Next Article in Journal
Endothelial Notch Signaling Regulates the Function of the Retinal Pigment Epithelial Barrier via EC Angiocrine Signaling
Previous Article in Journal
From Stress to Sick(le) and Back Again–Oxidative/Antioxidant Mechanisms, Genetic Modulation, and Cerebrovascular Disease in Children with Sickle Cell Anemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unleashing the Potential of Nrf2: A Novel Therapeutic Target for Pulmonary Vascular Remodeling

1
Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
2
Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2023, 12(11), 1978; https://doi.org/10.3390/antiox12111978
Submission received: 28 September 2023 / Revised: 22 October 2023 / Accepted: 5 November 2023 / Published: 7 November 2023
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)

Abstract

:
Pulmonary vascular remodeling, characterized by the thickening of all three layers of the blood vessel wall, plays a central role in the pathogenesis of pulmonary hypertension (PH). Despite the approval of several drugs for PH treatment, their long-term therapeutic effect remains unsatisfactory, as they mainly focus on vasodilation rather than addressing vascular remodeling. Therefore, there is an urgent need for novel therapeutic targets in the treatment of PH. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a vital transcription factor that regulates endogenous antioxidant defense and emerges as a novel regulator of pulmonary vascular remodeling. Growing evidence has suggested an involvement of Nrf2 and its downstream transcriptional target in the process of pulmonary vascular remodeling. Pharmacologically targeting Nrf2 has demonstrated beneficial effects in various diseases, and several Nrf2 inducers are currently undergoing clinical trials. However, the exact potential and mechanism of Nrf2 as a therapeutic target in PH remain unknown. Thus, this review article aims to comprehensively explore the role and mechanism of Nrf2 in pulmonary vascular remodeling associated with PH. Additionally, we provide a summary of Nrf2 inducers that have shown therapeutic potential in addressing the underlying vascular remodeling processes in PH. Although Nrf2-related therapies hold great promise, further research is necessary before their clinical implementation can be fully realized.

1. Introduction

Pulmonary hypertension (PH) is a progressive and life-threatening disease, characterized by an excessive proliferation of pulmonary vascular cells, leading to pulmonary vascular resistance, oxidative stress, right ventricular hypertrophy (RVH), and eventually heart failure [1,2]. PH is defined by a pulmonary vascular resistance > 3 Wood units, pulmonary artery wedge pressure ≤ 15 mmHg, and an increase in mean pulmonary arterial pressure ≥ 20 mmHg at rest [3,4]. PH can be divided into five major categories: (1) pulmonary arterial hypertension (PAH), including heritable, idiopathic, and drug/toxin-induced PH; (2) PH due to interstitial lung diseases and/or hypoxia, such as chronic obstructive pulmonary disease and high-altitude; (3) PH induced by left heart disease; (4) chronic thromboembolic pulmonary hypertension; and (5) PH with unclear and/or multifactorial origin, including systemic and hematologic disorders [5,6]. PH most frequently occurs at a young age (30 to 60 years), with a morbidity of over 30–50 million individuals every year and a severely reduced life expectancy [7]. Disease progression is inevitable in most PH patients, and mortality remains unacceptably high despite appropriate treatment [8]. Upon diagnosis, the average life expectancy of PH patients is approximately 7–10 years [9].
Despite great progress having been made in the treatment of PH, no drugs have shown satisfactory efficacy so far [10]. Pharmacological agents currently authorized for the treatment of PH include soluble guanylate cyclase stimulators, endothelin receptor antagonists, phosphodiesterase inhibitors, and nitric oxide donors to prostacyclin analogs and prostacyclin receptor agonists. Even though these agents prompt pulmonary vasodilation and improve symptoms, they do not significantly improve long-term prognosis, primarily due to their inability to address the underlying pulmonary vascular remodeling (PVR) process [11,12]. As a result, novel therapies targeting the pathogenesis of PH are urgently warranted. Vascular remodeling, a central pathogenic process in all types of PH, is associated with the dysfunction of the endothelium, the proliferation and hypertrophy of vascular smooth muscle cells (VSMCs), and the accumulation of extracellular matrix [13,14,15]. These processes lead to neointima formation, medial hypertrophy, muscularization, and plexiform lesions’ development, contributing to the obliteration of precapillary pulmonary arteries and sustained elevation of pulmonary arterial pressure (PAH) [16,17]. Notably, the pathogenesis of PVR remains elusive. However, it is widely considered that oxidative stress is a crucial factor in PVR [18,19,20,21]. Oxidative stress induces several pathophysiological processes such as the dysfunction of pulmonary arterial endothelial cells (PAEC), excessive proliferation of pulmonary arterial smooth muscle cells (PASMC), extracellular matrix protein deposition, distal pulmonary arterioles muscularization, autoimmune processes, and inflammation [22,23], all of which are involved in the pathogenesis of PVR. Therefore, oxidative stress may be a potential target for the treatment of PH.
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key transcriptional factor involved in the amplification of the antioxidant pathways associated with cardiovascular diseases [24,25,26]. It protects the cells against oxidative stress from exogenous or endogenous factors via activating antioxidant genes [18,27,28]. Additionally, Nrf2-regulated pathways play various roles in the pathophysiology and physiology of endothelial cells (ECs), vascular smooth muscle cells, and extravascular cells, exerting antioxidative, anti-inflammatory, and cytoprotective effects [29,30,31]. Recently, several studies have shown that Nrf2 inducers have potent antivascular remodeling effects in animal models of PH. This review summarizes the mechanistic role of Nrf2 in PVR and discusses the preclinical evidence of the therapeutic potential of Nrf2 inducers in PH.

2. Molecular Basis of Nrf2

Nrf2 was first discovered in the homolog of the hematopoietic transcription factor p45 NF-E2 [32]. It is encoded by the gene Nfe2l2 and belongs to a member of the Cap‘n’Collar (CNC) family, which regulates over 250 genes that contain the antioxidant response element (ARE) sequence [33]. They constitute a defense response to oxidation factors, including genes that encode for HO-1, superoxide dismutase (SOD), glutamate-cysteine ligase (GCL), glutathione S-transferase (GST), thioredoxin reductase (TXNRD)1, thioredoxin (TXN)1, glutathione peroxidase (GPx), and lowered glutathione (GSH) [34,35,36]. Additionally, this family has six members, including Nf-e2, Nrf1, Nrf2, Nrf3, Bach1, and Bach2 [37]. Nrf2 has seven conserved areas named Nrf2-ECH homology domains, which consist of Neh1 to Neh7 (Figure 1A). Among them, the Neh1, Neh3 to 5 domains are related to the transcriptional activation of Nrf2 by binding its co-activators, while the Neh2, Neh6 to 7 domains control the stability of Nrf2 by responding as a negative regulatory domain [38]. Stabilized Nrf2 translocates to the nucleus and binds with ARE on DNA to activate genes’ transcription, thus reducing or eliminating the production of reactive oxygen species (ROS) [28,39,40].

3. Signaling Transduction of Nrf2

Nrf2 is regulated by two specific mechanisms: the Kelch-like ECH-associated protein 1 (Keap1) and the participation of β-TrCP with the glycogen synthase kinase-3β (GSK)-3β [41]. Keap1 is one of the main regulators of Nrf2 [42]. Keap1 consists of five functional domains, namely, the N-terminal (NTR) domain, Broad complex/Tramtrack, Bric-a-Brac domain (BTB), a cysteine-rich intervening region (IVR), Kelch domain, and carboxyterminal (CTR) domain (Figure 1B). These domains of Keap1 play important roles in mediating Nrf2 ubiquitination and repression. Keap1 regulates Nrf2 protein expression in a redox-dependent manner [43,44]. Nrf2 is ubiquitinated and degraded by the Keap1-Cul3-Rbx1 complex under normal redox conditions. Under stress conditions, highly reactive cysteine residues of Keap1 are oxidized, which disrupts the binding of Keap1 to Nrf2 and promotes the nuclear translocation of Nrf2 (Figure 1C). The formation of Nrf2-sMaf heterodimers via its Neh1 domain in the nucleus induces gene expression by binding to the ARE sequences in the promoter regions of Nrf2 target genes [45]. The Nrf2/ARE signaling pathway is the major pathway for intracellular redox balance. ARE-genes encode a broad network of enzymes involved in antioxidant mechanisms encompassing nicotinamide adenine dinucleotide phosphate (NADPH), phase I, II, and III biotransformation reactions, lipid and iron catabolism, glutathione and thioredoxin-mediated reactions, among others. These pleiotropic functions contribute to significant cardiovascular protection effects. In addition, glycogen synthase kinase-3 was shown to prompt Nrf2 degradation by phosphorylating Nrf2 at serine 335 and 338. The poly ubiquitination of Nrf2 and its recognition by β-TrCP increase Nrf2 degradation [40,46]. Nrf2 controls the basal and inducible expression of an array of antioxidant and detoxification enzymes including the proteasome (Table 1). The Nrf2 signaling system has emerged as perhaps the most vital cellular defense and survival pathway against oxidative stress and toxicants [47].

4. Role of Nrf2 in Pulmonary Vascular Remodeling

Pulmonary vascular remodeling (PVR) refers to the thickening of blood vessels in the lungs, which involves all three layers of the blood vessel wall including the intima, the media, and the adventitia. Various pathological conditions, including pulmonary hypertension and chronic lung diseases, may cause PVR, as manifested by the thickening, narrowing, and stiffening of pulmonary arteries, capillaries, and veins. This remodeling process involves endothelial dysfunction, smooth muscle cell phenotypic switching and proliferation, and alterations of the extracellular matrix. These structural changes lead to increased pulmonary vascular resistance, reduced blood flow, and impaired gas exchange, ultimately leading to symptoms such as shortness of breath and exercise intolerance. Emerging evidence indicates that Nrf2 and its downstream transcriptional targets are involved in multiple processes of PVR. Understanding the role of Nrf2 in PVR could provide important insights into potential therapeutic strategies for managing the condition of PH.

4.1. Nrf2 and Endothelial Dysfunction in the Intima Remodeling

Although the exact causes of abnormal vascular remodeling in PH are still under investigation, emerging evidence suggests that EC dysfunction is among the primary triggers that initiate this process. EC dysfunction causes the activation of a series of cellular signaling pathways, leading to the uncontrolled proliferation of PAECs, SMCs, and fibroblasts, and eventually gives rise to vascular remodeling and even the occlusion of the pulmonary vessels. When the ECs are damaged, the barrier function of the endothelium and the muscle–endothelium interface is destroyed, and the concomitant loss of ECs regulation of SMCs leads to increased SMC proliferation and pulmonary vascular reconstruction [48,49]. BMPR2 mutations, epithelial–mesenchymal transition (EMT), thrombus formation, apoptosis, and inflammation are other factors connected with ECs’ dysfunction in PAH [50,51,52]. BMPR2 is mainly present in ECs of the vascular lumen in the lung and its expression is decreased in ECs from PH patients. Hence, mutated BMPR2 is thought to play an important role in ECs’ dysfunction in PAH [53,54]. Recently, BMP9 is also reported to be involved in ECs’ dysfunction in PAH [55,56]. EMT is a phenomenon in which ECs lose endothelial markers, which is accompanied by the acquisition of a lot of mesenchymal-like phenotypes and an increase in mesenchymal markers. Moreover, ECs change their morphology by losing cell–cell contact and obtaining a highly invasive and migratory phenotype, therefore losing the features of a normal endothelium [57,58]. Endothelial dysfunctions are characterized by both an increased production of vasoconstrictors, such as thromboxane, serotonin, and endothelin-1(ET-1), and a reduced secretion of potent vasodilators, such as prostacyclin (PGI2) and nitric oxide (NO) [12,59]. Studies in animal experiments and patients have demonstrated that NO deficiency is involved in the pathogenesis of PH [60,61]. A decreased PGI2 level is detected in various patients with different forms of PH, partially explaining the increase in SMC proliferation and pulmonary vasoconstriction in these patients. In animal models with PH, overexpressing PGI2 synthase is protected from developing chronic hypoxia-induced PAH [62,63]. Endothelium secretion of ET-1 increases in PH. ET-1 is mainly synthesized in ECs and its highest level in the overall body is in the lungs. It exerts its effects by binding to the receptors of ETA and ETB, which activate PASMC regulating proliferation, vasoconstriction, and vasorelaxation [64,65,66]. Thromboxane A2, produced by platelets and ECs, is increased in PH [62,67]. It creates an imbalance that may lead to vascular remodeling and excessive platelet aggregation noticed in PH. ECs′ apoptosis can also play a part in PH development via vascular dropout and selection pressure, which gives rise to the apoptosis-resistant phenotype of ECs in vascular lesions [68].
ROS-induced oxidative damage aggravates ECs’ dysfunction. The oxidative stress inducers activate Nrf2 and then exhibit protective effects in ECs by inducing its downstream antioxidant genes (Figure 2A and Table 1). In addition, Nrf2 has angiogenic and anti-inflammatory functions in ECs. Animal studies have shown that increased oxidative stress due to Nrf2 knockout impairs endothelial function and reduces functional congestion [69]. Genetic Nrf2 knockout also exacerbates endothelial dysfunction in the aorta, brain, and skeletal muscle microcirculation induced by obesity [70,71]. Emerging evidence demonstrates that Nrf2 and its downstream targets are related to PH. Lin et al. showed that activating Nrf2 could suppress hypoxia-induced pro-inflammatory ET-1 and enhance the production of vasodilatation factor NO and PGI2 expression in ECs [72,73]. Ji et al. showed that Nrf2 could suppress the protein level of HIF-1α expression and was thought to be a critical transcription factor for controlling angiogenesis [74]. Nrf2 may also play a part in the regulation of BMPR2 in PH. Diebold et al. showed that decreasing BMPR2 in PAECs by siRNA during re-oxygenation decreased Nrf2 expression, mitochondrial membrane potential, and ATP while inducing mitochondrial DNA deletion and apoptosis [75]. Chen et al. showed that Nrf2 activation attenuated PVR by inhibiting EMT [76]. Recent studies showed that many agents such as Z-Ligustilide, Sulfasalazine and even long non-coding RNA (lncRNA), protected vascular ECs from oxidative stress in PAH by activating the Nrf2 signaling pathways [77,78,79]. HO-1, one of the primary targets of Nrf2 activation, is a critical endogenous antioxidant [80]. HO-1 can adjust EMT [76], which is shown to be involved in PH pathobiology [1,81]. Yet et al. demonstrated the important role of HO-1 in an animal model of PH [82]. They exposed HO-1−/− mice to chronic hypoxia and verified that these mice developed right ventricular dilatation or failure and finally right ventricular infarcts. However, overexpressing HO-1 protected against hypoxia-induced lung inflammation and PAH [83]. A protective role of HO-1 was also documented in monocrotaline (MCT)-treated wild-type mice [84]. Furthermore, simvastatin could ameliorate PAH in MCT-treated rats via the induction of HO-1 expression [85]. Liang et al. also showed that bone-marrow-derived mesenchymal stem cells reduced chronic hypoxia-induced PAH via lung HO-1 expression [86]. In addition, HO-1 expression is substantial in lung tissue but decreased in the failing right ventricle in the Sugen 5416 and 10% chronic hypoxia (SuHx) model of PAH. Protandim, which can activate Nrf2 to upregulate the expression of genes encoding antioxidant enzymes, protects against the SuHx model of PAH in rats [87,88]. SOD is another downstream effector of Nrf2 activation. Hartney et al., who used pulmonary arteries acquired from calves with chronic hypoxic PAH, confirmed that the level of total SOD activity was reduced in the pulmonary arteries [89]. They showed that the overexpression of ECs SOD attenuated PH in mice. Van et al. also showed that overexpression of ECs SOD reversed bleomycin-induced lung fibrosis and nonangio-obliterative PH [90,91,92]. Additionally, SOD3, a subtype of SOD, is highly expressed in lung tissue [93]. Previous studies showed that the overexpression of SOD3 reduced MCT-induced PAH in rats and hypoxia-induced PAH in mice [94,95]. Xu et al. also showed that SOD3 defect in rats led to the development of PH, RVH, and vascular remodeling in response to hypoxia or MCT [96]. However, further experiments using endothelial or smooth-muscle-cell-specific knockout of SOD3 may be needed to investigate the cell-specific role of SOD3 in PAH. These results demonstrate that HO-1 and SOD, as vital downstream effectors of Nrf2, play crucial roles in the development of PH.

4.2. Nrf2 and Smooth Muscle Cell Phenotypic Switching in Media Remodeling

Rabinovitch et al. showed that BMPR2 mutations lower BMP signaling, contributing to the loss of the antiproliferative effects of BMP2 in PASMCs from PH patients [97]. In addition, ET-1 binds to the receptors of ETA and ETB, leading to complicated signaling pathways [98], involving the lowering of Potassium (K+) channel expression and activity, activation of Rho kinase (ROCK), and up-regulation of the [Ca2+] channel and Na+/H+ exchanger (NHE) [99,100]. Previous research showed that ET receptor inhibitors restrained or reversed vascular remodeling and PH in some animal models [101]. Thromboxane A2(TXA2) leads PH by inhibiting potassium channels, binding specific Gq/11 protein, and increasing intracellular calcium concentration ([Ca2+]i) [102,103,104]. It is revealed that PASMCs from PH patients were depolarized [105]. Reduced K+ channel activity and expression were subsequently confirmed as contributing factors to depolarizing PASMCs in hypoxic PH [106,107,108]. PASMCs express several K+ channel families, such as calcium-sensitive K(Ca), inward rectifier (Kir), voltage-gated (K(v)), ATP-sensitive K+ channel (KATP), and two-pore channels (K2P). Increasing evidence demonstrated that augmented K+ channel activity or expression lowered hypoxia-induced remodeling in animals and increased apoptosis in PASMCs from PH patients [109,110]. K(Ca) channels in VSMCs are targets for various physiological factors released from the endothelium, including NO and endothelium-derived hyperpolarizing factor (EDHF) [111,112]. The activation of voltage-gated calcium channels (VGCC) by agonists leads to PASMC proliferation [113,114]. Kir channels were first described in the SMCs of the coronary artery and were thought to express preferentially in small rather than large arteries [115]. KATP channels have numerous roles in PASMCs. They play an important role in maintaining membrane potential. Furthermore, the closure of KATP channels up-regulates the Ca2+ concentration in the cytoplasm, thus promoting the proliferation and contraction of PASMCs [116]. On the contrary, the opening of KATP channels results in an increase in membrane hyperpolarization, K+ efflux, inhibition of Ca2+ influx, and relaxation of the PASMC. Additionally, the EDHF and the NO can also activate KATP channels [117]. The K2P, also called potassium channel subfamily K member (KCNK), is a K+ channel in the subfamily K. It gathers different sub-families which are composed of several members [118]. KCNK3 is expressed in PASMC in humans. Furthermore, the knockdown of KCNK3 induced an obvious depolarization of resting membrane potential, demonstrating the significance of the KCNK3 channel in human PASMC resting membrane potential [119]. Increased [Ca2+], which has been verified in monocrotaline (MCT) and hypoxic animals treated with PH, is necessary for PASMC migration and growth. Additionally, the NHE is the primary contributor to the maintenance of PASMC pH homeostasis [120]. NHE activity is related to growth factor-induced proliferation. Lowering NHE activity via genetic deletion or pharmacological inhibition reduced hypoxia-induced vascular remodeling and PASMC migration and proliferation [121,122,123]. In addition, animal studies uncovered the role of HIF-1 in the development of hypoxia-induced PH [124,125,126]. Vascular remodeling and PH were improved in chronic hypoxia-stimulated Hif1a+/− mice, while hypoxia-induced proliferation in PASMCs extracted from Hif1a+/− mice was alleviated [124].
Pulmonary artery remodeling and vasoconstriction are key factors responsible for the vascular resistance observed in PH patients [127]. VSMCs’ excessive proliferation, hypertrophy, and apoptosis lead to the formation of the characteristic angio-proliferative lesions found in PH [128]. Oxidative stress plays a vital role in VSMC’s structure and function, and Nrf2 is one of the main antioxidant systems [129]. The expression of Nrf2 in rodent models of primary or secondary PH is decreased [130]. The selective delivery of Nrf2 activators to the injured vasculature has the potential to attenuate oxidative stress and decrease VSMC hyper-proliferation and migration towards the inner vessel wall [131]. Nrf2 gene transfer or Nrf2-inducing drugs such as sulforaphane, epigallocatechin gallate, cinnamic aldehyde, and exendin-4 have shown therapeutic applications in vascular diseases [132,133,134,135,136]. Previous research showed that the Nrf2/HO-1 pathway protected VSMCs from oxidative stress damage [137]. Another study showed that the activation of Nrf2 signaling alleviated VSMC phenotypic switching and vascular remodeling [138]. Recently, He et al. described a novel mechanism in which Nrf2 is a key regulator of VSMCs’ phenotypic switching and demonstrated a direct role for Nrf2 in the phenotypic switching of VSMCs during vascular remodeling [138]. Nrf2 exerts protective effects against PH development by mediating signaling pathways involved in their proliferation, migration, apoptosis, and phenotypic transition (Figure 2B). Of note, a previous study also showed that the antioxidant effects, which were mediated via the activation of the NO-cGMP-PKG-KATP channel signaling, relied on the Nrf2/HO-1 pathway [139]. Ko et al. showed that Nrf2 could suppress cell motility through RhoA-ROCK1 signaling [140]. Importantly, fasudil, a Rho-kinase inhibitor, mitigates pressure-overload-induced heart failure by activating Nrf2-mediated antioxidant responses [141]. This evidence demonstrates that Nrf2 is involved in the regulation of multiple signaling pathways of the occurrence and development of PH.
Pulmonary artery tension is mainly regulated by PASMC resting membrane potential and pulmonary artery endothelial function [142]. K+ channels are key regulators of vascular tone, cell proliferation, and apoptosis rates in PAH. PASMCs express several K+ channel families, including calcium-sensitive K(Ca), voltage-gated (K(v)), inward rectifier (Kir), ATP-sensitive K+channel (KATP), and K2P. The modulation of K+ channel activities by cellular oxidative stress has emerged as a significant determinant of vasomotor function in multiple disease states. Oxidative stress impairs K(v) channel function in persistent PH. Superoxide scavengers may improve pulmonary vasodilation in persistent PH of newborns in part by restoring K(v) channel function [143]. K(v) channel expression declined in PAH, resulting in elevations in cytosolic K+ and Ca2+ that decrease apoptosis and increase proliferation [144]. Pozeg et al. showed that K(v) in PASMCs is one of the reasons that led to pulmonary vasoconstriction even in PH [110]. K(v) channel activation significantly weakened the development of chronic hypoxia-induced PAH in mice and reversed spontaneous PAH [145]. In addition, during the formation process of left-to-right shunt-induced PAH, the function of the K(v) channel was inhibited, suggesting that K(v) channel may be the mechanism of PAH induced by left-to-right shunting [146]. One of the notable physiological functions of sequestosome1/p62 (SQSTM1) is the regulation of redox-sensitive voltage-gated potassium K(v) channels. Previous research confirmed that SQSTM1 enhanced the phosphorylation of K(v), which induced the suppression of pulmonary arterial Kv1.5 channels under acute hypoxia [147]. However, SQSTM1, regulated by the redox-sensitive transcription factor Nrf2, is an oxidative-stress-inducible protein. Therefore, Nrf2 may alleviate PAH by regulating K(v) channels. Cornfield et al. confirmed that reduced K(Ca) channel gene expression might lead to abnormal pulmonary vascular reactivity related to the persistent PH of the newborn [148]. Nrf2 is a vital determinant of the K(Ca) channels’ function and expression in VSMCs. Nrf2 promotes the expression of K(Ca) channels via a direct increase in gene transcriptions or other mechanisms [149]. Nrf2 is reported to be involved in KCNK3-regulated PAH vascular remodeling. KCNK3 expression and activity are strongly reduced in PASMCs in the PAH animal model. KCNK3 inhibition increased vasoconstriction, PASMC proliferation, and inflammation. Pharmacological activation of KCNK3 mitigated MCT-induced PAH in vivo [150]. Of note, Antigny et al. showed that the loss of function mutation of KCNK3 was responsible for the first channelopathy identified in PAH. They showed that the loss of KCNK3 expression resulted in the activation of the Nrf2/HO-1-mediated antioxidative stress response [151]. These results demonstrated that the loss of KCNK3 impaired PASMC function and a compensatory increase in Nrf2 could protect against it. Iptakalim (Ipt) is a new selective K(ATP) channel opener via electrophysiological, pharmacological, receptor binding tests and biochemical studies. In hypoxia-induced animal models, Ipt reduces the elevated mean pressure in pulmonary arteries and alleviates remodeling in the airways, right ventricle, and pulmonary arteries. Jin et al. showed that Ipt suppressed the effects of ET-1, decreased the intracellular calcium concentration, and restrained the proliferation of PASMCs [152]. Moreover, both the efficacy and safety of Ipt have been demonstrated in experimental animal models as well as in phase I clinical trials. Therefore, Ipt could be a potential candidate for hypoxic PH in the future [152]. Of note, Ipt protects against stress-induced oxidative stress through Nrf2-related pathways [153].

5. The Therapeutic Potential of Nrf2 Inducers in PH

In this section, we summarize the current potential therapeutic inducers targeting Nrf2 (Table 2) and discuss their potential applications for PH in preclinical studies.

5.1. SFN

SFN is the most widely used Nrf2 inducer in preclinical studies [190]. In addition to promoting the expression of endogenous antioxidants to regulate oxidative stress [191], it has significant anti-inflammatory effects and promotes cardiac protection in preclinical diabetic models. It has been reported that SFN protects against lung injury by activating the Nrf2 signaling pathway. Moreover, hypoxia-induced oxidation stress and pulmonary injury were significantly reduced by SFN in Nrf2+/+ mice but not in Nrf2−/− mice [192]. Zhang et al. showed that SFN prevented pulmonary damage by activating the Nrf2-defense response and subsequently ameliorating inflammatory responses by modulating cytokine production [193]. Keller et al. showed that SFN inhibited right ventricular injury and decreased PVR in PAH [154]. Male mice induced with SuHx for the PAH model were randomized to SFN treatment at a daily dose of 0.5 mg/kg, 5 days per week, for 4 weeks. The data showed that SFN reduced SuHx-induced PVR, fibrosis, and inflammation by upregulating Nrf2 expression. It also prevented SuHx-induced right ventricular dysfunction and remodeling. Another study demonstrated that the effect of SFN on vascular remodeling depends on Nrf2 [155]. Wild-type (WT) and Nrf2 knockout mice were induced by SuHx for PAH, followed by treatment with or without SFN for 4 weeks. The results showed that SFN partially or completely reversed SuHx-induced RV systolic/diastolic dysfunction in the WT mice, but not in the Nrf2 knockout mice. Although the role of SFN in PH has not been verified in clinical studies, these data suggest that SFN may be a candidate to prevent PAH. Very importantly, SFN has shown good tolerability, safety, and efficacy in clinical applications such as autism spectrum disorder [194].

5.2. Oltipraz

Oltipraz, an agonist of Nrf2, exerts a strong effect against oxidative stress in animal models or clinical patients with certain diseases [195]. Oltipraz can increase the binding activity of Nrf2 to the antioxidant response element, thereby increasing the production of phase II enzyme genes [196,197]. Induction of the expression of SOD and HO-1, downstream effectors of Nrf2, via Oltipraz administration in an animal model of chronic hypoxia suggests that Oltipraz may exert its beneficial effect via Nrf2 activation. Eba et al. showed that Oltipraz significantly reduced PVR and RVH associated with chronic hypoxia-induced PH [156]. They exposed Nrf2-deficient mice and Keap1 knockdown mice to hypoxia, and then treated the mice with different doses of Oltipraz (5, 50, or 500 mg/kg). They found that Nrf2-deficient mice exposed to hypoxia developed more pronounced RVH than WT mice, while Keap1-knockdown mice exposed to hypoxia displayed less PVR and RVH than WT mice, emphasizing the beneficial potency of Nrf2 activity against PH. They also showed a decreased expression of Nrf2-regulated antioxidant enzymes in chronic obstructive pulmonary disease-related PH patients. Moreover, pharmacologically inducing Nrf2 activity with Oltipraz significantly reduced PVR and RVH in the hypoxia-induced PH model. These data showed that Nrf2 activation exerted therapeutic efficacy against hypoxia-induced PH. Of note, the efficacy of Oltipraz highlights the therapeutic potential of Nrf2 activators for PH prevention in patients with hypoxic lung disease. However, further clinical studies are still needed to demonstrate the therapeutic potential of Oltipraz in PH.

5.3. Resveratrol

Resveratrol is a natural polyphenol found in red wine and grape skins. It has diverse biochemical and physiological actions including antiproliferative properties. It has been reported that treatment with resveratrol could restore the natural compound activator of Nrf2 and thus decrease oxidative stress [198]. Several studies have validated the protective role of resveratrol in animal models of PAH [157,158,159,160]. The role of Nrf2 induced by resveratrol in PAH has been extensively reviewed elsewhere [199].

5.4. Rosiglitazone

Rosiglitazone is an antidiabetic agent that belongs to the thiazolidinediones and is a nuclear hormone receptor peroxisome proliferator gamma (PPARγ) agonist [200,201]. Many studies showed that rosiglitazone alleviated hypoxia-induced pulmonary arterial remodeling [166,202], and an inhaled combination of sildenafil and rosiglitazone improved cardiac function, pulmonary hemodynamics, and arterial remodeling [203]. Rosiglitazone alleviated ET-1-induced vasoconstriction of pulmonary arteries in the rat model of PAH via the differential regulation of ET-1 receptors [167]. Previous studies showed that the expression of PPARγ was decreased in the lungs of PH patients, and PPARγ ligands were related to the release of vasoactive substances from vascular ECs and prevention of vascular remodeling. In a rat model of hypoxia-induced PH, oral administration of rosiglitazone (8 mg/kg, 5 days/week) for 4 weeks significantly reduced PVR and the development of PH. Rosiglitazone treatment restrained the hypoxia-induced reduction in PPARγ expression and restored VEGF and ET-1 expression almost to the levels of the normoxia group [168]. At the same time, another study also showed that rosiglitazone (10 mg/kg/d) prevented the development of PH at 3 weeks, and reversed established PH at 5 weeks [169]. Moreover, Wang et al. showed that the protective effects of rosiglitazone against MCT-induced PH were associated with drug dose and might be due to the inhibition of inflammation. In a rat model of MCT-induced PH, the administration of rosiglitazone (2.5 and 5 mg/kg) significantly attenuated the perivascular inflammation in the PH group. Compared with the low-dose rosiglitazone intervention group, all the above indices of the high-dose rosiglitazone intervention group appeared much lower [170]. Importantly, many studies showed that rosiglitazone was an activator of Nrf2 [204,205,206]. However, it is not clear whether the protecting effect of rosiglitazone against PH relies on Nrf2. Further studies are warranted to validate the role of Nrf2 in the therapeutic potential of rosiglitazone in PH. Considering that rosiglitazone is an FDA-approved treatment for diabetes, the preclinical studies of PH are expected to translate quickly into clinical trials.

5.5. Dimethyl Fumarate

Dimethyl Fumarate (DMF) is an FDA-approved antioxidative agent with good security. More and more data have confirmed that DMF is a powerful activator of Nrf2 [207,208]. Grzegorzewska et al. showed that DMF therapy is effective in reversing hemodynamic changes and reducing oxidative damage and lung fibrosis in the experimental models of PAH. They showed that DMF had pleiotropic modes of action that might improve PAH. DMF not only prevented the development of increased right ventricular systolic pressure (RVSP) and RVH in SuHx- and hypoxia-induced mice but also reversed pre-existing PH in a chronic hypoxia animal model [172]. Muralidharan et al. provided a rational design of advanced inhalable therapeutic dry powders containing DMF to treat pulmonary inflammation [209]. These data substantiate the beneficial effects of DMF on important molecular pathways leading to PAHs and support the treatment of patients with PAH. They also demonstrated a novel form of Nrf2 agonist for PH treatment, which could significantly reduce systemic side effects.

5.6. Rutin

Rutin is a bioflavonoid with a wide range of biological activities, including antioxidant and anti-inflammatory effects. Rutin reduced oxidative stress, vascular fibrosis, and cardiac hypertrophy by upregulating Nrf2 transcriptional activity and the expression levels of its downstream target genes [210]. Previous studies showed that rutin protected against lung inflammation and lung injury [211,212,213]. Bai et al. demonstrated that rutin could protect against bleomycin-induced lung fibrosis [212]. Li et al. showed that rutin also increased NO production in bovine PAECs, and exerts a vasodilator influence on the pulmonary artery [214]. In addition, rutin attenuated hypoxia-induced PASMC proliferation [215]. However, Shellenberger et al. showed that although rutin reduced the severity of hypoxia-induced PH, histological changes consistent with pulmonary arteriole remodeling were observed in some calves fed rutin. They used calves fed rutin under hypoxic conditions, which had lower mean PAH. Paradoxically, a larger proportion of calves fed rutin had more histological evidence of pulmonary arteriolar adventitial hyperplasia and medial hypertrophy than the controls did [173]. The controversial data may be due to the use of different experimental methods. These also suggest that rutin may play different roles in different cells or tissues, which requires further research.

5.7. Curcumin

Curcumin, a lipophilic polyphenol, has anticancer, antibiotic, anti-inflammatory, and antioxidant effects, as suggested by several experimental studies and clinical trials. Several studies have shown that curcumin may protect lungs from oxidative damage by activating the Nrf2 signaling pathway [216,217]. These have shown that curcumin could improve PAH, ameliorate pulmonary vessel remodeling, and reduce the deposition of collagen I in pulmonary arterioles [175]. Devadasu et al. attempted to exert the therapeutic benefits of curcumin in PH by encapsulating it in biodegradable polyacid nanoparticles. However, orally administered nanoparticulate curcumin failed to provide any protection against hypoxia-induced PH, and changes in RVH and vascular remodeling were similar to those in the untreated group. They found an obvious difference in the target tissue levels between normoxic and hypoxic rats. The target tissue levels of curcumin under hypoxia are much lower than those in normoxic rats. This might be due to the difference in particle dynamics, leading to the failure of treatment [218]. Kruangtip et al. showed that curcumin analogues were potential targets for developing efficacious and selective PDE5 inhibitors and treatment of PH [219]. Chen et al. suggested that curcumin could promote PASMC apoptosis, protect mitochondrial function, and reverse MCT-mediated PVR. It could suppress the PI3K/AKT pathway in PASMCs and regulate the expression of antiproliferative genes [174]. However, they also reported the side effects of curcumin in MCT-induced rats, which reminds us that the dosage should be used with caution and its toxicological mechanism should be further evaluated and studied.

5.8. Natural Products

Puerarin is primarily derived from Gegen (Pueraria lobata, Radix puerariae) and exhibits a variety of pharmacological activities by activating the Nrf2 signaling pathway [220,221]. Previous studies have demonstrated that puerarin prevents hypoxia-induced PVR and PH [177,178]. Li et al. showed that puerarin (20 mg/kg/d, 3 weeks) could improve PVR in rats with PH by inhibiting the deposition of collagen. They showed that puerarin increased NO and SOD, and reduced ET-1 and collagen I levels [176]. Tannins present in Terminalia bellirica have pharmacological activities such as oxidative stress resistance and anti-inflammatory effects [222,223]. Recent research has shown that it might reduce high-altitude PH by activating the Nrf2/HO-1 signaling pathway in rats [222]. Naringenin, a flavanone extracted from various plants, has potent vaso-protective effects likely related to the induction of Nrf2 and HO-1 [224]. Ahmed et al. showed that naringenin significantly added to the protective effect of L-arginine in PH induced by MCT in rats. The mechanism might be to increase the expression of eNOS and the expression of NO and to reduce the expression of iNOS [180]. Berberine is an isoquinone alkaloid which is derived from berberis aristata, berberis aquifolium and berberis vulgaris. It has all kinds of pharmacological effects, such as antioxidant, antitumor, antidiabetic, anti-inflammatory, and antimicrobial. Several studies have shown that the biological and therapeutic activities of berberine depend on Nrf2 activation [225,226]. Yu et al. also showed that it alleviated PH through the β-catenin and Trx1 signaling pathways in PASMCs [181]. Tanshinone IIA is the major antioxidant component in salvia miltiorrhiza. Nrf2 activation is the major regulatory pathway in tanshinone IIA-induced cytoprotective gene expression against oxidative stress and inhibition of SMCs proliferation [227]. Previous studies showed that tanshinone IIA ameliorated hypoxic PH by modulating K(v) currents and inhibited Ca2+ influx [189,228]. Another study suggested that tanshinone IIA markedly decreased right ventricular systolic pressure, mean RVSP, RVH index, and PVR in MCT-induced PH rats [184]. However, whether these natural products can inhibit the development of PH by directly acting on Nrf2 remains to be verified.

6. Concluding Remarks and Future Perspective

We discussed and summarized the therapeutic potential of Nrf2 inducers for PAH in preclinical studies in this article (Table 2). It is demonstrated that the activation of Nrf2 inhibits vascular remodeling and PH mainly by improving ROS-related pathological processes. Moreover, we found some knowledge gaps. Firstly, we do not know whether Nrf2 inducers have the same effects in other vascular remodeling-related diseases, such as aortic dissection and chronic obstructive pulmonary disease. Secondly, although some Nrf2 inducers have been reported to improve PVR and PH, it is not clear whether they act directly through Nrf2. Therefore, further studies are needed to validate their effect by using genetically Nrf2-deficient mice. Thirdly, the Nrf2 inducers used in these studies are not specific to Nrf2. Thus, transgenic mouse models, specific Nrf2 inducers, and cell-specific overexpression or knockout of Nrf2 should be used to dissect the role of Nrf2 in PH. Finally, although many preclinical studies have confirmed the treatment effect of Nrf2 inducer in PH, it has not been verified via clinical trials. Considering that some of the Nrf2 inducers such as rosiglitazone and DMF have already been approved by the FDA for clinical use in other diseases, they may be quickly translated into the clinic to explore the effect on PH. It is encouraging to note that these Nrf2 inducers have shown good efficacy, tolerability, and safety in other diseases such as neurodegenerative disease [229]. Taken together, Nrf2-based PH therapy shows great promise to address the underlying vascular remodeling processes in PH, but the real treatment effect by targeting Nrf2 specifically and the exact molecular mechanisms need to be further studied. Further research in this area may pave the way for the development of innovative treatment options for PH patients.

Author Contributions

Conceptualization, Q.F. and Y.B.; writing—original draft preparation, Q.F. and Y.B.; writing—review and editing, Y.W. and X.R.; visualization, S.H., J.D., L.L., M.D., J.Q. and L.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by grants from the National Natural Science Foundation of China (No. 82000340 and No. 82070383).

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.

Abbreviations

PHPulmonary hypertension
Nrf2Nuclear factor erythroid 2-related factor 2
VSMCvascular smooth muscle cell
PAECpulmonary arterial endothelial cell
PASMCpulmonary arterial smooth muscle cell
PAHPulmonary arterial hypertension
ROSreactive oxygen species
NOSnitric oxide synthase
NADPHnicotinamide adenine dinucleotide phosphate
SODsuperoxide dismutase
ET-1endothelin-1
PGI2prostacyclin
ECendothelial cell
SMCsmooth muscle cell
EMTepithelial-mesenchymal transition
NOnitric oxide
EDHFendothelium-derived hyperpolarizing factor
VGCCVoltagegated calcium channels
MCTmonocrotaline
NHENa+/H+ exchanger
AREantioxidant response element
EpREelectrophile response element
GSTglutathione S-transferase
GCLglutamate-cysteine ligase
GPxglutathione peroxidase
TXNRDthioredoxin reductase
TXNthioredoxin
GSHglutathione
sMAFsmall musculoaponeurotic fibrosarcoma
cAMPcyclic adenosine monophosphate
β-TrCPβ-transducin repeat-containing protein
RXR-αretinoic X receptor alpha
Keap1Kelch-like ECH-associated protein 1
BTBBric-a-Brac domain
IVRintervening region
LncRNAlong non-coding RNA
RVHright ventricular hypertrophy
SFNsulforaphane
KCNK3potassium channel subfamily K member 3
IptIptakalim
PPARγperoxisome proliferator gamma
DMFDimethyl Fumarate
RVSPright ventricular systolic pressure
SuHxSU5416 and 10% hypoxia
GCLglutamate-cysteine ligase
RV/(LV+S)right ventricular hypertrophy index

References

  1. Lambert, M.; Capuano, V.; Boet, A.; Tesson, L.; Bertero, T.; Nakhleh, M.K.; Remy, S.; Anegon, I.; Pechoux, C.; Hautefort, A.; et al. Characterization of Kcnk3-Mutated Rat, a Novel Model of Pulmonary Hypertension. Circ. Res. 2019, 125, 678–695. [Google Scholar] [CrossRef]
  2. Xue, X.; Zhang, S.; Jiang, W.; Wang, J.; Xin, Q.; Sun, C.; Li, K.; Qi, T.; Luan, Y. Protective effect of baicalin against pulmonary arterial hypertension vascular remodeling through regulation of TNF-alpha signaling pathway. Pharmacol. Res. Perspect. 2021, 9, e00703. [Google Scholar] [CrossRef] [PubMed]
  3. Galie, N.; Humbert, M.; Vachiery, J.L.; Gibbs, S.; Lang, I.; Torbicki, A.; Simonneau, G.; Peacock, A.; Vonk Noordegraaf, A.; Beghetti, M.; et al. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur. Respir. J. 2015, 46, 903–975. [Google Scholar] [CrossRef] [PubMed]
  4. Condon, D.F.; Nickel, N.P.; Anderson, R.; Mirza, S.; de Jesus Perez, V.A. The 6th World Symposium on Pulmonary Hypertension: What’s old is new. F1000Res 2019, 8, 888. [Google Scholar] [CrossRef] [PubMed]
  5. Galie, N.; Channick, R.N.; Frantz, R.P.; Grunig, E.; Jing, Z.C.; Moiseeva, O.; Preston, I.R.; Pulido, T.; Safdar, Z.; Tamura, Y.; et al. Risk stratification and medical therapy of pulmonary arterial hypertension. Eur. Respir. J. 2019, 53, 1801889. [Google Scholar] [CrossRef] [PubMed]
  6. Galie, N.; Hoeper, M.M.; Humbert, M.; Torbicki, A.; Vachiery, J.L.; Barbera, J.A.; Beghetti, M.; Corris, P.; Gaine, S.; Gibbs, J.S.; et al. Guidelines for the diagnosis and treatment of pulmonary hypertension: The Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS), endorsed by the International Society of Heart and Lung Transplantation (ISHLT). Eur. Heart J. 2009, 30, 2493–2537. [Google Scholar] [CrossRef]
  7. Humbert, M.; Sitbon, O.; Chaouat, A.; Bertocchi, M.; Habib, G.; Gressin, V.; Yaici, A.; Weitzenblum, E.; Cordier, J.F.; Chabot, F.; et al. Survival in patients with idiopathic, familial, and anorexigen-associated pulmonary arterial hypertension in the modern management era. Circulation 2010, 122, 156–163. [Google Scholar] [CrossRef]
  8. Benza, R.L.; Miller, D.P.; Barst, R.J.; Badesch, D.B.; Frost, A.E.; McGoon, M.D. An evaluation of long-term survival from time of diagnosis in pulmonary arterial hypertension from the REVEAL Registry. Chest 2012, 142, 448–456. [Google Scholar] [CrossRef] [PubMed]
  9. Lan, N.S.H.; Massam, B.D.; Kulkarni, S.S.; Lang, C.C. Pulmonary Arterial Hypertension: Pathophysiology and Treatment. Diseases 2018, 6, 38. [Google Scholar] [CrossRef] [PubMed]
  10. Thenappan, T.; Ormiston, M.L.; Ryan, J.J.; Archer, S.L. Pulmonary arterial hypertension: Pathogenesis and clinical management. BMJ 2018, 360, j5492. [Google Scholar] [CrossRef]
  11. Zhang, F.; Chen, A.; Pan, Y.; Wang, X.; Xu, Y.; Desai, A.A.; Tang, H.; Han, Y. Research Progress on Pulmonary Arterial Hypertension and the Role of the Angiotensin Converting Enzyme 2-Angiotensin-(1–7)-Mas Axis in Pulmonary Arterial Hypertension. Cardiovasc. Drugs Ther. 2022, 36, 363–370. [Google Scholar] [CrossRef] [PubMed]
  12. Humbert, M.; Lau, E.M.; Montani, D.; Jais, X.; Sitbon, O.; Simonneau, G. Advances in therapeutic interventions for patients with pulmonary arterial hypertension. Circulation 2014, 130, 2189–2208. [Google Scholar] [CrossRef] [PubMed]
  13. Evans, C.E.; Cober, N.D.; Dai, Z.; Stewart, D.J.; Zhao, Y.Y. Endothelial cells in the pathogenesis of pulmonary arterial hypertension. Eur. Respir. J. 2021, 58, 2003957. [Google Scholar] [CrossRef] [PubMed]
  14. Thenappan, T.; Chan, S.Y.; Weir, E.K. Role of extracellular matrix in the pathogenesis of pulmonary arterial hypertension. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H1322–H1331. [Google Scholar] [CrossRef] [PubMed]
  15. Satoh, K.; Satoh, T.; Kikuchi, N.; Omura, J.; Kurosawa, R.; Suzuki, K.; Sugimura, K.; Aoki, T.; Nochioka, K.; Tatebe, S.; et al. Basigin mediates pulmonary hypertension by promoting inflammation and vascular smooth muscle cell proliferation. Circ. Res. 2014, 115, 738–750. [Google Scholar] [CrossRef]
  16. Tuder, R.M. Pathology of pulmonary arterial hypertension. Semin. Respir. Crit. Care Med. 2009, 30, 376–385. [Google Scholar] [CrossRef] [PubMed]
  17. Rabinovitch, M. Molecular pathogenesis of pulmonary arterial hypertension. J. Clin. Investig. 2012, 122, 4306–4313. [Google Scholar] [CrossRef] [PubMed]
  18. Smukowska-Gorynia, A.; Rzymski, P.; Marcinkowska, J.; Poniedzialek, B.; Komosa, A.; Cieslewicz, A.; Slawek-Szmyt, S.; Janus, M.; Araszkiewicz, A.; Jankiewicz, S.; et al. Prognostic Value of Oxidative Stress Markers in Patients with Pulmonary Arterial or Chronic Thromboembolic Pulmonary Hypertension. Oxidative Med. Cell. Longev. 2019, 2019, 3795320. [Google Scholar] [CrossRef]
  19. Pena, E.; Brito, J.; El Alam, S.; Siques, P. Oxidative Stress, Kinase Activity and Inflammatory Implications in Right Ventricular Hypertrophy and Heart Failure under Hypobaric Hypoxia. Int. J. Mol. Sci. 2020, 21, 6421. [Google Scholar] [CrossRef]
  20. Turck, P.; Fraga, S.; Salvador, I.; Campos-Carraro, C.; Lacerda, D.; Bahr, A.; Ortiz, V.; Hickmann, A.; Koetz, M.; Bello-Klein, A.; et al. Blueberry extract decreases oxidative stress and improves functional parameters in lungs from rats with pulmonary arterial hypertension. Nutrition 2020, 70, 110579. [Google Scholar] [CrossRef]
  21. Rudyk, O.; Aaronson, P.I. Redox Regulation, Oxidative Stress, and Inflammation in Group 3 Pulmonary Hypertension. Adv. Exp. Med. Biol. 2021, 1303, 209–241. [Google Scholar] [CrossRef] [PubMed]
  22. Rabinovitch, M.; Guignabert, C.; Humbert, M.; Nicolls, M.R. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ. Res. 2014, 115, 165–175. [Google Scholar] [CrossRef] [PubMed]
  23. Frid, M.G.; Thurman, J.M.; Hansen, K.C.; Maron, B.A.; Stenmark, K.R. Inflammation, immunity, and vascular remodeling in pulmonary hypertension; Evidence for complement involvement? Glob. Cardiol. Sci. Pract. 2020, 2020, e202001. [Google Scholar] [CrossRef]
  24. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed]
  25. Tan, Y.; Ichikawa, T.; Li, J.; Si, Q.; Yang, H.; Chen, X.; Goldblatt, C.S.; Meyer, C.J.; Li, X.; Cai, L.; et al. Diabetic downregulation of Nrf2 activity via ERK contributes to oxidative stress-induced insulin resistance in cardiac cells in vitro and in vivo. Diabetes 2011, 60, 625–633. [Google Scholar] [CrossRef]
  26. Tian, C.; Gao, L.; Zimmerman, M.C.; Zucker, I.H. Myocardial infarction-induced microRNA-enriched exosomes contribute to cardiac Nrf2 dysregulation in chronic heart failure. Am. J. Physiol. Heart Circ. Physiol. 2018, 314, H928–H939. [Google Scholar] [CrossRef]
  27. Sova, M.; Saso, L. Design and development of Nrf2 modulators for cancer chemoprevention and therapy: A review. Drug Des. Dev. Ther. 2018, 12, 3181–3197. [Google Scholar] [CrossRef]
  28. Cores, A.; Piquero, M.; Villacampa, M.; Leon, R.; Menendez, J.C. NRF2 Regulation Processes as a Source of Potential Drug Targets against Neurodegenerative Diseases. Biomolecules 2020, 10, 904. [Google Scholar] [CrossRef]
  29. Ungvari, Z.; Tarantini, S.; Nyul-Toth, A.; Kiss, T.; Yabluchanskiy, A.; Csipo, T.; Balasubramanian, P.; Lipecz, A.; Benyo, Z.; Csiszar, A. Nrf2 dysfunction and impaired cellular resilience to oxidative stressors in the aged vasculature: From increased cellular senescence to the pathogenesis of age-related vascular diseases. Geroscience 2019, 41, 727–738. [Google Scholar] [CrossRef]
  30. Kim, S.L.; Choi, H.S.; Ko, Y.C.; Yun, B.S.; Lee, D.S. 5-Hydroxymaltol Derived from Beetroot Juice through Lactobacillus Fermentation Suppresses Inflammatory Effect and Oxidant Stress via Regulating NF-kB, MAPKs Pathway and NRF2/HO-1 Expression. Antioxidants 2021, 10, 1324. [Google Scholar] [CrossRef]
  31. Gallorini, M.; Berardi, A.C.; Gissi, C.; Cataldi, A.; Osti, L. Nrf2-mediated cytoprotective effect of four different hyaluronic acids by molecular weight in human tenocytes. J. Drug Target. 2020, 28, 212–224. [Google Scholar] [CrossRef]
  32. Itoh, K.; Igarashi, K.; Hayashi, N.; Nishizawa, M.; Yamamoto, M. Cloning and characterization of a novel erythroid cell-derived CNC family transcription factor heterodimerizing with the small Maf family proteins. Mol. Cell Biol. 1995, 15, 4184–4193. [Google Scholar] [CrossRef]
  33. 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]
  34. 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]
  35. Chan, J.Y.; Kwong, M. Impaired expression of glutathione synthetic enzyme genes in mice with targeted deletion of the Nrf2 basic-leucine zipper protein. Biochim. Biophys. Acta 2000, 1517, 19–26. [Google Scholar] [CrossRef] [PubMed]
  36. Hayes, J.D.; Chanas, S.A.; Henderson, C.J.; McMahon, M.; Sun, C.; Moffat, G.J.; Wolf, C.R.; Yamamoto, M. The Nrf2 transcription factor contributes both to the basal expression of glutathione S-transferases in mouse liver and to their induction by the chemopreventive synthetic antioxidants, butylated hydroxyanisole and ethoxyquin. Biochem. Soc. Trans. 2000, 28, 33–41. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, D.D. Mechanistic studies of the Nrf2-Keap1 signaling pathway. Drug Metab. Rev. 2006, 38, 769–789. [Google Scholar] [CrossRef] [PubMed]
  38. Panieri, E.; Telkoparan-Akillilar, P.; Suzen, S.; Saso, L. The NRF2/KEAP1 Axis in the Regulation of Tumor Metabolism: Mechanisms and Therapeutic Perspectives. Biomolecules 2020, 10, 791. [Google Scholar] [CrossRef] [PubMed]
  39. Zheng, F.; Goncalves, F.M.; Abiko, Y.; Li, H.; Kumagai, Y.; Aschner, M. Redox toxicology of environmental chemicals causing oxidative stress. Redox Biol. 2020, 34, 101475. [Google Scholar] [CrossRef] [PubMed]
  40. Shaw, P.; Chattopadhyay, A. Nrf2-ARE signaling in cellular protection: Mechanism of action and the regulatory mechanisms. J. Cell. Physiol. 2020, 235, 3119–3130. [Google Scholar] [CrossRef]
  41. Itoh, K.; Tong, K.I.; Yamamoto, M. Molecular mechanism activating Nrf2-Keap1 pathway in regulation of adaptive response to electrophiles. Free Radic. Biol. Med. 2004, 36, 1208–1213. [Google Scholar] [CrossRef] [PubMed]
  42. Song, M.Y.; Lee, D.Y.; Chun, K.S.; Kim, E.H. The Role of NRF2/KEAP1 Signaling Pathway in Cancer Metabolism. Int. J. Mol. Sci. 2021, 22, 4376. [Google Scholar] [CrossRef] [PubMed]
  43. Cullinan, S.B.; Gordan, J.D.; Jin, J.; Harper, J.W.; Diehl, J.A. The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase: Oxidative stress sensing by a Cul3-Keap1 ligase. Mol. Cell Biol. 2004, 24, 8477–8486. [Google Scholar] [CrossRef]
  44. Kobayashi, A.; Kang, M.I.; Okawa, H.; Ohtsuji, M.; Zenke, Y.; Chiba, T.; Igarashi, K.; Yamamoto, M. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell Biol. 2004, 24, 7130–7139. [Google Scholar] [CrossRef]
  45. Itoh, K.; Mimura, J.; Yamamoto, M. Discovery of the negative regulator of Nrf2, Keap1: A historical overview. Antioxid. Redox Signal. 2010, 13, 1665–1678. [Google Scholar] [CrossRef] [PubMed]
  46. Cuadrado, A. Structural and functional characterization of Nrf2 degradation by glycogen synthase kinase 3/β-TrCP. Free Radic. Biol. Med. 2015, 88, 147–157. [Google Scholar] [CrossRef] [PubMed]
  47. Ghosh, S.; Dutta, N.; Banerjee, P.; Gajbhiye, R.L.; Sareng, H.R.; Kapse, P.; Pal, S.; Burdelya, L.; Mandal, N.C.; Ravichandiran, V.; et al. Induction of monoamine oxidase A-mediated oxidative stress and impairment of NRF2-antioxidant defence response by polyphenol-rich fraction of Bergenia ligulata sensitizes prostate cancer cells in vitro and in vivo. Free Radic. Biol. Med. 2021, 172, 136–151. [Google Scholar] [CrossRef]
  48. Price, L.C.; Wort, S.J.; Perros, F.; Dorfmuller, P.; Huertas, A.; Montani, D.; Cohen-Kaminsky, S.; Humbert, M. Inflammation in pulmonary arterial hypertension. Chest 2012, 141, 210–221. [Google Scholar] [CrossRef] [PubMed]
  49. Quarck, R.; Nawrot, T.; Meyns, B.; Delcroix, M. C-reactive protein: A new predictor of adverse outcome in pulmonary arterial hypertension. J. Am. Coll. Cardiol. 2009, 53, 1211–1218. [Google Scholar] [CrossRef]
  50. Gong, J.; Feng, Z.; Peterson, A.L.; Carr, J.F.; Vang, A.; Braza, J.; Choudhary, G.; Dennery, P.A.; Yao, H. Endothelial to mesenchymal transition during neonatal hyperoxia-induced pulmonary hypertension. J. Pathol. 2020, 252, 411–422. [Google Scholar] [CrossRef]
  51. Wu, D.; Chen, Y.; Wang, W.; Li, H.; Yang, M.; Ding, H.; Lv, X.; Lian, N.; Zhao, J.; Deng, C. The role of inflammation in a rat model of chronic thromboembolic pulmonary hypertension induced by carrageenan. Ann. Transl. Med. 2020, 8, 492. [Google Scholar] [CrossRef] [PubMed]
  52. Ruffenach, G.; O’Connor, E.; Vaillancourt, M.; Hong, J.; Cao, N.; Sarji, S.; Moazeni, S.; Papesh, J.; Grijalva, V.; Cunningham, C.M.; et al. Oral 15-Hydroxyeicosatetraenoic Acid Induces Pulmonary Hypertension in Mice by Triggering T Cell-Dependent Endothelial Cell Apoptosis. Hypertension 2020, 76, 985–996. [Google Scholar] [CrossRef]
  53. Frump, A.; Prewitt, A.; de Caestecker, M.P. BMPR2 mutations and endothelial dysfunction in pulmonary arterial hypertension (2017 Grover Conference Series). Pulm. Circ. 2018, 8, 2045894018765840. [Google Scholar] [CrossRef] [PubMed]
  54. Atkinson, C.; Stewart, S.; Upton, P.D.; Machado, R.; Thomson, J.R.; Trembath, R.C.; Morrell, N.W. Primary pulmonary hypertension is associated with reduced pulmonary vascular expression of type II bone morphogenetic protein receptor. Circulation 2002, 105, 1672–1678. [Google Scholar] [CrossRef] [PubMed]
  55. Jiang, Q.; Liu, C.; Liu, S.; Lu, W.; Li, Y.; Luo, X.; Ma, R.; Zhang, C.; Chen, H.; Chen, Y.; et al. Dysregulation of BMP9/BMPR2/SMAD signalling pathway contributes to pulmonary fibrosis and pulmonary hypertension induced by bleomycin in rats. Br. J. Pharmacol. 2021, 178, 203–216. [Google Scholar] [CrossRef]
  56. Tu, L.; Desroches-Castan, A.; Mallet, C.; Guyon, L.; Cumont, A.; Phan, C.; Robert, F.; Thuillet, R.; Bordenave, J.; Sekine, A.; et al. Selective BMP-9 Inhibition Partially Protects Against Experimental Pulmonary Hypertension. Circ. Res. 2019, 124, 846–855. [Google Scholar] [CrossRef]
  57. Sanchez-Duffhues, G.; Garcia de Vinuesa, A.; Ten Dijke, P. Endothelial-to-mesenchymal transition in cardiovascular diseases: Developmental signaling pathways gone awry. Dev. Dyn. 2018, 247, 492–508. [Google Scholar] [CrossRef]
  58. Medici, D.; Kalluri, R. Endothelial-mesenchymal transition and its contribution to the emergence of stem cell phenotype. Semin. Cancer Biol. 2012, 22, 379–384. [Google Scholar] [CrossRef]
  59. Delaney, C.; Sherlock, L.; Fisher, S.; Maltzahn, J.; Wright, C.; Nozik-Grayck, E. Serotonin 2A receptor inhibition protects against the development of pulmonary hypertension and pulmonary vascular remodeling in neonatal mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 314, L871–L881. [Google Scholar] [CrossRef]
  60. Gredic, M.; Wu, C.Y.; Hadzic, S.; Pak, O.; Savai, R.; Kojonazarov, B.; Doswada, S.; Weiss, A.; Weigert, A.; Guenther, A.; et al. Myeloid-cell-specific deletion of inducible nitric oxide synthase protects against smoke-induced pulmonary hypertension in mice. Eur. Respir. J. 2022, 59, 2101153. [Google Scholar] [CrossRef]
  61. Wang, M.; Luo, P.; Shi, W.; Guo, J.; Huo, S.; Yan, D.; Peng, L.; Zhang, C.; Lv, J.; Lin, L.; et al. S-Nitroso-L-Cysteine Ameliorated Pulmonary Hypertension in the MCT-Induced Rats through Anti-ROS and Anti-Inflammatory Pathways. Oxidative Med. Cell. Longev. 2021, 2021, 6621232. [Google Scholar] [CrossRef]
  62. Budhiraja, R.; Tuder, R.M.; Hassoun, P.M. Endothelial dysfunction in pulmonary hypertension. Circulation 2004, 109, 159–165. [Google Scholar] [CrossRef]
  63. Geraci, M.W.; Gao, B.; Shepherd, D.C.; Moore, M.D.; Westcott, J.Y.; Fagan, K.A.; Alger, L.A.; Tuder, R.M.; Voelkel, N.F. Pulmonary prostacyclin synthase overexpression in transgenic mice protects against development of hypoxic pulmonary hypertension. J. Clin. Investig. 1999, 103, 1509–1515. [Google Scholar] [CrossRef] [PubMed]
  64. Chester, A.H.; Yacoub, M.H. The role of endothelin-1 in pulmonary arterial hypertension. Glob. Cardiol. Sci. Pract. 2014, 2014, 62–78. [Google Scholar] [CrossRef] [PubMed]
  65. Cinà, C.S.; Devereaux, P. Vascular viewpoint. Vasc. Med. 2006, 11, 61–63. [Google Scholar] [CrossRef] [PubMed]
  66. McFalls, E.O.; Ward, H.B.; Moritz, T.E.; Goldman, S.; Krupski, W.C.; Littooy, F.; Pierpont, G.; Santilli, S.; Rapp, J.; Hattler, B.; et al. Coronary-artery revascularization before elective major vascular surgery. N. Engl. J. Med. 2004, 351, 2795–2804. [Google Scholar] [CrossRef]
  67. Christman, B.W.; McPherson, C.D.; Newman, J.H.; King, G.A.; Bernard, G.R.; Groves, B.M.; Loyd, J.E. An imbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N. Engl. J. Med. 1992, 327, 70–75. [Google Scholar] [CrossRef]
  68. Tuder, R.M.; Archer, S.L.; Dorfmuller, P.; Erzurum, S.C.; Guignabert, C.; Michelakis, E.; Rabinovitch, M.; Schermuly, R.; Stenmark, K.R.; Morrell, N.W. Relevant issues in the pathology and pathobiology of pulmonary hypertension. J. Am. Coll. Cardiol. 2013, 62, D4–D12. [Google Scholar] [CrossRef]
  69. Tarantini, S.; Valcarcel-Ares, M.N.; Yabluchanskiy, A.; Tucsek, Z.; Hertelendy, P.; Kiss, T.; Gautam, T.; Zhang, X.A.; Sonntag, W.E.; de Cabo, R.; et al. Nrf2 Deficiency Exacerbates Obesity-Induced Oxidative Stress, Neurovascular Dysfunction, Blood-Brain Barrier Disruption, Neuroinflammation, Amyloidogenic Gene Expression, and Cognitive Decline in Mice, Mimicking the Aging Phenotype. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2018, 73, 853–863. [Google Scholar] [CrossRef]
  70. Ungvari, Z.; Bagi, Z.; Feher, A.; Recchia, F.A.; Sonntag, W.E.; Pearson, K.; de Cabo, R.; Csiszar, A. Resveratrol confers endothelial protection via activation of the antioxidant transcription factor Nrf2. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H18–H24. [Google Scholar] [CrossRef]
  71. Ungvari, Z.; Bailey-Downs, L.; Sosnowska, D.; Gautam, T.; Koncz, P.; Losonczy, G.; Ballabh, P.; de Cabo, R.; Sonntag, W.E.; Csiszar, A. Vascular oxidative stress in aging: A homeostatic failure due to dysregulation of NRF2-mediated antioxidant response. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H363–H372. [Google Scholar] [CrossRef] [PubMed]
  72. Lin, H.C.; Su, S.L.; Lu, C.Y.; Lin, A.H.; Lin, W.C.; Liu, C.S.; Yang, Y.C.; Wang, H.M.; Lii, C.K.; Chen, H.W. Andrographolide inhibits hypoxia-induced HIF-1alpha-driven endothelin 1 secretion by activating Nrf2/HO-1 and promoting the expression of prolyl hydroxylases 2/3 in human endothelial cells. Env. Toxicol. 2017, 32, 918–930. [Google Scholar] [CrossRef] [PubMed]
  73. 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]
  74. Ji, X.; Wang, H.; Zhu, J.; Zhu, L.; Pan, H.; Li, W.; Zhou, Y.; Cong, Z.; Yan, F.; Chen, S. Knockdown of Nrf2 suppresses glioblastoma angiogenesis by inhibiting hypoxia-induced activation of HIF-1α. Int. J. Cancer 2014, 135, 574–584. [Google Scholar] [CrossRef] [PubMed]
  75. Diebold, I.; Hennigs, J.K.; Miyagawa, K.; Li, C.G.; Nickel, N.P.; Kaschwich, M.; Cao, A.; Wang, L.; Reddy, S.; Chen, P.I.; et al. BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension. Cell Metab. 2015, 21, 596–608. [Google Scholar] [CrossRef]
  76. Chen, Y.; Yuan, T.; Zhang, H.; Yan, Y.; Wang, D.; Fang, L.; Lu, Y.; Du, G. Activation of Nrf2 Attenuates Pulmonary Vascular Remodeling via Inhibiting Endothelial-to-Mesenchymal Transition: An Insight from a Plant Polyphenol. Int. J. Biol. Sci. 2017, 13, 1067–1081. [Google Scholar] [CrossRef]
  77. 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]
  78. Li, W.W.; Cao, A.H.; Sun, F.Y. LncRNA MIAT stimulates oxidative stress in the hypoxic pulmonary hypertension model by sponging miR-29a-5p and inhibiting Nrf2 pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 9022–9029. [Google Scholar] [CrossRef]
  79. 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]
  80. Habtemariam, S. The Nrf2/HO-1 Axis as Targets for Flavanones: Neuroprotection by Pinocembrin, Naringenin, and Eriodictyol. Oxidative Med. Cell. Longev. 2019, 2019, 4724920. [Google Scholar] [CrossRef]
  81. Stenmark, K.R.; Frid, M.; Perros, F. Endothelial-to-Mesenchymal Transition: An Evolving Paradigm and a Promising Therapeutic Target in PAH. Circulation 2016, 133, 1734–1737. [Google Scholar] [CrossRef] [PubMed]
  82. Yet, S.F.; Perrella, M.A.; Layne, M.D.; Hsieh, C.M.; Maemura, K.; Kobzik, L.; Wiesel, P.; Christou, H.; Kourembanas, S.; Lee, M.E. Hypoxia induces severe right ventricular dilatation and infarction in heme oxygenase-1 null mice. J. Clin. Investig. 1999, 103, R23–R29. [Google Scholar] [CrossRef] [PubMed]
  83. Minamino, T.; Christou, H.; Hsieh, C.M.; Liu, Y.; Dhawan, V.; Abraham, N.G.; Perrella, M.A.; Mitsialis, S.A.; Kourembanas, S. Targeted expression of heme oxygenase-1 prevents the pulmonary inflammatory and vascular responses to hypoxia. Proc. Natl. Acad. Sci. USA 2001, 98, 8798–8803. [Google Scholar] [CrossRef]
  84. Goto, J.; Ishikawa, K.; Kawamura, K.; Watanabe, Y.; Matumoto, H.; Sugawara, D.; Maruyama, Y. Heme oxygenase-1 reduces murine monocrotaline-induced pulmonary inflammatory responses and resultant right ventricular overload. Antioxid. Redox Signal. 2002, 4, 563–568. [Google Scholar] [CrossRef]
  85. Zhang, W.H.; Zhang, Y.J.; Liu, C.P.; Yu, B.X.; Lu, W.X. Simvastatin protects against the development of monocrotaline-induced pulmonary hypertension in rats via a heme oxygenase-1-dependent pathway. Exp. Lung Res. 2011, 37, 492–499. [Google Scholar] [CrossRef]
  86. Liang, O.D.; Mitsialis, S.A.; Chang, M.S.; Vergadi, E.; Lee, C.; Aslam, M.; Fernandez-Gonzalez, A.; Liu, X.; Baveja, R.; Kourembanas, S. Mesenchymal stromal cells expressing heme oxygenase-1 reverse pulmonary hypertension. Stem Cells 2011, 29, 99–107. [Google Scholar] [CrossRef]
  87. Hybertson, B.M.; Gao, B.; Bose, S.K.; McCord, J.M. Oxidative stress in health and disease: The therapeutic potential of Nrf2 activation. Mol. Asp. Med. 2011, 32, 234–246. [Google Scholar] [CrossRef]
  88. Voelkel, N.F.; Bogaard, H.J.; Al Husseini, A.; Farkas, L.; Gomez-Arroyo, J.; Natarajan, R. Antioxidants for the treatment of patients with severe angioproliferative pulmonary hypertension? Antioxid. Redox Signal. 2013, 18, 1810–1817. [Google Scholar] [CrossRef] [PubMed]
  89. Hartney, T.; Birari, R.; Venkataraman, S.; Villegas, L.; Martinez, M.; Black, S.M.; Stenmark, K.R.; Nozik-Grayck, E. Xanthine oxidase-derived ROS upregulate Egr-1 via ERK1/2 in PA smooth muscle cells; model to test impact of extracellular ROS in chronic hypoxia. PLoS ONE 2011, 6, e27531. [Google Scholar] [CrossRef]
  90. Coates, D.E.; Zafar, S.; Milne, T.J. Quantitative Real-Time Gene Profiling of Human Alveolar Osteoblasts. Methods Mol. Biol. 2017, 1537, 447–459. [Google Scholar] [CrossRef]
  91. Farrow, K.N.; Lakshminrusimha, S.; Reda, W.J.; Wedgwood, S.; Czech, L.; Gugino, S.F.; Davis, J.M.; Russell, J.A.; Steinhorn, R.H. Superoxide dismutase restores eNOS expression and function in resistance pulmonary arteries from neonatal lambs with persistent pulmonary hypertension. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 295, L979–L987. [Google Scholar] [CrossRef]
  92. Van Rheen, Z.; Fattman, C.; Domarski, S.; Majka, S.; Klemm, D.; Stenmark, K.R.; Nozik-Grayck, E. Lung extracellular superoxide dismutase overexpression lessens bleomycin-induced pulmonary hypertension and vascular remodeling. Am. J. Respir. Cell Mol. Biol. 2011, 44, 500–508. [Google Scholar] [CrossRef]
  93. Sentman, M.L.; Granstrom, M.; Jakobson, H.; Reaume, A.; Basu, S.; Marklund, S.L. Phenotypes of mice lacking extracellular superoxide dismutase and copper- and zinc-containing superoxide dismutase. J. Biol. Chem. 2006, 281, 6904–6909. [Google Scholar] [CrossRef]
  94. Nozik-Grayck, E.; Suliman, H.B.; Majka, S.; Albietz, J.; Van Rheen, Z.; Roush, K.; Stenmark, K.R. Lung EC-SOD overexpression attenuates hypoxic induction of Egr-1 and chronic hypoxic pulmonary vascular remodeling. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 295, L422–L430. [Google Scholar] [CrossRef]
  95. Kamezaki, F.; Tasaki, H.; Yamashita, K.; Tsutsui, M.; Koide, S.; Nakata, S.; Tanimoto, A.; Okazaki, M.; Sasaguri, Y.; Adachi, T.; et al. Gene transfer of extracellular superoxide dismutase ameliorates pulmonary hypertension in rats. Am. J. Respir. Crit. Care Med. 2008, 177, 219–226. [Google Scholar] [CrossRef]
  96. Xu, D.; Guo, H.; Xu, X.; Lu, Z.; Fassett, J.; Hu, X.; Xu, Y.; Tang, Q.; Hu, D.; Somani, A.; et al. Exacerbated pulmonary arterial hypertension and right ventricular hypertrophy in animals with loss of function of extracellular superoxide dismutase. Hypertension 2011, 58, 303–309. [Google Scholar] [CrossRef] [PubMed]
  97. Li, D.; Shao, N.Y.; Moonen, J.R.; Zhao, Z.; Shi, M.; Otsuki, S.; Wang, L.; Nguyen, T.; Yan, E.; Marciano, D.P.; et al. ALDH1A3 Coordinates Metabolism With Gene Regulation in Pulmonary Arterial Hypertension. Circulation 2021, 143, 2074–2090. [Google Scholar] [CrossRef] [PubMed]
  98. Shimoda, L.A.; Sham, J.S.; Liu, Q.; Sylvester, J.T. Acute and chronic hypoxic pulmonary vasoconstriction: A central role for endothelin-1? Respir. Physiol. Neurobiol. 2002, 132, 93–106. [Google Scholar] [CrossRef]
  99. Undem, C.; Rios, E.J.; Maylor, J.; Shimoda, L.A. Endothelin-1 augments Na+/H+ exchange activity in murine pulmonary arterial smooth muscle cells via Rho kinase. PLoS ONE 2012, 7, e46303. [Google Scholar] [CrossRef] [PubMed]
  100. Whitman, E.M.; Pisarcik, S.; Luke, T.; Fallon, M.; Wang, J.; Sylvester, J.T.; Semenza, G.L.; Shimoda, L.A. Endothelin-1 mediates hypoxia-induced inhibition of voltage-gated K+ channel expression in pulmonary arterial myocytes. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 294, L309–L318. [Google Scholar] [CrossRef]
  101. Davie, N.J.; Schermuly, R.T.; Weissmann, N.; Grimminger, F.; Ghofrani, H.A. The science of endothelin-1 and endothelin receptor antagonists in the management of pulmonary arterial hypertension: Current understanding and future studies. Eur. J. Clin. Invest. 2009, 39 (Suppl. S2), 38–49. [Google Scholar] [CrossRef] [PubMed]
  102. Archer, S.; Rich, S. Primary pulmonary hypertension: A vascular biology and translational research “Work in progress”. Circulation 2000, 102, 2781–2791. [Google Scholar] [CrossRef] [PubMed]
  103. Ohkubo, S.; Nakahata, N.; Ohizumi, Y. Thromboxane A2-mediated shape change: Independent of Gq-phospholipase C-Ca2+ pathway in rabbit platelets. Br. J. Pharmacol. 1996, 117, 1095–1104. [Google Scholar] [CrossRef] [PubMed]
  104. Cogolludo, A.; Moreno, L.; Bosca, L.; Tamargo, J.; Perez-Vizcaino, F. Thromboxane A2-induced inhibition of voltage-gated K+ channels and pulmonary vasoconstriction: Role of protein kinase Czeta. Circ. Res. 2003, 93, 656–663. [Google Scholar] [CrossRef] [PubMed]
  105. Yuan, J.X.; Aldinger, A.M.; Juhaszova, M.; Wang, J.; Conte, J.V., Jr.; Gaine, S.P.; Orens, J.B.; Rubin, L.J. Dysfunctional voltage-gated K+ channels in pulmonary artery smooth muscle cells of patients with primary pulmonary hypertension. Circulation 1998, 98, 1400–1406. [Google Scholar] [CrossRef]
  106. Shimoda, L.A.; Polak, J. Hypoxia. 4. Hypoxia and ion channel function. Am. J. Physiol. Cell Physiol. 2011, 300, C951–C967. [Google Scholar] [CrossRef]
  107. Burg, E.D.; Remillard, C.V.; Yuan, J.X. Potassium channels in the regulation of pulmonary artery smooth muscle cell proliferation and apoptosis: Pharmacotherapeutic implications. Br. J. Pharmacol. 2008, 153 (Suppl. S1), S99–S111. [Google Scholar] [CrossRef]
  108. Michelakis, E.D.; McMurtry, M.S.; Wu, X.C.; Dyck, J.R.; Moudgil, R.; Hopkins, T.A.; Lopaschuk, G.D.; Puttagunta, L.; Waite, R.; Archer, S.L. Dichloroacetate, a metabolic modulator, prevents and reverses chronic hypoxic pulmonary hypertension in rats: Role of increased expression and activity of voltage-gated potassium channels. Circulation 2002, 105, 244–250. [Google Scholar] [CrossRef]
  109. Bonnet, S.; Rochefort, G.; Sutendra, G.; Archer, S.L.; Haromy, A.; Webster, L.; Hashimoto, K.; Bonnet, S.N.; Michelakis, E.D. The nuclear factor of activated T cells in pulmonary arterial hypertension can be therapeutically targeted. Proc. Natl. Acad. Sci. USA 2007, 104, 11418–11423. [Google Scholar] [CrossRef]
  110. Pozeg, Z.I.; Michelakis, E.D.; McMurtry, M.S.; Thebaud, B.; Wu, X.C.; Dyck, J.R.; Hashimoto, K.; Wang, S.; Moudgil, R.; Harry, G.; et al. In vivo gene transfer of the O2-sensitive potassium channel Kv1.5 reduces pulmonary hypertension and restores hypoxic pulmonary vasoconstriction in chronically hypoxic rats. Circulation 2003, 107, 2037–2044. [Google Scholar] [CrossRef]
  111. Bolotina, V.M.; Najibi, S.; Palacino, J.J.; Pagano, P.J.; Cohen, R.A. Nitric oxide directly activates calcium-dependent potassium channels in vascular smooth muscle. Nature 1994, 368, 850–853. [Google Scholar] [CrossRef] [PubMed]
  112. Fisslthaler, B.; Popp, R.; Kiss, L.; Potente, M.; Harder, D.R.; Fleming, I.; Busse, R. Cytochrome P450 2C is an EDHF synthase in coronary arteries. Nature 1999, 401, 493–497. [Google Scholar] [CrossRef]
  113. Luke, T.; Maylor, J.; Undem, C.; Sylvester, J.T.; Shimoda, L.A. Kinase-dependent activation of voltage-gated Ca2+ channels by ET-1 in pulmonary arterial myocytes during chronic hypoxia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 302, L1128–L1139. [Google Scholar] [CrossRef] [PubMed]
  114. Sacks, R.S.; Firth, A.L.; Remillard, C.V.; Agange, N.; Yau, J.; Ko, E.A.; Yuan, J.X. Thrombin-mediated increases in cytosolic [Ca2+] involve different mechanisms in human pulmonary artery smooth muscle and endothelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 295, L1048–L1055. [Google Scholar] [CrossRef]
  115. Quayle, J.M.; McCarron, J.G.; Brayden, J.E.; Nelson, M.T. Inward rectifier K+ currents in smooth muscle cells from rat resistance-sized cerebral arteries. Am. J. Physiol. 1993, 265, C1363–C1370. [Google Scholar] [CrossRef]
  116. Zuo, X.; Zong, F.; Wang, H.; Wang, Q.; Xie, W.; Wang, H. Iptakalim, a novel ATP-sensitive potassium channel opener, inhibits pulmonary arterial smooth muscle cell proliferation by downregulation of PKC-alpha. J. Biomed. Res. 2011, 25, 392–401. [Google Scholar] [CrossRef] [PubMed]
  117. Murphy, M.E.; Brayden, J.E. Nitric oxide hyperpolarizes rabbit mesenteric arteries via ATP-sensitive potassium channels. J. Physiol. 1995, 486 Pt 1, 47–58. [Google Scholar] [CrossRef]
  118. Kim, D.; Gnatenco, C. TASK-5, a new member of the tandem-pore K+ channel family. Biochem. Biophys. Res. Commun. 2001, 284, 923–930. [Google Scholar] [CrossRef]
  119. Olschewski, A.; Li, Y.; Tang, B.; Hanze, J.; Eul, B.; Bohle, R.M.; Wilhelm, J.; Morty, R.E.; Brau, M.E.; Weir, E.K.; et al. Impact of TASK-1 in human pulmonary artery smooth muscle cells. Circ. Res. 2006, 98, 1072–1080. [Google Scholar] [CrossRef]
  120. Huetsch, J.C.; Walker, J.; Undem, C.; Lade, J.; Yun, X.; Baksh, S.; Jiang, H.; Lai, N.; Shimoda, L.A. Rho kinase and Na+/H+ exchanger mediate endothelin-1-induced pulmonary arterial smooth muscle cell proliferation and migration. Physiol. Rep. 2018, 6, e13698. [Google Scholar] [CrossRef]
  121. Quinn, D.A.; Du, H.K.; Thompson, B.T.; Hales, C.A. Amiloride analogs inhibit chronic hypoxic pulmonary hypertension. Am. J. Respir. Crit. Care Med. 1998, 157, 1263–1268. [Google Scholar] [CrossRef] [PubMed]
  122. Antoniu, S.A. PDE5 inhibitors for cystic fibrosis: Can they also enhance chloride transport? Evaluation of: Lubamba B, Lecourt H, Lebacq J; et al. Preclinical evidence that sildenafil and vardenafil activate chloride transport in cystic fibrosis. Am J Respir Crit Care Med 2008;177(5):506–15. Expert Opin. Investig. Drugs 2008, 17, 965–968. [Google Scholar] [CrossRef] [PubMed]
  123. Yu, L.; Hales, C.A. Silencing of sodium-hydrogen exchanger 1 attenuates the proliferation, hypertrophy, and migration of pulmonary artery smooth muscle cells via E2F1. Am. J. Respir. Cell Mol. Biol. 2011, 45, 923–930. [Google Scholar] [CrossRef]
  124. Yu, A.Y.; Shimoda, L.A.; Iyer, N.V.; Huso, D.L.; Sun, X.; McWilliams, R.; Beaty, T.; Sham, J.S.; Wiener, C.M.; Sylvester, J.T.; et al. Impaired physiological responses to chronic hypoxia in mice partially deficient for hypoxia-inducible factor 1α. J. Clin. Investig. 1999, 103, 691–696. [Google Scholar] [CrossRef] [PubMed]
  125. Shimoda, L.A.; Semenza, G.L. HIF and the lung: Role of hypoxia-inducible factors in pulmonary development and disease. Am. J. Respir. Crit. Care Med. 2011, 183, 152–156. [Google Scholar] [CrossRef] [PubMed]
  126. Shimoda, L.A. 55th Bowditch Lecture: Effects of chronic hypoxia on the pulmonary circulation: Role of HIF-1. J. Appl. Physiol. 2012, 113, 1343–1352. [Google Scholar] [CrossRef]
  127. Lyle, M.A.; Davis, J.P.; Brozovich, F.V. Regulation of Pulmonary Vascular Smooth Muscle Contractility in Pulmonary Arterial Hypertension: Implications for Therapy. Front. Physiol. 2017, 8, 614. [Google Scholar] [CrossRef]
  128. He, H.; Chai, X.; Zhou, Y.; Pan, X.; Yang, G. Association of Lactate Dehydrogenase with In-Hospital Mortality in Patients with Acute Aortic Dissection: A Retrospective Observational Study. Int. J. Hypertens. 2020, 2020, 1347165. [Google Scholar] [CrossRef]
  129. Shi, Y.; Li, S.; Song, Y.; Liu, P.; Yang, Z.; Liu, Y.; Quan, K.; Yu, G.; Fan, Z.; Zhu, W. Nrf-2 signaling inhibits intracranial aneurysm formation and progression by modulating vascular smooth muscle cell phenotype and function. J. Neuroinflam. 2019, 16, 185. [Google Scholar] [CrossRef]
  130. Zhang, B.; Niu, W.; Xu, D.; Li, Y.; Liu, M.; Wang, Y.; Luo, Y.; Zhao, P.; Liu, Y.; Dong, M.; et al. Oxymatrine prevents hypoxia- and monocrotaline-induced pulmonary hypertension in rats. Free Radic. Biol. Med. 2014, 69, 198–207. [Google Scholar] [CrossRef]
  131. Cartaya, A.E.; Lutz, H.; Maiocchi, S.; Nalesnik, M.; Bahnson, E.M. Delivery of Cinnamic Aldehyde Antioxidant Response Activating nanoParticles (ARAPas) for Vascular Applications. Antioxidants 2021, 10, 709. [Google Scholar] [CrossRef] [PubMed]
  132. Levonen, A.L.; Inkala, M.; Heikura, T.; Jauhiainen, S.; Jyrkkanen, H.K.; Kansanen, E.; Maatta, K.; Romppanen, E.; Turunen, P.; Rutanen, J.; et al. Nrf2 gene transfer induces antioxidant enzymes and suppresses smooth muscle cell growth in vitro and reduces oxidative stress in rabbit aorta in vivo. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 741–747. [Google Scholar] [CrossRef] [PubMed]
  133. Shawky, N.M.; Pichavaram, P.; Shehatou, G.S.; Suddek, G.M.; Gameil, N.M.; Jun, J.Y.; Segar, L. Sulforaphane improves dysregulated metabolic profile and inhibits leptin-induced VSMC proliferation: Implications toward suppression of neointima formation after arterial injury in western diet-fed obese mice. J. Nutr. Biochem. 2016, 32, 73–84. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, P.L.; Liu, J.T.; Kuo, H.F.; Chong, I.W.; Hsieh, C.C. Epigallocatechin gallate attenuates proliferation and oxidative stress in human vascular smooth muscle cells induced by interleukin-1beta via heme oxygenase-1. Mediat. Inflamm. 2014, 2014, 523684. [Google Scholar] [CrossRef]
  135. 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]
  136. Zhou, T.; Zhang, M.; Zhao, L.; Li, A.; Qin, X. Activation of Nrf2 contributes to the protective effect of Exendin-4 against angiotensin II-induced vascular smooth muscle cell senescence. Am. J. Physiol. Cell Physiol. 2016, 311, C572–C582. [Google Scholar] [CrossRef]
  137. Qin, X.; Qiu, C.; Zhao, L. Maslinic acid protects vascular smooth muscle cells from oxidative stress through Akt/Nrf2/HO-1 pathway. Mol. Cell. Biochem. 2014, 390, 61–67. [Google Scholar] [CrossRef]
  138. 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]
  139. Manchope, M.F.; Calixto-Campos, C.; Coelho-Silva, L.; Zarpelon, A.C.; Pinho-Ribeiro, F.A.; Georgetti, S.R.; Baracat, M.M.; Casagrande, R.; Verri, W.A., Jr. Naringenin Inhibits Superoxide Anion-Induced Inflammatory Pain: Role of Oxidative Stress, Cytokines, Nrf-2 and the NO-cGMP-PKG-KATP Channel Signaling Pathway. PLoS ONE 2016, 11, e0153015. [Google Scholar] [CrossRef]
  140. Ko, E.; Kim, D.; Min, D.W.; Kwon, S.H.; Lee, J.Y. Nrf2 regulates cell motility through RhoA-ROCK1 signalling in non-small-cell lung cancer cells. Sci. Rep. 2021, 11, 1247. [Google Scholar] [CrossRef]
  141. Guan, P.; Liang, Y.; Wang, N. Fasudil alleviates pressure overload-induced heart failure by activating Nrf2-mediated antioxidant responses. J. Cell. Biochem. 2018, 119, 6452–6460. [Google Scholar] [CrossRef]
  142. Lambert, M.; Capuano, V.; Olschewski, A.; Sabourin, J.; Nagaraj, C.; Girerd, B.; Weatherald, J.; Humbert, M.; Antigny, F. Ion Channels in Pulmonary Hypertension: A Therapeutic Interest? Int. J. Mol. Sci. 2018, 19, 3162. [Google Scholar] [CrossRef] [PubMed]
  143. Konduri, G.G.; Bakhutashvili, I.; Eis, A.; Gauthier, K.M. Impaired voltage gated potassium channel responses in a fetal lamb model of persistent pulmonary hypertension of the newborn. Pediatr. Res. 2009, 66, 289–294. [Google Scholar] [CrossRef] [PubMed]
  144. Bonnet, S.; Archer, S.L. Potassium channel diversity in the pulmonary arteries and pulmonary veins: Implications for regulation of the pulmonary vasculature in health and during pulmonary hypertension. Pharmacol. Ther. 2007, 115, 56–69. [Google Scholar] [CrossRef] [PubMed]
  145. Morecroft, I.; Murray, A.; Nilsen, M.; Gurney, A.M.; MacLean, M.R. Treatment with the Kv7 potassium channel activator flupirtine is beneficial in two independent mouse models of pulmonary hypertension. Br. J. Pharmacol. 2009, 157, 1241–1249. [Google Scholar] [CrossRef] [PubMed]
  146. Han, Y.L.; Pang, Y.S.; Zeng, M. Change of voltage-gate potassium channel in pulmonary arterial smooth muscle cells of pulmonary hypertension induced by left-to-right shunt in rats. Zhonghua Er Ke Za Zhi = Chin. J. Pediatr. 2011, 49, 901–904. [Google Scholar]
  147. Ishii, T.; Warabi, E.; Siow, R.C.M.; Mann, G.E. Sequestosome1/p62: A regulator of redox-sensitive voltage-activated potassium channels, arterial remodeling, inflammation, and neurite outgrowth. Free Radic. Biol. Med. 2013, 65, 102–116. [Google Scholar] [CrossRef]
  148. Cornfield, D.N.; Resnik, E.R.; Herron, J.M.; Abman, S.H. Chronic intrauterine pulmonary hypertension decreases calcium-sensitive potassium channel mRNA expression. Am. J. Physiol. Lung Cell. Mol. Physiol. 2000, 279, L857–L862. [Google Scholar] [CrossRef]
  149. Sun, X.; Qian, L.L.; Li, Y.; Pfiefer, T.M.; Wang, X.L.; Lee, H.C.; Lu, T. Regulation of KCNMA1 transcription by Nrf2 in coronary arterial smooth muscle cells. J. Mol. Cell. Cardiol. 2020, 140, 68–76. [Google Scholar] [CrossRef]
  150. Antigny, F.; Hautefort, A.; Meloche, J.; Belacel-Ouari, M.; Manoury, B.; Rucker-Martin, C.; Pechoux, C.; Potus, F.; Nadeau, V.; Tremblay, E.; et al. Potassium Channel Subfamily K Member 3 (KCNK3) Contributes to the Development of Pulmonary Arterial Hypertension. Circulation 2016, 133, 1371–1385. [Google Scholar] [CrossRef]
  151. Le Ribeuz, H.; Dumont, F.; Ruellou, G.; Lambert, M.; Balliau, T.; Quatredeniers, M.; Girerd, B.; Cohen-Kaminsky, S.; Mercier, O.; Yen-Nicolay, S.; et al. Proteomic Analysis of KCNK3 Loss of Expression Identified Dysregulated Pathways in Pulmonary Vascular Cells. Int. J. Mol. Sci. 2020, 21, 7400. [Google Scholar] [CrossRef] [PubMed]
  152. Jin, Y.; Xie, W.P.; Wang, H. Hypoxic pulmonary hypertension and novel ATP-sensitive potassium channel opener: The new hope on the horizon. Zhongguo Ying Yong Sheng Li Xue Za Zhi = Zhongguo Yingyong Shenglixue Zazhi = Chin. J. Appl. Physiol. 2012, 28, 510–523. [Google Scholar]
  153. Zhao, X.J.; Zhao, Z.; Yang, D.D.; Cao, L.L.; Zhang, L.; Ji, J.; Gu, J.; Huang, J.Y.; Sun, X.L. Activation of ATP-sensitive potassium channel by iptakalim normalizes stress-induced HPA axis disorder and depressive behaviour by alleviating inflammation and oxidative stress in mouse hypothalamus. Brain Res. Bull. 2017, 130, 146–155. [Google Scholar] [CrossRef] [PubMed]
  154. Kang, Y.; Zhang, G.; Huang, E.C.; Huang, J.; Cai, J.; Cai, L.; Wang, S.; Keller, B.B. Sulforaphane prevents right ventricular injury and reduces pulmonary vascular remodeling in pulmonary arterial hypertension. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H853–H866. [Google Scholar] [CrossRef] [PubMed]
  155. Zhang, G.; Kang, Y.; Cathey, D.; LeBlanc, A.J.; Cai, J.; Cai, L.; Wang, S.; Huang, J.; Keller, B.B. Sulforaphane Does Not Protect Right Ventricular Systolic and Diastolic Functions in Nrf2 Knockout Pulmonary Artery Hypertension Mice. Cardiovasc. Drugs Ther. 2022, 36, 425–436. [Google Scholar] [CrossRef]
  156. Eba, S.; Hoshikawa, Y.; Moriguchi, T.; Mitsuishi, Y.; Satoh, H.; Ishida, K.; Watanabe, T.; Shimizu, T.; Shimokawa, H.; Okada, Y.; et al. The nuclear factor erythroid 2-related factor 2 activator oltipraz attenuates chronic hypoxia-induced cardiopulmonary alterations in mice. Am. J. Respir. Cell Mol. Biol. 2013, 49, 324–333. [Google Scholar] [CrossRef]
  157. Paffett, M.L.; Lucas, S.N.; Campen, M.J. Resveratrol reverses monocrotaline-induced pulmonary vascular and cardiac dysfunction: A potential role for atrogin-1 in smooth muscle. Vasc. Pharmacol. 2012, 56, 64–73. [Google Scholar] [CrossRef]
  158. Yang, D.L.; Zhang, H.G.; Xu, Y.L.; Gao, Y.H.; Yang, X.J.; Hao, X.Q.; Li, X.H. Resveratrol inhibits right ventricular hypertrophy induced by monocrotaline in rats. Clin. Exp. Pharmacol. Physiol. 2010, 37, 150–155. [Google Scholar] [CrossRef]
  159. Wilson, D.N.; Schacht, S.E.; Al-Nakkash, L.; Babu, J.R.; Broderick, T.L. Resveratrol prevents pulmonary trunk remodeling but not right ventricular hypertrophy in monocrotaline-induced pulmonary hypertension. Pathophysiology 2016, 23, 243–250. [Google Scholar] [CrossRef]
  160. Csiszar, A.; Labinskyy, N.; Olson, S.; Pinto, J.T.; Gupte, S.; Wu, J.M.; Hu, F.; Ballabh, P.; Podlutsky, A.; Losonczy, G.; et al. Resveratrol prevents monocrotaline-induced pulmonary hypertension in rats. Hypertension 2009, 54, 668–675. [Google Scholar] [CrossRef]
  161. Xu, D.; Li, Y.; Zhang, B.; Wang, Y.; Liu, Y.; Luo, Y.; Niu, W.; Dong, M.; Liu, M.; Dong, H.; et al. Resveratrol alleviate hypoxic pulmonary hypertension via anti-inflammation and anti-oxidant pathways in rats. Int. J. Med. Sci. 2016, 13, 942–954. [Google Scholar] [CrossRef] [PubMed]
  162. Li, C.; Peng, G.; Long, J.; Xiao, P.; Zeng, X.; Yang, H. Protective effects of resveratrol and SR1001 on hypoxia-induced pulmonary hypertension in rats. Clin. Exp. Hypertens. 2020, 42, 519–526. [Google Scholar] [CrossRef] [PubMed]
  163. Chen, B.; Xue, J.; Meng, X.; Slutzky, J.L.; Calvert, A.E.; Chicoine, L.G. Resveratrol prevents hypoxia-induced arginase II expression and proliferation of human pulmonary artery smooth muscle cells via Akt-dependent signaling. Am. J. Physiol. Lung Cell Mol. Physiol. 2014, 307, L317–L325. [Google Scholar] [CrossRef] [PubMed]
  164. Liu, Y.Y.; Zhang, W.Y.; Wang, C.G.; Huang, J.A.; Jiang, J.H.; Zeng, D.X. Resveratrol prevented experimental pulmonary vascular remodeling via miR-638 regulating NR4A3/cyclin D1 pathway. Microvasc. Res. 2020, 130, 103988. [Google Scholar] [CrossRef]
  165. Shi, W.; Zhai, C.; Feng, W.; Wang, J.; Zhu, Y.; Li, S.; Wang, Q.; Zhang, Q.; Yan, X.; Chai, L.; et al. Resveratrol inhibits monocrotaline-induced pulmonary arterial remodeling by suppression of SphK1-mediated NF-kappaB activation. Life Sci. 2018, 210, 140–149. [Google Scholar] [CrossRef]
  166. Gosemann, J.H.; Friedmacher, F.; Hofmann, A.; Zimmer, J.; Kuebler, J.F.; Rittinghausen, S.; Suttkus, A.; Lacher, M.; Alvarez, L.; Corcionivoschi, N.; et al. Prenatal treatment with rosiglitazone attenuates vascular remodeling and pulmonary monocyte influx in experimental congenital diaphragmatic hernia. PLoS ONE 2018, 13, e0206975. [Google Scholar] [CrossRef]
  167. Liu, Y.; Tian, X.Y.; Huang, Y.; Wang, N. Rosiglitazone Attenuated Endothelin-1-Induced Vasoconstriction of Pulmonary Arteries in the Rat Model of Pulmonary Arterial Hypertension via Differential Regulation of ET-1 Receptors. PPAR Res. 2014, 2014, 374075. [Google Scholar] [CrossRef] [PubMed]
  168. Kim, E.K.; Lee, J.H.; Oh, Y.M.; Lee, Y.S.; Lee, S.D. Rosiglitazone attenuates hypoxia-induced pulmonary arterial hypertension in rats. Respirology 2010, 15, 659–668. [Google Scholar] [CrossRef]
  169. Nisbet, R.E.; Bland, J.M.; Kleinhenz, D.J.; Mitchell, P.O.; Walp, E.R.; Sutliff, R.L.; Hart, C.M. Rosiglitazone attenuates chronic hypoxia-induced pulmonary hypertension in a mouse model. Am. J. Respir. Cell Mol. Biol. 2010, 42, 482–490. [Google Scholar] [CrossRef]
  170. Wang, X.F.; Lu, W.X.; Guo, J.; Li, G.; Zhang, Y.J. Protective effects of rosiglitazone intervention on monocrotaline-induced pulmonary arterial hypertension in rats and related inflammatory mechanisms. Zhonghua Yi Xue Za Zhi 2012, 92, 2144–2147. [Google Scholar]
  171. Crossno, J.T., Jr.; Garat, C.V.; Reusch, J.E.; Morris, K.G.; Dempsey, E.C.; McMurtry, I.F.; Stenmark, K.R.; Klemm, D.J. Rosiglitazone attenuates hypoxia-induced pulmonary arterial remodeling. Am. J. Physiol. Lung Cell Mol. Physiol. 2007, 292, L885–L897. [Google Scholar] [CrossRef] [PubMed]
  172. Grzegorzewska, A.P.; Seta, F.; Han, R.; Czajka, C.A.; Makino, K.; Stawski, L.; Isenberg, J.S.; Browning, J.L.; Trojanowska, M. Dimethyl Fumarate ameliorates pulmonary arterial hypertension and lung fibrosis by targeting multiple pathways. Sci. Rep. 2017, 7, 41605. [Google Scholar] [CrossRef] [PubMed]
  173. Shellenberger, N.W.; Collinsworth, K.K.; Subbiah, S.; Klein, D.; Neary, J.M. Hypoxia induces an increase in intestinal permeability and pulmonary arterial pressures in neonatal Holstein calves despite feeding the flavonoid rutin. J. Dairy Sci. 2020, 103, 2821–2828. [Google Scholar] [CrossRef] [PubMed]
  174. Chen, J.; Jiang, W.; Zhu, F.; Wang, Q.; Yang, H.; Wu, J. Curcumin Improves Pulmonary Hypertension Rats by Regulating Mitochondrial Function. Biomed. Res. Int. 2021, 2021, 1078019. [Google Scholar] [CrossRef] [PubMed]
  175. Lin, Q.; Wang, L.X.; Chen, S.X.; Zhou, X.F.; Huang, X.Y.; Fan, X.F. Effect of curcumin on pulmonary hypertension and wall collagen of pulmonary arterioles of chronic hypoxic hypercapnic rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi 2006, 22, 257–261. [Google Scholar]
  176. Li, J.W.; Chen, P.; Guan, X.Q.; Gong, Y.S.; Yang, P.L. Inhibition of puerarin on pulmonary hypertension in rats with hypoxia and hypercapnia. Zhongguo Zhong Yao Za Zhi 2008, 33, 544–549. [Google Scholar] [PubMed]
  177. Zhang, X.D.; Du, W.; Zhang, C.; Wang, S.J.; Sheng, J.J.; Li, Y.; Si, G.L.; Zhu, D.L. Effect of puerarin on hypoxic pulmonary hypertension and accompanying pulmonary fibrosi. Zhongguo Zhong Yao Za Zhi 2018, 43, 2618–2623. [Google Scholar] [CrossRef]
  178. Zhang, X.; Liu, Q.; Zhang, C.; Sheng, J.; Li, S.; Li, W.; Yang, X.; Wang, X.; He, S.; Bai, J.; et al. Puerarin prevents progression of experimental hypoxia-induced pulmonary hypertension via inhibition of autophagy. J. Pharmacol. Sci. 2019, 141, 97–105. [Google Scholar] [CrossRef]
  179. Chen, D.; Zhang, H.F.; Yuan, T.Y.; Sun, S.C.; Wang, R.R.; Wang, S.B.; Fang, L.H.; Lyu, Y.; Du, G.H. Puerarin-V prevents the progression of hypoxia- and monocrotaline-induced pulmonary hypertension in rodent models. Acta Pharmacol. Sin. 2022, 43, 2325–2339. [Google Scholar] [CrossRef]
  180. Ahmed, L.A.; Obaid, A.A.; Zaki, H.F.; Agha, A.M. Naringenin adds to the protective effect of L-arginine in monocrotaline-induced pulmonary hypertension in rats: Favorable modulation of oxidative stress, inflammation and nitric oxide. Eur. J. Pharm. Sci. 2014, 62, 161–170. [Google Scholar] [CrossRef]
  181. Wande, Y.; Jie, L.; Aikai, Z.; Yaguo, Z.; Linlin, Z.; Yue, G.; Hang, Z. Berberine alleviates pulmonary hypertension through Trx1 and beta-catenin signaling pathways in pulmonary artery smooth muscle cells. Exp. Cell Res. 2020, 390, 111910. [Google Scholar] [CrossRef] [PubMed]
  182. Luo, S.; Kan, J.; Zhang, J.; Ye, P.; Wang, D.; Jiang, X.; Li, M.; Zhu, L.; Gu, Y. Bioactive Compounds From Coptidis Rhizoma Alleviate Pulmonary Arterial Hypertension by Inhibiting Pulmonary Artery Smooth Muscle Cells’ Proliferation and Migration. J. Cardiovasc. Pharmacol. 2021, 78, 253–262. [Google Scholar] [CrossRef]
  183. Zhang, X.; Liu, S.; Sun, Y.; Li, G. Tanshinone IIA alleviates monocrotaline-induced pulmonary hypertension in rats through the PI3K/Akt-eNOS signaling pathway. Nan Fang Yi Ke Da Xue Xue Bao 2022, 42, 718–723. [Google Scholar] [CrossRef] [PubMed]
  184. Jiang, Q.; Chen, Y.; Chen, X.; Zhang, J.; Lu, W.; Wang, J. Tanshinone IIA sulfonate upregulated pulmonary artery smooth muscle peroxisome proliferator-activated receptor γ expression in monocrotaline induced pulmonary hypertension rat. Zhonghua Jie He He Hu Xi Za Zhi 2014, 37, 360–364. [Google Scholar] [PubMed]
  185. Zhang, N.; Dong, M.; Luo, Y.; Zhao, F.; Li, Y. Danshensu prevents hypoxic pulmonary hypertension in rats by inhibiting the proliferation of pulmonary artery smooth muscle cells via TGF-β-smad3-associated pathway. Eur. J. Pharmacol. 2018, 820, 1–7. [Google Scholar] [CrossRef]
  186. Wang, J.; Liu, W.; Lu, W.; Luo, X.; Lin, Y.; Liu, S.; Qian, J.; Zhang, C.; Chen, H.; Li, Y.; et al. Sodium tanshinone IIA sulfonate enhances the BMP9-BMPR2-Smad1/5/9 signaling pathway in rat pulmonary microvascular endothelial cells and human embryonic stem cell-derived endothelial cells. Biochem. Pharmacol. 2022, 199, 114986. [Google Scholar] [CrossRef]
  187. Jiang, Q.; Lu, W.; Yang, K.; Hadadi, C.; Fu, X.; Chen, Y.; Yun, X.; Zhang, J.; Li, M.; Xu, L.; et al. Sodium tanshinone IIA sulfonate inhibits hypoxia-induced enhancement of SOCE in pulmonary arterial smooth muscle cells via the PKG-PPAR-gamma signaling axis. Am. J. Physiol. Cell Physiol. 2016, 311, C136–C149. [Google Scholar] [CrossRef]
  188. Wang, J.; Jiang, Q.; Wan, L.; Yang, K.; Zhang, Y.; Chen, Y.; Wang, E.; Lai, N.; Zhao, L.; Jiang, H.; et al. Sodium tanshinone IIA sulfonate inhibits canonical transient receptor potential expression in pulmonary arterial smooth muscle from pulmonary hypertensive rats. Am. J. Respir. Cell Mol. Biol. 2013, 48, 125–134. [Google Scholar] [CrossRef]
  189. Zheng, L.; Liu, M.; Wei, M.; Liu, Y.; Dong, M.; Luo, Y.; Zhao, P.; Dong, H.; Niu, W.; Yan, Z.; et al. Tanshinone IIA attenuates hypoxic pulmonary hypertension via modulating KV currents. Respir. Physiol. Neurobiol. 2015, 205, 120–128. [Google Scholar] [CrossRef]
  190. Jabbarzadeh Kaboli, P.; Afzalipour Khoshkbejari, M.; Mohammadi, M.; Abiri, A.; Mokhtarian, R.; Vazifemand, R.; Amanollahi, S.; Yazdi Sani, S.; Li, M.; Zhao, Y.; et al. Targets and mechanisms of sulforaphane derivatives obtained from cruciferous plants with special focus on breast cancer-contradictory effects and future perspectives. Biomed. Pharmacother. = Biomed. Pharmacother. 2020, 121, 109635. [Google Scholar] [CrossRef]
  191. Bai, Y.; Wang, X.; Zhao, S.; Ma, C.; Cui, J.; Zheng, Y. Sulforaphane Protects against Cardiovascular Disease via Nrf2 Activation. Oxidative Med. Cell. Longev. 2015, 2015, 407580. [Google Scholar] [CrossRef]
  192. Cho, H.Y.; Miller-DeGraff, L.; Blankenship-Paris, T.; Wang, X.; Bell, D.A.; Lih, F.; Deterding, L.; Panduri, V.; Morgan, D.L.; Yamamoto, M.; et al. Sulforaphane enriched transcriptome of lung mitochondrial energy metabolism and provided pulmonary injury protection via Nrf2 in mice. Toxicol. Appl. Pharmacol. 2019, 364, 29–44. [Google Scholar] [CrossRef] [PubMed]
  193. Zheng, Y.; Tao, S.; Lian, F.; Chau, B.T.; Chen, J.; Sun, G.; Fang, D.; Lantz, R.C.; Zhang, D.D. Sulforaphane prevents pulmonary damage in response to inhaled arsenic by activating the Nrf2-defense response. Toxicol. Appl. Pharmacol. 2012, 265, 292–299. [Google Scholar] [CrossRef]
  194. Singh, K.; Connors, S.L.; Macklin, E.A.; Smith, K.D.; Fahey, J.W.; Talalay, P.; Zimmerman, A.W. Sulforaphane treatment of autism spectrum disorder (ASD). Proc. Natl. Acad. Sci. USA 2014, 111, 15550–15555. [Google Scholar] [CrossRef]
  195. Jiang, Z.; Bian, M.; Wu, J.; Li, D.; Ding, L.; Zeng, Q. Oltipraz Prevents High Glucose-Induced Oxidative Stress and Apoptosis in RSC96 Cells through the Nrf2/NQO1 Signalling Pathway. BioMed Res. Int. 2020, 2020, 5939815. [Google Scholar] [CrossRef]
  196. Choi, S.H.; Kim, Y.M.; Lee, J.M.; Kim, S.G. Antioxidant and mitochondrial protective effects of oxidized metabolites of oltipraz. Expert. Opin. Drug Metab. Toxicol. 2010, 6, 213–224. [Google Scholar] [CrossRef] [PubMed]
  197. Zhu, H.; Itoh, K.; Yamamoto, M.; Zweier, J.L.; Li, Y. Role of Nrf2 signaling in regulation of antioxidants and phase 2 enzymes in cardiac fibroblasts: Protection against reactive oxygen and nitrogen species-induced cell injury. FEBS Lett. 2005, 579, 3029–3036. [Google Scholar] [CrossRef] [PubMed]
  198. Javkhedkar, A.A.; Quiroz, Y.; Rodriguez-Iturbe, B.; Vaziri, N.D.; Lokhandwala, M.F.; Banday, A.A. Resveratrol restored Nrf2 function, reduced renal inflammation, and mitigated hypertension in spontaneously hypertensive rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 308, R840–R846. [Google Scholar] [CrossRef] [PubMed]
  199. Mirhadi, E.; Roufogalis, B.D.; Banach, M.; Barati, M.; Sahebkar, A. Resveratrol: Mechanistic and therapeutic perspectives in pulmonary arterial hypertension. Pharmacol. Res. 2021, 163, 105287. [Google Scholar] [CrossRef] [PubMed]
  200. Mingfeng, D.; Xiaodong, M.; Yue, L.; Taikui, P.; Lei, X.; Ming, L. Effects of PPAR-gamma agonist treatment on LPS-induced mastitis in rats. Inflammation 2014, 37, 1919–1924. [Google Scholar] [CrossRef]
  201. Xin, G.L.L.; Khee, Y.P.; Ying, T.Y.; Chellian, J.; Gupta, G.; Kunnath, A.P.; Nammi, S.; Collet, T.; Hansbro, P.M.; Dua, K.; et al. Current Status on Immunological Therapies for Type 1 Diabetes Mellitus. Curr. Diabetes Rep. 2019, 19, 22. [Google Scholar] [CrossRef]
  202. Rashid, J.; Alobaida, A.; Al-Hilal, T.A.; Hammouda, S.; McMurtry, I.F.; Nozik-Grayck, E.; Stenmark, K.R.; Ahsan, F. Repurposing rosiglitazone, a PPAR-gamma agonist and oral antidiabetic, as an inhaled formulation, for the treatment of PAH. J. Control Release 2018, 280, 113–123. [Google Scholar] [CrossRef] [PubMed]
  203. Rashid, J.; Nozik-Grayck, E.; McMurtry, I.F.; Stenmark, K.R.; Ahsan, F. Inhaled combination of sildenafil and rosiglitazone improves pulmonary hemodynamics, cardiac function, and arterial remodeling. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L119–L130. [Google Scholar] [CrossRef] [PubMed]
  204. Hernandez-Trujillo, Y.; Rodriguez-Esparragon, F.; Macias-Reyes, A.; Caballero-Hidalgo, A.; Rodriguez-Perez, J.C. Rosiglitazone but not losartan prevents Nrf-2 dependent CD36 gene expression up-regulation in an in vivo atherosclerosis model. Cardiovasc. Diabetol. 2008, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  205. Cho, R.L.; Yang, C.C.; Tseng, H.C.; Hsiao, L.D.; Lin, C.C.; Yang, C.M. Haem oxygenase-1 up-regulation by rosiglitazone via ROS-dependent Nrf2-antioxidant response elements axis or PPARgamma attenuates LPS-mediated lung inflammation. Br. J. Pharmacol. 2018, 175, 3928–3946. [Google Scholar] [CrossRef]
  206. Peng, Y.; Chen, L.; Qu, Y.; Wang, D.; Zhu, Y.; Zhu, Y. Rosiglitazone Prevents Autophagy by Regulating Nrf2-Antioxidant Response Element in a Rat Model of Lithium-pilocarpine-induced Status Epilepticus. Neuroscience 2021, 455, 212–222. [Google Scholar] [CrossRef] [PubMed]
  207. Cuadrado, A.; Kugler, S.; Lastres-Becker, I. Pharmacological targeting of GSK-3 and NRF2 provides neuroprotection in a preclinical model of tauopathy. Redox Biol. 2018, 14, 522–534. [Google Scholar] [CrossRef]
  208. Pascale, C.L.; Martinez, A.N.; Carr, C.; Sawyer, D.M.; Ribeiro-Alves, M.; Chen, M.; O’Donnell, D.B.; Guidry, J.J.; Amenta, P.S.; Dumont, A.S. Treatment with dimethyl fumarate reduces the formation and rupture of intracranial aneurysms: Role of Nrf2 activation. J. Cereb. Blood Flow. Metab. 2020, 40, 1077–1089. [Google Scholar] [CrossRef]
  209. Muralidharan, P.; Hayes, D., Jr.; Black, S.M.; Mansour, H.M. Microparticulate/Nanoparticulate Powders of a Novel Nrf2 Activator and an Aerosol Performance Enhancer for Pulmonary Delivery Targeting the Lung Nrf2/Keap-1 Pathway. Mol. Syst. Des. Eng. 2016, 1, 48–65. [Google Scholar] [CrossRef]
  210. El-Agamy, D.S.; El-Harbi, K.M.; Khoshhal, S.; Ahmed, N.; Elkablawy, M.A.; Shaaban, A.A.; Abo-Haded, H.M. Pristimerin protects against doxorubicin-induced cardiotoxicity and fibrosis through modulation of Nrf2 and MAPK/NF-kB signaling pathways. Cancer Manag. Res. 2019, 11, 47–61. [Google Scholar] [CrossRef]
  211. Verma, S.; Kalita, B.; Bajaj, S.; Prakash, H.; Singh, A.K.; Gupta, M.L. A Combination of Podophyllotoxin and Rutin Alleviates Radiation-Induced Pneumonitis and Fibrosis through Modulation of Lung Inflammation in Mice. Front. Immunol. 2017, 8, 658. [Google Scholar] [CrossRef]
  212. Bai, L.; Li, A.; Gong, C.; Ning, X.; Wang, Z. Protective effect of rutin against bleomycin induced lung fibrosis: Involvement of TGF-beta1/alpha-SMA/Col I and III pathway. Biofactors 2020, 46, 637–644. [Google Scholar] [CrossRef] [PubMed]
  213. Tosun, M.; Olmez, H.; Unver, E.; Arslan, Y.K.; Cimen, F.K.; Ozcicek, A.; Aktas, M.; Suleyman, H. Oxidative and pro-inflammatory lung injury induced by desflurane inhalation in rats and the protective effect of rutin. Adv. Clin. Exp. Med. 2021, 30, 941–948. [Google Scholar] [CrossRef] [PubMed]
  214. Li, Q.; Niu, S.; Wang, R.; Li, Y.; Zhang, R.; Zhu, D. Mechanisms that underlie the induction of vasodilation in pulmonary artery by rutin. Int. Angiol. 2012, 31, 557–564. [Google Scholar] [PubMed]
  215. Li, Q.; Qiu, Y.; Mao, M.; Lv, J.; Zhang, L.; Li, S.; Li, X.; Zheng, X. Antioxidant mechanism of Rutin on hypoxia-induced pulmonary arterial cell proliferation. Molecules 2014, 19, 19036–19049. [Google Scholar] [CrossRef] [PubMed]
  216. Balogun, E.; Hoque, M.; Gong, P.; Killeen, E.; Green, C.J.; Foresti, R.; Alam, J.; Motterlini, R. Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element. Biochem. J. 2003, 371, 887–895. [Google Scholar] [CrossRef]
  217. Shen, T.; Jiang, T.; Long, M.; Chen, J.; Ren, D.M.; Wong, P.K.; Chapman, E.; Zhou, B.; Zhang, D.D. A Curcumin Derivative That Inhibits Vinyl Carbamate-Induced Lung Carcinogenesis via Activation of the Nrf2 Protective Response. Antioxid. Redox Signal. 2015, 23, 651–664. [Google Scholar] [CrossRef]
  218. Devadasu, V.R.; Wadsworth, R.M.; Ravi Kumar, M.N. Tissue localization of nanoparticles is altered due to hypoxia resulting in poor efficacy of curcumin nanoparticles in pulmonary hypertension. Eur. J. Pharm. Biopharm. 2012, 80, 578–584. [Google Scholar] [CrossRef]
  219. Kruangtip, O.; Chootip, K.; Temkitthawon, P.; Changwichit, K.; Chuprajob, T.; Changtam, C.; Suksamrarn, A.; Khorana, N.; Scholfield, C.N.; Ingkaninan, K. Curcumin analogues inhibit phosphodiesterase-5 and dilate rat pulmonary arteries. J. Pharm. Pharmacol. 2015, 67, 87–95. [Google Scholar] [CrossRef]
  220. Wei, S.Y.; Chen, Y.; Xu, X.Y. Progress on the pharmacological research of puerarin: A review. Chin. J. Nat. Med. 2014, 12, 407–414. [Google Scholar] [CrossRef]
  221. Chen, X.; Huang, C.; Sun, H.; Hong, H.; Jin, J.; Bei, C.; Lu, Z.; Zhang, X. Puerarin suppresses inflammation and ECM degradation through Nrf2/HO-1 axis in chondrocytes and alleviates pain symptom in osteoarthritic mice. Food Funct. 2021, 12, 2075–2089. [Google Scholar] [CrossRef] [PubMed]
  222. Aimaier, S.; Tao, Y.; Lei, F.; Yupeng, Z.; Wenhui, S.; Aikemu, A.; Maimaitiyiming, D. Protective effects of the Terminalia bellirica tannin-induced Nrf2/HO-1 signaling pathway in rats with high-altitude pulmonary hypertension. BMC Complement. Med. Ther. 2023, 23, 150. [Google Scholar] [CrossRef] [PubMed]
  223. Chang, Z.; Zhang, Q.; Hu, Q.; Liu, Y.; Zhang, L.; Liu, R. Tannins in Terminalia bellirica inhibits hepatocellular carcinoma growth via re-educating tumor-associated macrophages and restoring CD8+T cell function. Biomed. Pharmacother. 2022, 154, 113543. [Google Scholar] [CrossRef]
  224. Feng, J.; Luo, J.; Deng, L.; Zhong, Y.; Wen, X.; Cai, Y.; Li, J. Naringenin-induced HO-1 ameliorates high glucose or free fatty acids-associated apoptosis via PI3K and JNK/Nrf2 pathways in human umbilical vein endothelial cells. Int. Immunopharmacol. 2019, 75, 105769. [Google Scholar] [CrossRef]
  225. Yang, X.J.; Liu, F.; Feng, N.; Ding, X.S.; Chen, Y.; Zhu, S.X.; Yang, L.C.; Feng, X.F. Berberine Attenuates Cholesterol Accumulation in Macrophage Foam Cells by Suppressing AP-1 Activity and Activation of the Nrf2/HO-1 Pathway. J. Cardiovasc. Pharmacol. 2020, 75, 45–53. [Google Scholar] [CrossRef] [PubMed]
  226. Ashrafizadeh, M.; Fekri, H.S.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Therapeutic and biological activities of berberine: The involvement of Nrf2 signaling pathway. J. Cell. Biochem. 2020, 121, 1575–1585. [Google Scholar] [CrossRef] [PubMed]
  227. Zhang, H.S.; Wang, S.Q. Nrf2 is involved in the effect of tanshinone IIA on intracellular redox status in human aortic smooth muscle cells. Biochem. Pharmacol. 2007, 73, 1358–1366. [Google Scholar] [CrossRef] [PubMed]
  228. Wang, J.; Dong, M.Q.; Liu, M.L.; Xu, D.Q.; Luo, Y.; Zhang, B.; Liu, L.L.; Xu, M.; Zhao, P.T.; Gao, Y.Q.; et al. Tanshinone IIA modulates pulmonary vascular response to agonist and hypoxia primarily via inhibiting Ca2+ influx and release in normal and hypoxic pulmonary hypertension rats. Eur. J. Pharmacol. 2010, 640, 129–138. [Google Scholar] [CrossRef] [PubMed]
  229. Clay, A.; Hearle, P.; Schadt, K.; Lynch, D.R. New developments in pharmacotherapy for Friedreich ataxia. Expert Opin. Pharmacother. 2019, 20, 1855–1867. [Google Scholar] [CrossRef]
Figure 1. (A). Nrf2 protein comprises seven conserved area domains Neh1 to Neh7. Neh1 domain is a CNC-bZIP domain which allows Nrf2 to bind ARE through interaction with other factors like small musculoaponeurotic fibrosarcoma (sMAF). The Neh2 domain negatively controls the Nrf2 through its DLG and ETGE motifs. The Neh3 domain recruits chromo-ATPase/helicase DNA-binding protein family member CDH6. The Neh4 and Neh5 domains can interact with the CH3 domain of cyclic adenosine monophosphate (cAMP) response element binding protein (CREB)-binding protein. The Neh6 domain has two motifs, DSGIS and DSAPGS of β-transducin repeat-containing protein (β-TrCP). Neh7 domain interacts with retinoic X receptor alpha (RXR-α). (B). Keap1 protein comprises five domains such as N-terminal region (NTR), Bric-a-Brac domain (BTB), a cysteine-rich intervening region (IVR), Kelch domain, and carboxy-terminal region (CTR). Neh, Nrf2-ECH homology; CNC, cap ‘‘n’’ collar; bZIP, basic-region leucine zipper; Nrf2, nuclear factor E2-related factor 2; RXR-α, retinoid X receptor α; Keap1, Kelch-like ECH associated protein 1. (C). A scheme of the Nrf2 signaling pathway.
Figure 1. (A). Nrf2 protein comprises seven conserved area domains Neh1 to Neh7. Neh1 domain is a CNC-bZIP domain which allows Nrf2 to bind ARE through interaction with other factors like small musculoaponeurotic fibrosarcoma (sMAF). The Neh2 domain negatively controls the Nrf2 through its DLG and ETGE motifs. The Neh3 domain recruits chromo-ATPase/helicase DNA-binding protein family member CDH6. The Neh4 and Neh5 domains can interact with the CH3 domain of cyclic adenosine monophosphate (cAMP) response element binding protein (CREB)-binding protein. The Neh6 domain has two motifs, DSGIS and DSAPGS of β-transducin repeat-containing protein (β-TrCP). Neh7 domain interacts with retinoic X receptor alpha (RXR-α). (B). Keap1 protein comprises five domains such as N-terminal region (NTR), Bric-a-Brac domain (BTB), a cysteine-rich intervening region (IVR), Kelch domain, and carboxy-terminal region (CTR). Neh, Nrf2-ECH homology; CNC, cap ‘‘n’’ collar; bZIP, basic-region leucine zipper; Nrf2, nuclear factor E2-related factor 2; RXR-α, retinoid X receptor α; Keap1, Kelch-like ECH associated protein 1. (C). A scheme of the Nrf2 signaling pathway.
Antioxidants 12 01978 g001
Figure 2. The mechanisms of activating of Nrf2 activity in PAEC dysfunction (A) and PASMC proliferation (B) in pulmonary hypertension. PAEC, pulmonary arterial endothelial cell; PASMC, pulmonary arterial smooth muscle cell.
Figure 2. The mechanisms of activating of Nrf2 activity in PAEC dysfunction (A) and PASMC proliferation (B) in pulmonary hypertension. PAEC, pulmonary arterial endothelial cell; PASMC, pulmonary arterial smooth muscle cell.
Antioxidants 12 01978 g002
Table 1. A list of Nrf2 target genes about antioxidants.
Table 1. A list of Nrf2 target genes about antioxidants.
FunctionGeneExtended Name
CATcatalase
GCLCglutamate-cysteine ligase catalytic subunit
GCLMglutamate-cysteine ligase modifier subunit
GGT1gamma-glutamyltransferase 1
GPX1glutathione peroxidase 1
GPX2glutathione peroxidase 2
GPX4glutathione peroxidase 4
GSR1glutathione reductase
AntioxidantsHO-1heme oxygenase 1
NQO1NAD(P)H quinone dehydrogenase 1
PRDX1peroxiredoxin 1
PRDX6peroxiredoxin 6
SLC7A11solute carrier family 7 member 11
SODsuperoxide dismutase
SRXN1sulfiredoxin 1
TXN1thioredoxin 1
TXNRD1thioredoxin reductase 1
Table 2. Therapeutic potential of Nrf2 inducers in pulmonary vascular remodeling in pulmonary hypertension.
Table 2. Therapeutic potential of Nrf2 inducers in pulmonary vascular remodeling in pulmonary hypertension.
CompoundChemical StructureModelTreatment StrategyEffectsReferences
SFNAntioxidants 12 01978 i001SuHx in miceSFN (0.5 mg/kg 5 days per week) for 28 daysPrevented SuHx-induced RV dysfunction and remodeling, reduced RV inflammation and fibrosis, reduced SuHx-induced pulmonary vascular remodeling, inflammation, and fibrosis.[154]
global Nrf2-knockout mice, SuHx in miceSFN (0.5 mg/kg 5 days per week) for 28 daysPartially or completely reversed SuHx-induced RV diastolic/systolic dysfunction and increased RV systolic pressure.[155]
OltiprazAntioxidants 12 01978 i002Hypoxia miceOltipraz (5, 50, or 500 mg/kg/day) for 3 daysDecreased RVH and pulmonary vascular remodeling.[156]
ResveratrolAntioxidants 12 01978 i003MCT ratresveratrol (3 mg/kg/day) for 14 daysAttenuated established MCT-induced PH indices, including right ventricular systolic pressure, right ventricular hypertrophy, and medial thickening of intrapulmonary.[157]
MCT ratresveratrol (10 and 30 mg/kg) twice daily for 21 days.Attenuated RV hypertrophy, swollen mitochrondria and cardiomyocyte apoptosis.[158]
MCT ratresveratrol (25 mg/kg/day) for 21 days.Reduced the thickness of the pulmonary trunk tunica media.[159]
MCT ratresveratrol (25 mg/kg/day) for 21 daysExerted anti-inflammatory, antioxidant, and antiproliferative effects.[160]
Hypoxia ratresveratrol (40 mg/kg/day) for 28 daysPrevented pulmonary hypertension through its antiproliferation, anti-inflammation and antioxidant effects.[161]
Hypoxia ratresveratrol (40 mg/kg/day) for 21 daysPrevented pulmonary hypertension and RVH.[162]
Hypoxia ratresveratrol (100 mg/kg/day) for 14 daysPrevented proliferation of human pulmonary artery smooth muscle cells and RVH.[163]
MCT ratresveratrol (25 mg/kg/day) for 28 daysPrevented pulmonary vascular remodeling.[164]
MCT ratresveratrol (25 mg/kg/day) for 28 daysInhibited pulmonary arterial remodeling.[165]
RosiglitazoneAntioxidants 12 01978 i004Pregnant rats were treated with nitrofenRosiglitazone (3 mg/kg/day) for 3 daysReduced pulmonary vascular remodeling.[166]
Hypoxia ratrosiglitazone (20 mg/kg per day) with oral gavage for 3 daysInhibited pulmonary artery vasoconstrictive.[167]
Hypoxia ratrosiglitazone (8 mg/kg orally, 5 days/week) for 28 daysReduced chronic hypoxic pulmonary hypertension.[168]
Hypoxia micerosiglitazone (10 mg/kg/d, gavage) for 35 daysReduced pulmonary vascular remodeling and hypertension.[169]
MCT ratrosiglitazone (5, 2.5 mg/kg/day) for 21 daysReduced perivascular inflammation.[170]
Hypoxia ratrosiglitazone (5 mg/kg/day) for 21 daysAttenuated hypoxia-induced pulmonary arterial remodeling.[171]
DMFAntioxidants 12 01978 i005Hypoxia miceDMF (90 mg/kg/day) for 21 daysReversed hemodynamic changes, reducing inflammation, oxidative damage, and fibrosis.[172]
RutinAntioxidants 12 01978 i006Hypoxia calvesglucorhamnoside rutin orally administered for 14 daysLed to pulmonary arteriolar medial hypertrophy and adventitial hyperplasia.[173]
CurcuminAntioxidants 12 01978 i007MCT ratcurcumin (30 mg/kg/day) for 18 daysImproved pulmonary vascular remodeling, promote PASMC apoptosis, and protect mitochondrial function.[174]
Hypoxia ratcurcumin (50 mg/kg/day) administrated for 28 daysDecreased pulmonary arterial pressure, improve pulmonary vessel remodeling and inhibit the deposition of collagen I in pulmonary arterioles.[175]
PuerarinAntioxidants 12 01978 i008Hypoxia ratpuerarin (20 mg/kg/day) for 21 daysImproved pulmonary vascular remodeling.[176]
Hypoxia ratpuerarin intraperitoneal injection, 20 mg/kg/d for 21 daysInhibition of vascular wall thickening pulmonary fibrosis.[177]
Hypoxia ratpuerarin (80 mg/kg/day, orally) for 21 daysReduced autophagy and suppressing cell proliferation.[178]
MCT rat or Hypoxia micepuerarin (10, 30, 100 mg/kg/d, i.g.) for 28 days or puerarin (60 mg/kg/d, i.g.) for 7 daysReduced RVSP and lung injury, improved pulmonary artery blood flow, inhibit inflammatory responses, improved resistance to apoptosis and abnormal proliferation, attenuate right ventricular injury and remodeling, and maintained normal function of the right ventricle.[179]
NaringeninAntioxidants 12 01978 i009MCT ratnaringenin (50 mg/kg) were orally administered daily for 21 daysAlleviated oxidative stress, inflammatory and apoptotic markers.[180]
BerberineAntioxidants 12 01978 i010SuHx in ratsberberine (100 mg per kg day) for 28 daysReversed right ventricular systolic pressure and right ventricular hypertrophy and decrease pulmonary vascular remodeling.[181]
MCT ratberberine (50 mg/kg/d) for 28 daysInhibited pulmonary artery smooth muscle cells’ proliferation and migration.[182]
Tanshinone IIAAntioxidants 12 01978 i011MCT rattanshinone IIA (10 mg/kg) for 14 daysInhibited pulmonary artery intimamedia thickening and muscularization of the pulmonary arterioles.[183]
MCT rattanshinone IIA (30 mg/kg/d) for 21 daysDecreased RVSP, MRVP, RV/(LV+S) and pulmonary vascular remodeling.[184]
Hypoxia rattanshinone IIA (160 mg/kg/d) for 28 daysInhibited proliferation of pulmonary artery smooth muscle cells.[185]
Hypoxia rattanshinone IIA (30 mg/kg/d) for 21 daysNormalized the RVSP and RVH, improved the cardiac output.[186]
Hypoxia rattanshinone IIA (30 mg/kg/d) for 21 daysPrevented hypoxia-mediated increases in intracellular calcium homeostasis and cell proliferation.[187]
MCT rat or Hypoxia micetanshinone IIA (10 mg/kg/d) for 21 daysRelieved RVSP and RVH.[188]
Hypoxia rattanshinone IIA (10 mg/kg/d) for 28 daysRestrained pulmonary artery wall remodeling.[189]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fang, Q.; Bai, Y.; Hu, S.; Ding, J.; Liu, L.; Dai, M.; Qiu, J.; Wu, L.; Rao, X.; Wang, Y. Unleashing the Potential of Nrf2: A Novel Therapeutic Target for Pulmonary Vascular Remodeling. Antioxidants 2023, 12, 1978. https://doi.org/10.3390/antiox12111978

AMA Style

Fang Q, Bai Y, Hu S, Ding J, Liu L, Dai M, Qiu J, Wu L, Rao X, Wang Y. Unleashing the Potential of Nrf2: A Novel Therapeutic Target for Pulmonary Vascular Remodeling. Antioxidants. 2023; 12(11):1978. https://doi.org/10.3390/antiox12111978

Chicago/Turabian Style

Fang, Qin, Yang Bai, Shuiqing Hu, Jie Ding, Lei Liu, Meiyan Dai, Jie Qiu, Lujin Wu, Xiaoquan Rao, and Yan Wang. 2023. "Unleashing the Potential of Nrf2: A Novel Therapeutic Target for Pulmonary Vascular Remodeling" Antioxidants 12, no. 11: 1978. https://doi.org/10.3390/antiox12111978

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