endothelial growth factors, and inflammatory factors [181,182].

3.3. PKC Pathway

PKC pathway plays a pivotal role in the cellular response to oxidative stress. The generation of ROS induced by oxidative stress directly activates various PKC isoforms through oxidative modifications of their regulatory domains [183]. In diabetes, hyperglycemia causes the intracellular diacylglycerol content to significantly increase, leading to the activation of the PKC pathway [184,185]. Furthermore, AGE/RAGE and polyol pathways can activate the PKC.
Upon activation, PKC may either promote cellular survival or instigate apoptosis, contingent upon the specific isoform and the cellular context. For instance, PKC-ε has been demonstrated to enhance antioxidant defenses by activating protective pathways, such as the Nrf2-mediated antioxidant response [186], whereas [187]. Furthermore, PKC can exacerbate oxidative stress by augmenting ROS production through the stimulation of NOX activity [188]. Through these diverse mechanisms, the PKC pathway serves as a critical mediator in determining cellular outcomes during oxidative stress, thereby influencing the development and progression of diseases, including cardiovascular disorders, neurodegenerative diseases, complications related to diabetes, and cancer [189].

3.4. Polyol Pathway

The polyol pathway plays a significant role in oxidative stress, particularly during chronic hyperglycemia, as seen in DM. Within this pathway, excess intracellular glucose is converted into sorbitol by the enzyme aldose reductase, which uses NADPH as a cofactor [190]. Sorbitol is then oxidized to fructose by sorbitol dehydrogenase, a reaction that produces NADH [166]. The activation of the polyol pathway under hyperglycemic conditions leads to several detrimental effects. Firstly, the consumption of NADPH reduces its availability for other vital antioxidant systems, especially for regenerating reduced glutathione (GSH), crucial for neutralizing ROS [191]. Secondly, the elevated NADH/NAD+ ratio from increased sorbitol dehydrogenase activity enhances mitochondrial superoxide production, worsening oxidative stress [192]. Additionally, sorbitol, which does not easily diffuse across cell membranes, accumulates within cells, leading to osmotic stress and subsequent damage to cellular structures [193]. Collectively, these changes create a cycle of oxidative damage and inflammation, significantly contributing to the development of diabetic complications like neuropathy, retinopathy, nephropathy, and CVD [104]. As a result, aldose reductase inhibitors have been investigated as potential therapies to reduce oxidative damage by regulating polyol pathway activity [194,195]. While experimental findings are promising, challenges remain in clinical applications, highlighting the complexity of oxidative stress mechanisms and the necessity for thorough strategies to address oxidative injury in diabetes [104].

3.5. Hexosamine Pathway

The hexosamine biosynthetic pathway (HBP) acts as a nutrient sensor and plays a key role in oxidative stress and cellular dysfunction in diabetes, obesity, and cardiovascular diseases. Normally, only a small fraction (2–5%) of glucose enters the HBP to produce UDP-N-acetylglucosamine (UDP-GlcNAc), which is used for O-linked β-N-acetylglucosamine (O-GlcNAc) modification of proteins in the nucleus, cytoplasm, and mitochondria [196]. Under hyperglycemic conditions, increased glucose flux through the HBP leads to excessive O-GlcNAcylation of regulatory proteins, altering gene expression, impairing mitochondrial function, and increasing oxidative stress [197,198]. For example, over-O-GlcNAcylation of the antioxidant transcription factor Nrf2 reduces its nuclear translocation and target gene activation, weakening antioxidant defenses and promoting ROS accumulation [199]. Additionally, HBP hyperactivation increases endoplasmic reticulum (ER) stress by promoting protein misfolding, which triggers the unfolded protein response and further ROS production [200]. Mitochondrial proteins are also subject to O-GlcNAcylation under high-glucose conditions, resulting in impaired ETC activity, altered mitochondrial dynamics, and elevated mtROS generation [201]. Notably, HBP-induced modification impairs insulin signaling pathways, which contribute to insulin resistance, a condition that serves as both a cause and a consequence of oxidative stress in metabolic tissues. Overall, HBP links metabolic overload to oxidative stress, inflammation, and organ dysfunction. Targeting HBP flux or enzymes regulating O-GlcNAc cycling, such as O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), holds promise for mitigating oxidative stress-related metabolic diseases.
Having discussed the mechanisms underlying oxidative stress in diabetes, it is important to consider how these processes translate into vascular tissue damage and disease-specific pathophysiology. Persistent oxidative stress in diabetes directly impairs endothelial function, disrupts vascular homeostasis, and promotes inflammatory and fibrotic responses, leading to structural and functional alterations within the vasculature. These oxidative stress-induced changes contribute to the development and progression of both microvascular and macrovascular complications in diabetes, including atherosclerosis, DR, and DKD. In the following sections, we will discuss how oxidative stress drives vascular dysfunction and specific pathophysiological mechanisms underlying these diabetes-related vascular complications.

4. Role of Oxidative Stress in Vascular Cells

Regulating vascular tone is essential for maintaining blood vessel homeostasis and ensuring adequate blood flow to peripheral organs [202]. Under physiological conditions, proper endothelial function allows for vasorelaxation through the release of vasoactive substances [203]. However, an imbalance in the production of protective and relaxing factors versus constricting substances by the endothelium indicates endothelial dysfunction, which often precedes various vascular diseases [204]. Oxidative stress plays a pivotal role in both the onset and development of endothelial dysfunction and vascular diseases that affect multiple cell types within the vascular wall [205]. Furthermore, oxidative stress within the vascular system triggers systemic inflammation through immune activation [206]. Immune cells that are activated migrate into blood vessels and release various factors such as ROS, metalloproteinases, cytokines, and chemokines, which contribute to endothelial and vascular smooth muscle cells (VSMCs) dysfunction and subsequent vascular injury, leading to vasoconstriction and remodeling of the vessels [205].

4.1. Oxidative Stress and Endothelial Cells

Endothelium is a monolayer of endothelial cells that lines the interior of blood vessels and plays a crucial role in maintaining vascular homeostasis [207,208]. In addition to this, healthy endothelial cells perform several other functions, such as the regulation of vascular tone, maintaining blood fluidity, modulating inflammation and immune responses, and facilitating the formation of new blood vessels (neovascularization) [30,209]. Impairment of the endothelium is a complex pathophysiological phenomenon that involves elevated activation of endothelial cells as well as the onset of endothelial dysfunction [30]. Endothelial activation refers to a pro-inflammatory and pro-coagulant state of these cells, associated with the expression of cell-surface adhesion molecules that are essential for recruiting and attaching inflammatory cells [210,211]. This activation is triggered by cytokines released by tissues and organs with inflammation. Studies reported that oxidative stress plays a significant role in mediating the production and secretion of cytokines, thus linking ROS to inflammation, as well as endothelial activation and dysfunction [212]. Endothelium-derived NO contributes to the maintenance of vascular homeostasis. A decrease in NO bioavailability, which can occur due to reduced NO production or increased degradation by superoxide anions, signals the initiation of endothelial dysfunction. Superoxide anions interact with nitric oxide (NO) to produce peroxynitrite (ONOO) [213,214,215]. Peroxynitrite is implicated in promoting protein nitration, leading to the dysfunction and apoptosis of endothelial cells [70,71,72]. NOX, xanthine oxidase, and uncoupled eNOS contribute to the production of superoxide anions. Furthermore, impairment in mitochondrial respiratory chain complexes significantly increases the production of oxidative stress-related substances. Pathobiological and metabolic factors, such as hyperglycemia, hyperlipidemia, hypertension [216], mental stress [217], aging [218], and drug exposure [219] can impair endothelial function by disturbing the molecular mechanisms behind NO generation. As a result, therapeutic interventions that target improving insulin sensitivity, glycemic control, lipid levels, and blood pressure can frequently mitigate endothelial dysfunction. Additionally, epigenetic factors such as histone acetylation and deacetylation, as well as modulation of microRNA expression, are implicated in influencing vascular homeostasis. The role of oxidative stress in the vascular endothelial cell is illustrated in Figure 2.

4.2. Oxidative Stress and Smooth Muscle Cells

Oxidative stress plays a critical role in the pathophysiology of vascular smooth muscle cells (VSMCs), which are key players in the maintenance of vascular tone, structure, and remodeling. In normal physiological conditions, VSMCs maintain a contractile phenotype essential for blood pressure regulation and vascular homeostasis [220]. However, under oxidative stress, there is a well-documented phenotypic switch from a contractile to a synthetic phenotype. This transformation is associated with increased VSMC proliferation, migration, extracellular matrix production, and a pro-inflammatory profile, all of which are implicated in vascular pathologies such as atherosclerosis, hypertension, and restenosis following vascular injury [221]. The major sources of ROS in VSMCs, include NOX, mitochondrial respiration, xanthine oxidase, and uncoupled eNOS [222]. Among these, the NOX family, particularly NOX1 and NOX4, are highly expressed in VSMCs and have been shown to mediate numerous redox-sensitive signaling pathways involved in vascular remodeling [223]. Elevated ROS levels influence multiple cellular processes by modifying redox-sensitive proteins and signaling molecules. For instance, ROS-induced activation of mitogen-activated protein kinases (MAPKs) and NFκB facilitates the expression of pro-inflammatory cytokines and matrix metalloproteinases (MMPs), which degrade the extracellular matrix and contribute to plaque instability in atherosclerosis [224]. Furthermore, oxidative stress is linked to the activation of transcription factors such as activator protein-1 (AP-1) and hypoxia-inducible factor-1α (HIF-1α). A recent study demonstrated that high-salt conditions enhance VSMC proliferation and migration by upregulating HIF-1α, which in turn increases oxidative stress and promotes vascular remodeling [225]. This indicates a feedback loop wherein oxidative stress both induces and is amplified by phenotypic switching of VSMCs. Another crucial consequence of oxidative stress is the induction of apoptosis in VSMCs, particularly in the context of aortic dissection and aneurysm formation. Apoptotic loss of VSMCs compromises the structural integrity of the vascular wall, rendering it prone to rupture. A study demonstrated that oxidative stress contributes to VSMC apoptosis via the mitochondrial pathway, with increased Bax/Bcl-2 ratios and cytochrome c release into the cytosol. This apoptotic cascade, if unregulated, leads to thinning of the medial layer and predisposes vessels to catastrophic events such as dissection [226]. Moreover, oxidative stress-induced apoptosis often coexists with the dysregulation of autophagy, which further compromises cellular survival and vascular homeostasis [227]. VSMCs are also affected by metabolic alterations driven by oxidative stress. Hyperuricemia, for instance, has emerged as a metabolic trigger that increases ROS production in VSMCs. A recent study reported that uric acid elevates intracellular oxidative stress by disrupting mitochondrial function, leading to decreased nitric oxide bioavailability and suppression of tumor suppressor p53, which is vital for maintaining VSMC quiescence and genomic stability [228]. These findings are particularly relevant in metabolic syndrome and gout, where elevated uric acid levels are common and contribute to vascular dysfunction through redox imbalance. The crosstalk between oxidative stress and other forms of cellular stress, such as endoplasmic reticulum (ER) stress, is also an area of increasing interest. ER stress occurs when misfolded proteins accumulate within the ER lumen, triggering the unfolded protein response (UPR). In oxidative stress, ROS can exacerbate ER stress, leading to VSMC inflammation and death. A recent study showed that hypertensive patients exhibit increased markers of both oxidative and ER stress in their vascular tissue, suggesting a synergistic pathogenic mechanism [229]. The convergence of these stress responses amplifies cellular damage and accelerates vascular disease progression. From a therapeutic perspective, targeting oxidative stress in VSMCs offers promising avenues for the prevention and treatment of vascular diseases. Antioxidant strategies include both pharmacological agents and natural compounds. Ginsenoside Rb1, a major component of ginseng, has been shown to exert vascular protective effects by modulating oxidative stress and inflammation in VSMCs. A study by Lu et al. demonstrated that Rb1 inhibits NOX4 expression and reduces ROS generation, thereby preventing VSMC phenotypic transformation and migration [230]. Another promising strategy involves activation of the Mas receptor (MasR) and particulate guanylyl cyclase A (pGCA), which enhance antioxidant responses in VSMCs. In a preclinical study, MasR and pGCA activation improved VSMC survival and attenuated ROS-mediated injury, suggesting their potential as drug targets [231]. Moreover, gene therapy approaches aiming to modulate redox-sensitive signaling molecules are under investigation. For instance, the delivery of catalase or SOD genes to vascular tissues has shown efficacy in reducing oxidative stress and mitigating VSMC dysfunction in animal models. Additionally, pharmacological inhibitors of NOX have been developed, with NOX1-specific inhibitors like GKT137831 demonstrating favorable effects in reducing oxidative damage and vascular inflammation [232]. However, clinical translation of these agents remains a challenge due to issues related to bioavailability, specificity, and off-target effects. In conclusion, oxidative stress plays a multifaceted and central role in the regulation of vascular smooth muscle cell function in normal physiology and pathological states. Through mechanisms involving phenotypic modulation, inflammation, apoptosis, and interaction with other cellular stress responses, ROS contribute to the pathogenesis of various vascular disorders. A deeper understanding of the redox signaling pathways in VSMCs is essential for the development of targeted therapeutic interventions. As research progresses, integrating redox biology into the clinical management of cardiovascular diseases may yield novel strategies to combat vascular dysfunction and improve patient outcomes.

4.3. Oxidative Stress and Vascular Fibroblast

Oxidative stress plays a critical role in the pathophysiology of vascular fibroblasts, influencing their behavior, phenotype, and contribution to cardiovascular diseases. Vascular fibroblasts, traditionally seen as passive structural components of the vessel wall, are now recognized as dynamic cells capable of responding to environmental stimuli, including ROS [233]. In vascular fibroblasts, oxidative stress activates redox-sensitive transcription factors such as NFκB, AP-1, and Nrf2, which in turn upregulate pro-inflammatory cytokines, MMPs, and adhesion molecules [46]. These molecules mediate fibroblast activation and transition into a myofibroblast phenotype, characterized by increased contractility, secretion of extracellular matrix (ECM) components such as collagen types I and III, and expression of α-smooth muscle actin (α-SMA) [234]. This phenotypic switching of vascular fibroblast contributes to pathological vascular remodeling by promoting fibrosis, vascular stiffening, and intimal hyperplasia [235]. Moreover, oxidative stress enhances the cross-linking of ECM proteins, further exacerbating vascular stiffness [236]. The profibrotic cytokine transforming growth factor-beta (TGF-β) is also upregulated under oxidative stress and acts synergistically with ROS to perpetuate fibroblast activation via the SMAD and non-SMAD pathways [237]. Importantly, ROS can also cause DNA damage, lipid peroxidation, and protein oxidation in fibroblasts, leading to cellular senescence, which paradoxically contributes to chronic inflammation through the senescence-associated secretory phenotype (SASP) [238]. These senescent fibroblasts remain metabolically active and secrete pro-inflammatory and pro-fibrotic mediators, sustaining a vicious cycle of oxidative stress and fibrosis [239]. Additionally, oxidative stress modulates fibroblast crosstalk with other vascular cells, including endothelial cells and smooth muscle cells. For example, ROS-induced endothelial dysfunction leads to reduced NO bioavailability and increased endothelial permeability, facilitating the infiltration of inflammatory cells and further stimulating fibroblast activation [240]. Crosstalk between fibroblasts and VSMCs under oxidative stress conditions may also contribute to VSMC phenotypic modulation and calcification [223]. Furthermore, mitochondrial dysfunction in fibroblasts under oxidative stress not only increases ROS production but also impairs cellular metabolism, shifting fibroblasts towards a glycolytic phenotype, which is often associated with fibrotic diseases [241]. In the context of vascular aging, oxidative stress speeds up fibroblast and endothelial cell senescence and contributes to age-related vascular stiffness and decreased compliance, making vessels more vulnerable to hemodynamic damage [242]. Therapeutically, targeting oxidative stress pathways in vascular fibroblasts represents a promising strategy for treating vascular diseases. In conclusion, oxidative stress is a central driver of vascular fibroblast dysfunction and contributes significantly to the pathogenesis of vascular fibrosis and remodeling. A better understanding of the redox-regulated mechanisms in fibroblasts may uncover novel therapeutic targets and improve the management of cardiovascular diseases associated with fibrosis and oxidative damage.

5. Role of Oxidative Stress in Vascular Diseases

5.1. The ROS-Aided Pathogenesis of Atherosclerosis

Oxidative stress-induced mechanisms are central to the pathogenesis of atherosclerosis, particularly in diabetes, where chronic hyperglycemia and insulin resistance enhance ROS production. Elevated ROS levels impair endothelial function by reducing NO bioavailability and increasing endothelial permeability, promoting monocyte adhesion and infiltration. ROS also oxidize LDL into oxLDL, which is taken up by macrophages to form foam cells, initiating fatty streaks. Additionally, oxidative stress activates redox-sensitive transcription factors, increasing pro-inflammatory cytokines, adhesion molecules, and matrix metalloproteinases, which drive chronic vascular inflammation, smooth muscle cell proliferation, and extracellular matrix remodeling. These processes collectively contribute to plaque formation, progression, and instability, underscoring oxidative stress as a key therapeutic target for preventing and mitigating atherosclerosis in diabetes.
There are several pathways that may contribute to the pathophysiology of oxidative stress-induced atherosclerosis (Figure 3).

5.1.1. Oxidation of Lipid

During atherogenesis, LDL builds up in the arterial wall, especially in areas with disturbed blood flow [243]. This LDL is then modified by ROS produced by NOX or uncoupled eNOS [244]. Oxidized LDL promotes atherosclerosis by altering endothelial NO production and upregulating the expression of leukocyte adhesion molecules [245]. Lowering LDL oxidation in the lipoxygenases knockout mouse model has been shown to decrease atherosclerosis [246], while heightened LDL oxidation expands the lesional area [247]. Mitochondrial lipids are also significantly affected by oxidative damage from ROS [248]. Cardiolipin is the signature lipid of the mitochondrial membrane, which comprises around 20% of the total mitochondrial membrane lipid [249], and is vulnerable to oxidative damage due to its high unsaturated fatty acid content [250]. A rodent model with ischemia-reperfusion injury demonstrated reduced cardiolipin levels and elevated lipid peroxidation in liver mitochondria [251]. This can be attributed to the oxidation of accumulated succinate upon post-ischemic restoration of ETC function and subsequent elevation of mtROS produced at Complex I [252]. Elevated levels of oxidized cardiolipin and endogenous anti-cardiolipin antibodies have been observed in atherosclerotic plaques [253]. Additionally, oxidized cardiolipin acts as a pro-inflammatory signaling molecule that promotes the expression of leukotriene and 5-lipoxygenase by leukocytes, as well as the expression of adhesion molecules such as ICAM-1 and VCAM-1 by endothelial cells [254].

5.1.2. Oxidation of Nucleic Acid

DNA is vulnerable to ROS-induced oxidative damage, including single-strand breaks, double-strand breaks, adduct formation, and deletions [255]. It is reported that patients suffering from CAD exhibit heightened chromosomal damage in their peripheral lymphocytes [256]. Elevated levels of 7,8-dihydro-8-oxo-2′-deoxyguanosine (8-oxo-dG), a biomarker of DNA damage in oxidative stress, are also found in plaque macrophages, smooth muscle cells, and endothelial cells [257]. Nuclear DNA damage plays an important role in the pathophysiology of atherosclerosis rather than being merely incidental. For instance, ApoE-deficient mice with haploinsufficient DNA repair enzyme kinase, ataxia telangiectasia mutated, demonstrated greater nuclear DNA damage, and accelerated atherosclerosis progression [255]. Enhanced base excision repair-induced inhibition of oxidative DNA damage significantly reduces plaque size [258]. Within mitochondria, the ROS-producing sites of Complex I are situated on the inner membrane’s matrix side, allowing superoxide to diffuse into the matrix [259]. Since mtDNA resides in the matrix and lacks protective histones, it is prone to oxidative damage from mtROS [260]. Indeed, mtDNA damage has been reported in the arteries and blood cells of patients with atherosclerosis [261,262]. Additionally, mtDNA damage worsens atherosclerosis in ApoE KO mice with impaired mtDNA polymerase proofreading activity, which correlates with signs of plaque vulnerability [261].

5.1.3. Endothelial Dysfunction

Endothelial dysfunction is characterized by increased leakage, overproduction of ROS, secretion of pro-inflammatory cytokines, upregulated expression of adhesion molecules, and decreased nitric oxide (NO) production are the essential inducers of atherogenesis [263]. This dysfunctional endothelium increases the likelihood of the low-density lipoprotein (LDL) trapped in the subendothelial space and subsequent oxidative modifications [264]. The endothelium acts as both a source and target of ROS [265]. Endothelial cells predominantly produce NO due to their continuous expression of endothelial nitric oxide synthase (eNOS) [266]. Furthermore, these cells also generate superoxide and peroxynitrite because of eNOS uncoupling during tetrahydrobipterin depletion [267]. Studies have demonstrated that the absence of eNOS lowers superoxide production in ApoE-deficient mice, suggesting that eNOS uncoupling occurs during atherosclerosis [268]. Prolonged exposure to superoxide triggers mtROS production in mouse vascular endothelial cells that results in diminished proliferation [269]. Oxidative stress switches the endothelium to a pro-inflammatory state, which is essential for the pathogenesis of atherosclerosis [270]. H2O2 exposure to human endothelial cells prolonged the expression of granule membrane protein 140, which results in enhanced neutrophil adhesion to the endothelial surface [271]. Additionally, oxidative stress activates NFκB, leading to increased expression of adhesion molecules like VCAM-1, ICAM-1, and E-selectin, as well as cytokines, including TNF [272]. This TNFα stimulates the production of mtROS, activation of NOX, and expression of iNOS in endothelial cells [272,273]. Consequently, oxidative stress in the endothelium fuels inflammation, which can promote further ROS generation. Furthermore, oxidized LDL has been shown to upregulate oxidized LDL receptor-1 (LOX-1) expression in endothelial cells which results in endothelial dysfunction by inducing apoptosis and inflammation [274]. Activation of LOX-1 with oxLDL mediates the downregulation of eNOS [275]. Notably, the expression of LOX-1 in endothelial cells is correlated with the progression of atherosclerosis [276]. ApoE deficient mice with elevated endothelial LOX-1 expression exhibited increased ROS production, plaque buildup, eNOS uncoupling, and macrophage infiltration [277].

5.1.4. Inflammation

ROS exerts its effects by activating various targets, including NFĸB, HIF-1α, and the NLRP3 inflammasome, to drive inflammation, a critical factor in atherogenesis [278]. NFĸB serves as a pivotal transcription factor that regulates the transcription of a large number of pro-inflammatory cytokines, including TNFα, IL-1β, and IL-18 [278,279]. In HUVECS, hydrogen peroxide treatment activates NFĸB through the tyrosine phosphorylation of IKB, the upstream target of NFĸB activation [280]. Additionally, superoxide generated by NOX can also activate NFĸB [281]. Lipopolysaccharide (LPS), a component of bacterial cell walls, binds to TLR4, which partners with NOX4 to promote ROS production and subsequent activation of NFĸB in HEK293T cells [282]. Endothelial NOX activation occurs under low shear stress conditions and subsequently enhancing NFĸB signaling in atherogenesis [59,283]. Bone morphogenic protein 4 is implicated to upregulate the expression of NOX1 mRNA following the production of H2O2 and superoxide [59]. Furthermore, HIF-1α has been shown to mediate ROS-induced expression of inflammatory cytokine [284]. Upon LPS stimulation, monocytes undergo a significant metabolic shift to differentiate into M1 macrophages, which depend on glycolysis for ATP production. This metabolic shift results in elevated glucose uptake and decreased reliance on mitochondrial oxidative phosphorylation (OXPHOS), both necessary for the expression of certain pro-inflammatory factors, such as IL-1β [285]. The diminished reliance on OXPHOS results in succinate accumulation, which can be oxidized by succinate dehydrogenase, spurring ROS production and stabilizing HIF-1α to boost IL-1β expression [286]. ROS also contribute to inflammation via the NLRP3 inflammasome [287]. The activation of the NLRP3 inflammasome is crucial for generating mature IL-1β and IL-18 [288]. The NLRP3 inflammasome consists of NLRP3, the ASC adapter protein, and caspase 1 that processes pro-IL-1β and pro-IL-18 into their active forms [289]. ROS derived from xanthine oxidase are pivotal for regulating the activation of NLRP3 inflammasome in macrophages [290]. ROS-induced NLRP3 activation increases IL-1β and IL-18 expression, whereas xanthine oxidase inhibition with febuxostat attenuated this effect [291]. Mitochondrial oxidative stress also plays a significant role in regulating the activity of NLRP3 inflammasome [292]. Increasing mtROS by inhibiting ETC enzymes or disrupting autophagy/mitophagy induces NLRP3 activation [293]. As a damage-associated molecular pattern, MtROS-induced oxidation of mtDNA activates the NLRP3 inflammasome [294]. Thus, ROS can enhance both the transcription and post-translational modifications of inflammatory cytokines. IL-1 expression is critical in atherosclerosis. IL-1β plays a crucial role in atherogenesis as it promotes the expression of adhesion molecules and chemokines, including ICAM-1, VCAM-1, and MCP-1, which recruit leukocytes and mononuclear phagocytes during the early stages of atherosclerosis development [295,296]. Furthermore, IL-1β acts as a potent mitogen for human SMC and induces further inflammation by auto-induction and IL-6 stimulation [297,298]. The significance of IL-1 in atherosclerosis was demonstrated in the CANTOS trial, where treatment with canakinumab, a monoclonal antibody targeting IL-1, reduced the primary endpoint of non-fatal myocardial infarction, non-fatal stroke, or cardiovascular death [299].

5.1.5. Destabilization of Fibrous Cap

Atherosclerotic plaque rupture results in thrombus formation that blocks blood vessels, leading to stroke and myocardial infarction [300]. This plaque rupture occurs due to the degradation of the fibrous cap that protects the plaque [301]. Destabilization of the plaques is determined by the high ratio of macrophages to VSMCs, a substantial lipid-rich necrotic core, and a thin fibrous cap [302]. The formation of fibrous caps involves the deposition of collagen-rich extracellular matrix (ECM) components along with the buildup of VSMCs [303]. Matrix metalloproteinases (MMPs) are pivotal in every phase of atherogenesis by regulating vascular inflammation, endothelial dysfunction, VSMCs migration, vascular calcification, extracellular matrix breakdown, as well as plaque activation and destabilization [304]. MMPs exert differential effects on plaque structure and development, potentially depending on the disease stage. For instance, upregulation of MMP-1, MMP-8, and MMP-13 is implicated in atherosclerotic plaque development and ECM collagen degradation [305]. Additionally, the upregulation of gelatinases MMP-2 and MMP-9 that cleave collagen is implicated in destabilized plaques [306,307]. Consistent with these findings, MMP-9 expressed in macrophages leads to plaque disruption, while MMP-12/ApoE double knockout mice exhibit increased VSMC content in plaques [308,309]. Contrary to the previous findings, MMP-3/ApoE double knockout mice showed larger plaque area, implying a protective function [309]. The expression and activity of MMPs are affected by ROS. OxLDL has been shown to upregulate the expression of MMP-1, MMP-2, and MMP-9 [310,311,312], whereas H2O2 activates MMP-1 and MMP-2 [313]. Superoxide derived from monocyte NOX correlates with plasma levels of MMP-9, enhances MMP-9 activity, and is linked to reduced plaque collagen content [314]. Therefore, the above studies suggest that vascular ROS promotes plaque stability by modulating the expression and activities of MMPs.

5.2. The Role of Oxidative Stress in the Pathophysiology of DR

DR is classified as a microvascular complication of diabetes, which poses significant risks to vision [315]. Approximately 400 million individuals globally are diagnosed with type 2 diabetes, with more than 45% of this population experiencing DR [316]. It is widely recognized that DR constitutes the primary cause of diabetes-related visual impairment or loss among working-age adults and the elderly across the globe [317]. Projections indicate that the number of patients suffering from DR may increase to 191.0 million by the year 2030 [318,319,320]. The primary pathophysiology of DR encompasses various alterations induced by hyperglycemia, which include the thickening of the retinal capillary basement membrane, elevated retinal vascular permeability, retinal tissue ischemia, and the upregulation of multiple vasoactive substances, resulting in neovascularization [321]. There are two major types of DR, including nonproliferative (mild, moderate or severe), and proliferative DR [322]. Non-proliferative DR is characterized by the absence of neovascularization, formation of microaneurysms and mild dilation of retinal blood vessels, which is associated with the earliest stages of DR progression [323,324], whereas proliferative DKD is characterized by the formation of new blood vessels on the retinal surface [325]. Due to the inherent instability of these newly formed blood vessels in proliferative DR, the constituents within the vessels, such as blood and extracellular fluids, are prone to leakage, leading to complications such as vitreous hemorrhage and retinal detachment that culminate in vision loss [326].
Oxidative stress is a key factor in the progression of DR. The excessive buildup of ROS damages the tissues surrounding the retinal microvasculature, ultimately contributing to the onset of DR [327]. Studies suggested four principal metabolic abnormalities that contribute to the pathophysiology of hyperglycemia-induced oxidative damage in the retina: (1) activation of the PKC pathway, (2) flux through the polyol pathway, (3) activation of the hexosamine pathway, and (4) intracellular formation of AGEs [328,329]. In addition to these metabolic issues, irregular epigenetic modifications [330], impaired activity of nuclear factors such as the upregulation of NFκB (nuclear factor κB) [331,332] and the downregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) activities [333,334], and mitochondrial dysfunction [335] have also been implicated in the overproduction of ROS in DR. Importantly, oxidative stress driven by epigenetic changes can persist over time, even after blood glucose levels normalized [336]. This phenomenon is known as “metabolic memory [336].” Moreover, hyperglycemia-induced oxidative stress contributes to apoptosis of retinal cells, inflammation, lipid peroxidation, and notable structural and functional changes, including microvascular complications and neurodegeneration in the eyes associated with DR [337]. There are several pathways that may contribute to the pathophysiology of oxidative stress-induced DR (Figure 4).

5.2.1. Mitochondrial Dysfunction

In hyperglycemia, oxidative stress-induced elevated ROS generation leads to mitochondrial dysfunction. The displacement-loop (D-loop) in mitochondrial DNA comprises a substantial non-coding sequence and is a highly vulnerable unwound region, containing essential elements for transcription as well as control regions for mitochondrial DNA (mtDNA) replication [338,339]. In DM, the D-loop experiences more significant impairments and mutations compared to other sections of mtDNA, leading to a reduction in its copy numbers. Furthermore, in DR, hyperglycemia-induced hypermethylation of mtDNA adversely affects its transcription, culminating in mitochondrial dysfunction, which ultimately promotes the apoptosis of retinal microvascular cells [340]. Epigenetic modification of mtDNA has also been corroborated as a latent factor contributing to base mismatch in mtDNA during the pathogenesis of DR [341]. mtDNA-encoded proteins are crucial for the normal functioning of the ETC and mitochondrial homeostasis [342]. In contrast to nuclear DNA, circular mtDNA is susceptible to oxidative stress-induced extensive and persistent damage, due to the absence of protective histones [343]. Damaged mtDNA compromises transcription and protein synthesis, further undermining electron transport and exacerbating ROS generation [340]. Additionally, the activation of matrix metalloproteinases (MMPs) is implicated in mitochondrial dysfunction in DR [344]. Oxidative stress in diabetes upregulates MMPs expression [345] through the activation of the Nox complex [346]. Oxidative stress in diabetes has been shown to increase the translocation of MMPs into the mitochondria [347]. This process involves the translocation and accumulation of redox-sensitive MMPs, specifically MMP-2 and MMP-9, within the retinal mitochondria. This translocation depends on the modulation of chaperones Hsp60 and Hsp70 [348,349]. Once inside the mitochondria, MMPs disrupt mitochondrial function and increase pore permeability by damaging connexin 43 [350,351]. The compromised lipid membrane of the mitochondria leads to mitochondrial swelling in the diabetic retinas [352] and promotes the leakage of cytochrome c (Cyt c) into the cytosol, which in turn triggers the assembly of the apoptosome platform to initiate caspase cascade [353,354]. Additionally, peroxynitrite, an extremely reactive molecule, is formed when superoxide reacts with nitric oxide (NO) [355]. Peroxynitrite oxidizes glutathione (GSH), cysteine, and tetrahydrobiopterin [356], and subsequently oxidizes membrane phospholipids, inactivates enzymes containing sulfhydryl groups, triggers the nitration of tyrosine residues, and exacerbates DNA fragmentation [357]. Furthermore, peroxynitrite causes irreversible damage to mitochondria by disrupting mitochondrial energy and calcium homeostasis and promoting the opening of the permeability transition pore, ultimately leading to cell apoptosis [358].

5.2.2. Cellular Apoptosis and Inflammation in the Retina

In DR, the apoptosis of retinal cells occurs early on. The accelerated death of retinal capillary cells is evident before any histopathological changes linked to this complication appear [214,359]. Hyperglycemia-induced oxidative stress in retinal endothelial cells and pericytes upregulates the activities of Caspase-3, NFκB, and other transcription factors that promote capillary cell apoptosis [329,360]. Caspases play an important role in apoptosis and are vulnerable to oxidative stress-induced damage [361,362]. Hyperglycemia-induced ROS generation in the mitochondria increases the organelle’s pore permeability [363,364], leading to the release of Cyt c and other pro-apoptotic factors to trigger apoptosis by activating caspases [365,366]. At the early stage in apoptosis, Cyt c in retinal capillary cells [367] activates Caspase-9 and subsequently activates Caspase-3, leading to DNA fragmentation [367,368]. Furthermore, NFκB plays an important role in hyperglycemia-induced inflammation and cell death due to its proinflammatory and proapoptotic properties [369,370]. The activity of retinal NFκB in diabetes is upregulated early in retinopathy development and remains active even as apoptosis in retinal capillary cells accelerates [371]. As a redox-sensitive nuclear transcription factor, NFκB significantly regulates inflammatory responses and inhibits antioxidant enzymes [372]. NFκB is implicated in starting proapoptotic processes in retinal pericytes in response to high glucose, explaining the early pericyte death in DR [373]. Upregulation of NFκB signaling pathways can activate MMP-9 [374], leading to increased mitochondrial MMP-9, which damages the mitochondrial gap junction protein connexin-43 and further enhances pore permeability. This contributes to the leakage of Cyt c into the cytosol [351]. Additionally, NFκB increases NO production by upregulating the expression of inducible nitric oxide synthase [375,376], which upregulates NFκB’ s transcriptional activity [377].

5.2.3. Lipid Peroxidation

Oxygen-derived free radicals, including hydroxyl and hydroperoxyl species, have been shown to oxidize phospholipids and other lipid components in the plasma membrane, resulting in lipid peroxidation. Tissues affected by oxidative stress-related retinal diseases have demonstrated a significant presence of metabolic products from lipid peroxidation, thereby indicating a correlation between oxidative stress and lipid peroxidation [378]. Hyperglycemia-induced oxidative stress in DM plays a pivotal role in augmenting lipid peroxidation in DR [379]. Furthermore, lipid peroxidation exhibits a positive association with the severity and duration of DM [380]. The high concentration of polyunsaturated fatty acids (PUFAs) in the retina enhances its susceptibility to oxidative stress due to the sensitivity of PUFAs to oxidation [381]. Intracameral administration of H2O2 into the rabbit retina resulted in an increase in lipid peroxidation within the membranes of iris epithelial cells [382]. Additionally, lipid metabolism contributes to the generation of ROS, such as H2O2, which facilitates the senescence of retinal pigment epithelial (RPE) cells, thereby exacerbating the progression of DR. Concurrently, lipid peroxidation products may promote the leakage of ROS from mitochondria [383]. Lipid peroxidation results in cellular membranes with compromised integrity and the formation of diffusible cytotoxic reactive aldehydes, including 4-hydroxy-2-nonenal (4HNE) and 4-hydroxyhexenal (HHE). 4HNE has been shown to activate the canonical WNT signaling pathway through oxidative stress that leads to the pathogenesis of DR [381]. Additionally, 4HNE induces apoptosis of RPE cells through p53 activation [384]. HHE has been shown to induce permeable transition pores in the mitochondrial membrane and augment NFκB-induced upregulation of pro-inflammatory genes [385]. Research on neurodegenerative models of the retina has shown an increase in lipid peroxidation that correlates with neuronal loss [386]. Hyperglycemia-induced ROS generation in mitochondria directly contributes to mitochondrial dysfunction. Consequently, ROS-mediated mitochondrial dysfunction results in the accumulation of lipid droplets within glial cells that may be peroxidized by ROS, leading to the initiation of neurodegeneration in the retina [387].

5.2.4. Changes in Retinal Microvasculature

Oxidative stress-induced metabolic impairments lead to various effects on the retinal microvasculature’s structure and function, including the thickening of the capillary basement membrane (CBM), disruption of the blood-retinal barrier (BRB), and the formation of acellular and occluded capillaries [388]. The thickening of the CBM is a consistent characteristic of DR, due to upregulated expression and decreased degeneration of extracellular matrix (ECM) proteins [389]. Hyperglycemia-induced oxidative stress and advanced glycation in diabetes contribute to the thickening of CBM in DR [390]. Hyperglycemia-induced ROS generation increases the activity of proinflammatory transcription factors that lead to thickening of the CBMs by upregulating the expression of ECM proteins, including fibronectin (FN) and collagen, in retinal endothelial cells [389,391]. AGE formation from ROS on collagen causes cross-linking, resulting in structural rigidity and limiting the transport of growth factors across membranes, which contributes to the loss of pericytes and endothelial cells [392]. Treatment with the AGE inhibitor aminoguanidine has shown protective effects against CBM thickening in diabetic rats [393]. Structural changes induced by oxidative stress, such as the loss of intercellular junctions and pericyte apoptosis, correlate with functional changes like alterations in blood flow and increased vessel permeability, both of which contribute to the pathogenesis of DR [394]. Oxidative stress is implicated in the damage of the BRB, a selective barrier that regulates substance exchange between the neural retina and circulating blood, ensuring retinal health by supplying nutrients and removing metabolic waste and toxins [395]. BRB breakdown leads to increased vascular permeability, leading to diabetic macular edema, a significant cause of vision loss among diabetic patients. ROS-induced elevation of VEGF levels plays a key role in BRB destruction [395], confirmed through intravitreal VEGF injections or implantation of VEGF-releasing pellets [396,397]. Occludin, claudin, and zonula occludens-1 (ZO-1) are intercellular junction proteins, essential for maintaining BRB integrity, and the breakdown of the BRB is closely linked to the dysfunction of these junction-associated proteins. For instance, oxidative stress upregulates VEGF expression, which in turn induces the expression of urokinase plasminogen activator receptor (uPAR), which results in the breakdown of the BRB. Inhibiting VEGF or uPAR expressions has been found to preserve BRB integrity in diabetic models [398]. Prolonged exposure to VEGF results in the loss of Claudin-1, jeopardizing BRB integrity [396]. Furthermore, oxidative stress-induced activation of NFκB in DR, regulates the transcription of various genes, including suppression of ZO-1 by this inflammatory regulator, which compromises BRB integrity [399]. Chronic hyperglycemia-induced activation of PKC-δ, initiating a signaling cascade that results in pericyte apoptosis [400]. As pericytes die, the structure of retinal microvessels is gradually altered, leading to BRB disruption [316]. Secretion of proangiogenic cytokines, growth factors, and proteases by proinflammatory cells has been shown to promote angiogenesis at inflammation sites in DR [401]. Oxidative stress-induced upregulation of prostaglandin E2 (PGE2) and Cyclooxygenase-2 (COX-2), promotes the expression of proangiogenic factor, VEGF, in diabetic retinas, which increases retinal neovascularization [402,403,404]. Oxidative stress-induced apoptosis of retinal neurons and pericytes in DR augments the growth of acellular and occluded capillaries, leading to microaneurysm formation and increased leukostasis, which contributes to CBM thickening [316]. Endothelin-1 is an important vasoactive peptide that regulates blood flow and permeability in the retinal microvasculature. ROS upregulates the transcription of endothelin-1 [405,406], which can promote CBM thickening by increasing the synthesis of extracellular matrix proteins [407,408].

5.3. The Role of Oxidative Stress in the Pathophysiology of DKD

DKD is attributed to complex interactions among various pathways, initiated by hyperglycemia and hemodynamic changes associated with diabetes [409]. These changes lead to albuminuria, deteriorating kidney function, renal fibrosis, and inflammation. Additionally, oxidative stress is recognized as a key factor connecting hyperglycemia to vascular complications of diabetes, especially DKD [410]. There are several pathways that may contribute to the pathophysiology of oxidative stress-induced DKD (Figure 5).

5.3.1. Pi3k/Akt Signaling Pathway

The phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway regulates cell proliferation, differentiation, and apoptosis. In response to extracellular stimuli, PI3K phosphorylates phosphatidylinositol 4,5-bisphosphate (PIP2), leading to the production of phosphatidylinositol-3,4,5-triphosphate (PIP3). The resulting PIP3 subsequently induces the translocation of Akt to the plasma membrane and activates Akt through the enzymatic action of phosphoinositide-dependent kinase-1 (PDK1) [411]. The activated Akt then in turn activates the downstream signaling molecules, including glycogen synthase kinase (GSK) 3β, mammalian target of rapamycin (mTOR), and forkhead box protein O1 (FoxO1) to show its pathophysiological effects [412].
GSK3β is one of the downstream targets of Akt. Activated Akt antagonizes the pathophysiological effects of GSK3β by phosphorylating it. Studies demonstrated that chronic high glucose exposure suppresses PI3K/Akt signaling pathway, resulting in decreased GSK3 activity, reduced Bcl-2, and increased Bax, which induces renal cell apoptosis [413]. An in vivo study in DKD mice demonstrated that phosphorylation of GSK3β increased inflammation by augmenting the activity of NFκB [414]. Another study indicated that diminished GSK3β activity elevated β-catenin levels and slowed the degradation of Snail, which enhanced the trans-differentiation of tubular epithelial cells and fostered renal interstitial fibrosis in diabetic rats [415].
mTOR has emerged as a key research target because of its strong association with Akt-mediated signaling pathways. When Akt is activated, it phosphorylates mTOR and its downstream pathways, affecting cellular functions such as proliferation, apoptosis, and glucose metabolism [416]. Elevated glucose levels have been shown to promote tubular epithelial NRK-52E cells to produce ROS, which in turn trigger the release of transforming growth factor-beta 1 (TGF-β1) and activated Akt. The activated Akt phosphorylates mTOR, facilitating the epithelial-mesenchymal transition (EMT) in NRK-52E cells, exacerbating diabetic kidney fibrosis [417]. Furthermore, high glucose levels are implicated in stimulating mTOR signaling, leading to a decrease in the LC3II/LC3I ratio and reduced Beclin levels, which inhibit autophagy and induce the progression of DKD [418].
FoxO1 consists of a set of highly conserved transcription factors found throughout the body, playing vital roles in oxidative stress, inflammation, autophagy, and apoptosis in response to elevated glucose levels [419]. Phosphorylation of FoxO1 by the PI3K/Akt signaling pathway results in its inactivation of nuclear translocation, which is essential for the onset of diabetic kidney injury [420]. In renal tissues, FoxO1 can aid in repairing the damaged glomerular filtration barrier and reducing apoptosis caused by the detachment of renal podocytes from the basement membrane due to impaired glucose metabolism [421]. An initial spike in glucose levels activates the TGF-β/Smad pathway, which helps mitigate kidney fibrosis in DKD [422]. Additionally, elevated phosphorylation of FoxO1 downregulated its activity and autophagy in DKD animal models, aggravating kidney damage [423].

5.3.2. NFκB Signaling Pathway

Under normal physiological conditions, NFκB is expressed throughout various cells in an inactive form. When stimulated by agonists, including proinflammatory cytokines, Toll-like receptors, p38-MAPK, HO-1, and ROS, NFκB dimers (p65 and p50) are translocated into the nucleus to regulate the expression of target genes, responsible for the body’s immune and inflammatory responses [424]. It is already well studied that upregulation of NFκB activity is linked to the pathophysiology of DKD. For instance, hyperglycemia-induced generation of ROS and inflammatory factors, including TNF and IL-6 activate the NFκB signaling pathway, and subsequently upregulates the transcription of proinflammatory cytokines, chemokines, adhesion molecules, and TGF-β1, which ultimately leads to cellular apoptosis, necrosis, and tissue fibrosis, all of which expedite the progression of DKD [425]. A study demonstrated that NFκB expression was significantly elevated in DKD patients compared to non-diabetic subjects, and this expression level correlated positively with the severity of proteinuria in those affected [426]. All these studies collectively suggest that, attenuating the upregulation of NFκB signaling pathway may offer a promising avenue for research aimed at preventing and treating DKD.

5.3.3. Nrf2/ARE Signaling Pathway

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a crucial transcription factor that enhances the body’s defense against oxidative stress. It regulates the transcription of several antioxidants, anti-inflammatory, anti-apoptotic, and anti-fibrotic factors, and its activity is negatively correlated with the severity of DKD. Under normal physiological circumstances, Nrf2 is found in the cytoplasm in an inactive form conjugated with its inhibitor, Kelch-like ECH-associated protein 1 (KEAP1). However, hyperglycemia-induced oxidative stress rescues Nrf2 from KEAP1, leading to its activation. Once activated, Nrf2 translocates into the nucleus and interacts with AREs to upregulate the expression of antioxidant factors, thereby protecting cells and alleviating the progression of kidney injury in DKD [427]. A study in mice revealed that Nrf2−/− mice experienced more severe kidney damage compared to wild-type control mice, suggesting the protective role of Nrf2 in kidney diseases [428]. Another study demonstrated that inhibiting Nrf2-mediated antioxidant signaling pathway downregulated the transcription of antioxidant enzymes, such as SOD, catalase (CAT), and glutathione peroxidase (GSH-Px), and ultimately worsened kidney injury in mice with spontaneous DKD [429]. Additionally, a study in rodents found that blocking Nrf2 signaling while activating TGFβ-smad signaling resulted in a delay in renal fibrosis in STZ-induced diabetic rats [430]. Furthermore, increasing Nrf2 levels are associated with a reduction in TNF, IL-6, Bax, and p38 levels, suppressed NFκB activation, and upregulated Bcl-2 expression, demonstrating anti-inflammatory and anti-apoptotic effects in STZ-induced DKD rats [431]. All these studies collectively suggest that Nrf2 could serve as a critical target for preventing DKD and preserving renal function.

5.3.4. TGFβ Signaling Pathway

Oxidative stress-induced activation of the TGF-β1 signaling pathway upregulates the expression of protein kinases or cytokines, followed by ECM accumulation and EMT, ultimately resulting in renal interstitial fibrosis and glomerulosclerosis in DKD [432]. A study in a human cohort indicated that serum TGF-β1 levels can be served as a potential biomarker for early detection of fibrosis in DKD [433]. Resveratrol, Salvia Root, Taxol, and Calcitriol are potential drugs that alleviate fibrosis in DKD by antagonizing the TGF-β1 signaling pathway [434]. One of the major downstream targets of TGF-β1-induced profibrotic activities in the pathogenesis of DKD is ERK. The activation of the TGF-β1/ERK pathway is implicated in the pathogenesis of kidney fibrosis. Multiple studies in animal model demonstrated an improvement in kidney fibrosis upon inhibition of the TGF-β1/ERK pathway [435].

5.3.5. JAK2/STAT3 Signaling Pathway

DKD is associated with an augmented activation of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway, whereas inhibiting this pathway ameliorate DKD progression [436]. A study in DKD rats showed elevated phosphorylation of JAK and STAT3 in renal tissue, which results in an upregulation of Bax, and a downregulation of Bcl-2 expression followed by an increased renal cell apoptosis [437]. Another study in mouse with diabetic nephropathy demonstrated that the activation of JAK2/STAT3 signaling pathway in the kidney residual macrophages induced the release of proinflammatory factors and ROS, worsening kidney damage [438]. CXCL6 may stimulate the production of TGF-β1, collagen I, collage III, MMP2, and MMP9 through the activation JAK/STAT pathway, and thereby accelerating the progress of renal fibrosis [439]. These findings collectively suggest that the JAK2/STAT3 signaling pathway primarily plays a role in governing immune response, inflammation, oxidative stress, and cell apoptosis throughout the development of DKD.

5.3.6. AMPK Signaling Pathway

AMPK is well-known as a cellular energy sensor and has garnered significant attention as a therapeutic target for obesity, diabetes, and diseases related to metabolic syndrome. Studies in humans demonstrated that the expression of phosphorylated AMPK (p-AMPK) protein levels is lower in the renal tissues of DKD patients. Furthermore, treatment with an AMPK agonist has been found to alleviate clinical symptoms, improve pathological changes, and reduce 24-h urinary protein levels in these patients [440]. Another study in diabetic rats showed that inactivation of AMPK in the renal cortex leads to NOX4 up-regulation, activation of TGFβ-1 signaling and increased ECM accumulation [441]. Upon activation, AMPK has been shown to increase the expression of SODs to diminish superoxide production and phosphorylates and activates FOXO to provide antioxidant effects [442]. Both in vivo and in vitro studies showed that AMPK, p-AMPK, SIRT1, NFκB levels were significantly reduced in STZ-induced DKD mice and in high-glucose-treated podocytes, triggering oxidative stress-induced pyroptosis [443]. High glucose-induced inhibition of AMPK phosphorylation upregulates the expression of PPAR-α, pro-inflammatory and profibrotic genes in proximal tubular epithelial cells (PTECs), promotes intrarenal lipid accumulation and apoptosis of PTECs [444]. Another study demonstrated that agonist-stimulated activation of AMPK increases mitophagy through the p-AMPK/Pink1/Parkin signaling pathway, thereby ameliorate renal oxidative stress and tubulointerstitial fibrosis in high-fat diet/STZ-induced diabetic mice [445].

6. Therapeutic Approaches to Tackle Oxidative Stress-Induced DVDs

Oxidative stress remains a central pathological mechanism underlying vascular complications in diabetes, driven primarily by persistent hyperglycemia, dyslipidemia, and insulin resistance, which collectively promote excessive production of ROS through mitochondrial dysfunction, NOX activation, and eNOS uncoupling. These pathways lead to endothelial dysfunction, vascular inflammation, and eventual macro- and microvascular complications [6]. As a result, therapeutic strategies have progressively shifted from conventional antioxidants to mechanistically targeted interventions. Among glucose-lowering agents, sodium glucose co-transporter 2 (SGLT2) inhibitors, including dapagliflozin, empagliflozin, and canagliflozin have demonstrated pleiotropic vascular protective effects beyond glycemic control. These agents mitigate mitochondrial ROS production, suppress NOX4 expression, activate the AMPK/SIRT1/PGC-1α axis, preserve mitochondrial membrane potential, and enhance eNOS phosphorylation, culminating in improved endothelial function and reduced arterial stiffness in both preclinical and human studies [446]. Similarly, thiazolidinediones (TZDs), such as pioglitazone and rosiglitazone, exert antioxidant effects by activating peroxisome proliferator-activated receptor gamma (PPARγ), reducing systemic and vascular oxidative stress, and attenuating NOX-mediated ROS production, partly through AMPK activation [447,448]. Another major class of therapeutic agents showing promise is glucagon-like peptide-1 receptor agonists (GLP-1 RAs), including liraglutide, semaglutide, and dulaglutide. These drugs reduce ROS generation through multiple mechanisms: by enhancing mitochondrial biogenesis and fusion (via PGC-1α), activating cAMP/PKA and PI3K/Akt pathways, increasing expression of antioxidant enzymes such as SOD2 and catalase, and restoring endothelial NO bioavailability [449,450]. Recent studies also demonstrate that GLP-1 RAs inhibit NOX activity and reduce inflammatory cytokine production in endothelial and vascular smooth muscle cells [451,452]. Importantly, large clinical trials like LEADER and SUSTAIN-6 have established cardiovascular benefits of GLP-1 RAs, correlating with improved vascular oxidative s tress biomarkers in patients with type 2 diabetes [453].
Beyond glucose-lowering agents, therapeutic approaches increasingly target endogenous antioxidant defense systems. Nrf2 activation via phytochemicals like sulforaphane or pharmacological agents such as bardoxolone methyl enhances transcription of AREs including heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), and glutathione peroxidase (GPx), thereby reducing vascular inflammation and oxidative injury [454,455]. Although bardoxolone methyl initially raised concerns regarding fluid retention in the BEACON trial, refined Nrf2 modulators (e.g., omaveloxolone) are currently in clinical evaluation with improved safety profiles [456]. NOX inhibitors have emerged as another promising avenue. For instance, GKT137831 selectively inhibits NOX1 and NOX4, reducing ROS generation and fibrotic signaling in diabetic vasculature [457]. These findings were corroborated in animal models of diabetic atherosclerosis and nephropathy, although human trials remain limited. Mitochondria-targeted antioxidants such as MitoQ, SS-31 (elamipretide), and MitoTEMPO represent advanced therapeutic tools capable of accumulating within the mitochondrial matrix to neutralize mtROS at the source. These agents restore mitochondrial membrane potential, suppress lipid peroxidation, and prevent mitochondrial DNA damage, ultimately preserving vascular endothelial function [458,459]. For instance, SS-31 has shown efficacy in reducing vascular stiffness and improving endothelial-dependent vasodilation in older adults with impaired mitochondrial energetics [460].
A novel frontier in redox biology is the regulation of ferroptosis, an iron-dependent, lipid peroxidation-driven form of regulated cell death which has been implicated in the progression of diabetic vasculopathy, nephropathy, and cardiomyopathy. High glucose environments promote ferroptosis by suppressing GPx4 activity and depleting glutathione, leading to accumulation of oxidized phospholipids and endothelial cell death [461]. Interventions such as ferrostatin-1, deferoxamine, or curcumin have shown protective effects against ferroptosis in vascular tissues under hyperglycemic conditions [462,463]. Nano-formulated curcumin exhibits enhanced bioavailability and ROS scavenging properties, suppressing ferroptotic gene expression and improving vascular remodeling in diabetic models [464,465,466]. In parallel, emerging nanomedicine platforms allow for targeted, ROS-responsive delivery of antioxidants or gene-editing cargo. Nanocarriers such as polymeric nanoparticles, lipid vesicles, or exosomes can deliver antioxidants like resveratrol, mangiferin, and metformin directly to sites of vascular inflammation, enhancing therapeutic efficacy while minimizing systemic toxicity [467,468,469].
Gene-based interventions are also being explored, including the delivery of plasmids or viral vectors encoding antioxidant enzymes (e.g., SOD2, catalase, Nrf2) directly to vascular tissues, or CRISPR-based activation of protective genes [470,471,472,473]. These strategies aim to restore redox balance in a durable, cell-type-specific manner. Additional therapeutic targets include epigenetic regulators of redox homeostasis. Histone deacetylase (HDAC) inhibitors and SIRT1 activators (such as resveratrol or nicotinamide mononucleotide) have demonstrated benefits in improving insulin sensitivity, suppressing inflammatory gene transcription, and enhancing mitochondrial antioxidant defenses [474,475,476]. Modulation of the AGE pathway via RAGE antagonists or pro-resolving mediators like Annexin A1 further reduces vascular ROS generation and leukocyte adhesion in diabetic endothelium [477,478]. Adjunct lifestyle interventions such as caloric restriction, time-restricted feeding, and Mediterranean diets rich in flavonoids also support vascular redox homeostasis by activating AMPK, Nrf2, and sirtuin pathways, while aerobic exercise upregulates endogenous antioxidant enzymes and improves mitochondrial function [479]. Importantly, combination therapies that integrate metabolic agents (e.g., SGLT2 inhibitors, GLP-1 RAs) with redox modulators (e.g., NOX inhibitors, ferroptosis blockers, or Nrf2 activators) show additive or synergistic effects in diabetic animal models and are being increasingly explored in early-phase clinical trials [480,481,482,483]. The pathogenic model of most DVDs centers heavily on oxidative stress as a major contributor. ROS, such as superoxide and hydrogen peroxide, mediate oxidative stress-induced endothelial dysfunction, oxLDL formation, vessel wall inflammation, and smooth muscle proliferation. In vitro and in vivo studies consistently support this model. Antioxidants ranging from vitamins C and E to flavonoids, coenzyme Q10, and mineral cofactors effectively quench ROS, enhance nitric oxide availability, and preserve endothelial function in experimental settings. These findings underpin the rationale for antioxidant therapies in human vascular disease [484].
However, this elegant mechanistic narrative starkly contrasts with the results of large-scale human trials. Multiple high-powered randomized clinical trials (RCTs) testing antioxidant vitamins in vascular disease have failed to demonstrate benefit and, in some cases, suggested harm. For instance, vitamins E, C, D, β-carotene showed no significant reduction in cardiovascular events in major secondary prevention trials [484,485]. Succinobucol, a potent lipoprotein-associated antioxidant did not reduce/improve events in the ARISE trial on 6144 patients with acute coronary syndrome [485]. The HOPE-TOO trial evaluated whether long-term (over 7 years) supplementation with vitamin E would reduce cardiovascular events in high-risk cardiovascular patients (n ≈ 7030). However, no significant reduction in major cardiovascular events, including myocardial infarction, stroke, and cardiovascular death was observed in the vitamin E group compared to placebo [486]. Coenzyme Q10 (CoQ10) [296], resveratrol [486], melatonin [487], N-Acetylcysteine (NAC) [488], mitochondria-targeted antioxidant (mitoQ) [489,490] in various hypertensive and cardiovascular trials showed occasional biomarker improvements (e.g., blood pressure, flow-mediated dilation) but no consistent clinical benefit. The failure of potential antioxidant therapy is stated in Table 2.
It is now well appreciated that the failure of large-scale antioxidant trials in the past such as the HOPE-TOO trial, which showed no cardiovascular benefit from vitamin E and C in high-risk patients was largely due to their non-specificity, failure to distinguish between physiological and pathological ROS, and lack of targeted delivery [495]. In contrast, the current precision-based strategies emphasize spatial and temporal control over oxidative stress, employing molecular diagnostics and biomarkers (e.g., oxidized LDL, 8-iso-PGF2α, or MDA levels) to guide treatment. This evolving therapeutic paradigm seeks not to eliminate ROS entirely, but rather to recalibrate redox signaling pathways that are aberrantly activated in diabetes. As our understanding of redox systems deepens, and as tools for targeted delivery and pathway-specific modulation continue to advance, there is renewed optimism that oxidative stress-induced vascular disease in diabetes may become more tractable. Future directions should focus on biomarker-guided clinical trials, combination regimens tailored to specific vascular phenotypes, and the integration of redox therapeutics with other emerging technologies such as single-cell transcriptomics and vascular imaging to assess real-time ROS dynamics. Together, these developments mark a significant leap forward in the quest to combat diabetes-associated vascular disease through redox-based precision medicine.

7. Conclusions

In conclusion, diabetes-induced oxidative stress plays a pivotal role in the pathogenesis of vascular diseases, contributing significantly to morbidity and mortality in diabetic populations. Chronic hyperglycemia drives excessive ROS production, leading to lipid, protein, and DNA damage and compromising vascular integrity. This oxidative environment induces endothelial dysfunction by reducing NO bioavailability, promoting vasoconstriction, inflammation, and thrombosis. Activation of redox-sensitive transcription factors further amplifies inflammation by upregulating adhesion molecules and cytokines, facilitating leukocyte infiltration into the endothelium and accelerating vascular complications in the heart, kidneys, and eyes. Emerging evidence suggests that targeting oxidative stress through antioxidant therapies, lifestyle interventions, and pharmacological agents may help preserve vascular function in diabetes. However, clinical translation remains challenging, underscoring the need for precise, personalized strategies to modulate redox balance. Addressing oxidative stress is essential for preventing vascular complications and improving outcomes in diabetic patients.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Dilworth, L.; Facey, A.; Omoruyi, F. Diabetes Mellitus and Its Metabolic Complications: The Role of Adipose Tissues. Int. J. Mol. Sci. 2021, 22, 7644. [Google Scholar] [CrossRef] [PubMed]
  2. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef] [PubMed]
  3. Khan, M.A.B.; Hashim, M.J.; King, J.K.; Govender, R.D.; Mustafa, H.; Al Kaabi, J. Epidemiology of Type 2 Diabetes—Global Burden of Disease and Forecasted Trends. J. Epidemiol. Glob. Health 2020, 10, 107–111. [Google Scholar] [CrossRef]
  4. Ruze, R.; Liu, T.; Zou, X.; Song, J.; Chen, Y.; Xu, R.; Yin, X.; Xu, Q. Obesity and type 2 diabetes mellitus: Connections in epidemiology, pathogenesis, and treatments. Front. Endocrinol. 2023, 14, 1161521. [Google Scholar] [CrossRef] [PubMed]
  5. Li, Y.; Liu, Y.; Liu, S.; Gao, M.; Wang, W.; Chen, K.; Huang, L.; Liu, Y. Diabetic vascular diseases: Molecular mechanisms and therapeutic strategies. Signal Transduct. Target. Ther. 2023, 8, 152. [Google Scholar] [CrossRef]
  6. Yang, D.R.; Wang, M.Y.; Zhang, C.L.; Wang, Y. Endothelial dysfunction in vascular complications of diabetes: A comprehensive review of mechanisms and implications. Front. Endocrinol. 2024, 15, 1359255. [Google Scholar] [CrossRef]
  7. Song, J.; Gao, N.; Chen, Z.; Xu, G.; Kong, M.; Wei, D.; Sun, Q.; Dong, A. Shared genetic etiology of vessel diseases: A genome-wide multi-traits association analysis. Thromb. Res. 2024, 241, 109102. [Google Scholar] [CrossRef]
  8. Hong, Y.; Boiti, A.; Vallone, D.; Foulkes, N.S. Reactive Oxygen Species Signaling and Oxidative Stress: Transcriptional Regulation and Evolution. Antioxidants 2024, 13, 312. [Google Scholar] [CrossRef]
  9. Jena, A.B.; Samal, R.R.; Bhol, N.K.; Duttaroy, A.K. Cellular Red-Ox system in health and disease: The latest update. Biomed. Pharmacother. 2023, 162, 114606. [Google Scholar] [CrossRef]
  10. Caturano, A.; D’Angelo, M.; Mormone, A.; Russo, V.; Mollica, M.P.; Salvatore, T.; Galiero, R.; Rinaldi, L.; Vetrano, E.; Marfella, R.; et al. Oxidative Stress in Type 2 Diabetes: Impacts from Pathogenesis to Lifestyle Modifications. Curr. Issues Mol. Biol. 2023, 45, 6651–6666. [Google Scholar] [CrossRef]
  11. Calcutt, N.A.; Cooper, M.E.; Kern, T.S.; Schmidt, A.M. Therapies for hyperglycaemia-induced diabetic complications: From animal models to clinical trials. Nat. Rev. Drug Discov. 2009, 8, 417–429. [Google Scholar] [CrossRef] [PubMed]
  12. Prattichizzo, F.; De Nigris, V.; Mancuso, E.; Spiga, R.; Giuliani, A.; Matacchione, G.; Lazzarini, R.; Marcheselli, F.; Recchioni, R.; Testa, R.; et al. Short-term sustained hyperglycaemia fosters an archetypal senescence-associated secretory phenotype in endothelial cells and macrophages. Redox Biol. 2018, 15, 170–181. [Google Scholar] [CrossRef] [PubMed]
  13. Rains, J.L.; Jain, S.K. Oxidative stress, insulin signaling, and diabetes. Free Radic. Biol. Med. 2011, 50, 567–575. [Google Scholar] [CrossRef] [PubMed]
  14. Dinic, S.; Arambasic Jovanovic, J.; Uskokovic, A.; Mihailovic, M.; Grdovic, N.; Tolic, A.; Rajic, J.; Dordevic, M.; Vidakovic, M. Oxidative stress-mediated beta cell death and dysfunction as a target for diabetes management. Front. Endocrinol. 2022, 13, 1006376. [Google Scholar] [CrossRef]
  15. Chen, X.; Xie, N.; Feng, L.; Huang, Y.; Wu, Y.; Zhu, H.; Tang, J.; Zhang, Y. Oxidative stress in diabetes mellitus and its complications: From pathophysiology to therapeutic strategies. Chin. Med. J. 2025, 138, 15–27. [Google Scholar] [CrossRef]
  16. Gonzalez, P.; Lozano, P.; Ros, G.; Solano, F. Hyperglycemia and Oxidative Stress: An Integral, Updated and Critical Overview of Their Metabolic Interconnections. Int. J. Mol. Sci. 2023, 24. [Google Scholar] [CrossRef]
  17. Ighodaro, O.M. Molecular pathways associated with oxidative stress in diabetes mellitus. Biomed. Pharmacother. 2018, 108, 656–662. [Google Scholar] [CrossRef]
  18. Andres, C.M.C.; Perez de la Lastra, J.M.; Andres Juan, C.; Plou, F.J.; Perez-Lebena, E. Superoxide Anion Chemistry-Its Role at the Core of the Innate Immunity. Int. J. Mol. Sci. 2023, 24, 1841. [Google Scholar] [CrossRef]
  19. Wong, H.S.; Dighe, P.A.; Mezera, V.; Monternier, P.A.; Brand, M.D. Production of superoxide and hydrogen peroxide from specific mitochondrial sites under different bioenergetic conditions. J. Biol. Chem. 2017, 292, 16804–16809. [Google Scholar] [CrossRef]
  20. Read, A.D.; Bentley, R.E.; Archer, S.L.; Dunham-Snary, K.J. Mitochondrial iron-sulfur clusters: Structure, function, and an emerging role in vascular biology. Redox Biol. 2021, 47, 102164. [Google Scholar] [CrossRef]
  21. Tirichen, H.; Yaigoub, H.; Xu, W.; Wu, C.; Li, R.; Li, Y. Mitochondrial Reactive Oxygen Species and Their Contribution in Chronic Kidney Disease Progression Through Oxidative Stress. Front. Physiol. 2021, 12, 627837. [Google Scholar] [CrossRef] [PubMed]
  22. Rauf, A.; Khalil, A.A.; Awadallah, S.; Khan, S.A.; Abu-Izneid, T.; Kamran, M.; Hemeg, H.A.; Mubarak, M.S.; Khalid, A.; Wilairatana, P. Reactive oxygen species in biological systems: Pathways, associated diseases, and potential inhibitors-A review. Food Sci. Nutr. 2024, 12, 675–693. [Google Scholar] [CrossRef] [PubMed]
  23. Checa, J.; Aran, J.M. Reactive Oxygen Species: Drivers of Physiological and Pathological Processes. J. Inflamm. Res. 2020, 13, 1057–1073. [Google Scholar] [CrossRef] [PubMed]
  24. Niemann, B.; Rohrbach, S.; Miller, M.R.; Newby, D.E.; Fuster, V.; Kovacic, J.C. Oxidative Stress and Cardiovascular Risk: Obesity, Diabetes, Smoking, and Pollution: Part 3 of a 3-Part Series. J. Am. Coll. Cardiol. 2017, 70, 230–251. [Google Scholar] [CrossRef]
  25. Alhayaza, R.; Haque, E.; Karbasiafshar, C.; Sellke, F.W.; Abid, M.R. The Relationship Between Reactive Oxygen Species and Endothelial Cell Metabolism. Front. Chem. 2020, 8, 592688. [Google Scholar] [CrossRef]
  26. Mauersberger, C.; Hinterdobler, J.; Schunkert, H.; Kessler, T.; Sager, H.B. Where the Action Is-Leukocyte Recruitment in Atherosclerosis. Front. Cardiovasc. Med. 2021, 8, 813984. [Google Scholar] [CrossRef]
  27. De Maranon, A.M.; Iannantuoni, F.; Abad-Jimenez, Z.; Canet, F.; Diaz-Pozo, P.; Lopez-Domenech, S.; Jover, A.; Morillas, C.; Marino, G.; Apostolova, N.; et al. Relationship between PMN-endothelium interactions, ROS production and Beclin-1 in type 2 diabetes. Redox Biol. 2020, 34, 101563. [Google Scholar] [CrossRef]
  28. Luna-Marco, C.; de Maranon, A.M.; Hermo-Argibay, A.; Rodriguez-Hernandez, Y.; Hermenejildo, J.; Fernandez-Reyes, M.; Apostolova, N.; Vila, J.; Sola, E.; Morillas, C.; et al. Effects of GLP-1 receptor agonists on mitochondrial function, inflammatory markers and leukocyte-endothelium interactions in type 2 diabetes. Redox Biol. 2023, 66, 102849. [Google Scholar] [CrossRef]
  29. Taverner, K.; Murad, Y.; Yasunaga, A.B.; Furrer, C.; Little, J.; Li, I.T.S. The effect of type-2 diabetes conditions on neutrophil rolling adhesion. BMC Res. Notes 2022, 15, 355. [Google Scholar] [CrossRef]
  30. Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc. Pharmacol. 2018, 100, 1–19. [Google Scholar] [CrossRef]
  31. Zhang, Z.; Huang, Q.; Zhao, D.; Lian, F.; Li, X.; Qi, W. The impact of oxidative stress-induced mitochondrial dysfunction on diabetic microvascular complications. Front. Endocrinol. 2023, 14, 1112363. [Google Scholar] [CrossRef] [PubMed]
  32. Nishikawa, T.; Edelstein, D.; Du, X.L.; Yamagishi, S.; Matsumura, T.; Kaneda, Y.; Yorek, M.A.; Beebe, D.; Oates, P.J.; Hammes, H.P.; et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000, 404, 787–790. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Z.; Zhao, Y.; Sun, R.; Sun, Y.; Liu, D.; Lin, M.; Chen, Z.; Zhou, J.; Lv, L.; Tian, X.; et al. circ-CBFB upregulates p66Shc to perturb mitochondrial dynamics in APAP-induced liver injury. Cell Death Dis. 2020, 11, 953. [Google Scholar] [CrossRef]
  34. Ji, L.; Zhang, X.; Chen, Z.; Wang, Y.; Zhu, H.; Nai, Y.; Huang, Y.; Lai, R.; Zhong, Y.; Yang, X.; et al. High glucose-induced p66Shc mitochondrial translocation regulates autophagy initiation and autophagosome formation in syncytiotrophoblast and extravillous trophoblast. Cell Commun. Signal 2024, 22, 234. [Google Scholar] [CrossRef]
  35. Orsini, F.; Migliaccio, E.; Moroni, M.; Contursi, C.; Raker, V.A.; Piccini, D.; Martin-Padura, I.; Pelliccia, G.; Trinei, M.; Bono, M.; et al. The life span determinant p66Shc localizes to mitochondria where it associates with mitochondrial heat shock protein 70 and regulates trans-membrane potential. J. Biol. Chem. 2004, 279, 25689–25695. [Google Scholar] [CrossRef]
  36. Giorgio, M.; Migliaccio, E.; Orsini, F.; Paolucci, D.; Moroni, M.; Contursi, C.; Pelliccia, G.; Luzi, L.; Minucci, S.; Marcaccio, M.; et al. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell 2005, 122, 221–233. [Google Scholar] [CrossRef]
  37. Laviola, L.; Orlando, M.R.; Incalza, M.A.; Caccioppoli, C.; Melchiorre, M.; Leonardini, A.; Cignarelli, A.; Tortosa, F.; Labarbuta, R.; Martemucci, S.; et al. TNFalpha signals via p66(Shc) to induce E-Selectin, promote leukocyte transmigration and enhance permeability in human endothelial cells. PLoS ONE 2013, 8, e81930. [Google Scholar] [CrossRef]
  38. Pagnin, E.; Fadini, G.; de Toni, R.; Tiengo, A.; Calo, L.; Avogaro, A. Diabetes induces p66shc gene expression in human peripheral blood mononuclear cells: Relationship to oxidative stress. J. Clin. Endocrinol. Metab. 2005, 90, 1130–1136. [Google Scholar] [CrossRef]
  39. Menini, S.; Amadio, L.; Oddi, G.; Ricci, C.; Pesce, C.; Pugliese, F.; Giorgio, M.; Migliaccio, E.; Pelicci, P.; Iacobini, C.; et al. Deletion of p66Shc longevity gene protects against experimental diabetic glomerulopathy by preventing diabetes-induced oxidative stress. Diabetes 2006, 55, 1642–1650. [Google Scholar] [CrossRef]
  40. Biondi, G.; Marrano, N.; Dipaola, L.; Borrelli, A.; Rella, M.; D’Oria, R.; Genchi, V.A.; Caccioppoli, C.; Porreca, I.; Cignarelli, A.; et al. The p66Shc Protein Mediates Insulin Resistance and Secretory Dysfunction in Pancreatic beta-Cells Under Lipotoxic Conditions. Diabetes 2022, 71, 1763–1771. [Google Scholar] [CrossRef]
  41. Paneni, F.; Mocharla, P.; Akhmedov, A.; Costantino, S.; Osto, E.; Volpe, M.; Luscher, T.F.; Cosentino, F. Gene silencing of the mitochondrial adaptor p66(Shc) suppresses vascular hyperglycemic memory in diabetes. Circ. Res. 2012, 111, 278–289. [Google Scholar] [CrossRef] [PubMed]
  42. Paneni, F.; Costantino, S.; Cosentino, F. p66(Shc)-induced redox changes drive endothelial insulin resistance. Atherosclerosis 2014, 236, 426–429. [Google Scholar] [CrossRef] [PubMed]
  43. Vermot, A.; Petit-Hartlein, 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]
  44. Cipriano, A.; Viviano, M.; Feoli, A.; Milite, C.; Sarno, G.; Castellano, S.; Sbardella, G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J. Med. Chem. 2023, 66, 11632–11655. [Google Scholar] [CrossRef]
  45. Thomas, S.R.; Witting, P.K.; Drummond, G.R. Redox control of endothelial function and dysfunction: Molecular mechanisms and therapeutic opportunities. Antioxid. Redox Signal 2008, 10, 1713–1765. [Google Scholar] [CrossRef]
  46. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal 2014, 20, 1126–1167. [Google Scholar] [CrossRef]
  47. Langbein, H.; Brunssen, C.; Hofmann, A.; Cimalla, P.; Brux, M.; Bornstein, S.R.; Deussen, A.; Koch, E.; Morawietz, H. NADPH oxidase 4 protects against development of endothelial dysfunction and atherosclerosis in LDL receptor deficient mice. Eur. Heart J. 2016, 37, 1753–1761. [Google Scholar] [CrossRef]
  48. Eid, S.A.; Savelieff, M.G.; Eid, A.A.; Feldman, E.L. Nox, Nox, Are You There? The Role of NADPH Oxidases in the Peripheral Nervous System. Antioxid. Redox Signal 2022, 37, 613–630. [Google Scholar] [CrossRef]
  49. Kim, Y.W.; Byzova, T.V. Oxidative stress in angiogenesis and vascular disease. Blood 2014, 123, 625–631. [Google Scholar] [CrossRef]
  50. Harel, S.; Mayaki, D.; Sanchez, V.; Hussain, S.N.A. NOX2, NOX4, and mitochondrial-derived reactive oxygen species contribute to angiopoietin-1 signaling and angiogenic responses in endothelial cells. Vasc. Pharmacol. 2017, 92, 22–32. [Google Scholar] [CrossRef]
  51. Datla, S.R.; Peshavariya, H.; Dusting, G.J.; Mahadev, K.; Goldstein, B.J.; Jiang, F. Important role of Nox4 type NADPH oxidase in angiogenic responses in human microvascular endothelial cells in vitro. Arter. Thromb. Vasc. Biol. 2007, 27, 2319–2324. [Google Scholar] [CrossRef] [PubMed]
  52. Park, H.S.; Chun, J.N.; Jung, H.Y.; Choi, C.; Bae, Y.S. Role of NADPH oxidase 4 in lipopolysaccharide-induced proinflammatory responses by human aortic endothelial cells. Cardiovasc. Res. 2006, 72, 447–455. [Google Scholar] [CrossRef] [PubMed]
  53. Amara, N.; Goven, D.; Prost, F.; Muloway, R.; Crestani, B.; Boczkowski, J. NOX4/NADPH oxidase expression is increased in pulmonary fibroblasts from patients with idiopathic pulmonary fibrosis and mediates TGFbeta1-induced fibroblast differentiation into myofibroblasts. Thorax 2010, 65, 733–738. [Google Scholar] [CrossRef] [PubMed]
  54. Pache, J.C.; Carnesecchi, S.; Deffert, C.; Donati, Y.; Herrmann, F.R.; Barazzone-Argiroffo, C.; Krause, K.H. NOX-4 is expressed in thickened pulmonary arteries in idiopathic pulmonary fibrosis. Nat. Med. 2011, 17, 31–32. [Google Scholar] [CrossRef]
  55. Sedeek, M.; Callera, G.; Montezano, A.; Gutsol, A.; Heitz, F.; Szyndralewiez, C.; Page, P.; Kennedy, C.R.; Burns, K.D.; Touyz, R.M.; et al. Critical role of Nox4-based NADPH oxidase in glucose-induced oxidative stress in the kidney: Implications in type 2 diabetic nephropathy. Am. J. Physiol. Ren. Physiol. 2010, 299, F1348–F1358. [Google Scholar] [CrossRef]
  56. Schroder, K.; Zhang, M.; Benkhoff, S.; Mieth, A.; Pliquett, R.; Kosowski, J.; Kruse, C.; Luedike, P.; Michaelis, U.R.; Weissmann, N.; et al. Nox4 is a protective reactive oxygen species generating vascular NADPH oxidase. Circ. Res. 2012, 110, 1217–1225. [Google Scholar] [CrossRef]
  57. Ray, R.; Murdoch, C.E.; Wang, M.; Santos, C.X.; Zhang, M.; Alom-Ruiz, S.; Anilkumar, N.; Ouattara, A.; Cave, A.C.; Walker, S.J.; et al. Endothelial Nox4 NADPH oxidase enhances vasodilatation and reduces blood pressure in vivo. Arter. Thromb. Vasc. Biol. 2011, 31, 1368–1376. [Google Scholar] [CrossRef]
  58. Sorescu, D.; Weiss, D.; Lassegue, B.; Clempus, R.E.; Szocs, K.; Sorescu, G.P.; Valppu, L.; Quinn, M.T.; Lambeth, J.D.; Vega, J.D.; et al. Superoxide production and expression of nox family proteins in human atherosclerosis. Circulation 2002, 105, 1429–1435. [Google Scholar] [CrossRef]
  59. Sorescu, G.P.; Song, H.; Tressel, S.L.; Hwang, J.; Dikalov, S.; Smith, D.A.; Boyd, N.L.; Platt, M.O.; Lassegue, B.; Griendling, K.K.; et al. Bone morphogenic protein 4 produced in endothelial cells by oscillatory shear stress induces monocyte adhesion by stimulating reactive oxygen species production from a nox1-based NADPH oxidase. Circ. Res. 2004, 95, 773–779. [Google Scholar] [CrossRef]
  60. Honjo, T.; Otsui, K.; Shiraki, R.; Kawashima, S.; Sawamura, T.; Yokoyama, M.; Inoue, N. Essential role of NOXA1 in generation of reactive oxygen species induced by oxidized low-density lipoprotein in human vascular endothelial cells. Endothelium 2008, 15, 137–141. [Google Scholar] [CrossRef]
  61. Drummond, G.R.; Sobey, C.G. Endothelial NADPH oxidases: Which NOX to target in vascular disease? Trends Endocrinol. Metab. 2014, 25, 452–463. [Google Scholar] [CrossRef] [PubMed]
  62. Wind, S.; Beuerlein, K.; Armitage, M.E.; Taye, A.; Kumar, A.H.; Janowitz, D.; Neff, C.; Shah, A.M.; Wingler, K.; Schmidt, H.H. Oxidative stress and endothelial dysfunction in aortas of aged spontaneously hypertensive rats by NOX1/2 is reversed by NADPH oxidase inhibition. Hypertension 2010, 56, 490–497. [Google Scholar] [CrossRef] [PubMed]
  63. Niu, X.L.; Madamanchi, N.R.; Vendrov, A.E.; Tchivilev, I.; Rojas, M.; Madamanchi, C.; Brandes, R.P.; Krause, K.H.; Humphries, J.; Smith, A.; et al. Nox activator 1: A potential target for modulation of vascular reactive oxygen species in atherosclerotic arteries. Circulation 2010, 121, 549–559. [Google Scholar] [CrossRef] [PubMed]
  64. Wendt, M.C.; Daiber, A.; Kleschyov, A.L.; Mulsch, A.; Sydow, K.; Schulz, E.; Chen, K.; Keaney, J.F., Jr.; Lassegue, B.; Walter, U.; et al. Differential effects of diabetes on the expression of the gp91phox homologues nox1 and nox4. Free Radic. Biol. Med. 2005, 39, 381–391. [Google Scholar] [CrossRef]
  65. Alderton, W.K.; Cooper, C.E.; Knowles, R.G. Nitric oxide synthases: Structure, function and inhibition. Biochem. J. 2001, 357, 593–615. [Google Scholar] [CrossRef]
  66. Balligand, J.L.; Feron, O.; Dessy, C. eNOS activation by physical forces: From short-term regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol. Rev. 2009, 89, 481–534. [Google Scholar] [CrossRef]
  67. Alp, N.J.; Channon, K.M. Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease. Arter. Thromb. Vasc. Biol. 2004, 24, 413–420. [Google Scholar] [CrossRef]
  68. Janaszak-Jasiecka, A.; Ploska, A.; Wieronska, J.M.; Dobrucki, L.W.; Kalinowski, L. Endothelial dysfunction due to eNOS uncoupling: Molecular mechanisms as potential therapeutic targets. Cell Mol. Biol. Lett. 2023, 28, 21. [Google Scholar] [CrossRef]
  69. Diers, A.R.; Broniowska, K.A.; Hogg, N. Nitrosative stress and redox-cycling agents synergize to cause mitochondrial dysfunction and cell death in endothelial cells. Redox Biol. 2013, 1, 1–7. [Google Scholar] [CrossRef]
  70. Liaudet, L.; Vassalli, G.; Pacher, P. Role of peroxynitrite in the redox regulation of cell signal transduction pathways. Front. Endocrinol. 2009, 14, 4809–4814. [Google Scholar] [CrossRef]
  71. Radi, R. Nitric oxide, oxidants, and protein tyrosine nitration. Proc. Natl. Acad. Sci. USA 2004, 101, 4003–4008. [Google Scholar] [CrossRef] [PubMed]
  72. Mathews, M.T.; Berk, B.C. PARP-1 inhibition prevents oxidative and nitrosative stress-induced endothelial cell death via transactivation of the VEGF receptor 2. Arter. Thromb. Vasc. Biol. 2008, 28, 711–717. [Google Scholar] [CrossRef] [PubMed]
  73. Laursen, J.B.; Somers, M.; Kurz, S.; McCann, L.; Warnholtz, A.; Freeman, B.A.; Tarpey, M.; Fukai, T.; Harrison, D.G. Endothelial regulation of vasomotion in apoE-deficient mice: Implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation 2001, 103, 1282–1288. [Google Scholar] [CrossRef] [PubMed]
  74. Heitzer, T.; Krohn, K.; Albers, S.; Meinertz, T. Tetrahydrobiopterin improves endothelium-dependent vasodilation by increasing nitric oxide activity in patients with Type II diabetes mellitus. Diabetologia 2000, 43, 1435–1438. [Google Scholar] [CrossRef]
  75. Higashi, Y.; Sasaki, S.; Nakagawa, K.; Fukuda, Y.; Matsuura, H.; Oshima, T.; Chayama, K. Tetrahydrobiopterin enhances forearm vascular response to acetylcholine in both normotensive and hypertensive individuals. Am. J. Hypertens. 2002, 15, 326–332. [Google Scholar] [CrossRef]
  76. Stroes, E.; Kastelein, J.; Cosentino, F.; Erkelens, W.; Wever, R.; Koomans, H.; Luscher, T.; Rabelink, T. Tetrahydrobiopterin restores endothelial function in hypercholesterolemia. J. Clin. Investig. 1997, 99, 41–46. [Google Scholar] [CrossRef]
  77. Sud, N.; Wells, S.M.; Sharma, S.; Wiseman, D.A.; Wilham, J.; Black, S.M. Asymmetric dimethylarginine inhibits HSP90 activity in pulmonary arterial endothelial cells: Role of mitochondrial dysfunction. Am. J. Physiol. Cell Physiol. 2008, 294, C1407–C1418. [Google Scholar] [CrossRef]
  78. Antoniades, C.; Shirodaria, C.; Leeson, P.; Antonopoulos, A.; Warrick, N.; Van-Assche, T.; Cunnington, C.; Tousoulis, D.; Pillai, R.; Ratnatunga, C.; et al. Association of plasma asymmetrical dimethylarginine (ADMA) with elevated vascular superoxide production and endothelial nitric oxide synthase uncoupling: Implications for endothelial function in human atherosclerosis. Eur. Heart J. 2009, 30, 1142–1150. [Google Scholar] [CrossRef]
  79. Vergnani, L.; Hatrik, S.; Ricci, F.; Passaro, A.; Manzoli, N.; Zuliani, G.; Brovkovych, V.; Fellin, R.; Malinski, T. Effect of native and oxidized low-density lipoprotein on endothelial nitric oxide and superoxide production: Key role of L-arginine availability. Circulation 2000, 101, 1261–1266. [Google Scholar] [CrossRef]
  80. Koshida, R.; Ou, J.; Matsunaga, T.; Chilian, W.M.; Oldham, K.T.; Ackerman, A.W.; Pritchard, K.A., Jr. Angiostatin: A negative regulator of endothelial-dependent vasodilation. Circulation 2003, 107, 803–806. [Google Scholar] [CrossRef]
  81. Topal, G.; Brunet, A.; Millanvoye, E.; Boucher, J.L.; Rendu, F.; Devynck, M.A.; David-Dufilho, M. Homocysteine induces oxidative stress by uncoupling of NO synthase activity through reduction of tetrahydrobiopterin. Free Radic. Biol. Med. 2004, 36, 1532–1541. [Google Scholar] [CrossRef] [PubMed]
  82. Cosentino, F.; Hishikawa, K.; Katusic, Z.S.; Luscher, T.F. High glucose increases nitric oxide synthase expression and superoxide anion generation in human aortic endothelial cells. Circulation 1997, 96, 25–28. [Google Scholar] [CrossRef] [PubMed]
  83. Zou, M.H.; Shi, C.; Cohen, R.A. Oxidation of the zinc-thiolate complex and uncoupling of endothelial nitric oxide synthase by peroxynitrite. J. Clin. Investig. 2002, 109, 817–826. [Google Scholar] [CrossRef] [PubMed]
  84. Thum, T.; Fraccarollo, D.; Schultheiss, M.; Froese, S.; Galuppo, P.; Widder, J.D.; Tsikas, D.; Ertl, G.; Bauersachs, J. Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes 2007, 56, 666–674. [Google Scholar] [CrossRef]
  85. Kuwabara, Y.; Nishino, T.; Okamoto, K.; Matsumura, T.; Eger, B.T.; Pai, E.F.; Nishino, T. Unique amino acids cluster for switching from the dehydrogenase to oxidase form of xanthine oxidoreductase. Proc. Natl. Acad. Sci. USA 2003, 100, 8170–8175. [Google Scholar] [CrossRef]
  86. Nishino, T.; Okamoto, K.; Eger, B.T.; Pai, E.F.; Nishino, T. Mammalian xanthine oxidoreductase—Mechanism of transition from xanthine dehydrogenase to xanthine oxidase. FEBS J. 2008, 275, 3278–3289. [Google Scholar] [CrossRef]
  87. Nishino, T.; Okamoto, K.; Kawaguchi, Y.; Hori, H.; Matsumura, T.; Eger, B.T.; Pai, E.F.; Nishino, T. Mechanism of the conversion of xanthine dehydrogenase to xanthine oxidase: Identification of the two cysteine disulfide bonds and crystal structure of a non-convertible rat liver xanthine dehydrogenase mutant. J. Biol. Chem. 2005, 280, 24888–24894. [Google Scholar] [CrossRef]
  88. Jankov, R.P.; Kantores, C.; Pan, J.; Belik, J. Contribution of xanthine oxidase-derived superoxide to chronic hypoxic pulmonary hypertension in neonatal rats. Am. J. Physiol. Lung Cell Mol. Physiol. 2008, 294, L233–L245. [Google Scholar] [CrossRef]
  89. Desco, M.C.; Asensi, M.; Marquez, R.; Martinez-Valls, J.; Vento, M.; Pallardo, F.V.; Sastre, J.; Vina, J. Xanthine oxidase is involved in free radical production in type 1 diabetes: Protection by allopurinol. Diabetes 2002, 51, 1118–1124. [Google Scholar] [CrossRef]
  90. Kuppusamy, U.R.; Indran, M.; Rokiah, P. Glycaemic control in relation to xanthine oxidase and antioxidant indices in Malaysian Type 2 diabetes patients. Diabet. Med. 2005, 22, 1343–1346. [Google Scholar] [CrossRef]
  91. McNally, J.S.; Davis, M.E.; Giddens, D.P.; Saha, A.; Hwang, J.; Dikalov, S.; Jo, H.; Harrison, D.G. Role of xanthine oxidoreductase and NAD(P)H oxidase in endothelial superoxide production in response to oscillatory shear stress. Am. J. Physiol. Heart Circ. Physiol. 2003, 285, H2290–H2297. [Google Scholar] [CrossRef]
  92. Landmesser, U.; Spiekermann, S.; Preuss, C.; Sorrentino, S.; Fischer, D.; Manes, C.; Mueller, M.; Drexler, H. Angiotensin II induces endothelial xanthine oxidase activation: Role for endothelial dysfunction in patients with coronary disease. Arter. Thromb. Vasc. Biol. 2007, 27, 943–948. [Google Scholar] [CrossRef] [PubMed]
  93. Battelli, M.G.; Bolognesi, A.; Polito, L. Pathophysiology of circulating xanthine oxidoreductase: New emerging roles for a multi-tasking enzyme. Biochim. Biophys. Acta 2014, 1842, 1502–1517. [Google Scholar] [CrossRef] [PubMed]
  94. Kelley, E.E.; Hock, T.; Khoo, N.K.; Richardson, G.R.; Johnson, K.K.; Powell, P.C.; Giles, G.I.; Agarwal, A.; Lancaster, J.R., Jr.; Tarpey, M.M. Moderate hypoxia induces xanthine oxidoreductase activity in arterial endothelial cells. Free Radic. Biol. Med. 2006, 40, 952–959. [Google Scholar] [CrossRef]
  95. Adachi, T.; Fukushima, T.; Usami, Y.; Hirano, K. Binding of human xanthine oxidase to sulphated glycosaminoglycans on the endothelial-cell surface. Biochem. J. 1993, 289, 523–527. [Google Scholar] [CrossRef]
  96. Farquharson, C.A.; Butler, R.; Hill, A.; Belch, J.J.; Struthers, A.D. Allopurinol improves endothelial dysfunction in chronic heart failure. Circulation 2002, 106, 221–226. [Google Scholar] [CrossRef]
  97. Butler, R.; Morris, A.D.; Belch, J.J.; Hill, A.; Struthers, A.D. Allopurinol normalizes endothelial dysfunction in type 2 diabetics with mild hypertension. Hypertension 2000, 35, 746–751. [Google Scholar] [CrossRef]
  98. Kluge, M.A.; Fetterman, J.L.; Vita, J.A. Mitochondria and endothelial function. Circ. Res. 2013, 112, 1171–1188. [Google Scholar] [CrossRef]
  99. Potashnik, R.; Bloch-Damti, A.; Bashan, N.; Rudich, A. IRS1 degradation and increased serine phosphorylation cannot predict the degree of metabolic insulin resistance induced by oxidative stress. Diabetologia 2003, 46, 639–648. [Google Scholar] [CrossRef]
  100. Tesfamariam, B.; Brown, M.L.; Cohen, R.A. Elevated glucose impairs endothelium-dependent relaxation by activating protein kinase C. J. Clin. Invest. 1991, 87, 1643–1648. [Google Scholar] [CrossRef]
  101. Sheetz, M.J.; King, G.L. Molecular understanding of hyperglycemia’s adverse effects for diabetic complications. JAMA 2002, 288, 2579–2588. [Google Scholar] [CrossRef] [PubMed]
  102. Lebiedzinska, M.; Karkucinska-Wieckowska, A.; Giorgi, C.; Karczmarewicz, E.; Pronicka, E.; Pinton, P.; Duszynski, J.; Pronicki, M.; Wieckowski, M.R. Oxidative stress-dependent p66Shc phosphorylation in skin fibroblasts of children with mitochondrial disorders. Biochim. Biophys. Acta 2010, 1797, 952–960. [Google Scholar] [CrossRef] [PubMed]
  103. Pinton, P.; Rizzuto, R. p66Shc, oxidative stress and aging: Importing a lifespan determinant into mitochondria. Cell Cycle 2008, 7, 304–308. [Google Scholar] [CrossRef] [PubMed]
  104. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef]
  105. Pal, R.; Bhadada, S.K. AGEs accumulation with vascular complications, glycemic control and metabolic syndrome: A narrative review. Bone 2023, 176, 116884. [Google Scholar] [CrossRef]
  106. Arora, D.K.; Machhadieh, B.; Matti, A.; Wadzinski, B.E.; Ramanadham, S.; Kowluru, A. High glucose exposure promotes activation of protein phosphatase 2A in rodent islets and INS-1 832/13 beta-cells by increasing the posttranslational carboxylmethylation of its catalytic subunit. Endocrinology 2014, 155, 380–391. [Google Scholar] [CrossRef]
  107. Fu, M.X.; Requena, J.R.; Jenkins, A.J.; Lyons, T.J.; Baynes, J.W.; Thorpe, S.R. The advanced glycation end product, Nepsilon-(carboxymethyl)lysine, is a product of both lipid peroxidation and glycoxidation reactions. J. Biol. Chem. 1996, 271, 9982–9986. [Google Scholar] [CrossRef]
  108. Thornalley, P.J.; Langborg, A.; Minhas, H.S. Formation of glyoxal, methylglyoxal and 3-deoxyglucosone in the glycation of proteins by glucose. Biochem. J. 1999, 344, 109–116. [Google Scholar] [CrossRef]
  109. Wells-Knecht, K.J.; Zyzak, D.V.; Litchfield, J.E.; Thorpe, S.R.; Baynes, J.W. Mechanism of autoxidative glycosylation: Identification of glyoxal and arabinose as intermediates in the autoxidative modification of proteins by glucose. Biochemistry 1995, 34, 3702–3709. [Google Scholar] [CrossRef]
  110. Scheijen, J.L.; Schalkwijk, C.G. Quantification of glyoxal, methylglyoxal and 3-deoxyglucosone in blood and plasma by ultra performance liquid chromatography tandem mass spectrometry: Evaluation of blood specimen. Clin. Chem. Lab. Med. 2014, 52, 85–91. [Google Scholar] [CrossRef]
  111. Goldin, A.; Beckman, J.A.; Schmidt, A.M.; Creager, M.A. Advanced glycation end products: Sparking the development of diabetic vascular injury. Circulation 2006, 114, 597–605. [Google Scholar] [CrossRef] [PubMed]
  112. Ishibashi, Y.; Matsui, T.; Takeuchi, M.; Yamagishi, S. Glucagon-like peptide-1 (GLP-1) inhibits advanced glycation end product (AGE)-induced up-regulation of VCAM-1 mRNA levels in endothelial cells by suppressing AGE receptor (RAGE) expression. Biochem. Biophys. Res. Commun. 2010, 391, 1405–1408. [Google Scholar] [CrossRef] [PubMed]
  113. Phalitakul, S.; Okada, M.; Hara, Y.; Yamawaki, H. Vaspin prevents methylglyoxal-induced apoptosis in human vascular endothelial cells by inhibiting reactive oxygen species generation. Acta Physiol. 2013, 209, 212–219. [Google Scholar] [CrossRef] [PubMed]
  114. Zhan, Y.; Sun, H.L.; Chen, H.; Zhang, H.; Sun, J.; Zhang, Z.; Cai, D.H. Glucagon-like peptide-1 (GLP-1) protects vascular endothelial cells against advanced glycation end products (AGEs)-induced apoptosis. Med. Sci. Monit. 2012, 18, BR286–BR291. [Google Scholar] [CrossRef]
  115. Chu, P.; Han, G.; Ahsan, A.; Sun, Z.; Liu, S.; Zhang, Z.; Sun, B.; Song, Y.; Lin, Y.; Peng, J.; et al. Phosphocreatine protects endothelial cells from Methylglyoxal induced oxidative stress and apoptosis via the regulation of PI3K/Akt/eNOS and NF-kappaB pathway. Vasc. Pharmacol. 2017, 91, 26–35. [Google Scholar] [CrossRef]
  116. Wu, Q.; Zhao, Y.; Duan, W.; Liu, Y.; Chen, X.; Zhu, M. Propofol inhibits high glucose-induced PP2A expression in human umbilical vein endothelial cells. Vasc. Pharmacol. 2017, 91, 18–25. [Google Scholar] [CrossRef]
  117. Weigert, C.; Brodbeck, K.; Staiger, H.; Kausch, C.; Machicao, F.; Haring, H.U.; Schleicher, E.D. Palmitate, but not unsaturated fatty acids, induces the expression of interleukin-6 in human myotubes through proteasome-dependent activation of nuclear factor-kappaB. J. Biol. Chem. 2004, 279, 23942–23952. [Google Scholar] [CrossRef]
  118. Kim, F.; Tysseling, K.A.; Rice, J.; Pham, M.; Haji, L.; Gallis, B.M.; Baas, A.S.; Paramsothy, P.; Giachelli, C.M.; Corson, M.A.; et al. Free fatty acid impairment of nitric oxide production in endothelial cells is mediated by IKKbeta. Arter. Thromb. Vasc. Biol. 2005, 25, 989–994. [Google Scholar] [CrossRef]
  119. Maloney, E.; Sweet, I.R.; Hockenbery, D.M.; Pham, M.; Rizzo, N.O.; Tateya, S.; Handa, P.; Schwartz, M.W.; Kim, F. Activation of NF-kappaB by palmitate in endothelial cells: A key role for NADPH oxidase-derived superoxide in response to TLR4 activation. Arter. Thromb. Vasc. Biol. 2009, 29, 1370–1375. [Google Scholar] [CrossRef]
  120. Choi, S.E.; Kang, Y.; Jang, H.J.; Shin, H.C.; Kim, H.E.; Kim, H.S.; Kim, H.J.; Kim, D.J.; Lee, K.W. Involvement of glycogen synthase kinase-3beta in palmitate-induced human umbilical vein endothelial cell apoptosis. J. Vasc. Res. 2007, 44, 365–374. [Google Scholar] [CrossRef]
  121. Chai, W.; Liu, Z. p38 mitogen-activated protein kinase mediates palmitate-induced apoptosis but not inhibitor of nuclear factor-kappaB degradation in human coronary artery endothelial cells. Endocrinology 2007, 148, 1622–1628. [Google Scholar] [CrossRef] [PubMed]
  122. Erdogdu, O.; Eriksson, L.; Xu, H.; Sjoholm, A.; Zhang, Q.; Nystrom, T. Exendin-4 protects endothelial cells from lipoapoptosis by PKA, PI3K, eNOS, p38 MAPK, and JNK pathways. J. Mol. Endocrinol. 2013, 50, 229–241. [Google Scholar] [CrossRef] [PubMed]
  123. Jiang, H.; Liang, C.; Liu, X.; Jiang, Q.; He, Z.; Wu, J.; Pan, X.; Ren, Y.; Fan, M.; Li, M.; et al. Palmitic acid promotes endothelial progenitor cells apoptosis via p38 and JNK mitogen-activated protein kinase pathways. Atherosclerosis 2010, 210, 71–77. [Google Scholar] [CrossRef] [PubMed]
  124. Holland, W.L.; Summers, S.A. Sphingolipids, insulin resistance, and metabolic disease: New insights from in vivo manipulation of sphingolipid metabolism. Endocr. Rev. 2008, 29, 381–402. [Google Scholar] [CrossRef]
  125. Zhang, D.X.; Zou, A.P.; Li, P.L. Ceramide-induced activation of NADPH oxidase and endothelial dysfunction in small coronary arteries. Am. J. Physiol. Heart Circ. Physiol. 2003, 284, H605–H612. [Google Scholar] [CrossRef]
  126. Li, H.; Junk, P.; Huwiler, A.; Burkhardt, C.; Wallerath, T.; Pfeilschifter, J.; Forstermann, U. Dual effect of ceramide on human endothelial cells: Induction of oxidative stress and transcriptional upregulation of endothelial nitric oxide synthase. Circulation 2002, 106, 2250–2256. [Google Scholar] [CrossRef]
  127. Rhee, S.G.; Chae, H.Z.; Kim, K. Peroxiredoxins: A historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic. Biol. Med. 2005, 38, 1543–1552. [Google Scholar] [CrossRef]
  128. Manevich, Y.; Sweitzer, T.; Pak, J.H.; Feinstein, S.I.; Muzykantov, V.; Fisher, A.B. 1-Cys peroxiredoxin overexpression protects cells against phospholipid peroxidation-mediated membrane damage. Proc. Natl. Acad. Sci. USA 2002, 99, 11599–11604. [Google Scholar] [CrossRef]
  129. Wang, Y.; Manevich, Y.; Feinstein, S.I.; Fisher, A.B. Adenovirus-mediated transfer of the 1-cys peroxiredoxin gene to mouse lung protects against hyperoxic injury. Am. J. Physiol. Lung Cell Mol. Physiol. 2004, 286, L1188–L1193. [Google Scholar] [CrossRef]
  130. Pak, J.H.; Manevich, Y.; Kim, H.S.; Feinstein, S.I.; Fisher, A.B. An antisense oligonucleotide to 1-cys peroxiredoxin causes lipid peroxidation and apoptosis in lung epithelial cells. J. Biol. Chem. 2002, 277, 49927–49934. [Google Scholar] [CrossRef]
  131. Kang, D.H.; Lee, D.J.; Lee, K.W.; Park, Y.S.; Lee, J.Y.; Lee, S.H.; Koh, Y.J.; Koh, G.Y.; Choi, C.; Yu, D.Y.; et al. Peroxiredoxin II is an essential antioxidant enzyme that prevents the oxidative inactivation of VEGF receptor-2 in vascular endothelial cells. Mol. Cell 2011, 44, 545–558. [Google Scholar] [CrossRef] [PubMed]
  132. Kumin, A.; Schafer, M.; Epp, N.; Bugnon, P.; Born-Berclaz, C.; Oxenius, A.; Klippel, A.; Bloch, W.; Werner, S. Peroxiredoxin 6 is required for blood vessel integrity in wounded skin. J. Cell Biol. 2007, 179, 747–760. [Google Scholar] [CrossRef] [PubMed]
  133. Mowbray, A.L.; Kang, D.H.; Rhee, S.G.; Kang, S.W.; Jo, H. Laminar shear stress up-regulates peroxiredoxins (PRX) in endothelial cells: PRX 1 as a mechanosensitive antioxidant. J. Biol. Chem. 2008, 283, 1622–1627. [Google Scholar] [CrossRef] [PubMed]
  134. Haendeler, J.; Popp, R.; Goy, C.; Tischler, V.; Zeiher, A.M.; Dimmeler, S. Cathepsin D and H2O2 stimulate degradation of thioredoxin-1: Implication for endothelial cell apoptosis. J. Biol. Chem. 2005, 280, 42945–42951. [Google Scholar] [CrossRef]
  135. Yamawaki, H.; Pan, S.; Lee, R.T.; Berk, B.C. Fluid shear stress inhibits vascular inflammation by decreasing thioredoxin-interacting protein in endothelial cells. J. Clin. Invest. 2005, 115, 733–738. [Google Scholar] [CrossRef]
  136. Schroeder, P.; Popp, R.; Wiegand, B.; Altschmied, J.; Haendeler, J. Nuclear redox-signaling is essential for apoptosis inhibition in endothelial cells--important role for nuclear thioredoxin-1. Arter. Thromb. Vasc. Biol. 2007, 27, 2325–2331. [Google Scholar] [CrossRef]
  137. Devarajan, A.; Bourquard, N.; Hama, S.; Navab, M.; Grijalva, V.R.; Morvardi, S.; Clarke, C.F.; Vergnes, L.; Reue, K.; Teiber, J.F.; et al. Paraoxonase 2 deficiency alters mitochondrial function and exacerbates the development of atherosclerosis. Antioxid. Redox Signal 2011, 14, 341–351. [Google Scholar] [CrossRef]
  138. Ng, C.J.; Bourquard, N.; Grijalva, V.; Hama, S.; Shih, D.M.; Navab, M.; Fogelman, A.M.; Lusis, A.J.; Young, S.; Reddy, S.T. Paraoxonase-2 deficiency aggravates atherosclerosis in mice despite lower apolipoprotein-B-containing lipoproteins: Anti-atherogenic role for paraoxonase-2. J. Biol. Chem. 2006, 281, 29491–29500. [Google Scholar] [CrossRef]
  139. Cui, Y.; Xu, X.; Bi, H.; Zhu, Q.; Wu, J.; Xia, X.; Qiushi, R.; Ho, P.C. Expression modification of uncoupling proteins and MnSOD in retinal endothelial cells and pericytes induced by high glucose: The role of reactive oxygen species in diabetic retinopathy. Exp. Eye Res. 2006, 83, 807–816. [Google Scholar] [CrossRef]
  140. Duval, C.; Negre-Salvayre, A.; Dogilo, A.; Salvayre, R.; Penicaud, L.; Casteilla, L. Increased reactive oxygen species production with antisense oligonucleotides directed against uncoupling protein 2 in murine endothelial cells. Biochem. Cell Biol. 2002, 80, 757–764. [Google Scholar] [CrossRef]
  141. Tian, X.Y.; Wong, W.T.; Xu, A.; Lu, Y.; Zhang, Y.; Wang, L.; Cheang, W.S.; Wang, Y.; Yao, X.; Huang, Y. Uncoupling protein-2 protects endothelial function in diet-induced obese mice. Circ. Res. 2012, 110, 1211–1216. [Google Scholar] [CrossRef] [PubMed]
  142. Sun, J.; Pu, Y.; Wang, P.; Chen, S.; Zhao, Y.; Liu, C.; Shang, Q.; Zhu, Z.; Liu, D. TRPV1-mediated UCP2 upregulation ameliorates hyperglycemia-induced endothelial dysfunction. Cardiovasc. Diabetol. 2013, 12, 69. [Google Scholar] [CrossRef] [PubMed]
  143. Lee, K.U.; Lee, I.K.; Han, J.; Song, D.K.; Kim, Y.M.; Song, H.S.; Kim, H.S.; Lee, W.J.; Koh, E.H.; Song, K.H.; et al. Effects of recombinant adenovirus-mediated uncoupling protein 2 overexpression on endothelial function and apoptosis. Circ. Res. 2005, 96, 1200–1207. [Google Scholar] [CrossRef] [PubMed]
  144. Fukai, T.; Folz, R.J.; Landmesser, U.; Harrison, D.G. Extracellular superoxide dismutase and cardiovascular disease. Cardiovasc. Res. 2002, 55, 239–249. [Google Scholar] [CrossRef]
  145. Foresman, E.L.; Miller, F.J., Jr. Extracellular but not cytosolic superoxide dismutase protects against oxidant-mediated endothelial dysfunction. Redox Biol. 2013, 1, 292–296. [Google Scholar] [CrossRef]
  146. Juarez, J.C.; Manuia, M.; Burnett, M.E.; Betancourt, O.; Boivin, B.; Shaw, D.E.; Tonks, N.K.; Mazar, A.P.; Donate, F. Superoxide dismutase 1 (SOD1) is essential for H2O2-mediated oxidation and inactivation of phosphatases in growth factor signaling. Proc. Natl. Acad. Sci. USA 2008, 105, 7147–7152. [Google Scholar] [CrossRef]
  147. Morikawa, K.; Shimokawa, H.; Matoba, T.; Kubota, H.; Akaike, T.; Talukder, M.A.; Hatanaka, M.; Fujiki, T.; Maeda, H.; Takahashi, S.; et al. Pivotal role of Cu,Zn-superoxide dismutase in endothelium-dependent hyperpolarization. J. Clin. Investig. 2003, 112, 1871–1879. [Google Scholar] [CrossRef]
  148. Dromparis, P.; Michelakis, E.D. Mitochondria in vascular health and disease. Annu. Rev. Physiol. 2013, 75, 95–126. [Google Scholar] [CrossRef]
  149. Connor, K.M.; Subbaram, S.; Regan, K.J.; Nelson, K.K.; Mazurkiewicz, J.E.; Bartholomew, P.J.; Aplin, A.E.; Tai, Y.T.; Aguirre-Ghiso, J.; Flores, S.C.; et al. Mitochondrial H2O2 regulates the angiogenic phenotype via PTEN oxidation. J. Biol. Chem. 2005, 280, 16916–16924. [Google Scholar] [CrossRef]
  150. He, T.; Peterson, T.E.; Holmuhamedov, E.L.; Terzic, A.; Caplice, N.M.; Oberley, L.W.; Katusic, Z.S. Human endothelial progenitor cells tolerate oxidative stress due to intrinsically high expression of manganese superoxide dismutase. Arter. Thromb. Vasc. Biol. 2004, 24, 2021–2027. [Google Scholar] [CrossRef]
  151. Ohashi, M.; Runge, M.S.; Faraci, F.M.; Heistad, D.D. MnSOD deficiency increases endothelial dysfunction in ApoE-deficient mice. Arter. Thromb. Vasc. Biol. 2006, 26, 2331–2336. [Google Scholar] [CrossRef] [PubMed]
  152. Afolayan, A.J.; Eis, A.; Teng, R.J.; Bakhutashvili, I.; Kaul, S.; Davis, J.M.; Konduri, G.G. Decreases in manganese superoxide dismutase expression and activity contribute to oxidative stress in persistent pulmonary hypertension of the newborn. Am. J. Physiol. Lung Cell Mol. Physiol. 2012, 303, L870–L879. [Google Scholar] [CrossRef] [PubMed]
  153. Marrotte, E.J.; Chen, D.D.; Hakim, J.S.; Chen, A.F. Manganese superoxide dismutase expression in endothelial progenitor cells accelerates wound healing in diabetic mice. J. Clin. Invest. 2010, 120, 4207–4219. [Google Scholar] [CrossRef] [PubMed]
  154. Fukai, T.; Ushio-Fukai, M. Superoxide dismutases: Role in redox signaling, vascular function, and diseases. Antioxid. Redox Signal 2011, 15, 1583–1606. [Google Scholar] [CrossRef]
  155. Brown, K.A.; Chu, Y.; Lund, D.D.; Heistad, D.D.; Faraci, F.M. Gene transfer of extracellular superoxide dismutase protects against vascular dysfunction with aging. Am. J. Physiol. Heart Circ. Physiol. 2006, 290, H2600–H2605. [Google Scholar] [CrossRef]
  156. Fennell, J.P.; Brosnan, M.J.; Frater, A.J.; Hamilton, C.A.; Alexander, M.Y.; Nicklin, S.A.; Heistad, D.D.; Baker, A.H.; Dominiczak, A.F. Adenovirus-mediated overexpression of extracellular superoxide dismutase improves endothelial dysfunction in a rat model of hypertension. Gene Ther. 2002, 9, 110–117. [Google Scholar] [CrossRef]
  157. Iida, S.; Chu, Y.; Francis, J.; Weiss, R.M.; Gunnett, C.A.; Faraci, F.M.; Heistad, D.D. Gene transfer of extracellular superoxide dismutase improves endothelial function in rats with heart failure. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H525–H532. [Google Scholar] [CrossRef]
  158. Hwang, I.; Lee, J.; Huh, J.Y.; Park, J.; Lee, H.B.; Ho, Y.S.; Ha, H. Catalase deficiency accelerates diabetic renal injury through peroxisomal dysfunction. Diabetes 2012, 61, 728–738. [Google Scholar] [CrossRef]
  159. Meilhac, O.; Zhou, M.; Santanam, N.; Parthasarathy, S. Lipid peroxides induce expression of catalase in cultured vascular cells. J. Lipid Res. 2000, 41, 1205–1213. [Google Scholar] [CrossRef]
  160. Forgione, M.A.; Weiss, N.; Heydrick, S.; Cap, A.; Klings, E.S.; Bierl, C.; Eberhardt, R.T.; Farber, H.W.; Loscalzo, J. Cellular glutathione peroxidase deficiency and endothelial dysfunction. Am. J. Physiol. Heart Circ. Physiol. 2002, 282, H1255–H1261. [Google Scholar] [CrossRef]
  161. Tang, X.; Luo, Y.X.; Chen, H.Z.; Liu, D.P. Mitochondria, endothelial cell function, and vascular diseases. Front. Physiol. 2014, 5, 175. [Google Scholar] [CrossRef] [PubMed]
  162. Zhang, Y.; Handy, D.E.; Loscalzo, J. Adenosine-dependent induction of glutathione peroxidase 1 in human primary endothelial cells and protection against oxidative stress. Circ. Res. 2005, 96, 831–837. [Google Scholar] [CrossRef] [PubMed]
  163. Oelze, M.; Kroller-Schon, S.; Steven, S.; Lubos, E.; Doppler, C.; Hausding, M.; Tobias, S.; Brochhausen, C.; Li, H.; Torzewski, M.; et al. Glutathione peroxidase-1 deficiency potentiates dysregulatory modifications of endothelial nitric oxide synthase and vascular dysfunction in aging. Hypertension 2014, 63, 390–396. [Google Scholar] [CrossRef]
  164. La Sala, L.; Cattaneo, M.; De Nigris, V.; Pujadas, G.; Testa, R.; Bonfigli, A.R.; Genovese, S.; Ceriello, A. Oscillating glucose induces microRNA-185 and impairs an efficient antioxidant response in human endothelial cells. Cardiovasc. Diabetol. 2016, 15, 71. [Google Scholar] [CrossRef]
  165. Srivastava, S.K.; Ramana, K.V.; Bhatnagar, A. Role of aldose reductase and oxidative damage in diabetes and the consequent potential for therapeutic options. Endocr. Rev. 2005, 26, 380–392. [Google Scholar] [CrossRef]
  166. Srikanth, K.K.; Orrick, J.A. Biochemistry, Polyol or Sorbitol Pathways; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  167. Dong, H.; Zhang, Y.; Huang, Y.; Deng, H. Pathophysiology of RAGE in inflammatory diseases. Front. Immunol. 2022, 13, 931473. [Google Scholar] [CrossRef]
  168. Chen, Y.; Meng, Z.; Li, Y.; Liu, S.; Hu, P.; Luo, E. Advanced glycation end products and reactive oxygen species: Uncovering the potential role of ferroptosis in diabetic complications. Mol. Med. 2024, 30, 141. [Google Scholar] [CrossRef]
  169. Rowan, S.; Bejarano, E.; Taylor, A. Mechanistic targeting of advanced glycation end-products in age-related diseases. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 3631–3643. [Google Scholar] [CrossRef]
  170. Koya, D.; King, G.L. Protein kinase C activation and the development of diabetic complications. Diabetes 1998, 47, 859–866. [Google Scholar] [CrossRef]
  171. Geraldes, P.; King, G.L. Activation of protein kinase C isoforms and its impact on diabetic complications. Circ. Res. 2010, 106, 1319–1331. [Google Scholar] [CrossRef]
  172. Xiao, Q.; Wang, D.; Li, D.; Huang, J.; Ma, F.; Zhang, H.; Sheng, Y.; Zhang, C.; Ha, X. Protein kinase C: A potential therapeutic target for endothelial dysfunction in diabetes. J. Diabetes Complicat. 2023, 37, 108565. [Google Scholar] [CrossRef] [PubMed]
  173. Paneque, A.; Fortus, H.; Zheng, J.; Werlen, G.; Jacinto, E. The Hexosamine Biosynthesis Pathway: Regulation and Function. Genes 2023, 14, 933. [Google Scholar] [CrossRef] [PubMed]
  174. Cairns, M.; Joseph, D.; Essop, M.F. The dual role of the hexosamine biosynthetic pathway in cardiac physiology and pathophysiology. Front. Endocrinol. 2022, 13, 984342. [Google Scholar] [CrossRef] [PubMed]
  175. Veluthakal, R.; Esparza, D.; Hoolachan, J.M.; Balakrishnan, R.; Ahn, M.; Oh, E.; Jayasena, C.S.; Thurmond, D.C. Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression. Int. J. Mol. Sci. 2024, 25, 1504. [Google Scholar] [CrossRef]
  176. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef]
  177. Bhatti, J.S.; Sehrawat, A.; Mishra, J.; Sidhu, I.S.; Navik, U.; Khullar, N.; Kumar, S.; Bhatti, G.K.; Reddy, P.H. Oxidative stress in the pathophysiology of type 2 diabetes and related complications: Current therapeutics strategies and future perspectives. Free Radic. Biol. Med. 2022, 184, 114–134. [Google Scholar] [CrossRef]
  178. Bastos, I.M.; Rebelo, S.; Silva, V.L.M. A review of poly(ADP-ribose)polymerase-1 (PARP1) role and its inhibitors bearing pyrazole or indazole core for cancer therapy. Biochem. Pharmacol. 2024, 221, 116045. [Google Scholar] [CrossRef]
  179. Shah, M.S.; Brownlee, M. Molecular and Cellular Mechanisms of Cardiovascular Disorders in Diabetes. Circ. Res. 2016, 118, 1808–1829. [Google Scholar] [CrossRef]
  180. Kawarada, Y.; Inoue, Y.; Kawasaki, F.; Fukuura, K.; Sato, K.; Tanaka, T.; Itoh, Y.; Hayashi, H. TGF-beta induces p53/Smads complex formation in the PAI-1 promoter to activate transcription. Sci. Rep. 2016, 6, 35483. [Google Scholar] [CrossRef]
  181. Khalid, M.; Petroianu, G.; Adem, A. Advanced Glycation End Products and Diabetes Mellitus: Mechanisms and Perspectives. Biomolecules 2022, 12, 542. [Google Scholar] [CrossRef]
  182. Schmidt, A.M.; Hori, O.; Chen, J.X.; Li, J.F.; Crandall, J.; Zhang, J.; Cao, R.; Yan, S.D.; Brett, J.; Stern, D. Advanced glycation endproducts interacting with their endothelial receptor induce expression of vascular cell adhesion molecule-1 (VCAM-1) in cultured human endothelial cells and in mice. A potential mechanism for the accelerated vasculopathy of diabetes. J. Clin. Investig. 1995, 96, 1395–1403. [Google Scholar] [CrossRef] [PubMed]
  183. Knapp, L.T.; Klann, E. Role of reactive oxygen species in hippocampal long-term potentiation: Contributory or inhibitory? J. Neurosci. Res. 2002, 70, 1–7. [Google Scholar] [CrossRef] [PubMed]
  184. Du, N.; Xu, Z.; Gao, M.; Liu, P.; Sun, B.; Cao, X. Combination of Ginsenoside Rg1 and Astragaloside IV reduces oxidative stress and inhibits TGF-beta1/Smads signaling cascade on renal fibrosis in rats with diabetic nephropathy. Drug Des. Dev. Ther. 2018, 12, 3517–3524. [Google Scholar] [CrossRef] [PubMed]
  185. Zhu, Y.; Zhu, C.; Yang, H.; Deng, J.; Fan, D. Protective effect of ginsenoside Rg5 against kidney injury via inhibition of NLRP3 inflammasome activation and the MAPK signaling pathway in high-fat diet/streptozotocin-induced diabetic mice. Pharmacol. Res. 2020, 155, 104746. [Google Scholar] [CrossRef]
  186. Dang, Y.F.; Qiu, T.X.; Song, D.W.; Liu, L. PMA-triggered PKCepsilon activity enhances Nrf2-mediated antiviral response on fish rhabdovirus infection. Fish. Shellfish. Immunol. 2019, 87, 871–878. [Google Scholar] [CrossRef]
  187. Liu, S.; Shen, M.; Li, C.; Wei, Y.; Meng, X.; Li, R.; Cao, Y.; Wu, W.; Liu, H. PKCdelta contributes to oxidative stress-induced apoptosis in porcine ovarian granulosa cells via activating JNK. Theriogenology 2019, 131, 89–95. [Google Scholar] [CrossRef]
  188. Ly, L.D.; Xu, S.; Choi, S.K.; Ha, C.M.; Thoudam, T.; Cha, S.K.; Wiederkehr, A.; Wollheim, C.B.; Lee, I.K.; Park, K.S. Oxidative stress and calcium dysregulation by palmitate in type 2 diabetes. Exp. Mol. Med. 2017, 49, e291. [Google Scholar] [CrossRef]
  189. Rybin, V.O.; Guo, J.; Sabri, A.; Elouardighi, H.; Schaefer, E.; Steinberg, S.F. Stimulus-specific differences in protein kinase C delta localization and activation mechanisms in cardiomyocytes. J. Biol. Chem. 2004, 279, 19350–19361. [Google Scholar] [CrossRef]
  190. Singh, M.; Kapoor, A.; Bhatnagar, A. Physiological and Pathological Roles of Aldose Reductase. Metabolites 2021, 11, 655. [Google Scholar] [CrossRef]
  191. Thorne, C.A.; Grey, A.C.; Lim, J.C.; Donaldson, P.J. The Synergistic Effects of Polyol Pathway-Induced Oxidative and Osmotic Stress in the Aetiology of Diabetic Cataracts. Int. J. Mol. Sci. 2024, 25, 9042. [Google Scholar] [CrossRef]
  192. Yan, L.J. Redox imbalance stress in diabetes mellitus: Role of the polyol pathway. Anim. Model. Exp. Med. 2018, 1, 7–13. [Google Scholar] [CrossRef] [PubMed]
  193. Godisela, K.K.; Reddy, S.S.; Reddy, P.Y.; Kumar, C.U.; Reddy, V.S.; Ayyagari, R.; Reddy, G.B. Role of sorbitol-mediated cellular stress response in obesity-associated retinal degeneration. Arch. Biochem. Biophys. 2020, 679, 108207. [Google Scholar] [CrossRef] [PubMed]
  194. Yabe-Nishimura, C. Aldose reductase in glucose toxicity: A potential target for the prevention of diabetic complications. Pharmacol. Rev. 1998, 50, 21–33. [Google Scholar] [CrossRef]
  195. Obrosova, I.G. Increased sorbitol pathway activity generates oxidative stress in tissue sites for diabetic complications. Antioxid. Redox Signal 2005, 7, 1543–1552. [Google Scholar] [CrossRef]
  196. Pierce, M.; Stanley, P. Deuterostomes. In Essentials of Glycobiology, 3rd ed.; Varki, A., Cummings, R.D., Esko, J.D., Stanley, P., Hart, G.W., Aebi, M., Darvill, A.G., Kinoshita, T., Packer, N.H., Prestegard, J.H., et al., Eds.; Cold Spring Harbor: New York, NY, USA, 2015; pp. 351–360. [Google Scholar]
  197. Marshall, S. Role of insulin, adipocyte hormones, and nutrient-sensing pathways in regulating fuel metabolism and energy homeostasis: A nutritional perspective of diabetes, obesity, and cancer. Sci. STKE 2006, 2006, re7. [Google Scholar] [CrossRef]
  198. Ma, Z.; Vosseller, K. Cancer metabolism and elevated O-GlcNAc in oncogenic signaling. J. Biol. Chem. 2014, 289, 34457–34465. [Google Scholar] [CrossRef]
  199. Yi, M.; Cruz Cisneros, L.; Cho, E.J.; Alexander, M.; Kimelman, F.A.; Swentek, L.; Ferrey, A.; Tantisattamo, E.; Ichii, H. Nrf2 Pathway and Oxidative Stress as a Common Target for Treatment of Diabetes and Its Comorbidities. Int. J. Mol. Sci. 2024, 25, 821. [Google Scholar] [CrossRef]
  200. Yuan, Y.H.; Yan, W.F.; Sun, J.D.; Huang, J.Y.; Mu, Z.; Chen, N.H. The molecular mechanism of rotenone-induced alpha-synuclein aggregation: Emphasizing the role of the calcium/GSK3beta pathway. Toxicol. Lett. 2015, 233, 163–171. [Google Scholar] [CrossRef]
  201. Ruan, H.B.; Dietrich, M.O.; Liu, Z.W.; Zimmer, M.R.; Li, M.D.; Singh, J.P.; Zhang, K.; Yin, R.; Wu, J.; Horvath, T.L.; et al. O-GlcNAc transferase enables AgRP neurons to suppress browning of white fat. Cell 2014, 159, 306–317. [Google Scholar] [CrossRef]
  202. Sandoo, A.; van Zanten, J.J.; Metsios, G.S.; Carroll, D.; Kitas, G.D. The endothelium and its role in regulating vascular tone. Open Cardiovasc. Med. J. 2010, 4, 302–312. [Google Scholar] [CrossRef]
  203. Kruger-Genge, A.; Blocki, A.; Franke, R.P.; Jung, F. Vascular Endothelial Cell Biology: An Update. Int. J. Mol. Sci. 2019, 20, 4411. [Google Scholar] [CrossRef] [PubMed]
  204. Verma, S.; Anderson, T.J. Fundamentals of endothelial function for the clinical cardiologist. Circulation 2002, 105, 546–549. [Google Scholar] [CrossRef] [PubMed]
  205. Sena, C.M.; Leandro, A.; Azul, L.; Seica, R.; Perry, G. Vascular Oxidative Stress: Impact and Therapeutic Approaches. Front. Physiol. 2018, 9, 1668. [Google Scholar] [CrossRef] [PubMed]
  206. Zhang, Z.; Zhao, L.; Zhou, X.; Meng, X.; Zhou, X. Role of inflammation, immunity, and oxidative stress in hypertension: New insights and potential therapeutic targets. Front. Immunol. 2022, 13, 1098725. [Google Scholar] [CrossRef]
  207. Cheng, C.K.; Huang, Y. Vascular endothelium: The interface for multiplex signal transduction. J. Mol. Cell Cardiol. 2024, 195, 97–102. [Google Scholar] [CrossRef]
  208. Bkaily, G.; Jacques, D. Morphological and Functional Remodeling of Vascular Endothelium in Cardiovascular Diseases. Int. J. Mol. Sci. 2023, 24, 1998. [Google Scholar] [CrossRef]
  209. Cahill, P.A.; Redmond, E.M. Vascular endothelium—Gatekeeper of vessel health. Atherosclerosis 2016, 248, 97–109. [Google Scholar] [CrossRef]
  210. Palomo, M.; Moreno-Castano, A.B.; Salas, M.Q.; Escribano-Serrat, S.; Rovira, M.; Guillen-Olmos, E.; Fernandez, S.; Ventosa-Capell, H.; Youssef, L.; Crispi, F.; et al. Endothelial activation and damage as a common pathological substrate in different pathologies and cell therapy complications. Front. Med. 2023, 10, 1285898. [Google Scholar] [CrossRef]
  211. Liao, J.K. Linking endothelial dysfunction with endothelial cell activation. J. Clin. Invest. 2013, 123, 540–541. [Google Scholar] [CrossRef]
  212. Scioli, M.G.; Storti, G.; D’Amico, F.; Rodriguez Guzman, R.; Centofanti, F.; Doldo, E.; Cespedes Miranda, E.M.; Orlandi, A. Oxidative Stress and New Pathogenetic Mechanisms in Endothelial Dysfunction: Potential Diagnostic Biomarkers and Therapeutic Targets. J. Clin. Med. 2020, 9, 1995. [Google Scholar] [CrossRef]
  213. Landmesser, U.; Dikalov, S.; Price, S.R.; McCann, L.; Fukai, T.; Holland, S.M.; Mitch, W.E.; Harrison, D.G. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J. Clin. Invest. 2003, 111, 1201–1209. [Google Scholar] [CrossRef] [PubMed]
  214. Al-Shabrawey, M.; Smith, S. Prediction of diabetic retinopathy: Role of oxidative stress and relevance of apoptotic biomarkers. EPMA J. 2010, 1, 56–72. [Google Scholar] [CrossRef] [PubMed]
  215. Wolin, M.S.; Gupte, S.A.; Neo, B.H.; Gao, Q.; Ahmad, M. Oxidant-redox regulation of pulmonary vascular responses to hypoxia and nitric oxide-cGMP signaling. Cardiol. Rev. 2010, 18, 89–93. [Google Scholar] [CrossRef] [PubMed]
  216. Dharmashankar, K.; Widlansky, M.E. Vascular endothelial function and hypertension: Insights and directions. Curr. Hypertens. Rep. 2010, 12, 448–455. [Google Scholar] [CrossRef]
  217. Toda, N.; Nakanishi-Toda, M. How mental stress affects endothelial function. Pflug. Arch. 2011, 462, 779–794. [Google Scholar] [CrossRef]
  218. Herrera, M.D.; Mingorance, C.; Rodriguez-Rodriguez, R.; Alvarez de Sotomayor, M. Endothelial dysfunction and aging: An update. Ageing Res. Rev. 2010, 9, 142–152. [Google Scholar] [CrossRef]
  219. Soultati, A.; Mountzios, G.; Avgerinou, C.; Papaxoinis, G.; Pectasides, D.; Dimopoulos, M.A.; Papadimitriou, C. Endothelial vascular toxicity from chemotherapeutic agents: Preclinical evidence and clinical implications. Cancer Treat. Rev. 2012, 38, 473–483. [Google Scholar] [CrossRef]
  220. Cao, G.; Xuan, X.; Hu, J.; Zhang, R.; Jin, H.; Dong, H. How vascular smooth muscle cell phenotype switching contributes to vascular disease. Cell Commun. Signal 2022, 20, 180. [Google Scholar] [CrossRef]
  221. Elmarasi, M.; Elmakaty, I.; Elsayed, B.; Elsayed, A.; Zein, J.A.; Boudaka, A.; Eid, A.H. Phenotypic switching of vascular smooth muscle cells in atherosclerosis, hypertension, and aortic dissection. J. Cell Physiol. 2024, 239, e31200. [Google Scholar] [CrossRef]
  222. Chen, Q.; Wang, Q.; Zhu, J.; Xiao, Q.; Zhang, L. Reactive oxygen species: Key regulators in vascular health and diseases. Br. J. Pharmacol. 2018, 175, 1279–1292. [Google Scholar] [CrossRef]
  223. Montezano, A.C.; Touyz, R.M. Reactive oxygen species, vascular Noxs, and hypertension: Focus on translational and clinical research. Antioxid. Redox Signal 2014, 20, 164–182. [Google Scholar] [CrossRef] [PubMed]
  224. Sun, S.; Liu, F.; Fan, F.; Chen, N.; Pan, X.; Wei, Z.; Zhang, Y. Exploring the mechanism of atherosclerosis and the intervention of traditional Chinese medicine combined with mesenchymal stem cells based on inflammatory targets. Heliyon 2023, 9, e22005. [Google Scholar] [CrossRef] [PubMed]
  225. Deng, W.; Huang, S.; Yu, L.; Gao, B.; Pan, Y.; Wang, X.; Li, L. HIF-1alpha knockdown attenuates phenotypic transformation and oxidative stress induced by high salt in human aortic vascular smooth muscle cells. Sci. Rep. 2024, 14, 28100. [Google Scholar] [CrossRef]
  226. Xu, S.; Han, X.; Wang, X.; Yu, Y.; Qu, C.; Liu, X.; Yang, B. The role of oxidative stress in aortic dissection: A potential therapeutic target. Front. Cardiovasc. Med. 2024, 11, 1410477. [Google Scholar] [CrossRef]
  227. Sharma, P.; Kaushal, N.; Saleth, L.R.; Ghavami, S.; Dhingra, S.; Kaur, P. Oxidative stress-induced apoptosis and autophagy: Balancing the contrary forces in spermatogenesis. Biochim. Biophys. Acta Mol. Basis Dis. 2023, 1869, 166742. [Google Scholar] [CrossRef]
  228. Dogru, S.; Yasar, E.; Yesilkaya, A. Effects of uric acid on oxidative stress in vascular smooth muscle cells. Biomed. Rep. 2024, 21, 171. [Google Scholar] [CrossRef]
  229. Camargo, L.L.; Wang, Y.; Rios, F.J.; McBride, M.; Montezano, A.C.; Touyz, R.M. Oxidative Stress and Endoplasmic Reticular Stress Interplay in the Vasculopathy of Hypertension. Can. J. Cardiol. 2023, 39, 1874–1887. [Google Scholar] [CrossRef]
  230. Lu, W.; Lin, Y.; Haider, N.; Moly, P.; Wang, L.; Zhou, W. Ginsenoside Rb1 protects human vascular smooth muscle cells against resistin-induced oxidative stress and dysfunction. Front. Cardiovasc. Med. 2023, 10, 1164547. [Google Scholar] [CrossRef]
  231. Ghatage, T.; Singh, S.; Mandal, K.; Dhar, A. MasR and pGCA receptor activation protects primary vascular smooth muscle cells and endothelial cells against oxidative stress via inhibition of intracellular calcium. J. Cell Biochem. 2023, 124, 943–960. [Google Scholar] [CrossRef]
  232. 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]
  233. Travers, J.G.; Kamal, F.A.; Robbins, J.; Yutzey, K.E.; Blaxall, B.C. Cardiac Fibrosis: The Fibroblast Awakens. Circ. Res. 2016, 118, 1021–1040. [Google Scholar] [CrossRef] [PubMed]
  234. Kong, P.; Christia, P.; Frangogiannis, N.G. The pathogenesis of cardiac fibrosis. Cell Mol. Life Sci. 2014, 71, 549–574. [Google Scholar] [CrossRef] [PubMed]
  235. Hua, R.; Gao, H.; He, C.; Xin, S.; Wang, B.; Zhang, S.; Gao, L.; Tao, Q.; Wu, W.; Sun, F.; et al. An emerging view on vascular fibrosis molecular mediators and relevant disorders: From bench to bed. Front. Cardiovasc. Med. 2023, 10, 1273502. [Google Scholar] [CrossRef] [PubMed]
  236. Eble, J.A.; de Rezende, F.F. Redox-relevant aspects of the extracellular matrix and its cellular contacts via integrins. Antioxid. Redox Signal. 2014, 20, 1977–1993. [Google Scholar] [CrossRef]
  237. Meng, X.M.; Nikolic-Paterson, D.J.; Lan, H.Y. TGF-beta: The master regulator of fibrosis. Nat. Rev. Nephrol. 2016, 12, 325–338. [Google Scholar] [CrossRef]
  238. Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef]
  239. Schafer, M.J.; White, T.A.; Iijima, K.; Haak, A.J.; Ligresti, G.; Atkinson, E.J.; Oberg, A.L.; Birch, J.; Salmonowicz, H.; Zhu, Y.; et al. Cellular senescence mediates fibrotic pulmonary disease. Nat. Commun. 2017, 8, 14532. [Google Scholar] [CrossRef]
  240. Cai, H.; Harrison, D.G. Endothelial dysfunction in cardiovascular diseases: The role of oxidant stress. Circ. Res. 2000, 87, 840–844. [Google Scholar] [CrossRef]
  241. Martinez-Reyes, I.; Chandel, N.S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat. Commun. 2020, 11, 102. [Google Scholar] [CrossRef]
  242. Hunt, M.; Torres, M.; Bachar-Wikstrom, E.; Wikstrom, J.D. Cellular and molecular roles of reactive oxygen species in wound healing. Commun. Biol. 2024, 7, 1534. [Google Scholar] [CrossRef]
  243. Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgozoglu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Primers 2019, 5, 56. [Google Scholar] [CrossRef] [PubMed]
  244. Forstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef] [PubMed]
  245. Jiang, H.; Zhou, Y.; Nabavi, S.M.; Sahebkar, A.; Little, P.J.; Xu, S.; Weng, J.; Ge, J. Mechanisms of Oxidized LDL-Mediated Endothelial Dysfunction and Its Consequences for the Development of Atherosclerosis. Front. Cardiovasc. Med. 2022, 9, 925923. [Google Scholar] [CrossRef] [PubMed]
  246. Cyrus, T.; Pratico, D.; Zhao, L.; Witztum, J.L.; Rader, D.J.; Rokach, J.; FitzGerald, G.A.; Funk, C.D. Absence of 12/15-lipoxygenase expression decreases lipid peroxidation and atherogenesis in apolipoprotein e-deficient mice. Circulation 2001, 103, 2277–2282. [Google Scholar] [CrossRef]
  247. Shih, D.M.; Xia, Y.R.; Wang, X.P.; Miller, E.; Castellani, L.W.; Subbanagounder, G.; Cheroutre, H.; Faull, K.F.; Berliner, J.A.; Witztum, J.L.; et al. Combined serum paraoxonase knockout/apolipoprotein E knockout mice exhibit increased lipoprotein oxidation and atherosclerosis. J. Biol. Chem. 2000, 275, 17527–17535. [Google Scholar] [CrossRef]
  248. Chen, S.; Li, Q.; Shi, H.; Li, F.; Duan, Y.; Guo, Q. New insights into the role of mitochondrial dynamics in oxidative stress-induced diseases. Biomed. Pharmacother. 2024, 178, 117084. [Google Scholar] [CrossRef]
  249. Ahmadpour, S.T.; Maheo, K.; Servais, S.; Brisson, L.; Dumas, J.F. Cardiolipin, the Mitochondrial Signature Lipid: Implication in Cancer. Int. J. Mol. Sci. 2020, 21, 8031. [Google Scholar] [CrossRef]
  250. Fuentes, J.M.; Morcillo, P. The Role of Cardiolipin in Mitochondrial Function and Neurodegenerative Diseases. Cells 2024, 13, 609. [Google Scholar] [CrossRef]
  251. Petrosillo, G.; Ruggiero, F.M.; Paradies, G. Role of reactive oxygen species and cardiolipin in the release of cytochrome c from mitochondria. FASEB J. 2003, 17, 2202–2208. [Google Scholar] [CrossRef]
  252. Chouchani, E.T.; Pell, V.R.; Gaude, E.; Aksentijevic, D.; Sundier, S.Y.; Robb, E.L.; Logan, A.; Nadtochiy, S.M.; Ord, E.N.J.; Smith, A.C.; et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 2014, 515, 431–435. [Google Scholar] [CrossRef]
  253. Shen, Z.; Ye, C.; McCain, K.; Greenberg, M.L. The Role of Cardiolipin in Cardiovascular Health. Biomed. Res. Int. 2015, 2015, 891707. [Google Scholar] [CrossRef] [PubMed]
  254. Wan, M.; Hua, X.; Su, J.; Thiagarajan, D.; Frostegard, A.G.; Haeggstrom, J.Z.; Frostegard, J. Oxidized but not native cardiolipin has pro-inflammatory effects, which are inhibited by Annexin A5. Atherosclerosis 2014, 235, 592–598. [Google Scholar] [CrossRef] [PubMed]
  255. Mercer, J.R.; Cheng, K.K.; Figg, N.; Gorenne, I.; Mahmoudi, M.; Griffin, J.; Vidal-Puig, A.; Logan, A.; Murphy, M.P.; Bennett, M. DNA damage links mitochondrial dysfunction to atherosclerosis and the metabolic syndrome. Circ. Res. 2010, 107, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
  256. Botto, N.; Rizza, A.; Colombo, M.G.; Mazzone, A.M.; Manfredi, S.; Masetti, S.; Clerico, A.; Biagini, A.; Andreassi, M.G. Evidence for DNA damage in patients with coronary artery disease. Mutat. Res. 2001, 493, 23–30. [Google Scholar] [CrossRef]
  257. Martinet, W.; Knaapen, M.W.; De Meyer, G.R.; Herman, A.G.; Kockx, M.M. Elevated levels of oxidative DNA damage and DNA repair enzymes in human atherosclerotic plaques. Circulation 2002, 106, 927–932. [Google Scholar] [CrossRef]
  258. Shah, A.; Gray, K.; Figg, N.; Finigan, A.; Starks, L.; Bennett, M. Defective Base Excision Repair of Oxidative DNA Damage in Vascular Smooth Muscle Cells Promotes Atherosclerosis. Circulation 2018, 138, 1446–1462. [Google Scholar] [CrossRef]
  259. St-Pierre, J.; Buckingham, J.A.; Roebuck, S.J.; Brand, M.D. Topology of superoxide production from different sites in the mitochondrial electron transport chain. J. Biol. Chem. 2002, 277, 44784–44790. [Google Scholar] [CrossRef]
  260. Yue, Y.; Ren, L.; Zhang, C.; Miao, K.; Tan, K.; Yang, Q.; Hu, Y.; Xi, G.; Luo, G.; Yang, M.; et al. Mitochondrial genome undergoes de novo DNA methylation that protects mtDNA against oxidative damage during the peri-implantation window. Proc. Natl. Acad. Sci. USA 2022, 119, e2201168119. [Google Scholar] [CrossRef]
  261. Yu, E.; Calvert, P.A.; Mercer, J.R.; Harrison, J.; Baker, L.; Figg, N.L.; Kumar, S.; Wang, J.C.; Hurst, L.A.; Obaid, D.R.; et al. Mitochondrial DNA damage can promote atherosclerosis independently of reactive oxygen species through effects on smooth muscle cells and monocytes and correlates with higher-risk plaques in humans. Circulation 2013, 128, 702–712. [Google Scholar] [CrossRef]
  262. Ballinger, S.W.; Patterson, C.; Knight-Lozano, C.A.; Burow, D.L.; Conklin, C.A.; Hu, Z.; Reuf, J.; Horaist, C.; Lebovitz, R.; Hunter, G.C.; et al. Mitochondrial integrity and function in atherogenesis. Circulation 2002, 106, 544–549. [Google Scholar] [CrossRef]
  263. Gimbrone, M.A., Jr.; Garcia-Cardena, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef] [PubMed]
  264. Jebari-Benslaiman, S.; Galicia-Garcia, U.; Larrea-Sebal, A.; Olaetxea, J.R.; Alloza, I.; Vandenbroeck, K.; Benito-Vicente, A.; Martin, C. Pathophysiology of Atherosclerosis. Int. J. Mol. Sci. 2022, 23, 3346. [Google Scholar] [CrossRef] [PubMed]
  265. Yan, R.; Zhang, X.; Xu, W.; Li, J.; Sun, Y.; Cui, S.; Xu, R.; Li, W.; Jiao, L.; Wang, T. ROS-Induced Endothelial Dysfunction in the Pathogenesis of Atherosclerosis. Aging Dis. 2024, 16, 250–268. [Google Scholar] [CrossRef]
  266. Janaszak-Jasiecka, A.; Siekierzycka, A.; Ploska, A.; Dobrucki, I.T.; Kalinowski, L. Endothelial Dysfunction Driven by Hypoxia-The Influence of Oxygen Deficiency on NO Bioavailability. Biomolecules 2021, 11, 982. [Google Scholar] [CrossRef]
  267. Hernandez-Navarro, I.; Botana, L.; Diez-Mata, J.; Tesoro, L.; Jimenez-Guirado, B.; Gonzalez-Cucharero, C.; Alcharani, N.; Zamorano, J.L.; Saura, M.; Zaragoza, C. Replicative Endothelial Cell Senescence May Lead to Endothelial Dysfunction by Increasing the BH2/BH4 Ratio Induced by Oxidative Stress, Reducing BH4 Availability, and Decreasing the Expression of eNOS. Int. J. Mol. Sci. 2024, 25, 9890. [Google Scholar] [CrossRef]
  268. Ponnuswamy, P.; Schrottle, A.; Ostermeier, E.; Gruner, S.; Huang, P.L.; Ertl, G.; Hoffmann, U.; Nieswandt, B.; Kuhlencordt, P.J. eNOS protects from atherosclerosis despite relevant superoxide production by the enzyme in apoE mice. PLoS ONE 2012, 7, e30193. [Google Scholar] [CrossRef]
  269. Shafique, E.; Torina, A.; Reichert, K.; Colantuono, B.; Nur, N.; Zeeshan, K.; Ravichandran, V.; Liu, Y.; Feng, J.; Zeeshan, K.; et al. Mitochondrial redox plays a critical role in the paradoxical effects of NAPDH oxidase-derived ROS on coronary endothelium. Cardiovasc. Res. 2017, 113, 234–246. [Google Scholar] [CrossRef]
  270. Higashi, Y. Roles of Oxidative Stress and Inflammation in Vascular Endothelial Dysfunction-Related Disease. Antioxidants 2022, 11, 1958. [Google Scholar] [CrossRef]
  271. Patel, K.D.; Zimmerman, G.A.; Prescott, S.M.; McEver, R.P.; McIntyre, T.M. Oxygen radicals induce human endothelial cells to express GMP-140 and bind neutrophils. J. Cell Biol. 1991, 112, 749–759. [Google Scholar] [CrossRef]
  272. Zhang, H.; Park, Y.; Wu, J.; Chen, X.; Lee, S.; Yang, J.; Dellsperger, K.C.; Zhang, C. Role of TNF-alpha in vascular dysfunction. Clin. Sci. 2009, 116, 219–230. [Google Scholar] [CrossRef]
  273. Corda, S.; Laplace, C.; Vicaut, E.; Duranteau, J. Rapid reactive oxygen species production by mitochondria in endothelial cells exposed to tumor necrosis factor-alpha is mediated by ceramide. Am. J. Respir. Cell Mol. Biol. 2001, 24, 762–768. [Google Scholar] [CrossRef] [PubMed]
  274. Li, D.; Mehta, J.L. Upregulation of endothelial receptor for oxidized LDL (LOX-1) by oxidized LDL and implications in apoptosis of human coronary artery endothelial cells: Evidence from use of antisense LOX-1 mRNA and chemical inhibitors. Arter. Thromb. Vasc. Biol. 2000, 20, 1116–1122. [Google Scholar] [CrossRef] [PubMed]
  275. Zhou, J.; Abid, M.D.; Xiong, Y.; Chen, Q.; Chen, J. ox-LDL downregulates eNOS activity via LOX-1-mediated endoplasmic reticulum stress. Int. J. Mol. Med. 2013, 32, 1442–1450. [Google Scholar] [CrossRef] [PubMed]
  276. Pothineni, N.V.K.; Karathanasis, S.K.; Ding, Z.; Arulandu, A.; Varughese, K.I.; Mehta, J.L. LOX-1 in Atherosclerosis and Myocardial Ischemia: Biology, Genetics, and Modulation. J. Am. Coll. Cardiol. 2017, 69, 2759–2768. [Google Scholar] [CrossRef]
  277. Akhmedov, A.; Rozenberg, I.; Paneni, F.; Camici, G.G.; Shi, Y.; Doerries, C.; Sledzinska, A.; Mocharla, P.; Breitenstein, A.; Lohmann, C.; et al. Endothelial overexpression of LOX-1 increases plaque formation and promotes atherosclerosis in vivo. Eur. Heart J. 2014, 35, 2839–2848. [Google Scholar] [CrossRef]
  278. Batty, M.; Bennett, M.R.; Yu, E. The Role of Oxidative Stress in Atherosclerosis. Cells 2022, 11, 3843. [Google Scholar] [CrossRef]
  279. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  280. Canty, T.G., Jr.; Boyle, E.M., Jr.; Farr, A.; Morgan, E.N.; Verrier, E.D.; Pohlman, T.H. Oxidative stress induces NF-kappaB nuclear translocation without degradation of IkappaBalpha. Circulation 1999, 100, II361–II364. [Google Scholar] [CrossRef]
  281. Clark, R.A.; Valente, A.J. Nuclear factor kappa B activation by NADPH oxidases. Mech. Ageing Dev. 2004, 125, 799–810. [Google Scholar] [CrossRef]
  282. Park, H.S.; Jung, H.Y.; Park, E.Y.; Kim, J.; Lee, W.J.; Bae, Y.S. Cutting edge: Direct interaction of TLR4 with NAD(P)H oxidase 4 isozyme is essential for lipopolysaccharide-induced production of reactive oxygen species and activation of NF-kappa B. J. Immunol. 2004, 173, 3589–3593. [Google Scholar] [CrossRef]
  283. Mohan, S.; Koyoma, K.; Thangasamy, A.; Nakano, H.; Glickman, R.D.; Mohan, N. Low shear stress preferentially enhances IKK activity through selective sources of ROS for persistent activation of NF-kappaB in endothelial cells. Am. J. Physiol. Cell Physiol. 2007, 292, C362–C371. [Google Scholar] [CrossRef] [PubMed]
  284. Tang, Y.Y.; Wang, D.C.; Wang, Y.Q.; Huang, A.F.; Xu, W.D. Emerging role of hypoxia-inducible factor-1alpha in inflammatory autoimmune diseases: A comprehensive review. Front. Immunol. 2022, 13, 1073971. [Google Scholar] [CrossRef]
  285. Tannahill, G.M.; Curtis, A.M.; Adamik, J.; Palsson-McDermott, E.M.; McGettrick, A.F.; Goel, G.; Frezza, C.; Bernard, N.J.; Kelly, B.; Foley, N.H.; et al. Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha. Nature 2013, 496, 238–242. [Google Scholar] [CrossRef] [PubMed]
  286. Mills, E.; O’Neill, L.A. Succinate: A metabolic signal in inflammation. Trends Cell Biol. 2014, 24, 313–320. [Google Scholar] [CrossRef]
  287. Abais, J.M.; Xia, M.; Zhang, Y.; Boini, K.M.; Li, P.L. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid. Redox Signal 2015, 22, 1111–1129. [Google Scholar] [CrossRef]
  288. Chen, Y.; Ye, X.; Escames, G.; Lei, W.; Zhang, X.; Li, M.; Jing, T.; Yao, Y.; Qiu, Z.; Wang, Z.; et al. The NLRP3 inflammasome: Contributions to inflammation-related diseases. Cell Mol. Biol. Lett. 2023, 28, 51. [Google Scholar] [CrossRef]
  289. Huang, Y.; Xu, W.; Zhou, R. NLRP3 inflammasome activation and cell death. Cell Mol. Immunol. 2021, 18, 2114–2127. [Google Scholar] [CrossRef]
  290. Ives, A.; Nomura, J.; Martinon, F.; Roger, T.; LeRoy, D.; Miner, J.N.; Simon, G.; Busso, N.; So, A. Xanthine oxidoreductase regulates macrophage IL1beta secretion upon NLRP3 inflammasome activation. Nat. Commun. 2015, 6, 6555. [Google Scholar] [CrossRef]
  291. Nomura, J.; Busso, N.; Ives, A.; Matsui, C.; Tsujimoto, S.; Shirakura, T.; Tamura, M.; Kobayashi, T.; So, A.; Yamanaka, Y. Xanthine oxidase inhibition by febuxostat attenuates experimental atherosclerosis in mice. Sci. Rep. 2014, 4, 4554. [Google Scholar] [CrossRef]
  292. Elliott, E.I.; Miller, A.N.; Banoth, B.; Iyer, S.S.; Stotland, A.; Weiss, J.P.; Gottlieb, R.A.; Sutterwala, F.S.; Cassel, S.L. Cutting Edge: Mitochondrial Assembly of the NLRP3 Inflammasome Complex Is Initiated at Priming. J. Immunol. 2018, 200, 3047–3052. [Google Scholar] [CrossRef]
  293. Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef] [PubMed]
  294. Shimada, K.; Crother, T.R.; Karlin, J.; Dagvadorj, J.; Chiba, N.; Chen, S.; Ramanujan, V.K.; Wolf, A.J.; Vergnes, L.; Ojcius, D.M.; et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 2012, 36, 401–414. [Google Scholar] [CrossRef] [PubMed]
  295. Libby, P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J. Am. Coll. Cardiol. 2017, 70, 2278–2289. [Google Scholar] [CrossRef] [PubMed]
  296. Bevilacqua, M.P.; Pober, J.S.; Wheeler, M.E.; Cotran, R.S.; Gimbrone, M.A., Jr. Interleukin-1 activation of vascular endothelium. Effects on procoagulant activity and leukocyte adhesion. Am. J. Pathol. 1985, 121, 394–403. [Google Scholar]
  297. Libby, P.; Warner, S.J.; Friedman, G.B. Interleukin 1: A mitogen for human vascular smooth muscle cells that induces the release of growth-inhibitory prostanoids. J. Clin. Investig. 1988, 81, 487–498. [Google Scholar] [CrossRef]
  298. Loppnow, H.; Libby, P. Proliferating or interleukin 1-activated human vascular smooth muscle cells secrete copious interleukin 6. J. Clin. Investig. 1990, 85, 731–738. [Google Scholar] [CrossRef]
  299. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef]
  300. Lusis, A.J. Atherosclerosis. Nature 2000, 407, 233–241. [Google Scholar] [CrossRef]
  301. Yuan, D.; Chu, J.; Qian, J.; Lin, H.; Zhu, G.; Chen, F.; Liu, X. New Concepts on the Pathophysiology of Acute Coronary Syndrome. Rev. Cardiovasc. Med. 2023, 24, 112. [Google Scholar] [CrossRef]
  302. Virmani, R.; Burke, A.P.; Farb, A.; Kolodgie, F.D. Pathology of the vulnerable plaque. J. Am. Coll. Cardiol. 2006, 47, C13–C18. [Google Scholar] [CrossRef]
  303. Yu, Y.; Cai, Y.; Yang, F.; Yang, Y.; Cui, Z.; Shi, D.; Bai, R. Vascular smooth muscle cell phenotypic switching in atherosclerosis. Heliyon 2024, 10, e37727. [Google Scholar] [CrossRef] [PubMed]
  304. Olejarz, W.; Lacheta, D.; Kubiak-Tomaszewska, G. Matrix Metalloproteinases as Biomarkers of Atherosclerotic Plaque Instability. Int. J. Mol. Sci. 2020, 21, 3946. [Google Scholar] [CrossRef] [PubMed]
  305. Lenglet, S.; Mach, F.; Montecucco, F. Role of matrix metalloproteinase-8 in atherosclerosis. Mediat. Inflamm. 2013, 2013, 659282. [Google Scholar] [CrossRef] [PubMed]
  306. Heo, S.H.; Cho, C.H.; Kim, H.O.; Jo, Y.H.; Yoon, K.S.; Lee, J.H.; Park, J.C.; Park, K.C.; Ahn, T.B.; Chung, K.C.; et al. Plaque rupture is a determinant of vascular events in carotid artery atherosclerotic disease: Involvement of matrix metalloproteinases 2 and 9. J. Clin. Neurol. 2011, 7, 69–76. [Google Scholar] [CrossRef]
  307. Newby, A.C. Metalloproteinases promote plaque rupture and myocardial infarction: A persuasive concept waiting for clinical translation. Matrix Biol. 2015, 44–46, 157–166. [Google Scholar] [CrossRef]
  308. Gough, P.J.; Gomez, I.G.; Wille, P.T.; Raines, E.W. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J. Clin. Investig. 2006, 116, 59–69. [Google Scholar] [CrossRef]
  309. Johnson, J.L.; George, S.J.; Newby, A.C.; Jackson, C.L. Divergent effects of matrix metalloproteinases 3, 7, 9, and 12 on atherosclerotic plaque stability in mouse brachiocephalic arteries. Proc. Natl. Acad. Sci. USA 2005, 102, 15575–15580. [Google Scholar] [CrossRef]
  310. Huang, Y.; Mironova, M.; Lopes-Virella, M.F. Oxidized LDL stimulates matrix metalloproteinase-1 expression in human vascular endothelial cells. Arter. Thromb. Vasc. Biol. 1999, 19, 2640–2647. [Google Scholar] [CrossRef]
  311. Valentin, F.; Bueb, J.L.; Kieffer, P.; Tschirhart, E.; Atkinson, J. Oxidative stress activates MMP-2 in cultured human coronary smooth muscle cells. Fundam. Clin. Pharmacol. 2005, 19, 661–667. [Google Scholar] [CrossRef]
  312. Xu, X.P.; Meisel, S.R.; Ong, J.M.; Kaul, S.; Cercek, B.; Rajavashisth, T.B.; Sharifi, B.; Shah, P.K. Oxidized low-density lipoprotein regulates matrix metalloproteinase-9 and its tissue inhibitor in human monocyte-derived macrophages. Circulation 1999, 99, 993–998. [Google Scholar] [CrossRef]
  313. Nelson, K.K.; Melendez, J.A. Mitochondrial redox control of matrix metalloproteinases. Free Radic. Biol. Med. 2004, 37, 768–784. [Google Scholar] [CrossRef] [PubMed]
  314. Zalba, G.; Fortuno, A.; Orbe, J.; San Jose, G.; Moreno, M.U.; Belzunce, M.; Rodriguez, J.A.; Beloqui, O.; Paramo, J.A.; Diez, J. Phagocytic NADPH oxidase-dependent superoxide production stimulates matrix metalloproteinase-9: Implications for human atherosclerosis. Arter. Thromb. Vasc. Biol. 2007, 27, 587–593. [Google Scholar] [CrossRef] [PubMed]
  315. Beli, E.; Yan, Y.; Moldovan, L.; Vieira, C.P.; Gao, R.; Duan, Y.; Prasad, R.; Bhatwadekar, A.; White, F.A.; Townsend, S.D.; et al. Restructuring of the Gut Microbiome by Intermittent Fasting Prevents Retinopathy and Prolongs Survival in db/db Mice. Diabetes 2018, 67, 1867–1879. [Google Scholar] [CrossRef]
  316. Calderon, G.D.; Juarez, O.H.; Hernandez, G.E.; Punzo, S.M.; De la Cruz, Z.D. Oxidative stress and diabetic retinopathy: Development and treatment. Eye 2017, 31, 1122–1130. [Google Scholar] [CrossRef]
  317. Wong, T.Y.; Cheung, C.M.; Larsen, M.; Sharma, S.; Simo, R. Diabetic retinopathy. Nat. Rev. Dis. Primers 2016, 2, 16012. [Google Scholar] [CrossRef]
  318. Lee, R.; Wong, T.Y.; Sabanayagam, C. Epidemiology of diabetic retinopathy, diabetic macular edema and related vision loss. Eye Vis. 2015, 2, 17. [Google Scholar] [CrossRef]
  319. Zheng, Y.; He, M.; Congdon, N. The worldwide epidemic of diabetic retinopathy. Indian. J. Ophthalmol. 2012, 60, 428–431. [Google Scholar] [CrossRef]
  320. Yau, J.W.; Rogers, S.L.; Kawasaki, R.; Lamoureux, E.L.; Kowalski, J.W.; Bek, T.; Chen, S.J.; Dekker, J.M.; Fletcher, A.; Grauslund, J.; et al. Global prevalence and major risk factors of diabetic retinopathy. Diabetes Care 2012, 35, 556–564. [Google Scholar] [CrossRef]
  321. Sheng, X.; Zhang, C.; Zhao, J.; Xu, J.; Zhang, P.; Ding, Q.; Zhang, J. Microvascular destabilization and intricated network of the cytokines in diabetic retinopathy: From the perspective of cellular and molecular components. Cell Biosci. 2024, 14, 85. [Google Scholar] [CrossRef]
  322. Lundeen, E.A.; Burke-Conte, Z.; Rein, D.B.; Wittenborn, J.S.; Saaddine, J.; Lee, A.Y.; Flaxman, A.D. Prevalence of Diabetic Retinopathy in the US in 2021. JAMA Ophthalmol. 2023, 141, 747–754. [Google Scholar] [CrossRef]
  323. Rodriguez, M.L.; Perez, S.; Mena-Molla, S.; Desco, M.C.; Ortega, A.L. Oxidative Stress and Microvascular Alterations in Diabetic Retinopathy: Future Therapies. Oxid. Med. Cell Longev. 2019, 2019, 4940825. [Google Scholar] [CrossRef] [PubMed]
  324. Andersen, N.; Hjortdal, J.O.; Schielke, K.C.; Bek, T.; Grauslund, J.; Laugesen, C.S.; Lund-Andersen, H.; Cerqueira, C.; Andresen, J. The Danish Registry of Diabetic Retinopathy. Clin. Epidemiol. 2016, 8, 613–619. [Google Scholar] [CrossRef] [PubMed]
  325. Kusuhara, S.; Fukushima, Y.; Ogura, S.; Inoue, N.; Uemura, A. Pathophysiology of Diabetic Retinopathy: The Old and the New. Diabetes Metab. J. 2018, 42, 364–376. [Google Scholar] [CrossRef] [PubMed]
  326. Nentwich, M.M.; Ulbig, M.W. Diabetic retinopathy—Ocular complications of diabetes mellitus. World J. Diabetes 2015, 6, 489–499. [Google Scholar] [CrossRef]
  327. Rusciano, D.; Bagnoli, P. Oxygen, the Paradox of Life and the Eye. Front. Biosci. 2024, 29, 319. [Google Scholar] [CrossRef]
  328. Hammes, H.P. Diabetic retinopathy: Hyperglycaemia, oxidative stress and beyond. Diabetologia 2018, 61, 29–38. [Google Scholar] [CrossRef]
  329. Kowluru, R.A.; Chan, P.S. Oxidative stress and diabetic retinopathy. Exp. Diabetes Res. 2007, 2007, 43603. [Google Scholar] [CrossRef]
  330. Kowluru, R.A. Retinopathy in a Diet-Induced Type 2 Diabetic Rat Model and Role of Epigenetic Modifications. Diabetes 2020, 69, 689–698. [Google Scholar] [CrossRef]
  331. Sui, A.; Chen, X.; Demetriades, A.M.; Shen, J.; Cai, Y.; Yao, Y.; Yao, Y.; Zhu, Y.; Shen, X.; Xie, B. Inhibiting NF-kappaB Signaling Activation Reduces Retinal Neovascularization by Promoting a Polarization Shift in Macrophages. Invest. Ophthalmol. Vis. Sci. 2020, 61, 4. [Google Scholar] [CrossRef]
  332. Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef]
  333. Miller, W.P.; Sunilkumar, S.; Giordano, J.F.; Toro, A.L.; Barber, A.J.; Dennis, M.D. The stress response protein REDD1 promotes diabetes-induced oxidative stress in the retina by Keap1-independent Nrf2 degradation. J. Biol. Chem. 2020, 295, 7350–7361. [Google Scholar] [CrossRef] [PubMed]
  334. Zhong, Q.; Mishra, M.; Kowluru, R.A. Transcription factor Nrf2-mediated antioxidant defense system in the development of diabetic retinopathy. Invest. Ophthalmol. Vis. Sci. 2013, 54, 3941–3948. [Google Scholar] [CrossRef] [PubMed]
  335. Wu, M.Y.; Yiang, G.T.; Lai, T.T.; Li, C.J. The Oxidative Stress and Mitochondrial Dysfunction during the Pathogenesis of Diabetic Retinopathy. Oxid. Med. Cell Longev. 2018, 2018, 3420187. [Google Scholar] [CrossRef] [PubMed]
  336. Miller, R.G.; Orchard, T.J. Understanding Metabolic Memory: A Tale of Two Studies. Diabetes 2020, 69, 291–299. [Google Scholar] [CrossRef]
  337. Kang, Q.; Yang, C. Oxidative stress and diabetic retinopathy: Molecular mechanisms, pathogenetic role and therapeutic implications. Redox Biol. 2020, 37, 101799. [Google Scholar] [CrossRef]
  338. Kang, E.; Wu, J.; Gutierrez, N.M.; Koski, A.; Tippner-Hedges, R.; Agaronyan, K.; Platero-Luengo, A.; Martinez-Redondo, P.; Ma, H.; Lee, Y.; et al. Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature 2016, 540, 270–275. [Google Scholar] [CrossRef]
  339. Jemt, E.; Persson, O.; Shi, Y.; Mehmedovic, M.; Uhler, J.P.; Davila Lopez, M.; Freyer, C.; Gustafsson, C.M.; Samuelsson, T.; Falkenberg, M. Regulation of DNA replication at the end of the mitochondrial D-loop involves the helicase TWINKLE and a conserved sequence element. Nucleic Acids Res. 2015, 43, 9262–9275. [Google Scholar] [CrossRef]
  340. Mishra, M.; Kowluru, R.A. Epigenetic Modification of Mitochondrial DNA in the Development of Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2015, 56, 5133–5142. [Google Scholar] [CrossRef]
  341. Mishra, M.; Kowluru, R.A. DNA Methylation-a Potential Source of Mitochondria DNA Base Mismatch in the Development of Diabetic Retinopathy. Mol. Neurobiol. 2019, 56, 88–101. [Google Scholar] [CrossRef]
  342. Kumar, J.; Mohammad, G.; Alka, K.; Kowluru, R.A. Mitochondrial Genome-Encoded Long Noncoding RNA and Mitochondrial Stability in Diabetic Retinopathy. Diabetes 2023, 72, 520–531. [Google Scholar] [CrossRef]
  343. Chen, X.J.; Butow, R.A. The organization and inheritance of the mitochondrial genome. Nat. Rev. Genet. 2005, 6, 815–825. [Google Scholar] [CrossRef] [PubMed]
  344. Santos, J.M.; Tewari, S.; Lin, J.Y.; Kowluru, R.A. Interrelationship between activation of matrix metalloproteinases and mitochondrial dysfunction in the development of diabetic retinopathy. Biochem. Biophys. Res. Commun. 2013, 438, 760–764. [Google Scholar] [CrossRef] [PubMed]
  345. Zhao, Y.; McLaughlin, D.; Robinson, E.; Harvey, A.P.; Hookham, M.B.; Shah, A.M.; McDermott, B.J.; Grieve, D.J. Nox2 NADPH oxidase promotes pathologic cardiac remodeling associated with Doxorubicin chemotherapy. Cancer Res. 2010, 70, 9287–9297. [Google Scholar] [CrossRef]
  346. Ago, T.; Kuroda, J.; Kamouchi, M.; Sadoshima, J.; Kitazono, T. Pathophysiological roles of NADPH oxidase/nox family proteins in the vascular system. -Rev. perspective. Circ. J. 2011, 75, 1791–1800. [Google Scholar] [CrossRef]
  347. Kowluru, R.A.; Kowluru, A.; Mishra, M.; Kumar, B. Oxidative stress and epigenetic modifications in the pathogenesis of diabetic retinopathy. Prog. Retin. Eye Res. 2015, 48, 40–61. [Google Scholar] [CrossRef]
  348. Kowluru, R.A.; Mohammad, G.; dos Santos, J.M.; Zhong, Q. Abrogation of MMP-9 gene protects against the development of retinopathy in diabetic mice by preventing mitochondrial damage. Diabetes 2011, 60, 3023–3033. [Google Scholar] [CrossRef]
  349. Mohammad, G.; Kowluru, R.A. Novel role of mitochondrial matrix metalloproteinase-2 in the development of diabetic retinopathy. Invest. Ophthalmol. Vis. Sci. 2011, 52, 3832–3841. [Google Scholar] [CrossRef]
  350. Kalani, A.; Kamat, P.K.; Tyagi, N. Diabetic Stroke Severity: Epigenetic Remodeling and Neuronal, Glial, and Vascular Dysfunction. Diabetes 2015, 64, 4260–4271. [Google Scholar] [CrossRef]
  351. Kowluru, R.A.; Mishra, M. Oxidative stress, mitochondrial damage and diabetic retinopathy. Biochim. Biophys. Acta 2015, 1852, 2474–2483. [Google Scholar] [CrossRef]
  352. Kanwar, M.; Chan, P.S.; Kern, T.S.; Kowluru, R.A. Oxidative damage in the retinal mitochondria of diabetic mice: Possible protection by superoxide dismutase. Invest. Ophthalmol. Vis. Sci. 2007, 48, 3805–3811. [Google Scholar] [CrossRef]
  353. Yi, X.; Guo, W.; Shi, Q.; Yang, Y.; Zhang, W.; Chen, X.; Kang, P.; Chen, J.; Cui, T.; Ma, J.; et al. SIRT3-Dependent Mitochondrial Dynamics Remodeling Contributes to Oxidative Stress-Induced Melanocyte Degeneration in Vitiligo. Theranostics 2019, 9, 1614–1633. [Google Scholar] [CrossRef] [PubMed]
  354. Guerra-Castellano, A.; Diaz-Quintana, A.; Perez-Mejias, G.; Elena-Real, C.A.; Gonzalez-Arzola, K.; Garcia-Maurino, S.M.; De la Rosa, M.A.; Diaz-Moreno, I. Oxidative stress is tightly regulated by cytochrome c phosphorylation and respirasome factors in mitochondria. Proc. Natl. Acad. Sci. USA 2018, 115, 7955–7960. [Google Scholar] [CrossRef] [PubMed]
  355. Ahmad, R.; Hussain, A.; Ahsan, H. Peroxynitrite: Cellular pathology and implications in autoimmunity. J. Immunoass. Immunochem. 2019, 40, 123–138. [Google Scholar] [CrossRef] [PubMed]
  356. Szabo, C.; Ischiropoulos, H.; Radi, R. Peroxynitrite: Biochemistry, pathophysiology and development of therapeutics. Nat. Rev. Drug Discov. 2007, 6, 662–680. [Google Scholar] [CrossRef]
  357. Radi, R. Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc. Natl. Acad. Sci. USA 2018, 115, 5839–5848. [Google Scholar] [CrossRef]
  358. Radi, R.; Cassina, A.; Hodara, R.; Quijano, C.; Castro, L. Peroxynitrite reactions and formation in mitochondria. Free Radic. Biol. Med. 2002, 33, 1451–1464. [Google Scholar] [CrossRef]
  359. Rask-Madsen, C.; King, G.L. Vascular complications of diabetes: Mechanisms of injury and protective factors. Cell Metab. 2013, 17, 20–33. [Google Scholar] [CrossRef]
  360. Wang, Y.; Eshwaran, R.; Beck, S.C.; Hammes, H.P.; Wieland, T.; Feng, Y. Contribution of the hexosamine biosynthetic pathway in the hyperglycemia-dependent and -independent breakdown of the retinal neurovascular unit. Mol. Metab. 2023, 73, 101736. [Google Scholar] [CrossRef]
  361. Van Opdenbosch, N.; Lamkanfi, M. Caspases in Cell Death, Inflammation, and Disease. Immunity 2019, 50, 1352–1364. [Google Scholar] [CrossRef]
  362. Riedl, S.J.; Shi, Y. Molecular mechanisms of caspase regulation during apoptosis. Nat. Rev. Mol. Cell Biol. 2004, 5, 897–907. [Google Scholar] [CrossRef]
  363. Dehdashtian, E.; Mehrzadi, S.; Yousefi, B.; Hosseinzadeh, A.; Reiter, R.J.; Safa, M.; Ghaznavi, H.; Naseripour, M. Diabetic retinopathy pathogenesis and the ameliorating effects of melatonin; involvement of autophagy, inflammation and oxidative stress. Life Sci. 2018, 193, 20–33. [Google Scholar] [CrossRef] [PubMed]
  364. Du, Y.; Miller, C.M.; Kern, T.S. Hyperglycemia increases mitochondrial superoxide in retina and retinal cells. Free Radic. Biol. Med. 2003, 35, 1491–1499. [Google Scholar] [CrossRef] [PubMed]
  365. Cao, R.; Li, L.; Ying, Z.; Cao, Z.; Ma, Y.; Mao, X.; Li, J.; Qi, X.; Zhang, Z.; Wang, X. A small molecule protects mitochondrial integrity by inhibiting mTOR activity. Proc. Natl. Acad. Sci. USA 2019, 116, 23332–23338. [Google Scholar] [CrossRef] [PubMed]
  366. Jiang, X.; Li, L.; Ying, Z.; Pan, C.; Huang, S.; Li, L.; Dai, M.; Yan, B.; Li, M.; Jiang, H.; et al. A Small Molecule That Protects the Integrity of the Electron Transfer Chain Blocks the Mitochondrial Apoptotic Pathway. Mol. Cell 2016, 63, 229–239. [Google Scholar] [CrossRef]
  367. Kowluru, R.A.; Abbas, S.N. Diabetes-induced mitochondrial dysfunction in the retina. Invest. Ophthalmol. Vis. Sci. 2003, 44, 5327–5334. [Google Scholar] [CrossRef]
  368. Jiang, X.; Jiang, H.; Shen, Z.; Wang, X. Activation of mitochondrial protease OMA1 by Bax and Bak promotes cytochrome c release during apoptosis. Proc. Natl. Acad. Sci. USA 2014, 111, 14782–14787. [Google Scholar] [CrossRef]
  369. Oeckinghaus, A.; Hayden, M.S.; Ghosh, S. Crosstalk in NF-kappaB signaling pathways. Nat. Immunol. 2011, 12, 695–708. [Google Scholar] [CrossRef]
  370. Moscat, J.; Diaz-Meco, M.T. p62 at the crossroads of autophagy, apoptosis, and cancer. Cell 2009, 137, 1001–1004. [Google Scholar] [CrossRef]
  371. Kowluru, R.A.; Koppolu, P.; Chakrabarti, S.; Chen, S. Diabetes-induced activation of nuclear transcriptional factor in the retina, and its inhibition by antioxidants. Free Radic. Res. 2003, 37, 1169–1180. [Google Scholar] [CrossRef]
  372. Nakajima, S.; Kitamura, M. Bidirectional regulation of NF-kappaB by reactive oxygen species: A role of unfolded protein response. Free Radic. Biol. Med. 2013, 65, 162–174. [Google Scholar] [CrossRef]
  373. Romeo, G.; Liu, W.H.; Asnaghi, V.; Kern, T.S.; Lorenzi, M. Activation of nuclear factor-kappaB induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes. Diabetes 2002, 51, 2241–2248. [Google Scholar] [CrossRef] [PubMed]
  374. Chen, K.; Qiu, P.; Yuan, Y.; Zheng, L.; He, J.; Wang, C.; Guo, Q.; Kenny, J.; Liu, Q.; Zhao, J.; et al. Pseurotin A Inhibits Osteoclastogenesis and Prevents Ovariectomized-Induced Bone Loss by Suppressing Reactive Oxygen Species. Theranostics 2019, 9, 1634–1650. [Google Scholar] [CrossRef] [PubMed]
  375. Altamirano, F.; Lopez, J.R.; Henriquez, C.; Molinski, T.; Allen, P.D.; Jaimovich, E. Increased resting intracellular calcium modulates NF-kappaB-dependent inducible nitric-oxide synthase gene expression in dystrophic mdx skeletal myotubes. J. Biol. Chem. 2012, 287, 20876–20887. [Google Scholar] [CrossRef] [PubMed]
  376. Lim, J.W.; Kim, H.; Kim, K.H. NF-kappaB, inducible nitric oxide synthase and apoptosis by Helicobacter pylori infection. Free Radic. Biol. Med. 2001, 31, 355–366. [Google Scholar] [CrossRef]
  377. Baig, M.S.; Zaichick, S.V.; Mao, M.; de Abreu, A.L.; Bakhshi, F.R.; Hart, P.C.; Saqib, U.; Deng, J.; Chatterjee, S.; Block, M.L.; et al. NOS1-derived nitric oxide promotes NF-kappaB transcriptional activity through inhibition of suppressor of cytokine signaling-1. J. Exp. Med. 2015, 212, 1725–1738. [Google Scholar] [CrossRef]
  378. Njie-Mbye, Y.F.; Kulkarni-Chitnis, M.; Opere, C.A.; Barrett, A.; Ohia, S.E. Lipid peroxidation: Pathophysiological and pharmacological implications in the eye. Front. Physiol. 2013, 4, 366. [Google Scholar] [CrossRef]
  379. Robles-Rivera, R.R.; Castellanos-Gonzalez, J.A.; Olvera-Montano, C.; Flores-Martin, R.A.; Lopez-Contreras, A.K.; Arevalo-Simental, D.E.; Cardona-Munoz, E.G.; Roman-Pintos, L.M.; Rodriguez-Carrizalez, A.D. Adjuvant Therapies in Diabetic Retinopathy as an Early Approach to Delay Its Progression: The Importance of Oxidative Stress and Inflammation. Oxid. Med. Cell Longev. 2020, 2020, 3096470. [Google Scholar] [CrossRef]
  380. Gupta, M.M.; Chari, S. Lipid peroxidation and antioxidant status in patients with diabetic retinopathy. Indian. J. Physiol. Pharmacol. 2005, 49, 187–192. [Google Scholar]
  381. Zhou, T.; Zhou, K.K.; Lee, K.; Gao, G.; Lyons, T.J.; Kowluru, R.; Ma, J.X. The role of lipid peroxidation products and oxidative stress in activation of the canonical wingless-type MMTV integration site (WNT) pathway in a rat model of diabetic retinopathy. Diabetologia 2011, 54, 459–468. [Google Scholar] [CrossRef]
  382. Ohia, S.E.; Opere, C.A.; Leday, A.M. Pharmacological consequences of oxidative stress in ocular tissues. Mutat. Res. 2005, 579, 22–36. [Google Scholar] [CrossRef]
  383. Chen, Q.; Tang, L.; Xin, G.; Li, S.; Ma, L.; Xu, Y.; Zhuang, M.; Xiong, Q.; Wei, Z.; Xing, Z.; et al. Oxidative stress mediated by lipid metabolism contributes to high glucose-induced senescence in retinal pigment epithelium. Free Radic. Biol. Med. 2019, 130, 48–58. [Google Scholar] [CrossRef] [PubMed]
  384. Sharma, A.; Sharma, R.; Chaudhary, P.; Vatsyayan, R.; Pearce, V.; Jeyabal, P.V.; Zimniak, P.; Awasthi, S.; Awasthi, Y.C. 4-Hydroxynonenal induces p53-mediated apoptosis in retinal pigment epithelial cells. Arch. Biochem. Biophys. 2008, 480, 85–94. [Google Scholar] [CrossRef] [PubMed]
  385. Bradley, M.A.; Xiong-Fister, S.; Markesbery, W.R.; Lovell, M.A. Elevated 4-hydroxyhexenal in Alzheimer’s disease (AD) progression. Neurobiol. Aging 2012, 33, 1034–1044. [Google Scholar] [CrossRef]
  386. Pena-Bautista, C.; Vento, M.; Baquero, M.; Chafer-Pericas, C. Lipid peroxidation in neurodegeneration. Clin. Chim. Acta 2019, 497, 178–188. [Google Scholar] [CrossRef]
  387. Liu, L.; Zhang, K.; Sandoval, H.; Yamamoto, S.; Jaiswal, M.; Sanz, E.; Li, Z.; Hui, J.; Graham, B.H.; Quintana, A.; et al. Glial lipid droplets and ROS induced by mitochondrial defects promote neurodegeneration. Cell 2015, 160, 177–190. [Google Scholar] [CrossRef]
  388. Madsen-Bouterse, S.A.; Kowluru, R.A. Oxidative stress and diabetic retinopathy: Pathophysiological mechanisms and treatment perspectives. Rev. Endocr. Metab. Disord. 2008, 9, 315–327. [Google Scholar] [CrossRef]
  389. Khan, Z.A.; Chakrabarti, S. Cellular signaling and potential new treatment targets in diabetic retinopathy. Exp. Diabetes Res. 2007, 2007, 31867. [Google Scholar] [CrossRef]
  390. Roy, S.; Kim, D. Retinal capillary basement membrane thickening: Role in the pathogenesis of diabetic retinopathy. Prog. Retin. Eye Res. 2021, 82, 100903. [Google Scholar] [CrossRef]
  391. Mason, R.M.; Wahab, N.A. Extracellular matrix metabolism in diabetic nephropathy. J. Am. Soc. Nephrol. 2003, 14, 1358–1373. [Google Scholar] [CrossRef]
  392. Goh, S.Y.; Cooper, M.E. Clinical review: The role of advanced glycation end products in progression and complications of diabetes. J. Clin. Endocrinol. Metab. 2008, 93, 1143–1152. [Google Scholar] [CrossRef]
  393. Gardiner, T.A.; Anderson, H.R.; Stitt, A.W. Inhibition of advanced glycation end-products protects against retinal capillary basement membrane expansion during long-term diabetes. J. Pathol. 2003, 201, 328–333. [Google Scholar] [CrossRef] [PubMed]
  394. Cheung, N.; Mitchell, P.; Wong, T.Y. Diabetic retinopathy. Lancet 2010, 376, 124–136. [Google Scholar] [CrossRef] [PubMed]
  395. Li, J.; Wang, J.J.; Yu, Q.; Chen, K.; Mahadev, K.; Zhang, S.X. Inhibition of reactive oxygen species by Lovastatin downregulates vascular endothelial growth factor expression and ameliorates blood-retinal barrier breakdown in db/db mice: Role of NADPH oxidase 4. Diabetes 2010, 59, 1528–1538. [Google Scholar] [CrossRef] [PubMed]
  396. Deissler, H.L.; Deissler, H.; Lang, G.K.; Lang, G.E. VEGF but not PlGF disturbs the barrier of retinal endothelial cells. Exp. Eye Res. 2013, 115, 162–171. [Google Scholar] [CrossRef]
  397. Iwase, T.; Oveson, B.C.; Hashida, N.; Lima e Silva, R.; Shen, J.; Krauss, A.H.; Gale, D.C.; Adamson, P.; Campochiaro, P.A. Topical pazopanib blocks VEGF-induced vascular leakage and neovascularization in the mouse retina but is ineffective in the rabbit. Invest. Ophthalmol. Vis. Sci. 2013, 54, 503–511. [Google Scholar] [CrossRef]
  398. El-Remessy, A.B.; Franklin, T.; Ghaley, N.; Yang, J.; Brands, M.W.; Caldwell, R.B.; Behzadian, M.A. Diabetes-induced superoxide anion and breakdown of the blood-retinal barrier: Role of the VEGF/uPAR pathway. PLoS ONE 2013, 8, e71868. [Google Scholar] [CrossRef]
  399. Jo, D.H.; Yun, J.H.; Cho, C.S.; Kim, J.H.; Kim, J.H.; Cho, C.H. Interaction between microglia and retinal pigment epithelial cells determines the integrity of outer blood-retinal barrier in diabetic retinopathy. Glia 2019, 67, 321–331. [Google Scholar] [CrossRef]
  400. Geraldes, P.; Hiraoka-Yamamoto, J.; Matsumoto, M.; Clermont, A.; Leitges, M.; Marette, A.; Aiello, L.P.; Kern, T.S.; King, G.L. Activation of PKC-delta and SHP-1 by hyperglycemia causes vascular cell apoptosis and diabetic retinopathy. Nat. Med. 2009, 15, 1298–1306. [Google Scholar] [CrossRef]
  401. Nawaz, I.M.; Rezzola, S.; Cancarini, A.; Russo, A.; Costagliola, C.; Semeraro, F.; Presta, M. Human vitreous in proliferative diabetic retinopathy: Characterization and translational implications. Prog. Retin. Eye Res. 2019, 72, 100756. [Google Scholar] [CrossRef]
  402. Gianni-Barrera, R.; Butschkau, A.; Uccelli, A.; Certelli, A.; Valente, P.; Bartolomeo, M.; Groppa, E.; Burger, M.G.; Hlushchuk, R.; Heberer, M.; et al. PDGF-BB regulates splitting angiogenesis in skeletal muscle by limiting VEGF-induced endothelial proliferation. Angiogenesis 2018, 21, 883–900. [Google Scholar] [CrossRef]
  403. Ayalasomayajula, S.P.; Amrite, A.C.; Kompella, U.B. Inhibition of cyclooxygenase-2, but not cyclooxygenase-1, reduces prostaglandin E2 secretion from diabetic rat retinas. Eur. J. Pharmacol. 2004, 498, 275–278. [Google Scholar] [CrossRef] [PubMed]
  404. Schoenberger, S.D.; Kim, S.J.; Sheng, J.; Rezaei, K.A.; Lalezary, M.; Cherney, E. Increased prostaglandin E2 (PGE2) levels in proliferative diabetic retinopathy, and correlation with VEGF and inflammatory cytokines. Invest. Ophthalmol. Vis. Sci. 2012, 53, 5906–5911. [Google Scholar] [CrossRef] [PubMed]
  405. Belaidi, E.; Morand, J.; Gras, E.; Pepin, J.L.; Godin-Ribuot, D. Targeting the ROS-HIF-1-endothelin axis as a therapeutic approach for the treatment of obstructive sleep apnea-related cardiovascular complications. Pharmacol. Ther. 2016, 168, 1–11. [Google Scholar] [CrossRef] [PubMed]
  406. D’Angelo, G.; Loria, A.S.; Pollock, D.M.; Pollock, J.S. Endothelin activation of reactive oxygen species mediates stress-induced pressor response in Dahl salt-sensitive prehypertensive rats. Hypertension 2010, 56, 282–289. [Google Scholar] [CrossRef]
  407. Majumdar, P.; Chen, S.; George, B.; Sen, S.; Karmazyn, M.; Chakrabarti, S. Leptin and endothelin-1 mediated increased extracellular matrix protein production and cardiomyocyte hypertrophy in diabetic heart disease. Diabetes Metab. Res. Rev. 2009, 25, 452–463. [Google Scholar] [CrossRef]
  408. Evans, T.; Deng, D.X.; Chen, S.; Chakrabarti, S. Endothelin receptor blockade prevents augmented extracellular matrix component mRNA expression and capillary basement membrane thickening in the retina of diabetic and galactose-fed rats. Diabetes 2000, 49, 662–666. [Google Scholar] [CrossRef]
  409. Pugliese, G.; Penno, G.; Natali, A.; Barutta, F.; Di Paolo, S.; Reboldi, G.; Gesualdo, L.; De Nicola, L. Diabetic kidney disease: New clinical and therapeutic issues. Joint position statement of the Italian Diabetes Society and the Italian Society of Nephrology on “The natural history of diabetic kidney disease and treatment of hyperglycemia in patients with type 2 diabetes and impaired renal function”. J. Nephrol. 2020, 33, 9–35. [Google Scholar] [CrossRef]
  410. Sugahara, M.; Pak, W.L.W.; Tanaka, T.; Tang, S.C.W.; Nangaku, M. Update on diagnosis, pathophysiology, and management of diabetic kidney disease. Nephrology 2021, 26, 491–500. [Google Scholar] [CrossRef]
  411. Wang, R.; Song, F.; Li, S.; Wu, B.; Gu, Y.; Yuan, Y. Salvianolic acid A attenuates CCl(4)-induced liver fibrosis by regulating the PI3K/AKT/mTOR, Bcl-2/Bax and caspase-3/cleaved caspase-3 signaling pathways. Drug Des. Devel Ther. 2019, 13, 1889–1900. [Google Scholar] [CrossRef]
  412. Hay, N. Interplay between FOXO, TOR, and Akt. Biochim. Biophys. Acta 2011, 1813, 1965–1970. [Google Scholar] [CrossRef]
  413. Ying, C.; Mao, Y.; Chen, L.; Wang, S.; Ling, H.; Li, W.; Zhou, X. Bamboo leaf extract ameliorates diabetic nephropathy through activating the AKT signaling pathway in rats. Int. J. Biol. Macromol. 2017, 105, 1587–1594. [Google Scholar] [CrossRef] [PubMed]
  414. Guo, J.; Liu, Z.; Gong, R. Long noncoding RNA: An emerging player in diabetes and diabetic kidney disease. Clin. Sci. 2019, 133, 1321–1339. [Google Scholar] [CrossRef] [PubMed]
  415. Bolos, V.; Peinado, H.; Perez-Moreno, M.A.; Fraga, M.F.; Esteller, M.; Cano, A. The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: A comparison with Snail and E47 repressors. J. Cell Sci. 2003, 116, 499–511. [Google Scholar] [CrossRef]
  416. Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvandi, P.; Geoerger, B.; et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef]
  417. Lu, Q.; Wang, W.W.; Zhang, M.Z.; Ma, Z.X.; Qiu, X.R.; Shen, M.; Yin, X.X. ROS induces epithelial-mesenchymal transition via the TGF-beta1/PI3K/Akt/mTOR pathway in diabetic nephropathy. Exp. Ther. Med. 2019, 17, 835–846. [Google Scholar] [CrossRef]
  418. Qing, Y.; Dong, X.; Hongli, L.; Yanhui, L. Berberine promoted myocardial protection of postoperative patients through regulating myocardial autophagy. Biomed. Pharmacother. 2018, 105, 1050–1053. [Google Scholar] [CrossRef]
  419. Kim, M.E.; Kim, D.H.; Lee, J.S. FoxO Transcription Factors: Applicability as a Novel Immune Cell Regulators and Therapeutic Targets in Oxidative Stress-Related Diseases. Int. J. Mol. Sci. 2022, 23, 11877. [Google Scholar] [CrossRef]
  420. Barthel, A.; Schmoll, D.; Unterman, T.G. FoxO proteins in insulin action and metabolism. Trends Endocrinol. Metab. 2005, 16, 183–189. [Google Scholar] [CrossRef]
  421. Li, W.; Wang, Q.; Du, M.; Ma, X.; Wu, L.; Guo, F.; Zhao, S.; Huang, F.; Wang, H.; Qin, G. Effects of overexpressing FoxO1 on apoptosis in glomeruli of diabetic mice and in podocytes cultured in high glucose medium. Biochem. Biophys. Res. Commun. 2016, 478, 612–617. [Google Scholar] [CrossRef]
  422. Qin, G.; Zhou, Y.; Guo, F.; Ren, L.; Wu, L.; Zhang, Y.; Ma, X.; Wang, Q. Overexpression of the FoxO1 Ameliorates Mesangial Cell Dysfunction in Male Diabetic Rats. Mol. Endocrinol. 2015, 29, 1080–1091. [Google Scholar] [CrossRef]
  423. Ma, X.; Ma, J.; Leng, T.; Yuan, Z.; Hu, T.; Liu, Q.; Shen, T. Advances in oxidative stress in pathogenesis of diabetic kidney disease and efficacy of TCM intervention. Ren. Fail. 2023, 45, 2146512. [Google Scholar] [CrossRef] [PubMed]
  424. Wen, C.; Ying, Y.; Zhao, H.; Jiang, Q.; Gan, X.; Wei, Y.; Wei, J.; Huang, X. Resistance exercise affects catheter-related thrombosis in rats through miR-92a-3p, oxidative stress and the MAPK/NF-kappaB pathway. BMC Cardiovasc. Disord. 2021, 21, 440. [Google Scholar] [CrossRef] [PubMed]
  425. Zoccali, C.; Mallamaci, F. Nonproteinuric progressive diabetic kidney disease. Curr. Opin. Nephrol. Hypertens. 2019, 28, 227–232. [Google Scholar] [CrossRef] [PubMed]
  426. Hofmann, M.A.; Schiekofer, S.; Isermann, B.; Kanitz, M.; Henkels, M.; Joswig, M.; Treusch, A.; Morcos, M.; Weiss, T.; Borcea, V.; et al. Peripheral blood mononuclear cells isolated from patients with diabetic nephropathy show increased activation of the oxidative-stress sensitive transcription factor NF-kappaB. Diabetologia 1999, 42, 222–232. [Google Scholar] [CrossRef]
  427. Zheng, H.; Whitman, S.A.; Wu, W.; Wondrak, G.T.; Wong, P.K.; Fang, D.; Zhang, D.D. Therapeutic potential of Nrf2 activators in streptozotocin-induced diabetic nephropathy. Diabetes 2011, 60, 3055–3066. [Google Scholar] [CrossRef]
  428. Jiang, T.; Huang, Z.; Lin, Y.; Zhang, Z.; Fang, D.; Zhang, D.D. The protective role of Nrf2 in streptozotocin-induced diabetic nephropathy. Diabetes 2010, 59, 850–860. [Google Scholar] [CrossRef]
  429. Ma, L.; Wu, F.; Shao, Q.; Chen, G.; Xu, L.; Lu, F. Baicalin Alleviates Oxidative Stress and Inflammation in Diabetic Nephropathy via Nrf2 and MAPK Signaling Pathway. Drug Des. Devel Ther. 2021, 15, 3207–3221. [Google Scholar] [CrossRef]
  430. Sen, Z.; Weida, W.; Jie, M.; Li, S.; Dongming, Z.; Xiaoguang, C. Coumarin glycosides from Hydrangea paniculata slow down the progression of diabetic nephropathy by targeting Nrf2 anti-oxidation and smad2/3-mediated profibrosis. Phytomedicine 2019, 57, 385–395. [Google Scholar] [CrossRef]
  431. Alshehri, A.S. Kaempferol attenuates diabetic nephropathy in streptozotocin-induced diabetic rats by a hypoglycaemic effect and concomitant activation of the Nrf-2/Ho-1/antioxidants axis. Arch. Physiol. Biochem. 2023, 129, 984–997. [Google Scholar] [CrossRef]
  432. Huang, W.; Liang, Y.; Dong, J.; Zhou, L.; Gao, C.; Jiang, C.; Chen, M.; Long, Y.; Xu, Y. SUMO E3 Ligase PIASy Mediates High Glucose-Induced Activation of NF-kappaB Inflammatory Signaling in Rat Mesangial Cells. Mediat. Inflamm. 2017, 2017, 1685194. [Google Scholar] [CrossRef]
  433. Iyengar, P.V. Regulation of Ubiquitin Enzymes in the TGF-beta Pathway. Int. J. Mol. Sci. 2017, 18, 877. [Google Scholar] [CrossRef] [PubMed]
  434. Iyengar, P.V.; Jaynes, P.; Rodon, L.; Lama, D.; Law, K.P.; Lim, Y.P.; Verma, C.; Seoane, J.; Eichhorn, P.J. USP15 regulates SMURF2 kinetics through C-lobe mediated deubiquitination. Sci. Rep. 2015, 5, 14733. [Google Scholar] [CrossRef] [PubMed]
  435. Wu, Y.; Wang, L.; Deng, D.; Zhang, Q.; Liu, W. Renalase Protects against Renal Fibrosis by Inhibiting the Activation of the ERK Signaling Pathways. Int. J. Mol. Sci. 2017, 18, 855. [Google Scholar] [CrossRef] [PubMed]
  436. Zhu, M.; Wang, H.; Chen, J.; Zhu, H. Sinomenine improve diabetic nephropathy by inhibiting fibrosis and regulating the JAK2/STAT3/SOCS1 pathway in streptozotocin-induced diabetic rats. Life Sci. 2021, 265, 118855. [Google Scholar] [CrossRef]
  437. Yu, J.; Wu, H.; Liu, Z.Y.; Zhu, Q.; Shan, C.; Zhang, K.Q. Advanced glycation end products induce the apoptosis of and inflammation in mouse podocytes through CXCL9-mediated JAK2/STAT3 pathway activation. Int. J. Mol. Med. 2017, 40, 1185–1193. [Google Scholar] [CrossRef]
  438. Chow, F.; Ozols, E.; Nikolic-Paterson, D.J.; Atkins, R.C.; Tesch, G.H. Macrophages in mouse type 2 diabetic nephropathy: Correlation with diabetic state and progressive renal injury. Kidney Int. 2004, 65, 116–128. [Google Scholar] [CrossRef]
  439. Liu, Y.; Wang, W.; Zhang, J.; Gao, S.; Xu, T.; Yin, Y. JAK/STAT signaling in diabetic kidney disease. Front. Cell Dev. Biol. 2023, 11, 1233259. [Google Scholar] [CrossRef]
  440. Dugan, L.L.; You, Y.H.; Ali, S.S.; Diamond-Stanic, M.; Miyamoto, S.; DeCleves, A.E.; Andreyev, A.; Quach, T.; Ly, S.; Shekhtman, G.; et al. AMPK dysregulation promotes diabetes-related reduction of superoxide and mitochondrial function. J. Clin. Investig. 2013, 123, 4888–4899. [Google Scholar] [CrossRef]
  441. Papadimitriou, A.; Peixoto, E.B.; Silva, K.C.; Lopes de Faria, J.M.; Lopes de Faria, J.B. Increase in AMPK brought about by cocoa is renoprotective in experimental diabetes mellitus by reducing NOX4/TGFbeta-1 signaling. J. Nutr. Biochem. 2014, 25, 773–784. [Google Scholar] [CrossRef]
  442. Guan, G.; Chen, Y.; Dong, Y. Unraveling the AMPK-SIRT1-FOXO Pathway: The In-Depth Analysis and Breakthrough Prospects of Oxidative Stress-Induced Diseases. Antioxidants 2025, 14, 70. [Google Scholar] [CrossRef]
  443. Li, F.; Chen, Y.; Li, Y.; Huang, M.; Zhao, W. Geniposide alleviates diabetic nephropathy of mice through AMPK/SIRT1/NF-kappaB pathway. Eur. J. Pharmacol. 2020, 886, 173449. [Google Scholar] [CrossRef] [PubMed]
  444. Wu, L.; Liu, C.; Chang, D.Y.; Zhan, R.; Zhao, M.; Man Lam, S.; Shui, G.; Zhao, M.H.; Zheng, L.; Chen, M. The Attenuation of Diabetic Nephropathy by Annexin A1 via Regulation of Lipid Metabolism Through the AMPK/PPARalpha/CPT1b Pathway. Diabetes 2021, 70, 2192–2203. [Google Scholar] [CrossRef] [PubMed]
  445. Han, Y.C.; Tang, S.Q.; Liu, Y.T.; Li, A.M.; Zhan, M.; Yang, M.; Song, N.; Zhang, W.; Wu, X.Q.; Peng, C.H.; et al. AMPK agonist alleviate renal tubulointerstitial fibrosis via activating mitophagy in high fat and streptozotocin induced diabetic mice. Cell Death Dis. 2021, 12, 925. [Google Scholar] [CrossRef]
  446. Zhou, Y.; Tai, S.; Zhang, N.; Fu, L.; Wang, Y. Dapagliflozin prevents oxidative stress-induced endothelial dysfunction via sirtuin 1 activation. Biomed. Pharmacother. 2023, 165, 115213. [Google Scholar] [CrossRef]
  447. Karunakaran, U.; Elumalai, S.; Moon, J.S.; Won, K.C. Pioglitazone-induced AMPK-Glutaminase-1 prevents high glucose-induced pancreatic beta-cell dysfunction by glutathione antioxidant system. Redox Biol. 2021, 45, 102029. [Google Scholar] [CrossRef]
  448. Xue, C.Y.; Zhou, M.Q.; Zheng, Q.Y.; Zhang, J.H.; Cheng, W.T.; Bai, X.H.; Zhou, F.; Wu, A.M.; Nie, B.; Liu, W.J.; et al. Thiazolidinediones play a positive role in the vascular endothelium and inhibit plaque progression in diabetic patients with coronary atherosclerosis: A systematic review and meta-analysis. Front. Cardiovasc. Med. 2022, 9, 1043406. [Google Scholar] [CrossRef]
  449. Pandey, S.; Mangmool, S.; Parichatikanond, W. Multifaceted Roles of GLP-1 and Its Analogs: A Review on Molecular Mechanisms with a Cardiotherapeutic Perspective. Pharmaceuticals 2023, 16, 836. [Google Scholar] [CrossRef]
  450. Oh, Y.S.; Jun, H.S. Effects of Glucagon-like Peptide-1 on Oxidative Stress and Nrf2 Signaling. Int. J. Mol. Sci. 2017, 19, 26. [Google Scholar] [CrossRef]
  451. Shiraki, A.; Oyama, J.; Komoda, H.; Asaka, M.; Komatsu, A.; Sakuma, M.; Kodama, K.; Sakamoto, Y.; Kotooka, N.; Hirase, T.; et al. The glucagon-like peptide 1 analog liraglutide reduces TNF-alpha-induced oxidative stress and inflammation in endothelial cells. Atherosclerosis 2012, 221, 375–382. [Google Scholar] [CrossRef]
  452. Hullon, D.; Subeh, G.K.; Volkova, Y.; Janiec, K.; Trach, A.; Mnevets, R. The role of glucagon-like peptide-1 receptor (GLP-1R) agonists in enhancing endothelial function: A potential avenue for improving heart failure with preserved ejection fraction (HFpEF). Cardiovasc. Diabetol. 2025, 24, 70. [Google Scholar] [CrossRef]
  453. Verges, B.; Charbonnel, B. After the LEADER trial and SUSTAIN-6, how do we explain the cardiovascular benefits of some GLP-1 receptor agonists? Diabetes Metab. 2017, 43 (Suppl. S1), 2S3–2S12. [Google Scholar] [CrossRef] [PubMed]
  454. Warady, B.A.; Pergola, P.E.; Agarwal, R.; Andreoli, S.; Appel, G.B.; Bangalore, S.; Block, G.A.; Chapman, A.B.; Chin, M.P.; Gibson, K.L.; et al. Effects of Bardoxolone Methyl in Alport Syndrome. Clin. J. Am. Soc. Nephrol. 2022, 17, 1763–1774. [Google Scholar] [CrossRef] [PubMed]
  455. Alves, I.; Araujo, E.M.Q.; Dalgaard, L.T.; Singh, S.; Borsheim, E.; Carvalho, E. Protective Effects of Sulforaphane Preventing Inflammation and Oxidative Stress to Enhance Metabolic Health: A Narrative Review. Nutrients 2025, 17, 428. [Google Scholar] [CrossRef] [PubMed]
  456. Gunther, K.; Profeta, V.; Keita, M.; Park, C.; Wells, M.; Sharma, S.; Schadt, K.; Lynch, D.R. Safety Monitoring of Omaveloxolone in Friedreich Ataxia: Results from One Year of Clinical Treatment. Neurol. Ther. 2025, 14, 1105–1114. [Google Scholar] [CrossRef]
  457. Gray, S.P.; Jha, J.C.; Kennedy, K.; van Bommel, E.; Chew, P.; Szyndralewiez, C.; Touyz, R.M.; Schmidt, H.; Cooper, M.E.; Jandeleit-Dahm, K.A.M. Combined NOX1/4 inhibition with GKT137831 in mice provides dose-dependent reno- and atheroprotection even in established micro- and macrovascular disease. Diabetologia 2017, 60, 927–937. [Google Scholar] [CrossRef]
  458. Xu, M.; Feng, P.; Yan, J.; Li, L. Mitochondrial quality control: A pathophysiological mechanism and potential therapeutic target for chronic obstructive pulmonary disease. Front. Pharmacol. 2024, 15, 1474310. [Google Scholar] [CrossRef]
  459. Fairley, L.H.; Das, S.; Dharwal, V.; Amorim, N.; Hegarty, K.J.; Wadhwa, R.; Mounika, G.; Hansbro, P.M. Mitochondria-Targeted Antioxidants as a Therapeutic Strategy for Chronic Obstructive Pulmonary Disease. Antioxidants 2023, 12, 973. [Google Scholar] [CrossRef]
  460. Tarantini, S.; Valcarcel-Ares, N.M.; Yabluchanskiy, A.; Fulop, G.A.; Hertelendy, P.; Gautam, T.; Farkas, E.; Perz, A.; Rabinovitch, P.S.; Sonntag, W.E.; et al. Treatment with the mitochondrial-targeted antioxidant peptide SS-31 rescues neurovascular coupling responses and cerebrovascular endothelial function and improves cognition in aged mice. Aging Cell 2018, 17, e12731. [Google Scholar] [CrossRef]
  461. Malaviya, P.; Kumar, J.; Kowluru, R.A. Role of ferroptosis in mitochondrial damage in diabetic retinopathy. Free Radic. Biol. Med. 2024, 225, 821–832. [Google Scholar] [CrossRef]
  462. Zhang, X.; Sun, J.; Wang, J.; Meng, T.; Yang, J.; Zhou, Y. The role of ferroptosis in diabetic cardiovascular diseases and the intervention of active ingredients of traditional Chinese medicine. Front. Pharmacol. 2023, 14, 1286718. [Google Scholar] [CrossRef]
  463. Jiang, Y.; Xing, S.; Ni, D.; Yang, B.; Kai, J.; Wang, T.; Yu, W.; Dai, Y. Curcumin Attenuates Ferroptosis and Ameliorates Erectile Function in Diabetic Rats by Activating Nrf2/HO-1 Pathway. Andrologia 2023, 2023, 7236816. [Google Scholar] [CrossRef]
  464. Guan, T.; Lu, Z.; Tai, R.; Guo, S.; Zhang, Z.; Deng, S.; Ye, J.; Chi, K.; Zhang, B.; Chen, H.; et al. Silicified curcumin microspheres Combats cardiovascular diseases via Nrf2/HO-1 pathway. Bioact. Mater. 2025, 49, 378–398. [Google Scholar] [CrossRef] [PubMed]
  465. Ataei, M.; Gumpricht, E.; Kesharwani, P.; Jamialahmadi, T.; Sahebkar, A. Recent advances in curcumin-based nanoformulations in diabetes. J. Drug Target. 2023, 31, 671–684. [Google Scholar] [CrossRef] [PubMed]
  466. Xue, M.; Tian, Y.; Zhang, H.; Dai, S.; Wu, Y.; Jin, J.; Chen, J. Curcumin nanocrystals ameliorate ferroptosis of diabetic nephropathy through glutathione peroxidase 4. Front. Pharmacol. 2024, 15, 1508312. [Google Scholar] [CrossRef]
  467. Li, C.; Wang, Z.; Lei, H.; Zhang, D. Recent progress in nanotechnology-based drug carriers for resveratrol delivery. Drug Deliv. 2023, 30, 2174206. [Google Scholar] [CrossRef]
  468. Hou, L.; Zhang, M.; Liu, L.; Zhong, Q.; Xie, M.; Zhao, G. Therapeutic applications of nanomedicine in metabolic diseases by targeting the endothelium. QJM 2023, 116, 493–501. [Google Scholar] [CrossRef]
  469. Mori, T.; Giovannelli, L.; Bilia, A.R.; Margheri, F. Exosomes: Potential Next-Generation Nanocarriers for the Therapy of Inflammatory Diseases. Pharmaceutics 2023, 15, 2276. [Google Scholar] [CrossRef]
  470. Yan, H.; Hu, Y.; Lyu, Y.; Akk, A.; Hirbe, A.C.; Wickline, S.A.; Pan, H.; Roberson, E.D.O.; Pham, C.T.N. Augmented expression of superoxide dismutase 2 mitigates progression and rupture of experimental abdominal aortic aneurysm. Theranostics 2025, 15, 4016–4032. [Google Scholar] [CrossRef]
  471. Liu, Z.; Gou, Y.; Zhang, H.; Zuo, H.; Zhang, H.; Liu, Z.; Yao, D. Estradiol improves cardiovascular function through up-regulation of SOD2 on vascular wall. Redox Biol. 2014, 3, 88–99. [Google Scholar] [CrossRef]
  472. He, L.; Chen, Q.; Wang, L.; Pu, Y.; Huang, J.; Cheng, C.K.; Luo, J.Y.; Kang, L.; Lin, X.; Xiang, L.; et al. Activation of Nrf2 inhibits atherosclerosis in ApoE(-/-) mice through suppressing endothelial cell inflammation and lipid peroxidation. Redox Biol. 2024, 74, 103229. [Google Scholar] [CrossRef]
  473. Kurzhagen, J.T.; Noel, S.; Lee, K.; Sadasivam, M.; Gharaie, S.; Ankireddy, A.; Lee, S.A.; Newman-Rivera, A.; Gong, J.; Arend, L.J.; et al. T Cell Nrf2/Keap1 Gene Editing Using CRISPR/Cas9 and Experimental Kidney Ischemia-Reperfusion Injury. Antioxid. Redox Signal 2023, 38, 959–973. [Google Scholar] [CrossRef] [PubMed]
  474. Wang, Y.M.; Huang, T.L.; Meng, C.; Zhang, J.; Fang, N.Y. SIRT1 deacetylates mitochondrial trifunctional enzyme alpha subunit to inhibit ubiquitylation and decrease insulin resistance. Cell Death Dis. 2020, 11, 821. [Google Scholar] [CrossRef] [PubMed]
  475. Yoshino, M.; Yoshino, J.; Kayser, B.D.; Patti, G.J.; Franczyk, M.P.; Mills, K.F.; Sindelar, M.; Pietka, T.; Patterson, B.W.; Imai, S.I.; et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science 2021, 372, 1224–1229. [Google Scholar] [CrossRef]
  476. De Picciotto, N.E.; Gano, L.B.; Johnson, L.C.; Martens, C.R.; Sindler, A.L.; Mills, K.F.; Imai, S.; Seals, D.R. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell 2016, 15, 522–530. [Google Scholar] [CrossRef] [PubMed]
  477. Naser, N.; Lonj, C.K.; Rikard-Bell, M.; Sandow, S.L.; Murphy, T.V. Advanced glycated end-products inhibit dilation through constitutive endothelial RAGE and Nox1/4 in rat isolated skeletal muscle arteries. Microcirculation 2024, 31, e12837. [Google Scholar] [CrossRef]
  478. Mozaffari, M.S. Therapeutic Potential of Annexin A1 Modulation in Kidney and Cardiovascular Disorders. Cells 2021, 10, 3420. [Google Scholar] [CrossRef]
  479. Surugiu, R.; Iancu, M.A.; Vintilescu, S.B.; Stepan, M.D.; Burdusel, D.; Genunche-Dumitrescu, A.V.; Dogaru, C.A.; Dumitra, G.G. Molecular Mechanisms of Healthy Aging: The Role of Caloric Restriction, Intermittent Fasting, Mediterranean Diet, and Ketogenic Diet-A Scoping Review. Nutrients 2024, 16, 2878. [Google Scholar] [CrossRef]
  480. Wang, Z.; Liu, S.; Shi, J.; Chen, D.; Li, S.; Yu, S.; Liu, S.; Yang, K.; Zhang, W.; Gao, X.; et al. Empagliflozin ameliorates RSL3-induced ferroptosis in vascular endothelial cells via the NRF2/HO-1 pathway. BMC Cardiovasc. Disord. 2025, 25, 437. [Google Scholar] [CrossRef]
  481. Lambadiari, V.; Thymis, J.; Kouretas, D.; Skaperda, Z.; Tekos, F.; Kousathana, F.; Kountouri, A.; Balampanis, K.; Parissis, J.; Andreadou, I.; et al. Effects of a 12-Month Treatment with Glucagon-like Peptide-1 Receptor Agonists, Sodium-Glucose Cotransporter-2 Inhibitors, and Their Combination on Oxidant and Antioxidant Biomarkers in Patients with Type 2 Diabetes. Antioxidants 2021, 10, 1379. [Google Scholar] [CrossRef]
  482. Martos-Guillami, N.; Vergara, A.; Llorens-Cebria, C.; Motto, A.E.; Martinez-Diaz, I.; Goncalves, F.; Garcias-Ramis, M.M.; Allo-Urzainqui, E.; Narvaez, A.; Bermejo, S.; et al. SGLT2i and GLP1-RA exert additive cardiorenal protection with a RAS blocker in uninephrectomized db/db mice. Front. Pharmacol. 2024, 15, 1415879. [Google Scholar] [CrossRef]
  483. Han, J.X.; Luo, L.L.; Wang, Y.C.; Miyagishi, M.; Kasim, V.; Wu, S.R. SGLT2 inhibitor empagliflozin promotes revascularization in diabetic mouse hindlimb ischemia by inhibiting ferroptosis. Acta Pharmacol. Sin. 2023, 44, 1161–1174. [Google Scholar] [CrossRef] [PubMed]
  484. Shihabi, A.; Li, W.G.; Miller, F.J., Jr.; Weintraub, N.L. Antioxidant therapy for atherosclerotic vascular disease: The promise and the pitfalls. Am. J. Physiol. Heart Circ. Physiol. 2002, 282, H797–H802. [Google Scholar] [CrossRef] [PubMed]
  485. Libby, P.; Everett, B.M. Novel Antiatherosclerotic Therapies. Arter. Thromb. Vasc. Biol. 2019, 39, 538–545. [Google Scholar] [CrossRef]
  486. Nunez-Selles, A.J.; Nunez-Musa, R.A.; Guillen-Marmolejos, R.A. Linking oxidative stress biomarkers to disease progression and antioxidant therapy in hypertension and diabetes mellitus. Front. Mol. Biosci. 2025, 12, 1611842. [Google Scholar] [CrossRef]
  487. Ramos Gonzalez, M.; Axler, M.R.; Kaseman, K.E.; Lobene, A.J.; Farquhar, W.B.; Witman, M.A.; Kirkman, D.L.; Lennon, S.L. Melatonin supplementation does not alter vascular function or oxidative stress in healthy normotensive adults on a high sodium diet. Physiol. Rep. 2023, 11, e15896. [Google Scholar] [CrossRef]
  488. Trimarchi, H.; Mongitore, M.R.; Baglioni, P.; Forrester, M.; Freixas, E.A.; Schropp, M.; Pereyra, H.; Alonso, M. N-acetylcysteine reduces malondialdehyde levels in chronic hemodialysis patients—A pilot study. Clin. Nephrol. 2003, 59, 441–446. [Google Scholar] [CrossRef]
  489. Rossman, M.J.; Santos-Parker, J.R.; Steward, C.A.C.; Bispham, N.Z.; Cuevas, L.M.; Rosenberg, H.L.; Woodward, K.A.; Chonchol, M.; Gioscia-Ryan, R.A.; Murphy, M.P.; et al. Chronic Supplementation with a Mitochondrial Antioxidant (MitoQ) Improves Vascular Function in Healthy Older Adults. Hypertension 2018, 71, 1056–1063. [Google Scholar] [CrossRef]
  490. Park, S.Y.; Pekas, E.J.; Headid, R.J., 3rd; Son, W.M.; Wooden, T.K.; Song, J.; Layec, G.; Yadav, S.K.; Mishra, P.K.; Pipinos, I.I. Acute mitochondrial antioxidant intake improves endothelial function, antioxidant enzyme activity, and exercise tolerance in patients with peripheral artery disease. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H456–H467. [Google Scholar] [CrossRef]
  491. Daei, S.; Ildarabadi, A.; Goodarzi, S.; Mohamadi-Sartang, M. Effect of Coenzyme Q10 Supplementation on Vascular Endothelial Function: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. High. Blood Press. Cardiovasc. Prev. 2024, 31, 113–126. [Google Scholar] [CrossRef]
  492. Li, J.J.; Dou, K.F.; Zhou, Z.G.; Zhao, D.; Ye, P.; Zhao, J.J.; Guo, L.X. Role of omega-3 fatty acids in the prevention and treatment of cardiovascular Diseases: A consensus statement from the Experts’ Committee of National Society of Cardiometabolic Medicine. Front. Pharmacol. 2022, 13, 1069992. [Google Scholar] [CrossRef]
  493. Kaur, G.; Mason, R.P.; Steg, P.G.; Bhatt, D.L. Omega-3 fatty acids for cardiovascular event lowering. Eur. J. Prev. Cardiol. 2024, 31, 1005–1014. [Google Scholar] [CrossRef] [PubMed]
  494. 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] [PubMed]
  495. Lonn, E.; Bosch, J.; Yusuf, S.; Sheridan, P.; Pogue, J.; Arnold, J.M.; Ross, C.; Arnold, A.; Sleight, P.; Probstfield, J.; et al. Effects of long-term vitamin E supplementation on cardiovascular events and cancer: A randomized controlled trial. JAMA 2005, 293, 1338–1347. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Sources of ROS. ROS production arises from NADPH oxidases (isoforms 1–5), exposure to radiation, oxidative phosphorylation within mitochondria, and endoplasmic reticulum (ER) stress with unfolded protein response. Additional sources include nitric oxide synthase activity, enzymatic reactions, and oxidative enzymes such as xanthine oxidase, cytochrome P450 (CYP450), and lipoxygenases. Metabolic reactions within peroxisomes and exposure to microbes, nanoparticles, and xenobiotics also contribute to ROS generation. Together, these diverse pathways highlight the multifactorial origins of ROS within biological systems, underscoring their central role in oxidative stress and cellular damage.
Figure 1. Sources of ROS. ROS production arises from NADPH oxidases (isoforms 1–5), exposure to radiation, oxidative phosphorylation within mitochondria, and endoplasmic reticulum (ER) stress with unfolded protein response. Additional sources include nitric oxide synthase activity, enzymatic reactions, and oxidative enzymes such as xanthine oxidase, cytochrome P450 (CYP450), and lipoxygenases. Metabolic reactions within peroxisomes and exposure to microbes, nanoparticles, and xenobiotics also contribute to ROS generation. Together, these diverse pathways highlight the multifactorial origins of ROS within biological systems, underscoring their central role in oxidative stress and cellular damage.
Medsci 13 00087 g001
Figure 2. Mechanisms of oxidative-stress-induced endothelial dysfunction in DM. Hyperglycemia increases ROS production via upregulation of the electron transport system, NADPH oxidase, xanthine oxidase, and uncoupled eNOS, while decreasing antioxidant defenses such as peroxiredoxins, thioredoxin, superoxide dismutase, uncoupling proteins, and catalase/peroxidases. Elevated ROS activates signaling pathways including oxidized LDL, MAPK, and PI3K/Ras/Akt, which in turn stimulate redox-sensitive transcription factors (e.g., NFκB, AP-1), leading to increased redox gene expression of adhesion molecules and chemokines (e.g., VCAM-1, ICAM-1, MCP-1). These molecular changes promote smooth muscle cell (SMC) growth, inflammation, thrombosis, vascular remodeling, apoptosis, and impaired vascular tone, culminating in endothelial dysfunction and contributing to the development of diseases associated with endothelial dysfunction in diabetes mellitus. NADPH, Nicotinamide adenine dinucleotide phosphate; eNOS, endothelial nitric oxide synthase; ROS, reactive oxygen species; LDL, low-density lipoprotein; MAPK, mitogen-activated protein kinase; PI3K, phosphoinositide 3-kinase; Ras, rat sarcoma virus; Akt, protein kinase B; NFκB, nuclear factor kappa B; AP-1, activator protein 1; VCAM-1, vascular cell adhesion molecule 1; ICAM-1, intercellular adhesion molecule 1; MCP-1, monocyte chemoattractant protein 1; SMC, smooth muscle cell. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
Figure 2. Mechanisms of oxidative-stress-induced endothelial dysfunction in DM. Hyperglycemia increases ROS production via upregulation of the electron transport system, NADPH oxidase, xanthine oxidase, and uncoupled eNOS, while decreasing antioxidant defenses such as peroxiredoxins, thioredoxin, superoxide dismutase, uncoupling proteins, and catalase/peroxidases. Elevated ROS activates signaling pathways including oxidized LDL, MAPK, and PI3K/Ras/Akt, which in turn stimulate redox-sensitive transcription factors (e.g., NFκB, AP-1), leading to increased redox gene expression of adhesion molecules and chemokines (e.g., VCAM-1, ICAM-1, MCP-1). These molecular changes promote smooth muscle cell (SMC) growth, inflammation, thrombosis, vascular remodeling, apoptosis, and impaired vascular tone, culminating in endothelial dysfunction and contributing to the development of diseases associated with endothelial dysfunction in diabetes mellitus. NADPH, Nicotinamide adenine dinucleotide phosphate; eNOS, endothelial nitric oxide synthase; ROS, reactive oxygen species; LDL, low-density lipoprotein; MAPK, mitogen-activated protein kinase; PI3K, phosphoinositide 3-kinase; Ras, rat sarcoma virus; Akt, protein kinase B; NFκB, nuclear factor kappa B; AP-1, activator protein 1; VCAM-1, vascular cell adhesion molecule 1; ICAM-1, intercellular adhesion molecule 1; MCP-1, monocyte chemoattractant protein 1; SMC, smooth muscle cell. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
Medsci 13 00087 g002
Figure 3. Pathophysiological mechanisms of oxidative stress-induced atherosclerosis. Oxidative stress leads to the oxidation of lipids, indicated by increased ROS and oxidized OxLDL, and the oxidation of nucleic acids, reflected by hydroxyl radicals, mitochondrial DNA damage, and oxidative DNA damage markers such as 8-oxo-dG. Endothelial dysfunction occurs due to decreased NO bioavailability, reduced endothelial repair capacity, and increased ROS from immune cells and mitochondria, leading to enhanced OxLDL uptake. Additionally, oxidative stress activates inflammation via NFκB and AP-1 transcription factors, increasing pro-inflammatory cytokines (IL-6, TNF, IL-1β), ROS from immune cells, and recruitment of neutrophils and monocytes, while influencing anti-inflammatory mechanisms (e.g., IL-10, Treg activity). Finally, oxidative stress destabilizes the fibrous cap through increased matrix metalloproteinases (MMP-2, MMP-9), ROS-induced smooth muscle cell apoptosis, and inflammation at the plaque shoulder, leading to reduced fibrous cap stability and collagen synthesis. Collectively, these processes contribute to the development and progression of atherosclerosis in the context of diabetes mellitus and oxidative stress. ROS, reactive oxygen species; •OH, hydroxyl radical; O2, superoxide anion (or superoxide radical); ONOO, Peroxynitrite anion; OxLDL, oxidized low-density lipoprotein; Vit E, vitamin E; SOD, Superoxide Dismutase; 8-oxo-dG, 8-oxo-2′-deoxyguanosine; NO, nitric oxide; eNOS, endothelial nitric oxide synthase; NFκB, nuclear factor kappa B; AP-1, activator protein 1; IL-6, interleukin 6; TNF, tumor necrosis factor; IL-1β, interleukin 1 beta; IL-10, interleukin 10; Treg, regulatory T cell; MMP-2, matrix metalloproteinases 2; MMP-9, matrix metalloproteinases 9; SMC, smooth muscle cell. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
Figure 3. Pathophysiological mechanisms of oxidative stress-induced atherosclerosis. Oxidative stress leads to the oxidation of lipids, indicated by increased ROS and oxidized OxLDL, and the oxidation of nucleic acids, reflected by hydroxyl radicals, mitochondrial DNA damage, and oxidative DNA damage markers such as 8-oxo-dG. Endothelial dysfunction occurs due to decreased NO bioavailability, reduced endothelial repair capacity, and increased ROS from immune cells and mitochondria, leading to enhanced OxLDL uptake. Additionally, oxidative stress activates inflammation via NFκB and AP-1 transcription factors, increasing pro-inflammatory cytokines (IL-6, TNF, IL-1β), ROS from immune cells, and recruitment of neutrophils and monocytes, while influencing anti-inflammatory mechanisms (e.g., IL-10, Treg activity). Finally, oxidative stress destabilizes the fibrous cap through increased matrix metalloproteinases (MMP-2, MMP-9), ROS-induced smooth muscle cell apoptosis, and inflammation at the plaque shoulder, leading to reduced fibrous cap stability and collagen synthesis. Collectively, these processes contribute to the development and progression of atherosclerosis in the context of diabetes mellitus and oxidative stress. ROS, reactive oxygen species; •OH, hydroxyl radical; O2, superoxide anion (or superoxide radical); ONOO, Peroxynitrite anion; OxLDL, oxidized low-density lipoprotein; Vit E, vitamin E; SOD, Superoxide Dismutase; 8-oxo-dG, 8-oxo-2′-deoxyguanosine; NO, nitric oxide; eNOS, endothelial nitric oxide synthase; NFκB, nuclear factor kappa B; AP-1, activator protein 1; IL-6, interleukin 6; TNF, tumor necrosis factor; IL-1β, interleukin 1 beta; IL-10, interleukin 10; Treg, regulatory T cell; MMP-2, matrix metalloproteinases 2; MMP-9, matrix metalloproteinases 9; SMC, smooth muscle cell. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
Medsci 13 00087 g003
Figure 4. Pathophysiological mechanisms of oxidative stress-induced DR. Chronic hyperglycemia in diabetes induces oxidative stress, which leads to mitochondrial dysfunction characterized by increased ROS production, mitochondrial DNA damage, altered fission/fusion dynamics (e.g., Drp1), loss of membrane potential, impaired ATP production, and reduced mitochondrial biogenesis and PGC-1α activity. Oxidative stress also promotes cellular apoptosis in retinal cells through upregulation of pro-apoptotic markers such as Bax, caspases 3 and 9, cytochrome c release, p53 activation, and DNA fragmentation, while downregulating anti-apoptotic proteins like Bcl-2 and survival pathways such as PI3K/AKT. Concurrently, inflammation is amplified via elevated TNF-α, IL-1β, IL-6, ICAM-1, MCP-1, and NF-κB activation, along with altered expression of tight junction and anti-inflammatory proteins. Oxidative stress also accelerates lipid peroxidation, evidenced by increased malondialdehyde, 4-hydroxynonenal, and oxidized LDL, and reduced antioxidant defenses including glutathione and enzymes like SOD and catalase. These cumulative changes lead to structural and functional alterations in the retinal microvasculature, including increased VEGF expression, vascular permeability, basement membrane thickening, pericyte loss, compromised tight junctions (e.g., occludin, claudin-5), reduced pericyte density, and impaired endothelial integrity. Collectively, these molecular and cellular disturbances culminate in the onset and progression of diabetic retinopathy. ROS, reactive oxygen species; Drp1, dynamin-related protein 1; ATP, adenosine triphosphate; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; Bax, Bcl-2–associated X; DNA, deoxyribonucleic acid; Bcl-2, B-cell lymphoma 2; PI3K, Phosphoinositide 3-kinases; Akt, protein kinase B; TNF, tumor necrosis factor; IL-1β, Interleukin-1 beta; IL-6, interleukin 6; NFκB, nuclear factor kappa B; ICAM-1, intercellular adhesion molecule 1; MCP-1, monocyte chemoattractant protein-1; IL-10, interleukin 10; LDL, low-density lipoprotein; SOD, Superoxide dismutase; VEGF, vascular endothelial growth factor. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
Figure 4. Pathophysiological mechanisms of oxidative stress-induced DR. Chronic hyperglycemia in diabetes induces oxidative stress, which leads to mitochondrial dysfunction characterized by increased ROS production, mitochondrial DNA damage, altered fission/fusion dynamics (e.g., Drp1), loss of membrane potential, impaired ATP production, and reduced mitochondrial biogenesis and PGC-1α activity. Oxidative stress also promotes cellular apoptosis in retinal cells through upregulation of pro-apoptotic markers such as Bax, caspases 3 and 9, cytochrome c release, p53 activation, and DNA fragmentation, while downregulating anti-apoptotic proteins like Bcl-2 and survival pathways such as PI3K/AKT. Concurrently, inflammation is amplified via elevated TNF-α, IL-1β, IL-6, ICAM-1, MCP-1, and NF-κB activation, along with altered expression of tight junction and anti-inflammatory proteins. Oxidative stress also accelerates lipid peroxidation, evidenced by increased malondialdehyde, 4-hydroxynonenal, and oxidized LDL, and reduced antioxidant defenses including glutathione and enzymes like SOD and catalase. These cumulative changes lead to structural and functional alterations in the retinal microvasculature, including increased VEGF expression, vascular permeability, basement membrane thickening, pericyte loss, compromised tight junctions (e.g., occludin, claudin-5), reduced pericyte density, and impaired endothelial integrity. Collectively, these molecular and cellular disturbances culminate in the onset and progression of diabetic retinopathy. ROS, reactive oxygen species; Drp1, dynamin-related protein 1; ATP, adenosine triphosphate; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; Bax, Bcl-2–associated X; DNA, deoxyribonucleic acid; Bcl-2, B-cell lymphoma 2; PI3K, Phosphoinositide 3-kinases; Akt, protein kinase B; TNF, tumor necrosis factor; IL-1β, Interleukin-1 beta; IL-6, interleukin 6; NFκB, nuclear factor kappa B; ICAM-1, intercellular adhesion molecule 1; MCP-1, monocyte chemoattractant protein-1; IL-10, interleukin 10; LDL, low-density lipoprotein; SOD, Superoxide dismutase; VEGF, vascular endothelial growth factor. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
Medsci 13 00087 g004
Figure 5. Pathophysiological mechanisms of oxidative stress-induced DKD. Hyperglycemia activates multiple metabolic pathways, including the polyol, AGE/RAGE, hexosamine, and PKC pathways—which collectively elevate ROS levels. ROS function as central mediators that disrupt key intracellular signaling cascades such as PI3K/AKT, NF-κB, Nrf2/ARE, JAK/STAT3, TGF-β1/MAPK, and AMPK pathways. These disruptions result in downstream effects including impaired antioxidant defense (via inhibition of Nrf2), enhanced inflammation (through NF-κB and JAK/STAT3), fibrotic signaling (via TGF-β1 and MAPK components p38, JNK, and ERK), and inhibition of autophagy (through suppressed AMPK and altered mTOR signaling). Collectively, these processes contribute to DNA damage, chronic inflammation, extracellular matrix deposition, apoptosis, and cellular dysfunction in renal tissue, ultimately culminating in the development and progression of diabetic kidney disease. AGE, advanced glycation end-products; RAGE, Receptor for AGE; PKC, protein kinase C; ROS, reactive oxygen species; PI3K, phosphoinositide 3-kinase; AKT, protein kinase B; AMPK, AMP-activated protein kinase; NFκB, nuclear factor kappa B; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; TGF-β1, transforming growth factor-beta 1; Nrf2, nuclear factor erythroid 2–related factor 2; ARE, antioxidant response element; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol (3,4,5)-trisphosphate.
Figure 5. Pathophysiological mechanisms of oxidative stress-induced DKD. Hyperglycemia activates multiple metabolic pathways, including the polyol, AGE/RAGE, hexosamine, and PKC pathways—which collectively elevate ROS levels. ROS function as central mediators that disrupt key intracellular signaling cascades such as PI3K/AKT, NF-κB, Nrf2/ARE, JAK/STAT3, TGF-β1/MAPK, and AMPK pathways. These disruptions result in downstream effects including impaired antioxidant defense (via inhibition of Nrf2), enhanced inflammation (through NF-κB and JAK/STAT3), fibrotic signaling (via TGF-β1 and MAPK components p38, JNK, and ERK), and inhibition of autophagy (through suppressed AMPK and altered mTOR signaling). Collectively, these processes contribute to DNA damage, chronic inflammation, extracellular matrix deposition, apoptosis, and cellular dysfunction in renal tissue, ultimately culminating in the development and progression of diabetic kidney disease. AGE, advanced glycation end-products; RAGE, Receptor for AGE; PKC, protein kinase C; ROS, reactive oxygen species; PI3K, phosphoinositide 3-kinase; AKT, protein kinase B; AMPK, AMP-activated protein kinase; NFκB, nuclear factor kappa B; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; TGF-β1, transforming growth factor-beta 1; Nrf2, nuclear factor erythroid 2–related factor 2; ARE, antioxidant response element; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol (3,4,5)-trisphosphate.
Medsci 13 00087 g005
Table 1. List of factors that are altered in diabetes-induced oxidative stress.
Table 1. List of factors that are altered in diabetes-induced oxidative stress.
PathwaysAltered Factors in Oxidative StressReferences
Polyol pathway
-
↑ Aldose reductase activity (glucose → sorbitol)
-
↓ NADPH, impairing antioxidant defense
-
↑ NADH, promoting ROS
-
↑ Osmotic and oxidative stress from sorbitol accumulation
[165,166]
AGE/RAGE pathway
-
↑ Formation of AGEs
-
↑ RAGE expression and activation
-
↑ Inflammatory cytokines (TNF, IL-6)
-
↑ ROS generation
-
↓ Glyoxalase detoxification
[167,168,169]
PKC pathway
-
↑ DAG production
-
↑ PKC isoform activation (PKC-β, PKC-δ)
-
PKC → NOX activation → ↑ ROS
-
Endothelial dysfunction, inflammation
[170,171,172]
Hexosamine pathway
-
↑ GFAT activity
-
↑ UDP-GlcNAc production
-
↑ Protein O-GlcNAcylation
-
Dysregulation of OGT/OGA balance
-
↑ ER stress, mitochondrial dysfunction
[173,174,175]
Abbreviations: AGE, advanced glycation end product; RAGE, receptor for AGE; ROS, reactive oxygen species; TNF, tumor necrosis factor; IL-6, interleukin-6; DAG, diacylglycerol; PKC, protein kinase C; NADPH, nicotinamide adenine dinucleotide phosphate; NADH, nicotinamide adenine dinucleotide; GFAT, glutamine:fructose-6-phosphate amidotransferase; UDP-GlcNAc, uridine uiphosphate N-acetylglucosamine; O-GlcNAcylation, O-linked β-N-acetylglucosaminylation; OGT, O-GlcNAc transferase; OGA, O-GlcNAcase; ER, endoplasmic reticulum. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
Table 2. Antioxidant therapy failure in RCTs.
Table 2. Antioxidant therapy failure in RCTs.
CompoundBiomarker EffectsClinical OutcomesReferences
CoQ10↑ FMD (+1.45%, p < 0.02) in meta-analysis of 12 RCTsNo reduction in cardiovascular events[491]
MitoQ↑ Brachial artery FMD by 42%,
↓ Ox-LDL and aortic stiffness in older adults
No clinical outcomes assessed[489]
Omega-3↓ BP (modest),
↓ triglycerides; favorable lipid profile
No consistent benefit for mortality or CV events[492,493]
MelatoninNo improvement in FMD or oxidative stress markers in high-salt intake studyNo vascular benefit observed[487]
NAC↓ MDA in hemodialysis patients (600 mg BID)No BP reduction; no hard CV outcomes assessed[488]
ResveratrolNo significant changes in BP or CV events in hypertensive patients across RCTsInconsistent or no benefit for CV endpoints[494]
Abbreviations: CoQ10, coenzyme Q10; FMD, Flow-mediated dilation; RCTs, randomized clinical trials; MitoQ, mitoquinone mesylate; BP, blood pressure; NAC, N-Acetylcysteine; MDA, Malondialdehyde; CV, cardiovascular. The upward arrow indicates an increase, and the downward arrow indicates a decrease.
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

Roy, B. Pathophysiological Mechanisms of Diabetes-Induced Macrovascular and Microvascular Complications: The Role of Oxidative Stress. Med. Sci. 2025, 13, 87. https://doi.org/10.3390/medsci13030087

AMA Style

Roy B. Pathophysiological Mechanisms of Diabetes-Induced Macrovascular and Microvascular Complications: The Role of Oxidative Stress. Medical Sciences. 2025; 13(3):87. https://doi.org/10.3390/medsci13030087

Chicago/Turabian Style

Roy, Bipradas. 2025. "Pathophysiological Mechanisms of Diabetes-Induced Macrovascular and Microvascular Complications: The Role of Oxidative Stress" Medical Sciences 13, no. 3: 87. https://doi.org/10.3390/medsci13030087

APA Style

Roy, B. (2025). Pathophysiological Mechanisms of Diabetes-Induced Macrovascular and Microvascular Complications: The Role of Oxidative Stress. Medical Sciences, 13(3), 87. https://doi.org/10.3390/medsci13030087

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