Next Article in Journal
Brain Endothelial Cells in Contrary to the Aortic Do Not Transport but Degrade Low-Density Lipoproteins via Both LDLR and ALK1
Next Article in Special Issue
Liraglutide Improves the Angiogenic Capability of EPC and Promotes Ischemic Angiogenesis in Mice under Diabetic Conditions through an Nrf2-Dependent Mechanism
Previous Article in Journal
Glucose 6-P Dehydrogenase—An Antioxidant Enzyme with Regulatory Functions in Skeletal Muscle during Exercise
Previous Article in Special Issue
Targeting on Nrf2/Sesn2 Signaling to Rescue Cardiac Dysfunction during High-Fat Diet-Induced Obesity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nrf2-Mediated Dichotomy in the Vascular System: Mechanistic and Therapeutic Perspective

1
School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
2
Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
3
Columbia VA Health System, Wm. Jennings Bryan Dorn VA Medical Center, Columbia, SC 29209, USA
*
Author to whom correspondence should be addressed.
Cells 2022, 11(19), 3042; https://doi.org/10.3390/cells11193042
Submission received: 2 August 2022 / Revised: 25 September 2022 / Accepted: 27 September 2022 / Published: 28 September 2022
(This article belongs to the Special Issue Nrf2 Signaling Pathway in Cardiovascular Health and Disease)

Abstract

:
Nuclear factor-erythroid 2-related factor 2 (Nrf2), a transcription factor, controls the expression of more than 1000 genes that can be clustered into different categories with distinct functions ranging from redox balance and metabolism to protein quality control in the cell. The biological consequence of Nrf2 activation can be either protective or detrimental in a context-dependent manner. In the cardiovascular system, most studies have focused on the protective properties of Nrf2, mainly as a key transcription factor of antioxidant defense. However, emerging evidence revealed an unexpected role of Nrf2 in mediating cardiovascular maladaptive remodeling and dysfunction in certain disease settings. Herein we review the role of Nrf2 in cardiovascular diseases with a focus on vascular disease. We discuss the negative effect of Nrf2 on the vasculature as well as the potential underlying mechanisms. We also discuss the clinical relevance of targeting Nrf2 pathways for the treatment of cardiovascular and other diseases.

1. Introduction

According to the statistical data of World Health Organization, cardiovascular disease (CVD) is the underlying cause of death for approximately 17.9 million global deaths each year. CVDs, such as hypertension, atherosclerosis, aneurysm, myocardial hypertrophy, and coronary heart disease, among others are the leading cause of death globally. Despite the advance in cardiovascular research, it is still far from a comprehensive understanding of CVD. Effective therapies for CVD are limited.
The causes of heart and vascular diseases may vary; however, most share an important pathological mechanism: oxidative stress [1,2,3,4], which is resulted from an imbalance between the production and detoxification of reactive oxygen species (ROS) [5]. Uncontrolled ROS cause arrhythmias and induce cardiac hypertrophy, apoptosis, and necrosis to promote cardiac pathological remodeling and dysfunction leading to heart failure [6,7]. Similarly, oxidative stress causes vascular damage and disease [8]. It is worthy to note that under physiological conditions, ROS also act as signaling molecules to regulate a variety of cellular functions in the cardiovascular system, such as endothelial cell (EC) and vascular smooth muscle cell (VSMC) proliferation, migration, and apoptosis, as well as angiogenesis, vascular tone, host defense, and genomic stability [9]. Anti-oxidative stress appears to be a therapeutic approach for the prevention and treatment of cardiovascular diseases; however, how to precisely eliminate oxidative stress without affecting physiological redox balance remains a challenge [1].
The nuclear factor-erythroid 2-related factor 2 (Nrf2), a multifunctional transcription factor, controls the basal and induced expression of more than 1000 genes that can be divided into several groups with different functions ranging from antioxidant defense and detoxification to metabolism and protein quality control [10]. Previously, Nrf2 has been identified as a principal transcription factor for antioxidant defense, thereby providing cardiovascular protection. However, Nrf2 can also act as a mediator of cardiovascular maladaptive remodeling and dysfunction, particularly in chronic conditions, such as sustained pressure overload and diabetes [11,12,13]. We have recently outlined and discussed the emerging dark side of Nrf2 in the heart and the potential mechanisms of Nrf2-mediated myocardial damage and dysfunction [14]. On the other hand, whilst most of the reported functions of Nrf2 in the vasculature appear to be positive, a handful of evidence reveal an adverse role of Nrf2 in promoting vascular disease. Nevertheless, the enthusiasm for developing Nrf2 activators to treat oxidative stress-associated diseases including CVD is still high. Therefore, we describe the Nrf2 signaling pathway and sort out the bidirectional role of Nrf2 signaling pathway in CVD, especially in vascular diseases. As a result, the outcome underscores the ‘dark’ side of Nrf2 while unveiling clues for investigating the nature of the Nrf2-mediated dichotomy in the cardiovascular system, thereby providing reliable and specific targets for clinical treatment.

2. Nrf2 Signaling

2.1. Basic Functions

Nrf2 was originally isolated as a homolog of the hematopoietic transcription factor NF-E2 (nuclear factor-erythroid factor 2) p45, as such Nrf2 was initially known as NF-E2-related factor 2 [15].
Both Nrf2 and NF-E2 belong to the Cap “n” Collar (CNC) family which contains basic a leucine zipper (bZip) structure. To date, six members of the family have been identified, namely: NF-E2, Nrf1-3, Bach 1 [broad-complex, tram-track, bric-a-brac (BTB), CNC homolog 1], and Bach 2.
The differences between these CNC family members have been extensively reviewed [14,16,17]. In general, NF-E2 and Nrf1-3 usually act as transcriptional activators, while Bach 1 and 2 function as transcriptional repressors. The differentiation of committed erythroid progenitor cells involves the transcription factor NF-E2 [18]. Megakaryocyte development and maturation are also governed by the activity of NF-E2 [19,20]. Nrf1-3 proteins are widely expressed in the body. Nrf1 helps regulate the basal expression levels of certain cytoprotective enzyme genes but does not affect their inducible expression [21]. Whilst Nrf3 plays a minor role in the regulation of Phase II enzyme genes [22,23], it has a crucial role in SMC differentiation from stem cells [24,25]. Nrf2 is widely expressed in oxygen-consuming organs, such as skeletal muscle, heart, blood vessels, liver, kidney, brain, lung, skin, and digestive tract [26]. Nrf2 orchestrates the basal and induced expression of genes by combining to a cis-acting enhancer with a core nucleotide sequence of 5′-RTGACNNNGC-3′23, which is referred to as the antioxidant response element (ARE). To date, over 1000 Nrf2 target genes have been identified. These genes can be clustered into several functional groups including antioxidant, detoxification, metabolism, transcription factors, proteasomal and autophagic degradation, cell proliferation, and cell survival [27,28,29]. Thus, this allows Nrf2 to exert not only antioxidant defense but also other multifunctional activities.

2.2. Protein Structural Domain of Nrf2

Nrf2 is the most potent member of the CNC transcription factor family. It contains 605 amino acids and is divided into 7 highly conserved erythroid-derived CNC homology (ECH) protein domains, ranging from Neh1 to Neh7, based on their functions [27,29,30]. From the N-terminal to the C-terminal, the Nrf2 protein comprises Neh2, Neh4-Neh5, Neh7, Neh6, Neh1, and Neh3 (Figure 1A). Neh2 contains a low-affinity DLG-binding motif and a high-affinity ETGE-binding motif [31], which bind to the double glycine repeat (DGR) domain of Kelch-like ECH associating protein 1 (Keap1) (Figure 1B), a major negative regulator of Nrf2 [32]. Neh2 acts as an intermediate in the formation of heterodimer Nrf2 and Keap1. Keap1 mediates the ubiquitination and degradation of Nrf2 and is a natural inhibitor of Nrf2 in the cytoplasm. Neh4 and Neh5 are independent of each other and jointly regulate the trans-activation of cytoprotective genes. Neh4 and Neh5 both independently and collaboratively bind to CBP [CREB (cAMP Responsive Element Binding protein) Binding Protein], thus attaining maximum activation of antioxidant gene expression [33]. Of note, abnormal activation of Nrf2 in cancer cells can confer cytoprotection to them, thereby reducing the therapeutic effect of chemotherapy and radiotherapy. The increased cytoprotection is resulted from the increased interaction between Nrf2 and CBP, that is mediated by the Neh4 and Neh5 domains of Nrf2. Disruption of this interplay through small-molecule therapeutics negate the aberrant activation of Nrf2 in cancer cells [34]. The Neh4 and Neh5 domains of Nrf2 also directly interact with the C/H3-containing C terminus of p300. Acetylation of Nrf2 by p300 during the antioxidant reaction enhances promoter-specific DNA binding of Nrf2 [35]. Nrf2 activation can be modulated and directly controlled through interactions between the Neh4 and Neh5 domains and the RAC3 protein [36]. The Neh7 domain is a newly discovered domain located between the Neh5 and Neh6 domains. The interplay between the Neh7 domain and 9-cis retinoic acid (RA) can suppress Nrf2 activity [37,38]. Like the Neh2 domain, the Neh6 domain contains two important motifs, the DSGIS and DSAPGS motifs. Unlike the Neh2 domain, these two motifs of the Neh6 domain mediate the keap1 independent degradation of Nrf2 in stressed cells. These two important motifs of Neh6 recruit the dimeric β–transducin repeat-containing protein (β-TrCP) [39,40]. Glycogen synthase kinase-3 (GSK-3) phosphorylates a particular serine residue in the Neh6 structural domain of Nrf2, which forms a degradable structural domain. This degradable structural domain is then recognized by the ubiquitin ligase adaptor β-TrCP for Nrf2 ubiquitination and subsequent proteasomal degradation by the Cullin1/ring-box 1 (RBX1) complex [41,42]. The Neh1 structural domain forms a heterodimer with the small muscle neurofibrosarcoma (sMaf) protein through the CNC-bZIP region and binds to AREs on target genes to regulate transcription [43,44]. It contains the functional nuclear localization signal (NLS) and nuclear export signal (NES) sequences [45,46]. The C-terminal Neh3 domain of Nrf2 is also important for its activity. It may function as a transcriptional activation structural domain and may be engaged in the interaction of transcriptional apparatus components that affect its transcriptional activity [47].

2.3. Nrf2 Transcription-Related

The aryl hydrocarbon receptor (AHR) directly binds to the xenobiotic response element (XRE)-like element located at the Nrf2 promoter and modulates Nrf2 gene transcription [48,49]. Notch signaling directly triggers the Nrf2 stress adaptation response pathway by recruiting the Notch intracellular structural domain (NICD) transcriptome to the conserved Rbpjκ site of the Nrf2 promoter [50]. TGFβ1-induced epithelial-mesenchymal transition occurs through the transcription of Notch4 via Nrf2-dependent promoter activation [51]. In addition, hypermethylation or single nucleotide polymorphism (SNP) in the Nrf2 promoter region results in reduced Nrf2 expression [52].

2.4. Ubiquitinated Degradation of Nrf2

The intracellular half-life of Nrf2 is less than 20 min. However, Keap1 plays a major role in the regulation of protein stability and transcriptional activity of Nrf2 [16,53]. Alternatively, GSK-3 and β-TrCP-mediated proteasomal degradation of Nrf2 is not associated with Keap1. A recent review provides a comprehensive discussion of these issues [14]. Thus, a concise summary is made as follows. The Keap1-Nrf2 pathway: Keap1 contains five domains including NTR (the N-terminal region), BTB, IVR (the intervening region), DGR or Kelch, and CTR (the C-terminal region) (Figure 1B). BTB and Kelch are two major domains. BTB is responsible for the interaction of the Keap1 homodimer with the Cul3-Rbx1-E3 ligase complex. Meanwhile, Kelch is combined with the DLG and ETGE motifs in Neh2 of Nrf2. Nrf2 binds to one of the Keap1 homodimers via ETGE motif and DLG motif, forming firstly an “open” conformation and then a “closed” conformation, with subsequent ubiquitination and degradation of Nrf2 by Clu3-Rbx1-E3 ligase and subsequent release of Keap1. Under normal conditions, Keap1 continuously targets Nrf2 for degradation (Figure 1C). However, under stressful conditions, some oxidizing or electrophilic molecules react with the cysteine residues of Keap1, leading to conformational changes in Keap1, thereby reducing Keap1-mediated ubiquitination and degradation of Nrf2 (Figure 1D). The GSK-3β-Nrf2 pathway: Under non-stressed conditions, AKT phosphorylates the N-terminal pseudo-substrate structural domain of GSK-3, thereby leaving GSK-3 in an inactive state. However, once AKT is inhibited or inactivated, GSK-3 is activated and then phosphorylates the Neh6 of Nrf2, which in turn recruits β-TrCP for β-TrCP-mediated the proteasomal degradation of the Nrf2. Typically, the Keap1-Nrf2 pathway happens in the cytosol, whereas the GSK-3β-Nrf2 pathway occurs in the nucleus. The specific mechanisms and relative importance of Keap1 and GSK-3-TrCP in regulating Nrf2 degradation remain poorly understood.

3. Nrf2 and Vascular Physiology and Pathology

3.1. Nrf2 and Endothelial Cells

ECs are one of the major cell types in the vascular wall. ECs sprouting from existing vessels forms new blood vessels, named angiogenesis, which is a critical event in embryonic development and multiple disease processes. Global and EC-specific knockout of Nrf2 in mice demonstrates a critical and cell-autonomous role of Nrf2 in ECs for promoting angiogenesis [54]. As ECs are constantly exposed to hemodynamic forces such as shear stress—the frictional force from the blood flow acting on the surface of the vascular lumen [55,56], they sense the shear stress to govern both short-term vascular tone and long-term vascular remodeling to adjust vessel diameters to tissue demand. In mechanobiology, disturbed flow and unidirectional laminar flow are two important types of shear stress that exert differential effects on the functions of ECs through mechanosensitive transcription factor-dependent gene expression [57]. Nrf2 is an important mechanosensitive transcription factor that is upregulated after exposure to unidirectional laminar flow [57,58,59,60]. In the sections below, we will review the specific role of Nrf2 in endothelial function.
In ECs, the laminar flow may use specific mechanoreceptor(s) to activate the Nrf2/ARE pathway. Nrf2 is activated by laminar shear stress through the PI3K-AKT signaling pathway [61,62]. However, when the shear stress on the vessel wall is disturbed, the oscillatory flow leads to reduced NO production and increased superoxide release [63]. This reduces Nrf2-mediated activation of ARE-linked cytoprotective genes [64]. In contrast, Nrf2 deletion or knockdown suppresses laminar shear stress-induced cytoprotective effects [58]. In addition to the known role of Nrf2 as a mechanosensitive transcription factor and oxidative stress inhibitor, Nrf2 is also involved in the inflammatory response of ECs. Nrf2 prevents ECs at atherosclerosis-protected sites from exhibiting a proinflammatory state by inhibiting the p38-VCAM-1 signaling pathway [65]. Protein kinase C (PKC)1 activates the Nrf2 signaling pathway and induces heme oxygenase (HO)-1 in the vascular endothelium to enhance resistance to inflammation to maintain vascular homeostasis [66]. Curcumin treatment of cultured human arterial ECs also induces HO-1 expression through activation of Nrf2 and inhibits acute vascular inflammation [67]. Angiotensin II (Ang II) is closely involved in endothelial dysfunction by induction of apoptosis and oxidative stress [68], and the induced endothelial dysfunction is a high-risk factor for CVD [69]. Sirtuin (SIRT) 6 protects vascular ECs from Ang II-induced apoptosis and oxidative stress via activating Nrf2/ARE signaling [70]. The SIRT6/Nrf2/ARE signaling pathway is a key regulator of redox homeostasis in vascular ECs. Ang II decreases antioxidant potential and increases oxidative stress and inflammatory responses, thereby causing damage to human umbilical cord vascular endothelial cells (HUVECs) by blunting the Nrf2/ERK1/2/Nox2 (NADPH oxidase 2) system.
Nrf2 has been extensively studied in cigarette smoke-induced emphysema and chronic obstructive pulmonary disease (COPD). Compared with wild-type mice, Nrf2-deficient mice had earlier-onset and more extensive cigarette smoke-induced emphysema associated with more pronounced oxidative stress [71,72]. Whilst Nrf2-mediated antioxidant defense has been well demonstrated in lung epithelial cells [72,73], the activation of a mitogen-activated protein kinase (MPAK)/Nrf2/HO-1 pathway was proposed to underlie ginkgo biloba extract-induced protection against cigarette smoke-induced oxidative stress and cell death in human pulmonary artery ECs [74]. These studies indicate that Nrf2-operated antioxidant defense is a common mechanism against smoking-induced damage in various types of lung cells including ECs. In addition, such a notion was extrapolated into smoking-induced atherosclerosis and cerebrovascular injury [75,76]. Knockdown of Nrf2 markedly enhanced cigarette smoke-induced ROS production and NLRP3 (NLR family pyrin domain containing 3) inflammasome activation in human aortic ECs [75]. Metformin could activate Nrf2 pathway and protect against cigarette smoke-induced damage to brain ECs and loss of blood–brain barrier [76]. Given that Nrf2 knockout increased susceptibility to brain edema and blood–brain barrier breakdown in a mouse model of subarachnoid hemorrhage [77], it is likely that Nrf2 is a critical mediator of metformin-induced brain protection. Taken together, these findings underscore a beneficial effect of Nrf2 activation in ECs.
Recently, several studies have reported that natural compounds can inhibit Ang II-induced ECs injury through the Nrf2 signaling pathway. Briefly, celastrol, known as tretinoin, is a functional ingredient of Trypterygiun wilfordii Hook F that has powerful antioxidant and anti-inflammatory properties. Celastrol effectively attenuates Ang II-mediated ECs injury by activating the Nrf2/ERK1/2/Nox2 pathway [78]. Epigallocatechin-3-gallate (EGCG) is the main chemical compound of green tea, which has anti-inflammatory, antioxidant, and anti-angiogenic effects. EGCG ameliorates Ang II-induced oxidative stress and apoptosis in HUVECs through activation of the Nrf2/Caspase-3 signaling pathway [79]. Schisandrin C (SchC) is a dibenzocyclooctadiene derivative of Schisandra Chinensis with antioxidant properties. SchC targets Keap1 to attenuate oxidative stress by activating the Nrf2 pathway in the Ang II-stimulated vascular endothelium [80]. Osthole is the main active component of the herbal fruit with anti-inflammatory and antioxidant activities. Osthole significantly attenuates Ang II-induced apoptosis in rat aorta endothelial cells (RAECs) by reducing inflammation and oxidative stress by targeting the NF-κB pathway and Keap-1/Nrf2 pathway [81]. In addition, tilapia by-product oligopeptides protect against Ang II-induced endothelial injury in HUVECs in vitro via the activation Nrf2/NF-κB signal pathway [82]. Synthetic drugs have also been demonstrated to activate the protective function of Nrf2 in vascular ECs. Memantine promotes the activation of Nrf2/HO-1 antioxidant signaling pathway and protects cardiac ECs from dysregulation in acute myocardial infarction [83]. Metformin also increases the expression level of Nrf2 and the nuclear accumulation level of Nrf2 in hyperglycemic HUVECs. In addition, Metformin can downregulate p65 to upregulate Nrf2 as a way to protect ECs function associated with gestational diabetes mellitus (GDM) [84]. Other drugs that target Nrf2 and protect ECs are detailed in Table 1.
Although less-studied, a handful of evidence has revealed the abnormalities associated with Nrf2 activation in ECs. An early showed that prolonged infection of ECs with Kaposi’s sarcoma-associated herpesvirus (KSHV) induces Nrf2 activation, thereby promoting Kaposi’s sarcoma pathogenesis [85]. A recent study further documented that cigarette smoke-induced oxidative stress activates an Nrf2/STAT3 pathway to interrupt fibronectin assembly and angiogenesis in human umbilical vein ECs and zebrafish [86]. Unlike the Nrf2-mediated beneficial effects in ECs which are validated by Nrf2 EC-specific knockout approach [54], the ‘dark’ side of Nrf2 remains to be fully established by genetic interrogation in vivo. Further investigation of Nrf2-mediated dichotomy in ECs is warranted, therefore providing novel insight into the therapeutic implications of targeting Nrf2 signaling.
Table 1. Therapeutic targeting Nrf2 signaling for EC protection.
Table 1. Therapeutic targeting Nrf2 signaling for EC protection.
ApproachNrf2 SignalingFindingReference
CompoundsAtherosclerosis (AS) Related
MelatoninNrf2/NLRP3Reduces cigarette smoke extract (CSE) treatment-induced pyroptosis in human aortic endothelial cells (HAECs) in vitro and cigarette smoke exposure-enhanced intimal hyperplasia in rat carotid arteries induced by balloon injury in vivo[75]
AstragalosideNrf2/HO-1Prevents oxidized low-density lipoprotein (oxLDL)-induced human umbilical vein endothelial cells (HUVECs) injury in vitro[87]
IsoflavoneNrf2/AERReduces oxLDL-induced oxidative stress damage in EA.hy926 cells in vitro and atherosclerosis in apolipoprotein E deficiency (ApoE−/−) mice fed with a high-fat diet in vivo[88]
Flavonoids from a Deep-Sea-Derived Arthrinium sp.AKT/Nrf2/HO-1Protects HUVECs against ox-LDL-induced oxidative stress in vitro[89]
Tanshinone IIANrf2Suppresses human coronary artery endothelial cells (HCAECs) ferroptosis in vitro[90]
Chalcone derivativeNrf2/HO-1Inhibits cholesterol efflux and suppresses inflammatory responses in HUVECs in vitro
Reduces lipid accumulation and plaque formation in LDL receptor knockout (Ldlr−/−) mice fed with a high-fat diet
[91]
Ilexgenin ANrf2/PSMB5Suppresses mitochondrial fission, and improves endothelial dysfunction induced by palmitate (PA) in vitro[92]
AcacetinNrf2Exerts antioxidant potential in ApoE−/− mice and in EA.hy926 cells induced by human oxLDL in vitro[93]
IrisinAKT/mTOR/S6K1/Nrf2Attenuates oxLDL-impaired angiogenesis of human microvascular ECs in vitro and in a chicken embryo membrane (CAM) model in vivo[94]
A peptide from microalgae Isochrysis zhanjiangensisNrf2Inhibits oxLDL-induced inflammation and apoptosis of HUVECs in vitro[95]
KaempferolPI3K/AKT/Nrf2Attenuates oxLDL-induced injury via activating G protein-coupled estrogen receptor (GPER) associated with upregulation of PI3K/AKT/Nrf2 signaling in HAECs in vitro
Suppresses atherosclerotic lesion formation in ovariectomized ApoE−/− mice fed with a high-fat diet in vivo
[96]
XanthoangelolNrf2/AREPrevents oxLDL-induced HUVECs injury in vitro[97]
EquolNrf2/t-BHP/CHOPAttenuates atherosclerosis in ApoE−/− mice fed with a high-fat diet in vivo and endoplasmic reticulum stress and apoptosis in HUVECs induced by tert-butyl hydroperoxide (t-BHP) and thapsigargin in vitro[98]
KirenolPI3K/AKT/Nrf2Prevents B[a]P-induced redox imbalance in HUVECs in vitro[99]
ResveratrolNrf2/ICAM-1Suppressive effects on pro-inflammatory responses in ECs and accelerated atherosclerosis in carotid arteries induced by ApoE−/− mice[100]
ZedoarondiolNrf2/HO-1Attenuates oxLDL-induced injury, oxidative stress, and inflammatory responses in HUVECs in vitro[101]
Dietary ellagic acidNrf2/HO-1Protective effects on damage in HAECs induced by hypochlorous acid (HOCl) in vitro and endothelial dysfunction in the mouse model of accelerated atherosclerosis in carotid arteries induced by partial ligation in vivo[102]
Salvianolic acid BNrf2/HO-1Inhibits tumor necrosis factor-alpha-induced NF-kappaB activation in HUVECs[103]
DihydromyricetinERK&AKT/Nrf2/HO-1Protects HUVECs from oxLDL-induced oxidative injury in vitro[104]
MiltironeNrf2/HO-1Protects human EA.hy926 ECs from oxidative stress-associated injury induced by t-BHP and oxLDL in vitro[105]
Z-LigustilideNrf2/AREProtects EA.hy926 cells from t-BHP-induced oxidative stress in vitro and attenuates atherogenesis in Ldlr−/− mice fed with a high-fat diet in vivo[106]
Nrf2/HO-1Attenuates inflammatory responses in HUVECs in vitro[107]
VitexinWnt/beta-catenin and Nrf2Protects HUVECs from high glucose-induced injury in vitro[108]
TheaflavinmiR-24/Nrf2/HO-1Alleviates oxidative injury and atherosclerosis progression in ApoE−/− mice fed with a high-fat diet in vivo and protects against cholesterol-induced oxidative injuries in HUVECs in vitro[109]
PI3K/AKT/Nrf2Attenuates t-BHP-induced oxidative stress in HUVECs in vitro and enhances vascularization in regenerated tissues and accelerates wound healing in vivo[110]
β-FarrerolGSK-3/Nrf2-AREProtect EA.hy926 cells against oxidative stress-induced injuries in vitro[111]
CompoundsDiabetes Related
MetforminNrf2Ameliorates the inhibitory effect of high glucose on migration and angiogenesis of HUVECs in vitro[84]
CarnosolNrf2/t-BHPProtects against t-BHP-induced human retinal microvascular endothelial cells (HRMECs) injury[112]
β-Buyang Huanwu DecoctionAKT/GSK3/Nrf2Enhances revascularization in a mouse model of diabetic hindlimb ischemia (HLI)[113]
Antrodin CNrf2/HO-1Prevents hyperglycemia-induced senescence and apoptosis in human ECs in vitro[114]
AllicinNrf2Alleviates aortic inflammatory responses associated with type 1 diabetes induced by intraperitoneal injection of streptozotocin (STZ) in mice and high glucose-induced growth inhibition and death in HUVECs in vitro[115]
CompoundsHypertension Related
Tilapia by-product oligopeptideNrf2/NF-κBProtects against Ang II-induced hypertensive injury in HUVECs in vitro[82]
A novel angiotensin-I-converting enzyme inhibitory peptide from microalgae Isochrysis zhanjiangensisNrf2Inhibits Ang II-induced vascular factor secretion, inflammatory responses, and apoptosis in HUVECs in vitro[116]
Hydrogen sulfideNrf2Ameliorates endothelial dysfunction associated with hypertension in spontaneously hypertensive rats (SHR) and Ang II-induced cellular damage and ROS formation in HUVECs in vitro[117]
CompoundsOther Diseases Related
CelastrolNrf2/ERK1/2/Nox2Attenuates Ang II-mediated HUVECs damage in vitro[78]
Schisandrin CKeap1/Nrf2As an antioxidative agent for the treatment of Ang II-induced vascular endothelial deficits in vitro[80]
OstholeKeap1/Nrf2Protects against Ang II-induced apoptosis of rat aortic endothelial cells (RAECs) in vitro[81]
MemantineNrf2/HO-1Protects against inflammatory responses and impaired endothelial tube formation induced by oxygen-glucose deprivation/reperfusion in HUVECs in vitro[83]
Cyanidin-3-O-glucosideNrf2/Bach1 and NF-κBImproves intracellular redox status of HUVECs exposed to palmitic acid (PA) in vitro[118]
Panax notoginseng SaponinPI3K/AKT/Nrf2Protects against cerebral ischemia/reperfusion (I/R)-induced blood–brain barrier disruption in cerebral microvascular endothelial cells (bEnd.3) in vitro[119]
Ginsenoside Rg3Nrf2/AREAntagonizes adriamycin-induced cardiotoxicity by improving endothelial dysfunction both in vivo and in vitro[120]
Procyanidin B2Nrf2/PPARγ/sFlt-1Ameliorates endothelial dysfunction and impaired angiogenesis of HUVECs in vitro and in a rat model of preeclampsia induced by uterine perfusion pressure (RUPP) in vivo[121]
Rice bran phenolic CompoundsNrf2/HO-1/NQO1/eNOSAn antioxidant/anti-inflammatory effect on HUVECs with induced oxidative stress in vitro[122]
Chlorogenic acidNrf2/HO-1Protective effects on Ang II infusion-induced vascular senescence in mice and H2O2-induced senescence in HUVECs in vitro[123]
PterostilbeneKeap1/Nrf2/HO-1Protects against uraemic serum (US)-mediated injury in HUVECs in vitro[124]
Lipoxin A4Nrf2/HO-1Attenuates H2O2-evoked cytotoxic injury in HUVECs in vitro[125]
Hydrogen sulfideNrf2/HIF-1αPrevents balloon injury-induced neointimal hyperplasia in carotid arteries of rats in vivo and enhances HUVECs tube formation and migration in vitro[126]
PaeoniflorinNrf2/HO-1Alleviates t-BHP-stimulated HUVECs cellular dysfunction and apoptosis in vitro and enhances the vascularization of regenerated tissues and promotes flap survival in vivo[127]
IrigeninNrf2Alleviates Ang II-induced oxidative stress and apoptosis in HUVECs in vitro[128]
CinnamaldehydeNrf2/HO-1Protects against H2O2 or TNFα-induced inflammatory responses in HUVECs and inhibits lipopolysaccharide (LPS)-induced inflammatory cell infiltration in vivo[129]
ProtandimNrf2Protects against salt-induced vascular dysfunction in vivo by restoring redox homeostasis in the vasculature[130]
Magnesium lithospermate BPI3K/AKT/Nrf2Protects against LPS-induced endothelial dysfunction in a murine acute inflammation model and in human dermal microvascular endothelial cells (HMECs-1) in vitro[131]
Ginkgo biloba extractNrf2/HO-1Reduces leukocyte adherence to injury arteries, enhances HO-1 expression in circulating monocytes and arteries after wire injury, and reduces TNF-alpha-stimulated endothelial adhesiveness[132]
Brazilian Green PropolisPI3K/AKT/mTOR/Nrf2/HO-1Inhibits oxLDL-stimulated oxidative stress in HUVECs in vitro[133]
Unripe Carica papaya fruit extractNrf2Protects ECs challenged with H2O2 in vitro[134]
LycopeneNrf2/HO-1Inhibits cyclic strain-induced ET-1 gene expression of HUVECs in vitro[135]
Hippocampus abdominalis-derived peptidesNrf2/HO-1Protects H2O2-induced cell death through antiapoptotic action in vitro[136]
Aspirin eugenol esterNrf2Attenuates oxidative injury in a hamster model of atherosclerosis induced by a high-fat diet and H2O2-induced apoptosis in HUVECs, an in vitro model of oxidative stress[137]
miRNAs
miR-24Nrf2/HO-1Promotes endothelial repair caused by oxidative stress after balloon injury in diabetic rats[138]
miR-200aNrf2Thymosin beta-4 (Tbeta4) attenuates H/R-induced cardiac microvascular endothelial cells (CMECs) injury by miR-200a-Nrf2 signaling in vitro[139]
miR-140-5pNrf2 and Sirt2Upregulation of miR-140-5p in the atherosclerotic aorta in ApoE−/− mice
Increases oxidative stress in HUVECs in vitro
[140]

3.2. Nrf2 and Vascular Smooth Muscle Cells

Vascular smooth muscle cells (VSMCs) are not terminally differentiated, instead, they are highly plastic. Under pathological conditions, mature quiescent and contractile VSMCs can be converted into a synthetic type of VSMCs characterized by decreased expression of SMC contractile proteins and increased capabilities of cellular proliferation, migration, and synthesis and secretion of extracellular matrix, calcific cells, or even stem cells, a process known as VSMC phenotypic transition or dedifferentiation which contributes to the development and progression of many vascular diseases [141,142,143,144,145].
Single-cell RNA sequencing shows that Nrf2 is a key regulator of VSMC dedifferentiation [141]. Nrf2 is required for choline-induced inhibition of VSMC dedifferentiation and vascular protection [146]. Other evidence reveals that downregulation of Nrf2 expression and activity contributes to the dedifferentiation of coronary arterial smooth muscle cells due to the genetic deficiency of CD38 [147]. Whilst Nrf2 depletion enhances platelet-derived growth factor (PDGF)-stimulated migration but not the proliferation of rat aortic SMCs, global Nrf2 knockout inhibits intimal hyperplasia of wire-injured femoral arteries in mice [148]. Accordingly, Nrf2 signaling becomes the target of botanical medicine for the treatment of vascular disease (Table 2) [149,150,151,152,153,154,155]. For example, the Keap1-Nrf2-ARE antioxidant system was demonstrated to mediate the inhibitory effects of rosmarinic acid, a hydroxylated compound frequently found in herbal plants, on platelet aggregation, VSMC dedifferentiation, and neointima formation [149]. Nrf2 activation is associated with andrographolide-induced suppression of pulmonary arterial SMC growth in vitro and pulmonary arterial pathological remodeling and hypertension in vivo [150]. Similarly, cinnamic aldehyde, an electrophilic Nrf2 activator, can inhibit VSMC growth and intimal hyperplasia after balloon injury in a rat model of diabetic restenosis [152]. In addition, such beneficial activation of Nrf2 is implicated in the potential efficacy of other plant extracts in treating vascular diseases, including prunella vulgaris, sulfiredoxin-1, and physalin B [103,151,156,157,158,159,160]. Synthetic drugs have also been documented to activate the protective function of Nrf2 in VSMCs. For example, coenzyme Q10 (CoQ10), a commonly used nutritional supplement, promotes the expression of Nrf2 and reduces oxidative stress in VSMCs, and attenuates intracranial aneurysm formation and rupture in mice [161]. Canagliflozin, a new sodium-glucose co-transport protein 2 (SGLT2) inhibitor, reduces CVD and mortality in patients with type 2 diabetes presumably via the activation of ROS/Nrf2/HO-1 path way in VSMCs [162]. An axis of ERK5/Nrf2 activation induced by statins such as fluvastatin reduces advanced glycation endproduct-induced VSMCs proliferation and migration, thus supporting the therapeutic potential of targeting the ERK5-Nrf2 signaling module in treating vascular lesions associated with diabetes [163]. Other functional synthetic drugs that protect VSMCs by activating Nrf2 are listed in Table 2.
Vascular calcification, a common complication of CVD, is driven in part by VSMC dedifferentiation into calcific cells [170]. Interestingly, several studies demonstrated that Nrf2-operated antioxidant defense [171,172] is critical for suppressing VSMC dedifferentiation into calcific cells in vitro. In addition, the gastransmitter hydrogen sulfide (H2S) attenuates VSMC calcification through activation of Keap1/Nrf2/NQO1 [164] while the rosmarinic acid- or mitoquinone-induced suppression of VSMC calcification depends on Nrf2 activation [166,168]. In contrast, O-linked N-acetylglucosamine transferase (OGT)-mediated Keap1 glycosylation accelerates the degradation of Nrf2 and suppresses autophagy. thereby leading to hyperphosphatemia-induced vascular calcification in chronic kidney disease [173].
However, emerging evidence indicated that activation of Nrf2 likely underlies palmitate-induced pro-inflammatory responses in human coronary artery SMCs in vitro [174]. In addition, Nrf2 deficiency can attenuate atherosclerosis by reducing lectin-like oxidized low-density lipoprotein receptor (LOX)-1-mediated VSMC proliferation and migration [143]. Whether such Nrf2-operated pathological signaling is restricted to VSMCs in a proatherosclerotic setting remains unknown.
Clearly, activation of Nrf2 activation can be either beneficial or detrimental to VSMCs, thereby contributing to either vascular repair or disease. Further studies of molecular mechanisms driving the Nrf2-operated dichotomy in diverse pathophysiological settings may shed light on the complexity of Nrf2 signaling in the vasculature.

3.3. Nrf2 and Vascular Disease

Atherosclerosis is a chronic vascular disease of the arteries. Oxidative stress is an important factor in the development of atherosclerosis, and the Nrf2 signaling pathway as the main antioxidant pathway has become an important target for atherosclerosis prevention and treatment [175]. Intriguingly, it has been demonstrated that Nrf2 plays a dual role in the development and progression of atherosclerosis (Table 3). Nrf2 knockout approach revealed that Nrf2 upregulates the expression of receptor CD36 and enhances uptake of oxLDL and foam cell formation in macrophages, thereby promoting atherosclerosis development [176,177,178]. Nrf2 exacerbated atherosclerosis by enhancing IL-1-mediated vascular inflammation [179]. In addition, Nrf2 participated in the development and progression of atherosclerosis by promoting the polarization of macrophages [180,181]. Of note, these Nrf2-mediated detrimental actions occur in the setting of ApoE deficiency. However, in hypercholesterolemic mice, Nrf2 can mediate both pro- and anti-atherosclerotic effects. Although the Nrf2-CD36 pathway promotes atherosclerosis, Nrf2-mediated induction of antioxidant genes may contribute to a reduction in atherosclerotic lesion development [182,183].
Abdominal aortic aneurysm (AAA) is a degenerative disease that is one of the principal causes of death in people over 65 years of age. The increasing ROS and oxidative stress seem to play a key role in the development of AAA [184]. Many drugs are designed to treat AAA by targeting Nrf2-operated antioxidant defense [185,186]. For example, calcitriol supplementation reduces the severity of AAA by reactivating Nrf2 and inhibiting apoptotic pathways. Additionally, betanin prevents experimental AAA progression by modulating the Nrf2/HO-1 pathways. Several small molecules and metabolites also protect AAA progression by targeting Nrf2. For example, itaconate prevents abdominal aortic aneurysm formation by inhibiting inflammation through the activation of Nrf2 [187]. Sestrin2 attenuates Ang II-induced apoptosis in VSMCs via the Nrf2 pathway of AAA [188]. Carbon monoxide-induced Nrf2/HO-1 alleviates inflammatory responses to Ang II by inhibiting NADPH oxidase- and mitochondria-derived ROS in AAA [189]. However, simvastatin treatment increases HO-1 protein levels in AAA, but independently of Nrf2 [190]. When the Nrf2 transcriptional activity is lacking, simvastatin may further reduce AAA formation [191].
Table 3. Nrf2 signaling cascades in vascular damage and dysfunction.
Table 3. Nrf2 signaling cascades in vascular damage and dysfunction.
Nrf2 SignalingAnimal ModelPathological
Setting
TreatmentPhenotypeProposed
Mechanism
Reference
Nrf2/CD36Nrf2−/−,
ApoE−/−
AtherosclerosisNoneNrf2 knockout decreases susceptibility to atherosclerotic plaque formation in ApoE−/− mice.Nrf2 upregulates CD36 thereby promoting foam cell formation and the pathogenesis of atherosclerosis.[177]
Nrf2/CD36Nrf2−/−,
ApoE−/−
AtherosclerosisNoneNrf2 expression promotes atherosclerotic lesion formation.Nrf2 deficiency results in decreased cholesterol
Influx, correlated with lower CD36 expression.
[178]
Nrf2/IL-1Nrf2−/−,
ApoE−/−
AtherosclerosisNoneNrf2-deficient ApoE−/− mice are highly protected against diet-induced atherogenesis.Nrf2 aggravates atherosclerosis by enhancing IL-1-mediated vascular inflammation.[179]
Nrf2Nrf2−/−,
ApoE−/−
AtherosclerosisNoneNrf2 deletion in bone marrow-derived cells is protective against atherosclerosis.Nrf2−/−; ApoE−/− decreases expression of macrophage M1-subtype genes in mice.[180]
Nrf2ApoE−/−AtherosclerosisPCB29-pQPCB29-pQ driven CD163+ macrophage accumulated in the aortic valve.Nrf2 activation is the main reason for macrophage differentiation.[181]
Nrf2 AtherosclerosisVitamin ENrf2 can mediate both pro-and anti-atherosclerotic effects.Nrf2 activation can promote CD36-mediated cholesterol uptake by macrophages, increase induction of Nrf2-mediated antioxidant genes, and is likely to contribute to decreased lesion progression.[182]
Nrf2Nrf2−/−,
Ldlr−/−,
Apob100/100
AtherosclerosisNoneNrf2 deficiency impairs atherosclerotic lesion development but promotes the features of plaque instability in hyper-cholesterolemic mice.Nrf2 deficiency in Ldlr−/− mice reduces total plasma cholesterol and triglycerides in Ldlr−/−ApoB100/100 mice and aggravates aortic plaque maturation as it increased plaque calcification.[183]
Nrf2Nrf2−/−Abdominal aortic aneurysmNoneLacking Nrf2 transcriptional activity attenuates AAA formation under simvastatin treatment.The inhibition of Nrf2 transcriptional activity facilitates AAA formation in mice, which can be prevented by simvastatin.[191]

4. Therapeutic Potential of Targeting Nrf2 Pathway

Nrf2 can be a potential therapeutic target for a variety of diseases, including CVDs, thus the development of drugs that modulate Nrf2 activity or pathways of actions appears to be an important approach for the future clinical treatment of CVDs. This notion is indeed supported by a mounting number of previous studies showing Nrf2-mediated cardiovascular protection in diverse pathological settings. However, as we recently highlighted in the heart [14], Nrf2 activation may not always be beneficial in the vasculature aforementioned. Of note, in ApoE−/− mice, knockdown of Nrf2 has a protective effect against atherosclerosis [177,178,179,180]. While it is generally accepted that Nrf2-mediated vascular protection is due to Nrf2-operated antioxidative defense, it remains unclear how Nrf2 exacerbates vascular disease. A series of our studies demonstrated that in the heart, Nrf2 functions protectively when autophagy is normal, but when autophagy is impaired, the Nrf2-mediated cardiac protection is lost, and Nrf2-operated myocardial damage appears [11,12,13]. Therefore, it is most likely that inhibition of autophagy is responsible for the activation of Nrf2-mediated cardiac injury. Of interest, several studies documented that autophagy is inhibited in atherosclerosis [192] and autophagy activation suppresses atherosclerosis in ApoE−/− mice [193,194]. Accordingly, it is possible that autophagy also plays a critical role in determining the biological consequences of Nrf2 activation in the vasculature. Taken together, these findings reveal Nrf2-mediated dichotomy in the cardiovascular system. Special precautions should be taken to avoid the detrimental activation of Nrf2.
Notably, since methyl Bardoxolone, a potent Nrf2 activator, increases the incidence of cardiovascular events, including heart failure and death, the clinical phase III trial testing its efficacy in the treatment of chronic kidney disease associated with type 2 diabetes was terminated [195]. Although the precise reasons remain to be determined, a possibility is that Bardoxolone methyl activates a yet unrecognized Nrf2 detrimental signaling in such a pathological setting. Unfortunately, the pathological activation of Nrf2 is still far from a comprehensive understanding. Whilst it is important to study the endogenous signaling pathways underlying Nrf2-mediated cardiovascular protection such as Notch-Nrf2 axis and PI3K-AKT-Nrf2 pathway in the cardiovascular system [50,196], it is crucial to dissect the Nrf2 signaling that causes cardiovascular damage and dysfunction. Accordingly, the outcome could provide guidance for the rationale, design, and screen of a novel class of Nrf2 modulators that can selectively activate Nrf2-mediated cardiovascular protection while turning off the Nrf2-operating pathological program.

5. Closing Remark

Since the discovery of Nrf2 as a master transcription factor of antioxidant defense, the Nrf2 signaling pathway has become not only a hot topic in the field of anti-oxidative stress research but also a therapeutic target to treat various diseases. It is clear that Nrf2 is a multifunctional transcription factor and executes diverse biological actions in a context-dependent manner, resulting in either protective or detrimental effects on the cardiovascular system. However, the molecular mechanisms which turn on or off the Nrf2-operating dichotomous actions are poorly understood. Nevertheless, there are still a lot of efforts in developing Nrf2 activators to treat several non-CVDs [197]. Therefore, it is necessary to further investigate the molecular mechanisms of Nrf2-mediated cardiovascular damage and dysfunction, thereby providing crucial information for precisely targeting Nrf2 signaling to treat cardiovascular and other diseases.

Author Contributions

Writing—original draft preparation, W.W., A.H. and S.N.; writing—review and editing, T.C.; supervision and funding acquisition, T.C.; All authors have read and agreed to the published version of the manuscript.

Funding

This work is partly supported by grants from the National Heart, Lung, and Blood Institute of NIH to T.C. (R01 HL160541) and from the US Department of Veterans Affairs to T.C. (I01 CX002062).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhang, Y.; Murugesan, P.; Huang, K.; Cai, H. NADPH oxidases and oxidase crosstalk in cardiovascular diseases: Novel therapeutic targets. Nat. Rev. Cardiol. 2020, 17, 170–194. [Google Scholar] [CrossRef] [PubMed]
  2. Sack, M.N.; Fyhrquist, F.Y.; Saijonmaa, O.J.; Fuster, V.; Kovacic, J.C. Basic Biology of Oxidative Stress and the Cardiovascular System: Part 1 of a 3-Part Series. J. Am. Coll. Cardiol. 2017, 70, 196–211. [Google Scholar] [CrossRef] [PubMed]
  3. Munzel, T.; Camici, G.G.; Maack, C.; Bonetti, N.R.; Fuster, V.; Kovacic, J.C. Impact of Oxidative Stress on the Heart and Vasculature: Part 2 of a 3-Part Series. J. Am. Coll. Cardiol. 2017, 70, 212–229. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. Munzel, T.; Gori, T.; Bruno, R.M.; Taddei, S. Is oxidative stress a therapeutic target in cardiovascular disease? Eur. Heart J. 2010, 31, 2741–2748. [Google Scholar] [CrossRef]
  6. Burgoyne, J.R.; Mongue-Din, H.; Eaton, P.; Shah, A.M. Redox signaling in cardiac physiology and pathology. Circ. Res. 2012, 111, 1091–1106. [Google Scholar] [CrossRef]
  7. Burgoyne, J.R.; Din, H.M.; Eaton, P.; Shah, A.M. Response to Detailed aspects of redox signaling in cardiac physiology and pathology. Circ. Res. 2013, 112, e2. [Google Scholar] [CrossRef]
  8. Stocker, R.; Keaney, J.F., Jr. Role of oxidative modifications in atherosclerosis. Physiol. Rev. 2004, 84, 1381–1478. [Google Scholar] [CrossRef]
  9. Freed, J.K.; Gutterman, D.D. Mitochondrial reactive oxygen species and vascular function: Less is more. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 673–675. [Google Scholar] [CrossRef]
  10. Zu, X.; Yu, L.; Sun, Y.; Tian, J.; Liu, F.; Sun, Q.; He, S.; Sun, G.; Luo, W.; Jiang, Y. Global mapping of ZBTB7A transcription factor binding sites in HepG2 cells. Cell. Mol. Biol. Lett. 2010, 15, 260–271. [Google Scholar] [CrossRef]
  11. Qin, Q.; Qu, C.; Niu, T.; Zang, H.; Qi, L.; Lyu, L.; Wang, X.; Nagarkatti, M.; Nagarkatti, P.; Janicki, J.S.; et al. Nrf2-Mediated Cardiac Maladaptive Remodeling and Dysfunction in a Setting of Autophagy Insufficiency. Hypertension 2016, 67, 107–117. [Google Scholar] [CrossRef] [PubMed]
  12. Zang, H.; Wu, W.; Qi, L.; Tan, W.; Nagarkatti, P.; Nagarkatti, M.; Wang, X.; Cui, T. Autophagy Inhibition Enables Nrf2 to Exaggerate the Progression of Diabetic Cardiomyopathy in Mice. Diabetes 2020, 69, 2720–2734. [Google Scholar] [CrossRef] [PubMed]
  13. Wu, W.; Qin, Q.; Ding, Y.; Zang, H.; Li, D.S.; Nagarkatti, M.; Nagarkatti, P.; Wang, W.; Wang, X.; Cui, T. Autophagy Controls Nrf2-Mediated Dichotomy in Pressure Overloaded Hearts. Front. Physiol. 2021, 12, 673145. [Google Scholar] [CrossRef] [PubMed]
  14. Zang, H.; Mathew, R.O.; Cui, T. The Dark Side of Nrf2 in the Heart. Front. Physiol. 2020, 11, 722. [Google Scholar] [CrossRef]
  15. Yamamoto, M.; Kensler, T.W.; Motohashi, H. The KEAP1-NRF2 System: A Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol. Rev. 2018, 98, 1169–1203. [Google Scholar] [CrossRef]
  16. Li, J.; Ichikawa, T.; Janicki, J.S.; Cui, T. Targeting the Nrf2 pathway against cardiovascular disease. Expert Opin. Ther. Targets 2009, 13, 785–794. [Google Scholar] [CrossRef]
  17. Maher, J.; Yamamoto, M. The rise of antioxidant signaling—The evolution and hormetic actions of Nrf2. Toxicol. Appl. Pharmacol. 2010, 244, 4–15. [Google Scholar] [CrossRef]
  18. Chan, J.Y.; Kwong, M.; Lo, M.; Emerson, R.; Kuypers, F.A. Reduced oxidative-stress response in red blood cells from p45NFE2-deficient mice. Blood 2001, 97, 2151–2158. [Google Scholar] [CrossRef]
  19. Tiwari, S.; Italiano, J.E., Jr.; Barral, D.C.; Mules, E.H.; Novak, E.K.; Swank, R.T.; Seabra, M.C.; Shivdasani, R.A. A role for Rab27b in NF-E2-dependent pathways of platelet formation. Blood 2003, 102, 3970–3979. [Google Scholar] [CrossRef]
  20. Lecine, P.; Italiano, J.E., Jr.; Kim, S.W.; Villeval, J.L.; Shivdasani, R.A. Hematopoietic-specific beta 1 tubulin participates in a pathway of platelet biogenesis dependent on the transcription factor NF-E2. Blood 2000, 96, 1366–1373. [Google Scholar] [CrossRef] [Green Version]
  21. Leung, L.; Kwong, M.; Hou, S.; Lee, C.; Chan, J.Y. Deficiency of the Nrf1 and Nrf2 transcription factors results in early embryonic lethality and severe oxidative stress. J. Biol. Chem. 2003, 278, 48021–48029. [Google Scholar] [CrossRef] [PubMed]
  22. Kobayashi, A.; Ito, E.; Toki, T.; Kogame, K.; Takahashi, S.; Igarashi, K.; Hayashi, N.; Yamamoto, M. Molecular cloning and functional characterization of a new Cap‘n’ collar family transcription factor Nrf3. J. Biol. Chem. 1999, 274, 6443–6452. [Google Scholar] [CrossRef] [PubMed]
  23. Derjuga, A.; Gourley, T.S.; Holm, T.M.; Heng, H.H.; Shivdasani, R.A.; Ahmed, R.; Andrews, N.C.; Blank, V. Complexity of CNC transcription factors as revealed by gene targeting of the Nrf3 locus. Mol. Cell. Biol. 2004, 24, 3286–3294. [Google Scholar] [CrossRef] [PubMed]
  24. Xiao, Q.; Pepe, A.E.; Wang, G.; Luo, Z.; Zhang, L.; Zeng, L.; Zhang, Z.; Hu, Y.; Ye, S.; Xu, Q. Nrf3-Pla2g7 interaction plays an essential role in smooth muscle differentiation from stem cells. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 730–744. [Google Scholar] [CrossRef]
  25. Pepe, A.E.; Xiao, Q.; Zampetaki, A.; Zhang, Z.; Kobayashi, A.; Hu, Y.; Xu, Q. Crucial role of nrf3 in smooth muscle cell differentiation from stem cells. Circ. Res. 2010, 106, 870–879. [Google Scholar] [CrossRef]
  26. Oyake, T.; Itoh, K.; Motohashi, H.; Hayashi, N.; Hoshino, H.; Nishizawa, M.; Yamamoto, M.; Igarashi, K. Bach proteins belong to a novel family of BTB-basic leucine zipper transcription factors that interact with MafK and regulate transcription through the NF-E2 site. Mol. Cell. Biol. 1996, 16, 6083–6095. [Google Scholar] [CrossRef]
  27. Hayes, J.D.; Dinkova-Kostova, A.T. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 2014, 39, 199–218. [Google Scholar] [CrossRef]
  28. Cui, T.; Lai, Y.; Janicki, J.S.; Wang, X. Nuclear factor erythroid-2 related factor 2 (Nrf2)-mediated protein quality control in cardiomyocytes. Front. Biosci. (Landmark Ed.) 2016, 21, 192–202. [Google Scholar] [CrossRef]
  29. Kopacz, A.; Kloska, D.; Forman, H.J.; Jozkowicz, A.; Grochot-Przeczek, A. Beyond repression of Nrf2: An update on Keap1. Free Radic. Biol. Med. 2020, 157, 63–74. [Google Scholar] [CrossRef]
  30. Silva-Islas, C.A.; Maldonado, P.D. Canonical and non-canonical mechanisms of Nrf2 activation. Pharmacol. Res. 2018, 134, 92–99. [Google Scholar] [CrossRef]
  31. Fukutomi, T.; Takagi, K.; Mizushima, T.; Ohuchi, N.; Yamamoto, M. Kinetic, thermodynamic, and structural characterizations of the association between Nrf2-DLGex degron and Keap1. Mol. Cell. Biol. 2014, 34, 832–846. [Google Scholar] [CrossRef] [PubMed]
  32. Tian, W.; Rojo de la Vega, M.; Schmidlin, C.J.; Ooi, A.; Zhang, D.D. Kelch-like ECH-associated protein 1 (KEAP1) differentially regulates nuclear factor erythroid-2-related factors 1 and 2 (NRF1 and NRF2). J. Biol. Chem. 2018, 293, 2029–2040. [Google Scholar] [CrossRef] [PubMed]
  33. Katoh, Y.; Itoh, K.; Yoshida, E.; Miyagishi, M.; Fukamizu, A.; Yamamoto, M. Two domains of Nrf2 cooperatively bind CBP, a CREB binding protein, and synergistically activate transcription. Genes Cells 2001, 6, 857–868. [Google Scholar] [CrossRef] [PubMed]
  34. Chang, M.; Wilson, C.J.; Karunatilleke, N.C.; Moselhy, M.H.; Karttunen, M.; Choy, W.Y. Exploring the Conformational Landscape of the Neh4 and Neh5 Domains of Nrf2 Using Two Different Force Fields and Circular Dichroism. J. Chem. Theory Comput. 2021, 17, 3145–3156. [Google Scholar] [CrossRef] [PubMed]
  35. Sun, Z.; Chin, Y.E.; Zhang, D.D. Acetylation of Nrf2 by p300/CBP augments promoter-specific DNA binding of Nrf2 during the antioxidant response. Mol. Cell. Biol. 2009, 29, 2658–2672. [Google Scholar] [CrossRef] [PubMed]
  36. Kim, J.H.; Yu, S.; Chen, J.D.; Kong, A.N. The nuclear cofactor RAC3/AIB1/SRC-3 enhances Nrf2 signaling by interacting with transactivation domains. Oncogene 2013, 32, 514–527. [Google Scholar] [CrossRef]
  37. Wang, H.; Liu, K.; Geng, M.; Gao, P.; Wu, X.; Hai, Y.; Li, Y.; Li, Y.; Luo, L.; Hayes, J.D.; et al. RXRalpha inhibits the NRF2-ARE signaling pathway through a direct interaction with the Neh7 domain of NRF2. Cancer Res. 2013, 73, 3097–3108. [Google Scholar] [CrossRef]
  38. Jiang, S.; Yang, Y.; Li, T.; Ma, Z.; Hu, W.; Deng, C.; Fan, C.; Lv, J.; Sun, Y.; Yi, W. An overview of the mechanisms and novel roles of Nrf2 in cardiovascular diseases. Expert Opin. Ther. Targets 2016, 20, 1413–1424. [Google Scholar] [CrossRef]
  39. Rada, P.; Rojo, A.I.; Chowdhry, S.; McMahon, M.; Hayes, J.D.; Cuadrado, A. SCF/{beta}-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol. Cell. Biol. 2011, 31, 1121–1133. [Google Scholar] [CrossRef]
  40. Chowdhry, S.; Zhang, Y.; McMahon, M.; Sutherland, C.; Cuadrado, A.; Hayes, J.D. Nrf2 is controlled by two distinct beta-TrCP recognition motifs in its Neh6 domain, one of which can be modulated by GSK-3 activity. Oncogene 2013, 32, 3765–3781. [Google Scholar] [CrossRef] [Green Version]
  41. Cuadrado, A. Structural and functional characterization of Nrf2 degradation by glycogen synthase kinase 3/beta-TrCP. Free Radic. Biol. Med. 2015, 88, 147–157. [Google Scholar] [CrossRef] [PubMed]
  42. Rojo, A.I.; Medina-Campos, O.N.; Rada, P.; Zuniga-Toala, A.; Lopez-Gazcon, A.; Espada, S.; Pedraza-Chaverri, J.; Cuadrado, A. Signaling pathways activated by the phytochemical nordihydroguaiaretic acid contribute to a Keap1-independent regulation of Nrf2 stability: Role of glycogen synthase kinase-3. Free Radic. Biol. Med. 2012, 52, 473–487. [Google Scholar] [CrossRef] [PubMed]
  43. Moi, P.; Chan, K.; Asunis, I.; Cao, A.; Kan, Y.W. Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region. Proc. Natl. Acad. Sci. USA 1994, 91, 9926–9930. [Google Scholar] [CrossRef]
  44. Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem. Biophys. Res. Commun. 1997, 236, 313–322. [Google Scholar] [CrossRef] [PubMed]
  45. Li, W.; Jain, M.R.; Chen, C.; Yue, X.; Hebbar, V.; Zhou, R.; Kong, A.N. Nrf2 Possesses a redox-insensitive nuclear export signal overlapping with the leucine zipper motif. J. Biol. Chem. 2005, 280, 28430–28438. [Google Scholar] [CrossRef]
  46. Jain, A.K.; Bloom, D.A.; Jaiswal, A.K. Nuclear import and export signals in control of Nrf2. J. Biol. Chem. 2005, 280, 29158–29168. [Google Scholar] [CrossRef] [PubMed]
  47. Nioi, P.; Nguyen, T.; Sherratt, P.J.; Pickett, C.B. The carboxy-terminal Neh3 domain of Nrf2 is required for transcriptional activation. Mol. Cell. Biol. 2005, 25, 10895–10906. [Google Scholar] [CrossRef]
  48. Miao, W.; Hu, L.; Scrivens, P.J.; Batist, G. Transcriptional regulation of NF-E2 p45-related factor (NRF2) expression by the aryl hydrocarbon receptor-xenobiotic response element signaling pathway: Direct cross-talk between phase I and II drug-metabolizing enzymes. J. Biol. Chem. 2005, 280, 20340–20348. [Google Scholar] [CrossRef]
  49. Kohle, C.; Bock, K.W. Coordinate regulation of Phase I and II xenobiotic metabolisms by the Ah receptor and Nrf2. Biochem. Pharmacol. 2007, 73, 1853–1862. [Google Scholar] [CrossRef]
  50. Wakabayashi, N.; Skoko, J.J.; Chartoumpekis, D.V.; Kimura, S.; Slocum, S.L.; Noda, K.; Palliyaguru, D.L.; Fujimuro, M.; Boley, P.A.; Tanaka, Y.; et al. Notch-Nrf2 axis: Regulation of Nrf2 gene expression and cytoprotection by notch signaling. Mol. Cell. Biol. 2014, 34, 653–663. [Google Scholar] [CrossRef] [Green Version]
  51. Yazaki, K.; Matsuno, Y.; Yoshida, K.; Sherpa, M.; Nakajima, M.; Matsuyama, M.; Kiwamoto, T.; Morishima, Y.; Ishii, Y.; Hizawa, N. ROS-Nrf2 pathway mediates the development of TGF-beta1-induced epithelial-mesenchymal transition through the activation of Notch signaling. Eur. J. Cell Biol. 2021, 100, 151181. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, Y.; Gordon, G.B. A strategy for cancer prevention: Stimulation of the Nrf2-ARE signaling pathway. Mol. Cancer Ther. 2004, 3, 885–893. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, Q.M.; Maltagliati, A.J. Nrf2 at the heart of oxidative stress and cardiac protection. Physiol. Genom. 2018, 50, 77–97. [Google Scholar] [CrossRef] [PubMed]
  54. Wei, Y.; Gong, J.; Thimmulappa, R.K.; Kosmider, B.; Biswal, S.; Duh, E.J. Nrf2 acts cell-autonomously in endothelium to regulate tip cell formation and vascular branching. Proc. Natl. Acad. Sci. USA 2013, 110, E3910–E3918. [Google Scholar] [CrossRef]
  55. Baeyens, N.; Bandyopadhyay, C.; Coon, B.G.; Yun, S.; Schwartz, M.A. Endothelial fluid shear stress sensing in vascular health and disease. J. Clin. Investig. 2016, 126, 821–828. [Google Scholar] [CrossRef]
  56. Chiu, J.J.; Chien, S. Effects of disturbed flow on vascular endothelium: Pathophysiological basis and clinical perspectives. Physiol. Rev. 2011, 91, 327–387. [Google Scholar] [CrossRef]
  57. Niu, N.; Xu, S.; Xu, Y.; Little, P.J.; Jin, Z.G. Targeting Mechanosensitive Transcription Factors in Atherosclerosis. Trends Pharmacol. Sci. 2019, 40, 253–266. [Google Scholar] [CrossRef]
  58. Takabe, W.; Warabi, E.; Noguchi, N. Anti-atherogenic effect of laminar shear stress via Nrf2 activation. Antioxid. Redox Signal. 2011, 15, 1415–1426. [Google Scholar] [CrossRef]
  59. Nigro, P.; Abe, J.; Berk, B.C. Flow shear stress and atherosclerosis: A matter of site specificity. Antioxid. Redox Signal. 2011, 15, 1405–1414. [Google Scholar] [CrossRef]
  60. McSweeney, S.R.; Warabi, E.; Siow, R.C. Nrf2 as an Endothelial Mechanosensitive Transcription Factor: Going with the Flow. Hypertension 2016, 67, 20–29. [Google Scholar] [CrossRef]
  61. Chen, X.L.; Varner, S.E.; Rao, A.S.; Grey, J.Y.; Thomas, S.; Cook, C.K.; Wasserman, M.A.; Medford, R.M.; Jaiswal, A.K.; Kunsch, C. Laminar flow induction of antioxidant response element-mediated genes in endothelial cells. A novel anti-inflammatory mechanism. J. Biol. Chem. 2003, 278, 703–711. [Google Scholar] [CrossRef] [PubMed]
  62. Dai, G.; Vaughn, S.; Zhang, Y.; Wang, E.T.; Garcia-Cardena, G.; Gimbrone, M.A., Jr. Biomechanical forces in atherosclerosis-resistant vascular regions regulate endothelial redox balance via phosphoinositol 3-kinase/Akt-dependent activation of Nrf2. Circ. Res. 2007, 101, 723–733. [Google Scholar] [CrossRef] [PubMed]
  63. Hosoya, T.; Maruyama, A.; Kang, M.I.; Kawatani, Y.; Shibata, T.; Uchida, K.; Warabi, E.; Noguchi, N.; Itoh, K.; Yamamoto, M. Differential responses of the Nrf2-Keap1 system to laminar and oscillatory shear stresses in endothelial cells. J. Biol. Chem. 2005, 280, 27244–27250. [Google Scholar] [CrossRef]
  64. Cheng, X.; Siow, R.C.; Mann, G.E. Impaired redox signaling and antioxidant gene expression in endothelial cells in diabetes: A role for mitochondria and the nuclear factor-E2-related factor 2-Kelch-like ECH-associated protein 1 defense pathway. Antioxid. Redox Signal. 2011, 14, 469–487. [Google Scholar] [CrossRef] [PubMed]
  65. Zakkar, M.; Van der Heiden, K.; Luong le, A.; Chaudhury, H.; Cuhlmann, S.; Hamdulay, S.S.; Krams, R.; Edirisinghe, I.; Rahman, I.; Carlsen, H.; et al. Activation of Nrf2 in endothelial cells protects arteries from exhibiting a proinflammatory state. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 1851–1857. [Google Scholar] [CrossRef] [PubMed]
  66. Mylroie, H.; Dumont, O.; Bauer, A.; Thornton, C.C.; Mackey, J.; Calay, D.; Hamdulay, S.S.; Choo, J.R.; Boyle, J.J.; Samarel, A.M.; et al. PKCepsilon-CREB-Nrf2 signalling induces HO-1 in the vascular endothelium and enhances resistance to inflammation and apoptosis. Cardiovasc. Res. 2015, 106, 509–519. [Google Scholar] [CrossRef]
  67. Xiao, Y.; Xia, J.; Wu, S.; Lv, Z.; Huang, S.; Huang, H.; Su, X.; Cheng, J.; Ke, Y. Curcumin Inhibits Acute Vascular Inflammation through the Activation of Heme Oxygenase-1. Oxid. Med. Cell. Longev. 2018, 2018, 3295807. [Google Scholar] [CrossRef]
  68. Li, P.; Guo, X.; Lei, P.; Shi, S.; Luo, S.; Cheng, X. PI3K/Akt/uncoupling protein 2 signaling pathway may be involved in cell senescence and apoptosis induced by angiotensin II in human vascular endothelial cells. Mol. Biol. Rep. 2014, 41, 6931–6937. [Google Scholar] [CrossRef]
  69. Bendall, J.K.; Rinze, R.; Adlam, D.; Tatham, A.L.; de Bono, J.; Wilson, N.; Volpi, E.; Channon, K.M. Endothelial Nox2 overexpression potentiates vascular oxidative stress and hemodynamic response to angiotensin II: Studies in endothelial-targeted Nox2 transgenic mice. Circ. Res. 2007, 100, 1016–1025. [Google Scholar] [CrossRef]
  70. Yang, Y.; Tian, T.; Wang, Y.; Li, Z.; Xing, K.; Tian, G. SIRT6 protects vascular endothelial cells from angiotensin II-induced apoptosis and oxidative stress by promoting the activation of Nrf2/ARE signaling. Eur. J. Pharmacol. 2019, 859, 172516. [Google Scholar] [CrossRef]
  71. Iizuka, T.; Ishii, Y.; Itoh, K.; Kiwamoto, T.; Kimura, T.; Matsuno, Y.; Morishima, Y.; Hegab, A.E.; Homma, S.; Nomura, A.; et al. Nrf2-deficient mice are highly susceptible to cigarette smoke-induced emphysema. Genes Cells 2005, 10, 1113–1125. [Google Scholar] [CrossRef] [PubMed]
  72. Rangasamy, T.; Cho, C.Y.; Thimmulappa, R.K.; Zhen, L.; Srisuma, S.S.; Kensler, T.W.; Yamamoto, M.; Petrache, I.; Tuder, R.M.; Biswal, S. Genetic ablation of Nrf2 enhances susceptibility to cigarette smoke-induced emphysema in mice. J. Clin. Investig. 2004, 114, 1248–1259. [Google Scholar] [CrossRef] [PubMed]
  73. Singh, A.; Rangasamy, T.; Thimmulappa, R.K.; Lee, H.; Osburn, W.O.; Brigelius-Flohe, R.; Kensler, T.W.; Yamamoto, M.; Biswal, S. Glutathione peroxidase 2, the major cigarette smoke-inducible isoform of GPX in lungs, is regulated by Nrf2. Am. J. Respir. Cell. Mol. Biol. 2006, 35, 639–650. [Google Scholar] [CrossRef] [PubMed]
  74. Hsu, C.L.; Wu, Y.L.; Tang, G.J.; Lee, T.S.; Kou, Y.R. Ginkgo biloba extract confers protection from cigarette smoke extract-induced apoptosis in human lung endothelial cells: Role of heme oxygenase-1. Pulm. Pharmacol. Ther. 2009, 22, 286–296. [Google Scholar] [CrossRef]
  75. Zhao, Z.; Wang, X.; Zhang, R.; Ma, B.; Niu, S.; Di, X.; Ni, L.; Liu, C. Melatonin attenuates smoking-induced atherosclerosis by activating the Nrf2 pathway via NLRP3 inflammasomes in endothelial cells. Aging (Albany NY) 2021, 13, 11363–11380. [Google Scholar] [CrossRef]
  76. Prasad, S.; Sajja, R.K.; Kaisar, M.A.; Park, J.H.; Villalba, H.; Liles, T.; Abbruscato, T.; Cucullo, L. Role of Nrf2 and protective effects of Metformin against tobacco smoke-induced cerebrovascular toxicity. Redox. Biol. 2017, 12, 58–69. [Google Scholar] [CrossRef]
  77. Li, T.; Wang, H.; Ding, Y.; Zhou, M.; Zhou, X.; Zhang, X.; Ding, K.; He, J.; Lu, X.; Xu, J.; et al. Genetic elimination of Nrf2 aggravates secondary complications except for vasospasm after experimental subarachnoid hemorrhage in mice. Brain Res. 2014, 1558, 90–99. [Google Scholar] [CrossRef]
  78. Li, M.; Liu, X.; He, Y.; Zheng, Q.; Wang, M.; Wu, Y.; Zhang, Y.; Wang, C. Celastrol attenuates angiotensin II mediated human umbilical vein endothelial cells damage through activation of Nrf2/ERK1/2/Nox2 signal pathway. Eur. J. Pharmacol. 2017, 797, 124–133. [Google Scholar] [CrossRef]
  79. Zhou, X.; Liang, L.; Zhao, Y.; Zhang, H. Epigallocatechin-3-Gallate Ameliorates Angiotensin II-Induced Oxidative Stress and Apoptosis in Human Umbilical Vein Endothelial Cells through the Activation of Nrf2/Caspase-3 Signaling. J. Vasc. Res. 2017, 54, 299–308. [Google Scholar] [CrossRef]
  80. Han, J.; Shi, X.; Du, Y.; Shi, F.; Zhang, B.; Zheng, Z.; Xu, J.; Jiang, L. Schisandrin C targets Keap1 and attenuates oxidative stress by activating Nrf2 pathway in Ang II-challenged vascular endothelium. Phytother. Res. 2019, 33, 779–790. [Google Scholar] [CrossRef]
  81. Tao, L.; Gu, X.; Xu, E.; Ren, S.; Zhang, L.; Liu, W.; Lin, X.; Yang, J.; Chen, C. Osthole protects against Ang II-induced endotheliocyte death by targeting NF-kappaB pathway and Keap-1/Nrf2 pathway. Am. J. Transl. Res. 2019, 11, 142–159. [Google Scholar] [PubMed]
  82. Chen, J.; Gong, F.; Chen, M.F.; Li, C.; Hong, P.; Sun, S.; Zhou, C.; Qian, Z.J. In Vitro Vascular-Protective Effects of a Tilapia By-Product Oligopeptide on Angiotensin II-Induced Hypertensive Endothelial Injury in HUVEC by Nrf2/NF-kappaB Pathways. Mar. Drugs 2019, 17, 431. [Google Scholar] [CrossRef] [PubMed]
  83. Lv, X.; Li, Q.; Mao, S.; Qin, L.; Dong, P. The protective effects of memantine against inflammation and impairment of endothelial tube formation induced by oxygen-glucose deprivation/reperfusion. Aging (Albany NY) 2020, 12, 21469–21480. [Google Scholar] [CrossRef] [PubMed]
  84. Sun, C.C.; Lai, Y.N.; Wang, W.H.; Xu, X.M.; Li, X.Q.; Wang, H.; Zheng, J.Y.; Zheng, J.Q. Metformin Ameliorates Gestational Diabetes Mellitus-Induced Endothelial Dysfunction via Downregulation of p65 and Upregulation of Nrf2. Front. Pharmacol. 2020, 11, 575390. [Google Scholar] [CrossRef] [PubMed]
  85. Gjyshi, O.; Flaherty, S.; Veettil, M.V.; Johnson, K.E.; Chandran, B.; Bottero, V. Kaposi’s sarcoma-associated herpesvirus induces Nrf2 activation in latently infected endothelial cells through SQSTM1 phosphorylation and interaction with polyubiquitinated Keap1. J. Virol. 2015, 89, 2268–2286. [Google Scholar] [CrossRef] [PubMed]
  86. Xue, J.; Liao, Q.; Luo, M.; Hua, C.; Zhao, J.; Yu, G.; Chen, X.; Li, X.; Zhang, X.; Ran, R.; et al. Cigarette smoke-induced oxidative stress activates NRF2 to mediate fibronectin disorganization in vascular formation. Open Biol. 2022, 12, 210310. [Google Scholar] [CrossRef]
  87. Zhu, Z.; Li, J.; Zhang, X. Astragaloside IV Protects Against Oxidized Low-Density Lipoprotein (ox-LDL)-Induced Endothelial Cell Injury by Reducing Oxidative Stress and Inflammation. Med. Sci. Monit. 2019, 25, 2132–2140. [Google Scholar] [CrossRef]
  88. Guo, J.; Ma, J.; Cai, K.; Chen, H.; Xie, K.; Xu, B.; Quan, D.; Du, J. Isoflavones from Semen Sojae Preparatum Improve Atherosclerosis and Oxidative Stress by Modulating Nrf2 Signaling Pathway through Estrogen-Like Effects. Evid. Based. Complement. Altern. Med. 2022, 2022, 4242099. [Google Scholar] [CrossRef]
  89. Hou, J.R.; Wang, Y.H.; Zhong, Y.N.; Che, T.T.; Hu, Y.; Bao, J.; Meng, N. Protective Effect of Flavonoids from a Deep-Sea-Derived Arthrinium sp. against ox-LDL-Induced Oxidative Injury through Activating the AKT/Nrf2/HO-1 Pathway in Vascular Endothelial Cells. Mar. Drugs 2021, 19, 712. [Google Scholar] [CrossRef]
  90. He, L.; Liu, Y.Y.; Wang, K.; Li, C.; Zhang, W.; Li, Z.Z.; Huang, X.Z.; Xiong, Y. Tanshinone IIA protects human coronary artery endothelial cells from ferroptosis by activating the NRF2 pathway. Biochem. Biophys. Res. Commun. 2021, 575, 1–7. [Google Scholar] [CrossRef]
  91. Chen, L.W.; Tsai, M.C.; Chern, C.Y.; Tsao, T.P.; Lin, F.Y.; Chen, S.J.; Tsui, P.F.; Liu, Y.W.; Lu, H.J.; Wu, W.L.; et al. A chalcone derivative, 1m-6, exhibits atheroprotective effects by increasing cholesterol efflux and reducing inflammation-induced endothelial dysfunction. Br. J. Pharmacol. 2020, 177, 5375–5392. [Google Scholar] [CrossRef] [PubMed]
  92. Zhu, Y.; Li, M.; Lu, Y.; Li, J.; Ke, Y.; Yang, J. Ilexgenin A inhibits mitochondrial fission and promote Drp1 degradation by Nrf2-induced PSMB5 in endothelial cells. Drug Dev. Res. 2019, 80, 481–489. [Google Scholar] [CrossRef] [PubMed]
  93. Wu, Y.; Song, F.; Li, Y.; Li, J.; Cui, Y.; Hong, Y.; Han, W.; Wu, W.; Lakhani, I.; Li, G.; et al. Acacetin exerts antioxidant potential against atherosclerosis through Nrf2 pathway in ApoE−/− Mice. J. Cell. Mol. Med. 2021, 25, 521–534. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, M.; Xu, Y.; Jiang, L. Irisin attenuates oxidized low-density lipoprotein impaired angiogenesis through AKT/mTOR/S6K1/Nrf2 pathway. J. Cell. Physiol. 2019, 234, 18951–18962. [Google Scholar] [CrossRef]
  95. Pei, Y.; Lui, Y.; Cai, S.; Zhou, C.; Hong, P.; Qian, Z.J. A Novel Peptide Isolated from Microalgae Isochrysis zhanjiangensis Exhibits Anti-apoptosis and Anti-inflammation in Ox-LDL Induced HUVEC to Improve Atherosclerosis. Plant Foods Hum. Nutr. 2022, 77, 181–189. [Google Scholar] [CrossRef]
  96. Feng, Z.; Wang, C.; Yue, J.; Meng, Q.; Wu, J.; Sun, H. Kaempferol-induced GPER upregulation attenuates atherosclerosis via the PI3K/AKT/Nrf2 pathway. Pharm. Biol. 2021, 59, 1106–1116. [Google Scholar] [CrossRef]
  97. Yan, R.; Yan, J.; Chen, X.; Yu, Y.; Sun, T. Xanthoangelol Prevents Ox-LDL-Induced Endothelial Cell Injury by Activating Nrf2/ARE Signaling. J. Cardiovasc. Pharmacol. 2019, 74, 162–171. [Google Scholar] [CrossRef]
  98. Zhang, T.; Hu, Q.; Shi, L.; Qin, L.; Zhang, Q.; Mi, M. Equol Attenuates Atherosclerosis in Apolipoprotein E-Deficient Mice by Inhibiting Endoplasmic Reticulum Stress via Activation of Nrf2 in Endothelial Cells. PLoS ONE 2016, 11, e0167020. [Google Scholar] [CrossRef]
  99. Rajendran, P.; Alzahrani, A.M.; Ahmed, E.A.; Veeraraghavan, V.P. Kirenol Inhibits B[a]P-Induced Oxidative Stress and Apoptosis in Endothelial Cells via Modulation of the Nrf2 Signaling Pathway. Oxid. Med. Cell. Longev. 2021, 2021, 5585303. [Google Scholar] [CrossRef]
  100. Seo, Y.; Park, J.; Choi, W.; Ju Son, D.; Sung Kim, Y.; Kim, M.K.; Yoon, B.E.; Pyee, J.; Tae Hong, J.; Go, Y.M.; et al. Antiatherogenic Effect of Resveratrol Attributed to Decreased Expression of ICAM-1 (Intercellular Adhesion Molecule-1). Arterioscler. Thromb. Vasc. Biol. 2019, 39, 675–684. [Google Scholar] [CrossRef] [Green Version]
  101. Mao, H.; Tao, T.; Wang, X.; Liu, M.; Song, D.; Liu, X.; Shi, D. Zedoarondiol Attenuates Endothelial Cells Injury Induced by Oxidized Low-Density Lipoprotein via Nrf2 Activation. Cell. Physiol. Biochem. 2018, 48, 1468–1479. [Google Scholar] [CrossRef] [PubMed]
  102. Ding, Y.; Zhang, B.; Zhou, K.; Chen, M.; Wang, M.; Jia, Y.; Song, Y.; Li, Y.; Wen, A. Dietary ellagic acid improves oxidant-induced endothelial dysfunction and atherosclerosis: Role of Nrf2 activation. Int. J. Cardiol. 2014, 175, 508–514. [Google Scholar] [CrossRef] [PubMed]
  103. Lee, H.J.; Seo, M.; Lee, E.J. Salvianolic acid B inhibits atherogenesis of vascular cells through induction of Nrf2-dependent heme oxygenase-1. Curr. Med. Chem. 2014, 21, 3095–3106. [Google Scholar] [CrossRef]
  104. Luo, Y.; Lu, S.; Dong, X.; Xu, L.; Sun, G.; Sun, X. Dihydromyricetin protects human umbilical vein endothelial cells from injury through ERK and Akt mediated Nrf2/HO-1 signaling pathway. Apoptosis 2017, 22, 1013–1024. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, L.; Zhang, H.; Li, X.; Jia, B.; Yang, Y.; Zhou, P.; Li, P.; Chen, J. Miltirone protects human EA.hy926 endothelial cells from oxidized low-density lipoprotein-derived oxidative stress via a heme oxygenase-1 and MAPK/Nrf2 dependent pathway. Phytomedicine 2016, 23, 1806–1813. [Google Scholar] [CrossRef] [PubMed]
  106. Zhu, Y.; Zhang, Y.; Huang, X.; Xie, Y.; Qu, Y.; Long, H.; Gu, N.; Jiang, W. Z-Ligustilide protects vascular endothelial cells from oxidative stress and rescues high fat diet-induced atherosclerosis by activating multiple NRF2 downstream genes. Atherosclerosis 2019, 284, 110–120. [Google Scholar] [CrossRef] [PubMed]
  107. Choi, E.S.; Yoon, J.J.; Han, B.H.; Jeong, D.H.; Lee, Y.J.; Kang, D.G.; Lee, H.S. Ligustilide attenuates vascular inflammation and activates Nrf2/HO-1 induction and, NO synthesis in HUVECs. Phytomedicine 2018, 38, 12–23. [Google Scholar] [CrossRef]
  108. Zhang, S.; Jin, S.; Zhang, S.; Li, Y.Y.; Wang, H.; Chen, Y.; Lu, H. Vitexin protects against high glucose-induced endothelial cell apoptosis and oxidative stress via Wnt/beta-catenin and Nrf2 signalling pathway. Arch. Physiol. Biochem. 2022, 7, 1–10. [Google Scholar] [CrossRef]
  109. Zeng, J.; Deng, Z.; Zou, Y.; Liu, C.; Fu, H.; Gu, Y.; Chang, H. Theaflavin alleviates oxidative injury and atherosclerosis progress via activating microRNA-24-mediated Nrf2/HO-1 signal. Phytother. Res. 2021, 35, 3418–3427. [Google Scholar] [CrossRef]
  110. Chen, D.; Wu, Z.; Wu, L.N.; Jiang, J.; Hu, G.N. Theaflavin Attenuates TBHP-Induced Endothelial Cells Oxidative Stress by Activating PI3K/AKT/Nrf2 and Accelerates Wound Healing in Rats. Front. Bioeng. Biotechnol. 2022, 10, 830574. [Google Scholar] [CrossRef]
  111. Yan, C.; Zhang, X.; Miao, J.; Yuan, H.; Liu, E.; Liang, T.; Li, Q. Farrerol Directly Targets GSK-3beta to Activate Nrf2-ARE Pathway and Protect EA.hy926 Cells against Oxidative Stress-Induced Injuries. Oxid. Med. Cell. Longev. 2020, 2020, 5967434. [Google Scholar] [CrossRef] [PubMed]
  112. Ren, Y.; Liu, Y.; Liu, K.; Shu, Z.; Lv, T.; Chen, Z.; Feng, W.; Zhang, Y. A transcriptomics and molecular biology based investigation reveals the protective effect and mechanism of carnosol on t-BHP induced HRMECs via Nrf2 signaling pathway. Eur. J. Pharmacol. 2022, 923, 174933. [Google Scholar] [CrossRef] [PubMed]
  113. Bao, X.Y.; Deng, L.H.; Huang, Z.J.; Daror, A.S.; Wang, Z.H.; Jin, W.J.; Zhuang, Z.; Tong, Q.; Zheng, G.Q.; Wang, Y. Buyang Huanwu Decoction Enhances Revascularization via Akt/GSK3beta/NRF2 Pathway in Diabetic Hindlimb Ischemia. Oxid. Med. Cell. Longev. 2021, 2021, 1470829. [Google Scholar] [CrossRef]
  114. Senthil, K.K.J.; Gokila, V.M.; Wang, S.Y. Activation of Nrf2-mediated anti-oxidant genes by antrodin C prevents hyperglycemia-induced senescence and apoptosis in human endothelial cells. Oncotarget 2017, 8, 96568–96587. [Google Scholar] [CrossRef] [PubMed]
  115. Li, C.L.; Liu, X.H.; Qiao, Y.; Ning, L.N.; Li, W.J.; Sun, Y.S.; Liu, D.S.; Gao, W.; Ma, C.M. Allicin alleviates inflammation of diabetic macroangiopathy via the Nrf2 and NF-kB pathway. Eur. J. Pharmacol. 2020, 876, 173052. [Google Scholar] [CrossRef]
  116. Chen, J.; Tan, L.; Li, C.; Zhou, C.; Hong, P.; Sun, S.; Qian, Z.J. Mechanism Analysis of a Novel Angiotensin-I-Converting Enzyme Inhibitory Peptide from Isochrysis zhanjiangensis Microalgae for Suppressing Vascular Injury in Human Umbilical Vein Endothelial Cells. J. Agric. Food Chem. 2020, 68, 4411–4423. [Google Scholar] [CrossRef] [PubMed]
  117. Li, J.; Teng, X.; Jin, S.; Dong, J.; Guo, Q.; Tian, D.; Wu, Y. Hydrogen sulfide improves endothelial dysfunction by inhibiting the vicious cycle of NLRP3 inflammasome and oxidative stress in spontaneously hypertensive rats. J. Hypertens. 2019, 37, 1633–1643. [Google Scholar] [CrossRef]
  118. Fratantonio, D.; Speciale, A.; Ferrari, D.; Cristani, M.; Saija, A.; Cimino, F. Palmitate-induced endothelial dysfunction is attenuated by cyanidin-3-O-glucoside through modulation of Nrf2/Bach1 and NF-kappaB pathways. Toxicol. Lett. 2015, 239, 152–160. [Google Scholar] [CrossRef]
  119. Hu, S.; Wu, Y.; Zhao, B.; Hu, H.; Zhu, B.; Sun, Z.; Li, P.; Du, S. Panax notoginseng Saponins Protect Cerebral Microvascular Endothelial Cells against Oxygen-Glucose Deprivation/Reperfusion-Induced Barrier Dysfunction via Activation of PI3K/Akt/Nrf2 Antioxidant Signaling Pathway. Molecules 2018, 23, 2781. [Google Scholar] [CrossRef]
  120. Wang, X.; Chen, L.; Wang, T.; Jiang, X.; Zhang, H.; Li, P.; Lv, B.; Gao, X. Ginsenoside Rg3 antagonizes adriamycin-induced cardiotoxicity by improving endothelial dysfunction from oxidative stress via upregulating the Nrf2-ARE pathway through the activation of akt. Phytomedicine 2015, 22, 875–884. [Google Scholar] [CrossRef]
  121. Liu, L.; Wang, R.; Xu, R.; Chu, Y.; Gu, W. Procyanidin B2 ameliorates endothelial dysfunction and impaired angiogenesis via the Nrf2/PPARgamma/sFlt-1 axis in preeclampsia. Pharmacol. Res. 2022, 177, 106127. [Google Scholar] [CrossRef] [PubMed]
  122. Saji, N.; Francis, N.; Blanchard, C.L.; Schwarz, L.J.; Santhakumar, A.B. Rice Bran Phenolic Compounds Regulate Genes Associated with Antioxidant and Anti-Inflammatory Activity in Human Umbilical Vein Endothelial Cells with Induced Oxidative Stress. Int. J. Mol. Sci. 2019, 20, 4715. [Google Scholar] [CrossRef] [PubMed]
  123. Hada, Y.; Uchida, H.A.; Otaka, N.; Onishi, Y.; Okamoto, S.; Nishiwaki, M.; Takemoto, R.; Takeuchi, H.; Wada, J. The Protective Effect of Chlorogenic Acid on Vascular Senescence via the Nrf2/HO-1 Pathway. Int. J. Mol. Sci. 2020, 21, 4527. [Google Scholar] [CrossRef]
  124. Chen, Z.W.; Miu, H.F.; Wang, H.P.; Wu, Z.N.; Wang, W.J.; Ling, Y.J.; Xu, X.H.; Sun, H.J.; Jiang, X. Pterostilbene protects against uraemia serum-induced endothelial cell damage via activation of Keap1/Nrf2/HO-1 signaling. Int. Urol. Nephrol. 2018, 50, 559–570. [Google Scholar] [CrossRef] [PubMed]
  125. Yang, S.; Zheng, Y.; Hou, X. Lipoxin A4 restores oxidative stress-induced vascular endothelial cell injury and thrombosis-related factor expression by its receptor-mediated activation of Nrf2-HO-1 axis. Cell. Signal. 2019, 60, 146–153. [Google Scholar] [CrossRef] [PubMed]
  126. Ling, K.; Xu, A.; Chen, Y.; Chen, X.; Li, Y.; Wang, W. Protective effect of a hydrogen sulfide donor on balloon injury-induced restenosis via the Nrf2/HIF-1alpha signaling pathway. Int. J. Mol. Med. 2019, 43, 1299–1310. [Google Scholar] [CrossRef]
  127. Jiang, J.; Dong, C.; Zhai, L.; Lou, J.; Jin, J.; Cheng, S.; Chen, Z.; Guo, X.; Lin, D.; Ding, J.; et al. Paeoniflorin Suppresses TBHP-Induced Oxidative Stress and Apoptosis in Human Umbilical Vein Endothelial Cells via the Nrf2/HO-1 Signaling Pathway and Improves Skin Flap Survival. Front. Pharmacol. 2021, 12, 735530. [Google Scholar] [CrossRef]
  128. Zhang, Q.; Xue, T.; Guan, J.; Wang, W.; Shi, J.; Lu, J.; Jiang, X. Irigenin alleviates angiotensin II-induced oxidative stress and apoptosis in HUVEC cells by activating Nrf2 pathway. Drug Dev. Res. 2021, 82, 999–1007. [Google Scholar] [CrossRef]
  129. Kim, N.Y.; Trinh, N.T.; Ahn, S.G.; Kim, S.A. Cinnamaldehyde protects against oxidative stress and inhibits the TNFalphainduced inflammatory response in human umbilical vein endothelial cells. Int. J. Mol. Med. 2020, 46, 449–457. [Google Scholar] [CrossRef]
  130. Priestley, J.R.C.; Fink, K.E.; McCord, J.M.; Lombard, J.H. NRF2 activation with Protandim attenuates salt-induced vascular dysfunction and microvascular rarefaction. Microcirculation 2019, 26, e12575. [Google Scholar] [CrossRef]
  131. Gao, F.; Li, J.M.; Xi, C.; Li, H.H.; Liu, Y.L.; Wang, Y.P.; Xuan, L.J. Magnesium lithospermate B protects the endothelium from inflammation-induced dysfunction through activation of Nrf2 pathway. Acta Pharmacol. Sin. 2019, 40, 867–878. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, J.S.; Huang, P.H.; Wang, C.H.; Lin, F.Y.; Tsai, H.Y.; Wu, T.C.; Lin, S.J.; Chen, J.W. Nrf-2 mediated heme oxygenase-1 expression, an antioxidant-independent mechanism, contributes to anti-atherogenesis and vascular protective effects of Ginkgo biloba extract. Atherosclerosis 2011, 214, 301–309. [Google Scholar] [CrossRef] [PubMed]
  133. Yuan, W.; Chang, H.; Liu, X.; Wang, S.; Liu, H.; Xuan, H. Brazilian Green Propolis Inhibits Ox-LDL-Stimulated Oxidative Stress in Human Umbilical Vein Endothelial Cells Partly through PI3K/Akt/mTOR-Mediated Nrf2/HO-1 Pathway. Evid.-Based. Complement. Altern. Med. 2019, 2019, 5789574. [Google Scholar] [CrossRef] [PubMed]
  134. Jarisarapurin, W.; Sanrattana, W.; Chularojmontri, L.; Kunchana, K.; Wattanapitayakul, S.K. Antioxidant Properties of Unripe Carica papaya Fruit Extract and Its Protective Effects against Endothelial Oxidative Stress. Evid.-Based. Complement. Altern. Med. 2019, 2019, 4912631. [Google Scholar] [CrossRef]
  135. Sung, L.C.; Chao, H.H.; Chen, C.H.; Tsai, J.C.; Liu, J.C.; Hong, H.J.; Cheng, T.H.; Chen, J.J. Lycopene inhibits cyclic strain-induced endothelin-1 expression through the suppression of reactive oxygen species generation and induction of heme oxygenase-1 in human umbilical vein endothelial cells. Clin. Exp. Pharmacol. Physiol. 2015, 42, 632–639. [Google Scholar] [CrossRef]
  136. Oh, Y.; Ahn, C.B.; Je, J.Y. Cytoprotective Role of Edible Seahorse (Hippocampus abdominalis)-Derived Peptides in H2O2-Induced Oxidative Stress in Human Umbilical Vein Endothelial Cells. Mar. Drugs 2021, 19, 86. [Google Scholar] [CrossRef]
  137. Huang, M.Z.; Yang, Y.J.; Liu, X.W.; Qin, Z.; Li, J.Y. Aspirin eugenol ester attenuates oxidative injury of vascular endothelial cells by regulating NOS and Nrf2 signalling pathways. Br. J. Pharmacol. 2019, 176, 906–918. [Google Scholar] [CrossRef]
  138. Zhang, J.; Cai, W.; Fan, Z.; Yang, C.; Wang, W.; Xiong, M.; Ma, C.; Yang, J. MicroRNA-24 inhibits the oxidative stress induced by vascular injury by activating the Nrf2/Ho-1 signaling pathway. Atherosclerosis 2019, 290, 9–18. [Google Scholar] [CrossRef]
  139. Li, Y.; Zhu, X.; Liu, X.; Du, A.; Yu, B. miR-200a mediates protection of thymosin beta-4 in cardiac microvascular endothelial cells as a novel mechanism under hypoxia-reoxygenation injury. J. Cell. Biochem. 2019, 120, 19098–19106. [Google Scholar] [CrossRef]
  140. Liu, Q.Q.; Ren, K.; Liu, S.H.; Li, W.M.; Huang, C.J.; Yang, X.H. MicroRNA-140-5p aggravates hypertension and oxidative stress of atherosclerosis via targeting Nrf2 and Sirt2. Int. J. Mol. Med. 2019, 43, 839–849. [Google Scholar] [CrossRef] [Green Version]
  141. Pan, H.; Xue, C.; Auerbach, B.J.; Fan, J.; Bashore, A.C.; Cui, J.; Yang, D.Y.; Trignano, S.B.; Liu, W.; Shi, J.; et al. Single-Cell Genomics Reveals a Novel Cell State During Smooth Muscle Cell Phenotypic Switching and Potential Therapeutic Targets for Atherosclerosis in Mouse and Human. Circulation 2020, 142, 2060–2075. [Google Scholar] [CrossRef] [PubMed]
  142. Herring, B.P.; Hoggatt, A.M.; Burlak, C.; Offermanns, S. Previously differentiated medial vascular smooth muscle cells contribute to neointima formation following vascular injury. Vasc. Cell. 2014, 6, 21. [Google Scholar] [CrossRef] [PubMed]
  143. Li, H.; Zhuang, W.; Xiong, T.; Park, W.S.; Zhang, S.; Zha, Y.; Yao, J.; Wang, F.; Yang, Y.; Chen, Y.; et al. Nrf2 deficiency attenuates atherosclerosis by reducing LOX-1-mediated proliferation and migration of vascular smooth muscle cells. Atherosclerosis 2022, 347, 1–16. [Google Scholar] [CrossRef] [PubMed]
  144. Wu, W.; Wang, C.; Zang, H.; Qi, L.; Azhar, M.; Nagarkatti, M.; Nagarkatti, P.; Cai, G.; Weiser-Evans, M.C.M.; Cui, T. Mature Vascular Smooth Muscle Cells, but Not Endothelial Cells, Serve as the Major Cellular Source of Intimal Hyperplasia in Vein Grafts. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1870–1890. [Google Scholar] [CrossRef]
  145. Miano, J.M.; Fisher, E.A.; Majesky, M.W. Fate and State of Vascular Smooth Muscle Cells in Atherosclerosis. Circulation 2021, 143, 2110–2116. [Google Scholar] [CrossRef] [PubMed]
  146. He, X.; Deng, J.; Yu, X.J.; Yang, S.; Yang, Y.; Zang, W.J. Activation of M3AChR (Type 3 Muscarinic Acetylcholine Receptor) and Nrf2 (Nuclear Factor Erythroid 2-Related Factor 2) Signaling by Choline Alleviates Vascular Smooth Muscle Cell Phenotypic Switching and Vascular Remodeling. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2649–2664. [Google Scholar] [CrossRef] [PubMed]
  147. Xu, M.; Li, X.X.; Wang, L.; Wang, M.; Zhang, Y.; Li, P.L. Contribution of Nrf2 to Atherogenic Phenotype Switching of Coronary Arterial Smooth Muscle Cells Lacking CD38 Gene. Cell. Physiol. Biochem. 2015, 37, 432–444. [Google Scholar] [CrossRef]
  148. Ashino, T.; Yamamoto, M.; Yoshida, T.; Numazawa, S. Redox-sensitive transcription factor Nrf2 regulates vascular smooth muscle cell migration and neointimal hyperplasia. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 760–768. [Google Scholar] [CrossRef]
  149. Chen, C.; Ma, J.; Xu, Z.; Chen, L.; Sun, B.; Shi, Y.; Miao, Y.; Wu, T.; Qin, M.; Zhang, Y.; et al. Rosmarinic Acid Inhibits Platelet Aggregation and Neointimal Hyperplasia In Vivo and Vascular Smooth Muscle Cell Dedifferentiation, Proliferation, and Migration In Vitro via Activation of the Keap1-Nrf2-ARE Antioxidant System. J. Agric. Food Chem. 2022, 70, 7420–7440. [Google Scholar] [CrossRef]
  150. Nie, X.; Shen, C.; Tan, J.; Yang, X.; Wang, W.; Dai, Y.; Sun, H.; Wu, Z.; Chen, J. Andrographolide Attenuates Established Pulmonary Hypertension via Rescue of Vascular Remodeling. Biomolecules 2021, 11, 1801. [Google Scholar] [CrossRef]
  151. Hwang, S.M.; Lee, Y.J.; Lee, Y.P.; Yoon, J.J.; Lee, S.M.; Cha, J.D.; Choi, K.M.; Kang, D.G.; Lee, H.S. Anti-Proliferative Effect of an Aqueous Extract of Prunella vulgaris in Vascular Smooth Muscle Cells. Evid.-Based. Complement. Altern. Med. 2013, 2013, 936463. [Google Scholar] [CrossRef] [PubMed]
  152. Buglak, N.E.; Jiang, W.; Bahnson, E.S.M. Cinnamic aldehyde inhibits vascular smooth muscle cell proliferation and neointimal hyperplasia in Zucker Diabetic Fatty rats. Redox Biol. 2018, 19, 166–178. [Google Scholar] [CrossRef] [PubMed]
  153. Zhang, M.; Xu, Y.; Qiu, Z.; Jiang, L. Sulforaphane Attenuates Angiotensin II-Induced Vascular Smooth Muscle Cell Migration via Suppression of NOX4/ROS/Nrf2 Signaling. Int. J. Biol. Sci. 2019, 15, 148–157. [Google Scholar] [CrossRef] [PubMed]
  154. Zghonda, N.; Yoshida, S.; Araki, M.; Kusunoki, M.; Mliki, A.; Ghorbel, A.; Miyazaki, H. Greater effectiveness of epsilon-viniferin in red wine than its monomer resveratrol for inhibiting vascular smooth muscle cell proliferation and migration. Biosci. Biotechnol. Biochem. 2011, 75, 1259–1267. [Google Scholar] [CrossRef]
  155. Kim, J.W.; Lim, S.C.; Lee, M.Y.; Lee, J.W.; Oh, W.K.; Kim, S.K.; Kang, K.W. Inhibition of neointimal formation by trans-resveratrol: Role of phosphatidyl inositol 3-kinase-dependent Nrf2 activation in heme oxygenase-1 induction. Mol. Nutr. Food Res. 2010, 54, 1497–1505. [Google Scholar] [CrossRef] [PubMed]
  156. Jiang, H.; Zhao, Y.; Feng, P.; Liu, Y. Sulfiredoxin-1 Inhibits PDGF-BB-Induced Vascular Smooth Muscle Cell Proliferation and Migration by Enhancing the Activation of Nrf2/ARE Signaling. Int. Heart J. 2022, 63, 113–121. [Google Scholar] [CrossRef]
  157. Qiu, L.; Hu, L.; Liu, X.; Li, W.; Zhang, X.; Xia, H.; Zhang, C. Physalin B inhibits PDGF-BB-induced VSMC proliferation, migration and phenotypic transformation by activating the Nrf2 pathway. Food Funct. 2021, 12, 10950–10966. [Google Scholar] [CrossRef]
  158. Choi, S.H.; Park, S.; Oh, C.J.; Leem, J.; Park, K.G.; Lee, I.K. Dipeptidyl peptidase-4 inhibition by gemigliptin prevents abnormal vascular remodeling via NF-E2-related factor 2 activation. Vascul. Pharmacol. 2015, 73, 11–19. [Google Scholar] [CrossRef]
  159. Kim, N.; Hwangbo, C.; Lee, S.; Lee, J.H. Eupatolide inhibits PDGF-induced proliferation and migration of aortic smooth muscle cells through ROS-dependent heme oxygenase-1 induction. Phytother. Res. 2013, 27, 1700–1707. [Google Scholar] [CrossRef]
  160. Lee, S.; Seo, J.; Ryoo, S.; Cuong, T.D.; Min, B.S.; Lee, J.H. Malabaricone C inhibits PDGF-induced proliferation and migration of aortic smooth muscle cells through induction of heme oxygenase-1. J. Cell. Biochem. 2012, 113, 2866–2876. [Google Scholar] [CrossRef]
  161. Huang, J.; Zhang, H.; You, L.; Zhang, J.; Jiang, Z. Coenzyme Q10 inhibits intracranial aneurysm formation and progression in a mouse model. Pediatr. Res. 2022, 91, 839–845. [Google Scholar] [CrossRef] [PubMed]
  162. Behnammanesh, G.; Durante, G.L.; Khanna, Y.P.; Peyton, K.J.; Durante, W. Canagliflozin inhibits vascular smooth muscle cell proliferation and migration: Role of heme oxygenase-1. Redox. Biol. 2020, 32, 101527. [Google Scholar] [CrossRef] [PubMed]
  163. Hwang, A.R.; Han, J.H.; Lim, J.H.; Kang, Y.J.; Woo, C.H. Fluvastatin inhibits AGE-induced cell proliferation and migration via an ERK5-dependent Nrf2 pathway in vascular smooth muscle cells. PLoS ONE 2017, 12, e0178278. [Google Scholar] [CrossRef] [PubMed]
  164. Aghagolzadeh, P.; Radpour, R.; Bachtler, M.; van Goor, H.; Smith, E.R.; Lister, A.; Odermatt, A.; Feelisch, M.; Pasch, A. Hydrogen sulfide attenuates calcification of vascular smooth muscle cells via KEAP1/NRF2/NQO1 activation. Atherosclerosis 2017, 265, 78–86. [Google Scholar] [CrossRef]
  165. Ma, W.Q.; Sun, X.J.; Zhu, Y.; Liu, N.F. Metformin attenuates hyperlipidaemia-associated vascular calcification through anti-ferroptotic effects. Free Radic. Biol. Med. 2021, 165, 229–242. [Google Scholar] [CrossRef]
  166. Cui, L.; Zhou, Q.; Zheng, X.; Sun, B.; Zhao, S. Mitoquinone attenuates vascular calcification by suppressing oxidative stress and reducing apoptosis of vascular smooth muscle cells via the Keap1/Nrf2 pathway. Free Radic. Biol. Med. 2020, 161, 23–31. [Google Scholar] [CrossRef]
  167. Chen, Y.; Huang, C.; Zhu, S.Y.; Zou, H.C.; Xu, C.Y.; Chen, Y.X. Overexpression of HOTAIR attenuates Pi-induced vascular calcification by inhibiting Wnt/beta-catenin through regulating miR-126/Klotho/SIRT1 axis. Mol. Cell. Biochem. 2021, 476, 3551–3561. [Google Scholar] [CrossRef]
  168. Ji, R.; Sun, H.; Peng, J.; Ma, X.; Bao, L.; Fu, Y.; Zhang, X.; Luo, C.; Gao, C.; Jin, Y.; et al. Rosmarinic acid exerts an antagonistic effect on vascular calcification by regulating the Nrf2 signalling pathway. Free Radic. Res. 2019, 53, 187–197. [Google Scholar] [CrossRef]
  169. Kim, J.Y.; Cho, H.J.; Sir, J.J.; Kim, B.K.; Hur, J.; Youn, S.W.; Yang, H.M.; Jun, S.I.; Park, K.W.; Hwang, S.J.; et al. Sulfasalazine induces haem oxygenase-1 via ROS-dependent Nrf2 signalling, leading to control of neointimal hyperplasia. Cardiovasc. Res. 2009, 82, 550–560. [Google Scholar] [CrossRef]
  170. Durham, A.L.; Speer, M.Y.; Scatena, M.; Giachelli, C.M.; Shanahan, C.M. Role of smooth muscle cells in vascular calcification: Implications in atherosclerosis and arterial stiffness. Cardiovasc. Res. 2018, 114, 590–600. [Google Scholar] [CrossRef]
  171. Jin, D.; Lin, L.; Xie, Y.; Jia, M.; Qiu, H.; Xun, K. NRF2-suppressed vascular calcification by regulating the antioxidant pathway in chronic kidney disease. FASEB J. 2022, 36, e22098. [Google Scholar] [CrossRef] [PubMed]
  172. Wei, R.; Enaka, M.; Muragaki, Y. Activation of KEAP1/NRF2/P62 signaling alleviates high phosphate-induced calcification of vascular smooth muscle cells by suppressing reactive oxygen species production. Sci. Rep. 2019, 9, 10366. [Google Scholar] [CrossRef] [PubMed]
  173. Xu, T.H.; Du, Y.; Sheng, Z.; Li, Y.; Qiu, X.; Tian, B.; Yao, L. OGT-Mediated KEAP1 Glycosylation Accelerates NRF2 Degradation Leading to High Phosphate-Induced Vascular Calcification in Chronic Kidney Disease. Front. Physiol. 2020, 11, 1092. [Google Scholar] [CrossRef] [PubMed]
  174. Girona, J.; Rosales, R.; Saavedra, P.; Masana, L.; Vallve, J.C. Palmitate decreases migration and proliferation and increases oxidative stress and inflammation in smooth muscle cells: Role of the Nrf2 signaling pathway. Am. J. Physiol. Cell. Physiol. 2019, 316, C888–C897. [Google Scholar] [CrossRef] [PubMed]
  175. Alonso-Pineiro, J.A.; Gonzalez-Rovira, A.; Sanchez-Gomar, I.; Moreno, J.A.; Duran-Ruiz, M.C. Nrf2 and Heme Oxygenase-1 Involvement in Atherosclerosis Related Oxidative Stress. Antioxidants 2021, 10, 1463. [Google Scholar] [CrossRef]
  176. Ishii, T.; Itoh, K.; Ruiz, E.; Leake, D.S.; Unoki, H.; Yamamoto, M.; Mann, G.E. Role of Nrf2 in the regulation of CD36 and stress protein expression in murine macrophages: Activation by oxidatively modified LDL and 4-hydroxynonenal. Circ. Res. 2004, 94, 609–616. [Google Scholar] [CrossRef]
  177. Sussan, T.E.; Jun, J.; Thimmulappa, R.; Bedja, D.; Antero, M.; Gabrielson, K.L.; Polotsky, V.Y.; Biswal, S. Disruption of Nrf2, a key inducer of antioxidant defenses, attenuates ApoE-mediated atherosclerosis in mice. PLoS ONE 2008, 3, e3791. [Google Scholar] [CrossRef] [PubMed]
  178. Barajas, B.; Che, N.; Yin, F.; Rowshanrad, A.; Orozco, L.D.; Gong, K.W.; Wang, X.; Castellani, L.W.; Reue, K.; Lusis, A.J.; et al. NF-E2-related factor 2 promotes atherosclerosis by effects on plasma lipoproteins and cholesterol transport that overshadow antioxidant protection. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 58–66. [Google Scholar] [CrossRef]
  179. Freigang, S.; Ampenberger, F.; Spohn, G.; Heer, S.; Shamshiev, A.T.; Kisielow, J.; Hersberger, M.; Yamamoto, M.; Bachmann, M.F.; Kopf, M. Nrf2 is essential for cholesterol crystal-induced inflammasome activation and exacerbation of atherosclerosis. Eur. J. Immunol. 2011, 41, 2040–2051. [Google Scholar] [CrossRef]
  180. Harada, N.; Ito, K.; Hosoya, T.; Mimura, J.; Maruyama, A.; Noguchi, N.; Yagami, K.; Morito, N.; Takahashi, S.; Maher, J.M.; et al. Nrf2 in bone marrow-derived cells positively contributes to the advanced stage of atherosclerotic plaque formation. Free Radic. Biol. Med. 2012, 53, 2256–2262. [Google Scholar] [CrossRef]
  181. Liu, J.; Yang, B.; Wang, Y.; Wu, Y.; Fan, B.; Zhu, S.; Song, E.; Song, Y. Polychlorinated biphenyl quinone promotes macrophage polarization to CD163(+) cells through Nrf2 signaling pathway. Environ. Pollut. 2020, 257, 113587. [Google Scholar] [CrossRef] [PubMed]
  182. Bozaykut, P.; Karademir, B.; Yazgan, B.; Sozen, E.; Siow, R.C.; Mann, G.E.; Ozer, N.K. Effects of vitamin E on peroxisome proliferator-activated receptor gamma and nuclear factor-erythroid 2-related factor 2 in hypercholesterolemia-induced atherosclerosis. Free Radic. Biol. Med. 2014, 70, 174–181. [Google Scholar] [CrossRef] [PubMed]
  183. Ruotsalainen, A.K.; Lappalainen, J.P.; Heiskanen, E.; Merentie, M.; Sihvola, V.; Napankangas, J.; Lottonen-Raikaslehto, L.; Kansanen, E.; Adinolfi, S.; Kaarniranta, K.; et al. Nuclear factor E2-related factor 2 deficiency impairs atherosclerotic lesion development but promotes features of plaque instability in hypercholesterolaemic mice. Cardiovasc. Res. 2019, 115, 243–254. [Google Scholar] [CrossRef]
  184. McCormick, M.L.; Gavrila, D.; Weintraub, N.L. Role of oxidative stress in the pathogenesis of abdominal aortic aneurysms. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 461–469. [Google Scholar] [CrossRef] [PubMed]
  185. Jreije, A.; Medlej-Hashim, M.; Hajal, J.; Saliba, Y.; Chacar, S.; Fares, N.; Khouzami, L. Calcitriol Supplementation Protects Against Apoptosis and Alleviates the Severity of Abdominal Aortic Aneurysm Induced by Angiotensin II and Anti-TGFβ. J. Cardiovasc. Transl. Res. 2022; ahead of print. [Google Scholar] [CrossRef]
  186. Qiu, R.; Chen, S.; Hua, F.; Bian, S.; Chen, J.; Li, G.; Wu, X. Betanin Prevents Experimental Abdominal Aortic Aneurysm Progression by Modulating the TLR4/NF-kappaB and Nrf2/HO-1 Pathways. Biol. Pharm. Bull. 2021, 44, 1254–1262. [Google Scholar] [CrossRef] [PubMed]
  187. Song, H.; Xu, T.; Feng, X.; Lai, Y.; Yang, Y.; Zheng, H.; He, X.; Wei, G.; Liao, W.; Liao, Y.; et al. Itaconate prevents abdominal aortic aneurysm formation through inhibiting inflammation via activation of Nrf2. EBioMedicine 2020, 57, 102832. [Google Scholar] [CrossRef] [PubMed]
  188. Xiao, T.; Zhang, L.; Huang, Y.; Shi, Y.; Wang, J.; Ji, Q.; Ye, J.; Lin, Y.; Liu, H. Sestrin2 increases in aortas and plasma from aortic dissection patients and alleviates angiotensin II-induced smooth muscle cell apoptosis via the Nrf2 pathway. Life Sci. 2019, 218, 132–138. [Google Scholar] [CrossRef] [PubMed]
  189. Hsu, C.Y.; Vo, T.T.T.; Lee, C.W.; Chen, Y.L.; Lin, W.N.; Cheng, H.C.; Vo, Q.C.; Lee, I.T. Carbon monoxide releasing molecule-2 attenuates angiotensin II-induced IL-6/Jak2/Stat3-associated inflammation by inhibiting NADPH oxidase- and mitochondria-derived ROS in human aortic smooth muscle cells. Biochem. Pharmacol. 2022, 198, 114978. [Google Scholar] [CrossRef]
  190. Piechota-Polanczyk, A.; Kopacz, A.; Kloska, D.; Zagrapan, B.; Neumayer, C.; Grochot-Przeczek, A.; Huk, I.; Brostjan, C.; Dulak, J.; Jozkowicz, A. Simvastatin Treatment Upregulates HO-1 in Patients with Abdominal Aortic Aneurysm but Independently of Nrf2. Oxid. Med. Cell. Longev. 2018, 2018, 2028936. [Google Scholar] [CrossRef] [Green Version]
  191. Kopacz, A.; Werner, E.; Grochot-Przeczek, A.; Kloska, D.; Hajduk, K.; Neumayer, C.; Jozkowicz, A.; Piechota-Polanczyk, A. Simvastatin Attenuates Abdominal Aortic Aneurysm Formation Favoured by Lack of Nrf2 Transcriptional Activity. Oxid. Med. Cell. Longev. 2020, 2020, 6340190. [Google Scholar] [CrossRef]
  192. Pan, B.; Zhang, H.; Cui, T.; Wang, X. TFEB activation protects against cardiac proteotoxicity via increasing autophagic flux. J. Mol. Cell. Cardiol. 2017, 113, 51–62. [Google Scholar] [CrossRef] [PubMed]
  193. Dai, S.; Wang, B.; Li, W.; Wang, L.; Song, X.; Guo, C.; Li, Y.; Liu, F.; Zhu, F.; Wang, Q.; et al. Systemic application of 3-methyladenine markedly inhibited atherosclerotic lesion in ApoE−/− mice by modulating autophagy, foam cell formation and immune-negative molecules. Cell Death Dis. 2016, 7, e2498. [Google Scholar] [CrossRef] [PubMed]
  194. Tian, J.; Popal, M.S.; Liu, Y.; Gao, R.; Lyu, S.; Chen, K.; Liu, Y. Ginkgo Biloba Leaf Extract Attenuates Atherosclerosis in Streptozotocin-Induced Diabetic ApoE−/− Mice by Inhibiting Endoplasmic Reticulum Stress via Restoration of Autophagy through the mTOR Signaling Pathway. Oxid. Med. Cell. Longev. 2019, 2019, 8134678. [Google Scholar] [CrossRef] [PubMed]
  195. de Zeeuw, D.; Akizawa, T.; Audhya, P.; Bakris, G.L.; Chin, M.; Christ-Schmidt, H.; Goldsberry, A.; Houser, M.; Krauth, M.; Lambers Heerspink, H.J.; et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N. Engl. J. Med. 2013, 369, 2492–2503. [Google Scholar] [CrossRef] [PubMed]
  196. Mitsuishi, Y.; Taguchi, K.; Kawatani, Y.; Shibata, T.; Nukiwa, T.; Aburatani, H.; Yamamoto, M.; Motohashi, H. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 2012, 22, 66–79. [Google Scholar] [CrossRef] [PubMed]
  197. Robledinos-Anton, N.; Fernandez-Gines, R.; Manda, G.; Cuadrado, A. Activators and Inhibitors of NRF2: A Review of Their Potential for Clinical Development. Oxid. Med. Cell. Longev. 2019, 2019, 9372182. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Detailed schematic diagram of Nrf2-related signaling pathway. Structural characteristics of Nrf2 (A) and Keap1 (B). The spatial patterns of interaction between Nrf2 and Keap1 under physiological conditions (C) and stressful conditions (D). The degradation of Nrf2 depends on different proteins under different conditions. Under physiological conditions, it depends on Keap1 (C); under stressful conditions, it depends on GSK-3 (E). Ub, ubiquitin; P, Phosphorylation; SUMO, small ubiquitin-like modifiers, SUMOylation; Ac, acetylation.
Figure 1. Detailed schematic diagram of Nrf2-related signaling pathway. Structural characteristics of Nrf2 (A) and Keap1 (B). The spatial patterns of interaction between Nrf2 and Keap1 under physiological conditions (C) and stressful conditions (D). The degradation of Nrf2 depends on different proteins under different conditions. Under physiological conditions, it depends on Keap1 (C); under stressful conditions, it depends on GSK-3 (E). Ub, ubiquitin; P, Phosphorylation; SUMO, small ubiquitin-like modifiers, SUMOylation; Ac, acetylation.
Cells 11 03042 g001
Table 2. Therapeutic targeting Nrf2 signaling for VSMC protection.
Table 2. Therapeutic targeting Nrf2 signaling for VSMC protection.
ApproachNrf2 SignalingFindingReference
CompoundsAtherosclerosis (AS) Related
Salvianolic acid BNrf2/HO-1Inhibits PDGF-induced proliferation and migration of VSMCs
Activates Nrf2-mediated HO-1 expression and suppression of PDGF-induced neointimal hyperplasia in cultured arterial rings isolated from mice ex vivo
[103]
EupatolideNrf2/HO-1Inhibits PDGF-induced proliferation and migration of aortic SMCs in vitro[159]
CompoundsDiabetes Related
Aqueous extract of Prunella vulgarisNrf2/HO-1Exhibits inhibitory effects on high glucose-stimulated VSMCs proliferation and migration, and invasion activities in vitro[151]
Cinnamic aldehydeNrf2Inhibits neointimal hyperplasia after carotid artery balloon injury in the Zucker Diabetic Fatty (ZDF) rats and inhibits proliferation of ZDF VSMCs in vitro[152]
CanagliflozinNrf2/HO-1Stimulates HO-1 expression in mice and human VSMCs through the ROS-Nrf2 pathway, and inhibits VSMCs proliferation and migration in vitro[162]
FluvastatinERK5/Nrf2Activates ERK5-dependent Nrf2 pathway and inhibits cellular proliferation and migration in VSMCs in vitro[163]
CompoundsVascular Calcification Related
Hydrogen sulfideKeap1/Nrf2/NQO1Attenuates circulating calciprotein particles (CPP)-induced VSMCs calcification in vitro[164]
MetforminNrf2Inhibits hyperlipidemia-associated calcium deposition in the rat aortic tissue of hyperlipidemia-related vascular calcification model in vivo and attenuates ferroptosis with increased calcium deposition in VSMCs with PA treatment[165]
MitoquinoneKeap1/Nrf2Attenuates vascular calcification by suppressing oxidative stress and reducing apoptosis in adenine-induced calcification in rats and inorganic phosphate-induced calcification in VSMCs in vitro[166]
miR-126Sirt1/Nrf2Attenuates calcification, in human aortic smooth muscle cells (HASMCs) in vitro and in a mouse calcification model in vivo [167]
Rosmarinic acidKeap1/Nrf2/AREInhibits VSMCs proliferation, migration, and calcification in a rat model of vascular calcification model induced by high-fat diet and vitamin D3 injection as well as β-glyerophosphate-induced calcification in rat aortic SMCs in vitro[168]
CompoundsVascular Occlusive Disease and Other Diseases Related
Rosmarinic acidKeap1/Nrf2/AREInhibits platelet aggregation and neointimal hyperplasia in vivo and VSMCs dedifferentiation, proliferation, and migration in vitro [149]
AndrographolideNOX/Nrf2Reverses pulmonary vascular remodeling through modulation of NOX/Nrf2-mediated oxidative stress and NF-κB-mediated inflammation in both chronic hypoxia and Sugen5416/hypoxia mouse pulmonary hypertension (PH) models and in cultured human PASMCs isolated from either healthy donors or PH patients[150]
SulforaphaneNOX4/ROS/Nrf2Attenuates Ang II-induced human VSMCs migration in vitro[153]
Trans-resveratrolNrf2/HO-1Suppresses intimal hyperplasia in a mouse model of wire-injured femoral artery injury by oral administration
Inhibits PDGF-stimulated DNA synthesis and cell proliferation in VSMCs in vitro
[155]
Sulfiredoxin-1Nrf2/AREInhibits PDGF-BB-induced VSMCs proliferation and migration in vitro[156]
Physalin BNrf2/HO-1Inhibits PDGF-BB-induced VSMCs proliferation, migration, and phenotypic transformation in vitro and prevents intimal hyperplasia in a mouse model of carotid artery injury induced by ligation[157]
GemigliptinNrf2/HO-1Exerts a preventative effect on ligation injury-induced neointimal hyperplasia in vivo and inhibits VSMCs proliferation and migration in vitro[158]
Malabaricone CNrf2/HO-1Inhibits PDGF-induced proliferation and migration of aortic SMCs in vitro[160]
Coenzyme Q10Nrf2/HO-1Attenuates intracranial aneurysm formation and rupture in mice and reduces H2O2-induced oxidative stress in VSMCs in vitro[161]
SulfasalazineNrf2/HO-1Suppresses VSMCs growth in vitro and prevents neointimal hyperplasia in rat carotid arteries induced by balloon in vivo[169]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, W.; Hendrix, A.; Nair, S.; Cui, T. Nrf2-Mediated Dichotomy in the Vascular System: Mechanistic and Therapeutic Perspective. Cells 2022, 11, 3042. https://doi.org/10.3390/cells11193042

AMA Style

Wu W, Hendrix A, Nair S, Cui T. Nrf2-Mediated Dichotomy in the Vascular System: Mechanistic and Therapeutic Perspective. Cells. 2022; 11(19):3042. https://doi.org/10.3390/cells11193042

Chicago/Turabian Style

Wu, Weiwei, Andrew Hendrix, Sharad Nair, and Taixing Cui. 2022. "Nrf2-Mediated Dichotomy in the Vascular System: Mechanistic and Therapeutic Perspective" Cells 11, no. 19: 3042. https://doi.org/10.3390/cells11193042

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