Open access peer-reviewed chapter - ONLINE FIRST

The Role of NRF2 Transcription Factor in Metabolic Syndrome

Written By

Danyelly Bruneska Gondim Martins, Thaysa Walleria Aragão Santos, Maria Helena Menezes Estevam Alves and Rosângela Ferreira Frade de Araújo

Submitted: 06 March 2024 Reviewed: 11 March 2024 Published: 21 May 2024

DOI: 10.5772/intechopen.1005035

The Role of NRF2 Transcription Factor IntechOpen
The Role of NRF2 Transcription Factor Edited by Jose Antonio Morales-Gonzalez

From the Edited Volume

The Role of NRF2 Transcription Factor [Working Title]

Dr. Jose Antonio Morales-Gonzalez, Dr. Eduardo Osiris Madrigal-Santillan, Prof. Marvin A. Soriano-Ursúa and Dr. Ángel Morales-González

Chapter metrics overview

17 Chapter Downloads

View Full Metrics

Abstract

Metabolic syndrome includes several diseases that are associated with metabolic abnormalities such as obesity, dyslipidemia, hypertension, type 2 diabetes, obesity, cardiac diseases, and insulin resistance. In order to maintain cellular homeostasis, it is necessary to regulate the signaling pathways involved in controlling oxidative stress. Nuclear factor erythroid-2 factor 2 (NRF2) is a transcription factor largely expressed in several tissues and cells and participates in the oxidative stress regulation signaling pathways. NRF2 also mediates transcriptional regulation of a variety of target genes to signalize and regulate acute and chronic stress pathways in metabolic syndrome. Deregulation of NRF2 could contribute to a worst prognosis/profile of individuals with metabolic syndrome. Therefore, NRF2 and its activators might play a role in its treatment, highlighted as targets for modulation by pharmacological agents.

Keywords

  • oxidative stress
  • obesity
  • inflammation
  • cardiovascular diseases
  • hypertension

1. Introduction

Metabolic syndrome (MetS) is associated with an increased risk of developing cardiovascular diseases (CVDs) and type 2 diabetes (T2D) and is characterized by several metabolic changes, among which the following stand out: obesity, insulin resistance, hypertension, high levels of triglycerides and fasting glucose, and decreased levels of high-density cholesterol (HDL-c) [1, 2]. The MetS global prevalence can vary from 12.5 to 31.4% being higher in the Eastern Mediterranean Region and Americas [3]. MetS was also associated with a higher risk of incidence of cardiovascular mortality in prospective analyses, increasing for each additional characteristic of MetS observed in the patient [4]. Cardiovascular diseases account for most noncommunicable diseases (NCD) deaths, about 17.9 million people annually, followed by cancers (9.3 million), chronic respiratory diseases (4.1 million), and diabetes (2.0 million including kidney disease deaths caused by diabetes) [5].

Inflammation and oxidative stress are precursors to several complications associated with the findings of MetS, mainly due to increased adiposity, insulin resistance, hypertension, and hyperlipidemia. Oxidative stress is an imbalance between oxidant and antioxidant molecules, leading to changes in signaling pathways, control of redox processes, and cellular damage [6]. The imbalance between prooxidant and antioxidant factors in the body can lead to an increased production of reactive oxygen species (ROS) and nitrogen species (RNS) [7]. ROS play important signaling roles under physiological conditions, once cells deliberately generate ROS through the respiratory chain and other metabolic processes, activating molecular signaling and immune defense mechanisms [8]. Also, ROS regulate several cellular functions such as cell growth, contraction, dilation, migration of vascular cells, and muscle tone [9]. Due to their importance in various physiological functions, certain ROS levels have beneficial effects such as in the process of glucose metabolism, adipogenesis, and adipocyte differentiation [10]. However, increased ROS leads to mitochondrial dysfunction, lipid peroxidation, protein, and DNA damage, compromising antioxidation pathways in metabolic syndrome [11]. Nuclear factor erythroid 2-related factor 2 (NRF2) is known as a master regulator of transcriptional activation of components of antioxidant systems. NRF2 transduces chemical signals to regulate a battery of cytoprotective genes [12]. Target genes include superoxide dismutase (SOD), catalase, glutathione S-transferase (GST), nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase, quinone oxidoreductase 1 (NQO1), γ-glutamate cysteine ligase (GCL), and heme oxygenase-1 (HO-1) [10].

Changes in metabolism and gene expression of redox homeostasis activate NRF2 and other stress response pathways. Furthermore, NRF2 has been identified as a regulator of drug or xenobiotic detoxification, target genes involved in proteasomal and autophagic function, iron, lipid, and carbohydrate metabolism, and DNA repair [13]. In homeostatic conditions, NRF2 binds to its main inhibitor, Kelch-like ECH-associated protein 1 (Keap1), with two specific coupling sites form, a cyclic E3-ubiquitin ligase and Cullin-3/Rbx1 complex. So, NRF2 goes into the ubiquitin proteasome to be degraded. In addition to being an NRF2 inhibitor, Keap 1 acts as a cysteine ​​thiol-rich redox sensor [12]. In the process of oxidative stress, oxidative factors can act and alter the chemical conformation of Keap1 cysteine, thus inhibiting ubiquitination and leading to excessive activation of NRF2 [14]. NRF2 can be phosphorylated by several enzymes, such as protein kinase C (PKC), which phosphorylates it at the Ser40 position, disrupting its interaction with Keap1. ROS can also increase NRF2 translocation to the nucleus, through promotion of the oxidation of cysteine residues in Keap1, allowing the release of NRF2, which will initiate the antioxidant transcription process [15].

Several studies correlate NRF2 with various regulatory functions of genes involved in cell proliferation and differentiation, inflammatory response, and lipid metabolism [12]. It has already confirmed the association between the etiology of MetS, oxidative stress, and chronic inflammation, which are under important control of NRF2 [11]. These data show the importance of investigating the role of NRF2 in this context, which involves changes in metabolic pathways in different organs.

Advertisement

2. NRF2 and metabolic syndrome

The involvement of NRF2 in the development of MetS is based on the interaction of NRF2 with other transcription factors or with the regulation of the expression of molecules that play a role in metabolism. Therefore, the impacts of Nrf2 on MetS require a systemic influence, rather than being controlled by a specific organ. Alterations in NRF2 have been shown to impair adipogenesis through peroxisome proliferator-activated receptor (PPAR), a master regulator of adipocyte differentiation and adipogenesis involved in MetS-related diseases, such as obesity and dyslipidemia [16]. It is known that oxidative stress plays a significant role in inducing insulin resistance, especially in association with the intake of high-fat diets (HFD) and obesity. On the other hand, in pancreatic β cells, excessive oxidative stress suppresses the transcription of insulin-related genes. Once NRF2 regulates redox homeostasis, many studies have been focused on deletion of the NRF2 gene, as well as its overexpression and its pharmacological activators [10].

Liu et al. [17] investigated about the role of NRF2 in nonalcoholic fatty liver disease, a hepatic manifestation of MetS, and suggested that deletion of NRF2 in mice may lead to hepatic insulin resistance due to activation of nuclear factor kappa B (NF-κB), a molecule involved in the phosphorylation of insulin receptor substrates. Consequently, NRF2 activation mitigated the progression of the disease and proved to be a therapeutic strategy. A significant decrease in mitochondrial fatty acid oxidation was observed in the absence of NRF2 and accelerated when NRF2 was constitutively active. These findings have implications for chronic disease conditions, including MetS [18].

2.1 Obesity and dyslipidemia

Commonly recognized for its energy storage function, adipose tissue is also responsible for endocrine functions. Adipocytes can release active biological compounds (adipokines), such as leptin, which act in the control of body weight and play inflammatory functions. This tissue also has different types of cells, such as immune and endothelial cells, which produce different types of hormones, with different functions such as glucose control and lipid metabolism, thus acting as a regulator of metabolic homeostasis [19].

Adipose tissue serves as a pivotal factor in the generation of systemic oxidative stress. The oxidation of fatty acids, within mitochondria and peroxisomes, stands as a significant contributor to oxidative stress, catalyzing the production of ROS. The overconsumption of oxygen also triggers the generation of free radicals within the mitochondrial respiratory chain, intrinsically linked with oxidative phosphorylation [20]. So, it is clear that deregulation in the adipose metabolism can lead to ROS impairment and break down the homeostasis.

Obesity is correlated with low-grade chronic and systemic inflammatory processes. Different studies associate these inflammation mechanisms with severe damage to various organs and tissues and are directly linked to several metabolic diseases [21]. Obese individuals or those with marked weight gain present a phenotypic change in white adipose tissue (WAT) with the appearance of inflamed and dysfunctional adipocytes that secrete pro-inflammatory cytokines [21, 22, 23]. These individuals may also have dyslipidemia, which is characterized by a lipid disorder changes in plasma lipoproteins. These alterations in the lipid profile include reduced levels of high-density lipoprotein (HDL) cholesterol, elevated small dense low-density lipoprotein (LDL), and hypertriglyceridemia [24].

Nonalcoholic fatty liver disease (NAFLD), associated with obesity and T2D, is characterized by liver damage caused by a dysfunctional diet, associated with the excess triglycerides in hepatocytes. So, in obese individuals, factors, such as NAFLD, induce oxidative stress in hepatocytes, thus increasing cellular damage. In addition, hepatic oxidative stress was attenuated with an antioxidant diet in rats with NAFLD, through modulation of the KEAP1/NRF2 pathway [14, 25]. Ma et al. [26] noted that wine devoid of sulfur dioxide effectively alleviated dyslipidemia induced by a HFD, influencing transcription factors, such as NRF2. Then, NRF2 activation enhances lipid metabolism and fatty acid β-oxidation by upregulating crucial genes, such as carnitine palmitoyltransferase I and PARP alpha. Ginsenoside Rg1 (antioxidant molecule extracted from ginseng) significantly improved chronic hepatic inflammatory injuries and fibrosis induced by lipopolysaccharides. This process involves the dissociation of NRF2 from Keap1, subsequently activating NRF2 pathway, which further inhibits NLRP3, NLRP1, and AIM2 inflammasomes. It is important to highlight that the activation of inflammasomes can be mediated by an increase in ROS, contributing to the inflammatory process also present in CVDs [27].

2.2 Hypertension

Reactive oxygen species and changes in radical production can break down redox-sensitive signaling and play an important role in the development of cardiovascular diseases and hypertension, through alterations in the renin-angiotensin system (RAS) and the increased RAS activation and Ang II production lead to an imbalance of redox pathways and to the activation of ROS-producing sources [28].

The harmful effects of the oxidative stress manifest themselves in tissues, inducing endothelial injuries and promoting the enlargement and thickening of the heart walls, especially in the left ventricle. This phenomenon contributes to the development of hypertension and other cardiovascular diseases (CVD) [29]. Increased peripheral resistance, a hallmark of arterial hypertension, arises mainly from structural and functional changes in small resistance arteries, manifested through vascular remodeling and endothelial dysfunction. These changes can result in narrowing the lumen due to a reduced outer diameter (eutrophic remodeling) or an invasion of the lumen by a thicker layer (hypertrophic remodeling) [30].

The interconnection between endothelial function and vascular remodeling is primarily attributed to the decreased availability of nitric oxide (NO), resulting in the promotion of predominantly hypertrophic vascular remodeling [31]. NO is an important vasodilator produced in the endothelium by the catalytic conversion of L-arginine into L-citrulline, through the enzyme endothelial nitric oxide synthase (eNOS). A series of cofactors are involved in this reaction, and their disruption leads to a monomeric form of the enzyme, which is uncoupled, and, instead of NO, superoxide is formed. In both hypertension and atherosclerosis, this uncoupled form of eNOS has been found. Increasingly, evidence indicates that NRF2 activation can trigger vasoprotective effects, reducing the bioavailability of ROS and increasing NO levels by eNOS [15]. In a study using rat pheochromocytoma cells, it was observed that NO can activate NRF2 by S-nitrosylation of Keap1 and, alternatively, by PKC-catalyzed phosphorylation of NRF2 [32]. Therefore, downregulation of NRF2 in hypertension may contribute to decreased antioxidant capacity, resulting in increased oxidative stress and consequent vascular dysfunction.

2.3 Diabetes

Oxidative stress is also causal in the development of β cell dysfunction, diabetes and insulin resistance [33, 34]. Diabetic patients have been shown increased cellular levels of ROS and ROS-induced DNA damage through a variety of mechanisms. An increased glycemic load in diabetes overwhelms the Krebs cycle, resulting in the inhibition of electron transfer within the mitochondrial membrane. Then, the accumulation of free radicals and ROS could lead to diabetic alterations such as the depletion of natural antioxidant molecules and damage to vascular cells, as well as alterations in gene and protein expression, blood flow, and endothelial cell permeability [33, 35, 36].

The excess of glucose is related to ROS dependent and alterations in NRF2 concentration are involved in the diabetes physiopathology and its complications. In both mouse placental tissue and human placenta cells, both increase and decrease of NRF2 were linked to gestational diabetes and implications for the health of the mother and offspring [37]. Proteins involved in oxidative stress, NRF2-linked phase II enzyme detoxification, and glutathione synthesis showed alterations in gestational diabetes, leading to increased oxidative stress, mitochondrial superoxide generation, protein carbonylation, and DNA damage [38]. In addition, alterations to NRF2 function have been implicated in the onset and progression of type 1 diabetes, through inhibition of the Keap1/NRF2 signaling pathway [39].

Regarding T2DM, the NRF2 has been shown to play a role in mediating diabetic organ-related complications. An important organ-related complication is the pancreatic β cell dysfunction and death induced by ROS. In mice, the genetic NRF2 induction reduced β cell damage, whereas the genetic NRF2 depletion markedly enhanced β cell damage [40, 41]. In addition to the pancreas, NRF2 has a role in diabetes in adipose tissue, skeletal muscle, heart, liver, and kidneys, with the complication of diabetic nephropathy [42].

The treatment of patients with T2DM is challenging since there is no cure available, but glycemia can be controlled by pharmacological therapy [43]. The NRF2 activation results in the activation of other antioxidant enzymes and cytoprotective genes, making it an attractive therapeutic target for treatment of diabetic complications [44]. Antioxidant therapies are comparatively more effective for treating diabetes from which NRF2 and its related factors are one of the potential activators. Moreover, antioxidant-based treatments serve as potential therapeutics for protection against the enhanced oxidative stress, as well as inflammation in diabetes mellitus [45, 46, 47, 48, 49]. The diabetic cardiomyopathy induced by T2DM has two main pathogenesis: the oxidative stress status in cardiomyocytes and lipid toxicity [50]. In the late diabetic cardiomyopathy stage, the Nrf2 is significantly downregulated, and then therapies focused on restoring the expression and activity of NRF2 could prevent diabetes-associated injuries in cardiomyocytes [50, 51, 52].

2.4 Coronary artery disease

Coronary artery disease (CAD) presents several common complications such as arrhythmias, acute coronary syndrome, congestive heart failure, mitral regurgitation, pericarditis, aneurysm formation, and mural thrombi. Recent research has challenged the protective effects of HDL due to difficulties in raising serum levels and is now focusing on triglyceride-rich lipoproteins (TGRL), in addition to LDL, as causes of atherosclerosis. Traditional risk factors for coronary artery disease have been well combated over the years, such as smoking, LDL levels, and high blood pressure, for example, due to population guidelines and low-cost therapies. However, TGRL are examples of emerging targets as they have greater correlations with the inflammatory state than LDL itself. TGRL cause inflammation, considering, for example, the content of one of its components, apolipoprotein C-III [53, 54].

The pathophysiology of CAD involves both the inflammatory response of the vascular endothelium and oxidative stress. Activation of the NRF2 pathway is associated with a reduction in myocardial infarct size and a delay in the progression to heart failure. The use of NRF2 activation also demonstrated protective effects against ischemia/reperfusion injury. Oxidative stress and inflammation are closely interconnected processes with oxidative stress-induced inflammation predominantly driven by NF-κB activation. It plays a crucial role in regulating the inflammatory response by controlling the gene expression of inflammatory cytokines, which are the main contributors to cardiovascular diseases in humans [54, 55].

Several mechanisms lead to the complex connection between the NRF2 and NF-kB pathways, which occur by inhibiting each other. IκB-α is an inhibitor of the NF-κB transcription factor as it prevents, for example, the binding of NF-kB to DNA, making its action impossible. NRF2 prevents proteasomal degradation of IκB-α and inhibits nuclear translocation of NF-κB. This occurs due to the increase in HO-1 levels stimulated by NRF2 and subsequent increase in the expression of phase II enzymes that block IκB degradation. NRF2 acts to decrease the expression of pro-inflammatory cytokines such as vascular cell adhesion molecule-1 (VCAM-1), monocyte chemoattractant protein-1 (MCP-1), and tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and some interleukins, all involved in atherosclerosis. Understanding this intricate interaction is essential to unravel the inflammatory mechanisms involved in the pathogenesis of CAD and develop more specific therapeutic interventions [56, 57, 58].

Advertisement

3. NRF2 and therapy

Most pharmacological activators of NRF2 are electrophilic substances capable of modifying cysteine residues present in Keap1, ultimately resulting in changes in its conformation through oxidation or alkylation processes [15]. Some of these new therapies and medications have been considered to control MetS through regulation of Keap1 protein and NRF2 (Figure 1).

Figure 1.

Effect of oxidative stress and antioxidant drugs used in the treatment of metabolic syndrome.

Cannabidiol (CBD) is a nontoxic compound derived from cannabis, known for its anti-inflammatory, anti-apoptotic, and antioxidant properties. Interest in the use of CBD has grown, including studies focusing on cardiovascular diseases. In mice, it was observed increased translocation of NRF2 from the cytoplasm to the nucleus via Keap1 inhibition, recognized as a beneficial effect of CBD, preventing acute myocardial infarction after exhaustive exercise [59].

In an environment of oxidative stress NRF2 is released from Keap1, allowing the nuclear translocation of NRF2 and the increase of expression of antioxidant proteins. The action of drugs on MetS components (obesity, diabetes, hypertension, and coronary artery disease) may lead to the decrease of Keap1, thus increasing the activation of NRF2 and HO-1, NRF2 nuclear translocation, and activation of antioxidant protein expression. The drugs also act to reduce inflammatory cytokines and NF-Kb, which in normal oxidative conditions act to inhibit NRF2, impairing translocation.

Resveratrol, administered to hypertensive rats, improved NRF2 activity by combating oxidative stress in proximal tubular epithelial cells, improving inflammation, and attenuating the development of hypertension [60]. Lopes et al. [30] observed a vasoprotective function of NRF2 in hypertension when evaluating hypertensive rats under conditions of vascular oxidative stress related to vascular dysfunction. Bardoxolone and L-sulforaphane activate NRF2 when administered to animals, reducing the formation of ROS that are induced by angiotensin II, and reestablishing the endothelial function in the cells. The impact of oxidative stress, induced by ANG II infusion, was also investigated by Wang et al. [61], in mice, analyzing the effects of treatment with tert-butylhydroquinone (tBHQ), that triggered the activation of NRF2, preventing increased oxidative stress, microvascular ROS, endothelial dysfunction, enhanced microvascular remodeling, contractility, and hypertension. In a study by Gao et al. [62], Qing brick tea (QBT), a traditional and popular Chinese drink, was administered to mice with obesity induced by monosodium glutamate, proving effective in protecting against metabolic syndrome. QBT treatment attenuated oxidative stress and IR and markedly stimulated the NRF2/HO-1 and insulin signaling pathways. A study based on the transcriptome network analysis after using methylglyoxal, conducted on the visceral adipose tissue of hereditary hypertriglyceridemic rats, a nonobese model of metabolic syndrome, revealed that the downregulation of NRF2 expression resulted in the over-representation of genes associated with insulin signaling, lipid metabolism, and angiogenesis [63].

Some drugs already used to control glycemia act regulating the activity of NRF2. Among these, Sitagliptin is a drug indicated for use combined with metformin or sulfonylurea, for treating T2DM. Recent studies indicate that Sitagliptin can also have a regulatory effect on autophagy and protect the cardiovascular system. In the cell, it acts to reduce NF-kB, which, in turn, regulates inflammatory cytokines and helps to inhibit the overexpression of NRF2 [64, 65]. Metformin is the first-line pharmacotherapy for T2DM. Studies carried out with mice to understand the extent of the drug’s use have shown that its long-term use not only helps in the treatment of T2DM but also acts as a cardioprotector against heart diseases associated with MetS. These studies indicate that Metformin increases the expression of Nrf2, inducing an improvement in the nuclear translocation of Nrf2 [65, 66]. It reduces excessive ROS generation, limiting cell apoptosis, and increasing signaling through the ROS-mediated PI3K/Akt and NRF2/HO-1 pathways and the levels of antioxidants [66].

Diabetic nephropathy is an important diabetes complication characterized by inflammatory response and renal dysfunction [48]. Sulforaphane (SFN) is an organosulfur compound that exhibits anticancer and antidiabetic properties and has an indirect antioxidant effect inducing expression of several enzymes via the Keap1/NRF2 pathway [67, 68]. Then, SFN has particular interest in the role via induction of NRF2 of preventing diabetic nephropathy and renal damage [69]. The upregulation of NRF2 of diabetic nephropathy mice model, by treatment with fenofibrate, led to regulation of other antioxidant molecules and delayed the progression of diabetic nephropathy [46]. Baicalin (BAI), a flavonoid component extracted from the traditional Chinese medicine S. baicalensis, showed to inhibit oxidative stress in hyperglycemic environments through regulation of NRF2 pathways. It also decreases blood lipids and inflammation markers and suppresses the excessive production of ROS in cardiovascular diseases [50, 70, 71].

Advertisement

4. Conclusion

Considering the role of NRF2 in activating genes involved in combating oxidative stress and inflammation, it is clear that its inhibition, which often occurs due to increased Keap1 activity, can significantly increase the chances of developing metabolic syndrome. This association is justified by the participation of these genes, activated by this important transcription factor, in several metabolic pathways related especially to endothelial function, insulin signaling, and lipid catabolism. So, it impacts the set of clinical findings that constitute metabolic syndrome. Therefore, several drugs that act on NRF2 activation are promising for improving or preventing chronic metabolic diseases that are not only common but also increasing, assumed as one of the diseases of the century.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Simmons RK, Alberti KG, Gale EA, Colagiuri S, Tuomilehto J, Qiao Q , et al. The metabolic syndrome: Useful concept or clinical tool? Report of a WHO expert consultation. Diabetologia. 2010;53(4):600-605. DOI: 10.1007/s00125-009-1620-4
  2. 2. Saklayen MG. The global epidemic of the metabolic syndrome. Current Hypertension Reports. 2018;20(2):12. DOI: 10.1007/s11906-018-0812-z
  3. 3. Noubiap JJ, Nansseu JR, Lontchi-Yimagou E, Nkeck JR, Nyaga UF, Ngouo AT, et al. Geographic distribution of metabolic syndrome and its components in the general adult population: A meta-analysis of global data from 28 million individuals. Diabetes Research and Clinical Practice. 2022;188:109924. DOI: 10.1016/j.diabres.2022.109924
  4. 4. Guembe MJ, Fernandez-Lazaro CI, Sayon-Orea C, Toledo E, Moreno-Iribas C, RIVANA Study Investigators. Risk for cardiovascular disease associated with metabolic syndrome and its components: A 13-year prospective study in the RIVANA cohort. Cardiovascular Diabetology. 2020;19(1):195. DOI: 10.1186/s12933-020-01166-6
  5. 5. WHO. World Health Organization (WHO). 2023. Available from: https://www.who.int/news-room/fact-sheets/detail/noncommunicable-diseases [Accessed: Jan 20, 2024]
  6. 6. Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The role of sirtuins in antioxidant and redox signaling. Antioxidants & Redox Signaling. 2018;28(8):643-661. DOI: 10.1089/ars.2017.7290
  7. 7. Cadenas S. Mitochondrial uncoupling, ROS generation and cardioprotection. Biochimica et Biophysica Acta - Bioenergetics. 2018;1859(9):940-950. DOI: 10.1016/j.bbabio.2018.05.019
  8. 8. Goszcz K, Deakin SJ, Duthie GG, Stewart D, Leslie SJ, Megson IL. Antioxidants in cardiovascular therapy: Panacea or false hope? Frontiers in Cardiovascular Medicine. 2015;2:29. DOI: 10.3389/fcvm.2015.00029
  9. 9. Montezano AC, Tsiropoulou S, Dulak-Lis M, Harvey A, Camargo Lde L, Touyz RM. Redox signaling, Nox5 and vascular remodeling in hypertension. Current Opinion in Nephrology and Hypertension. 2015;24(5):425-433. DOI: 10.1097/MNH.0000000000000153
  10. 10. Zhang Z, Zhou S, Jiang X, Wang YH, Li F, Wang YG, et al. The role of the Nrf2/Keap1 pathway in obesity and metabolic syndrome. Reviews in Endocrine & Metabolic Disorders. 2015;16(1):35-45. DOI: 10.1007/s11154-014-9305-9
  11. 11. Masenga SK, Kabwe LS, Chakulya M, Kirabo A. Mechanisms of oxidative stress in metabolic syndrome. International Journal of Molecular Sciences. 2023;24(9):7898. DOI: 10.3390/ijms24097898
  12. 12. Yamamoto M, Kensler TW, Motohashi H. The KEAP1-NRF2 system: A thiol-based sensor-effector apparatus for maintaining redox homeostasis. Physiological Reviews. 2018;98(3):1169-1203. DOI: 10.1152/physrev.00023.2017
  13. 13. Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biology. 2019;23:101107. DOI: 10.1016/j.redox.2019.101107. Epub 2019 Jan 11
  14. 14. Lu L, Fu J, Sun J, Liu D, Chen C, Wang H, et al. Is Nrf2-ARE a potential target in NAFLD mitigation? Current Opinion in Toxicology. 2019;13:35-44. DOI: 10.1016/j.cotox.2018.12.005
  15. 15. Tanase DM, Apostol AG, Costea CF, Tarniceriu CC, Tudorancea I, Maranduca MA, et al. Oxidative stress in arterial hypertension (HTN): The nuclear factor erythroid factor 2-related factor 2 (Nrf2) pathway, implications and future perspectives. Pharmaceutics. 2022;14(3):534. DOI: 10.3390/pharmaceutics14030534
  16. 16. Pi J, Leung L, Xue P, Wang W, Hou Y, Liu D, et al. Deficiency in the nuclear factor E2-related factor-2 transcription factor results in impaired adipogenesis and protects against diet-induced obesity. The Journal of Biological Chemistry. 2010;285(12):9292-9300. DOI: 10.1074/jbc.M109.093955
  17. 17. Liu Z, Dou W, Ni Z, Wen Q , Zhang R, Qin M, et al. Deletion of Nrf2 leads to hepatic insulin resistance via the activation of NF-κB in mice fed a high-fat diet. Molecular Medicine Reports. 2016;14(2):1323-1331. DOI: 10.3892/mmr.2016.5393
  18. 18. Ludtmann MH, Angelova PR, Zhang Y, Abramov AY, Dinkova-Kostova AT. Nrf2 affects the efficiency of mitochondrial fatty acid oxidation. The Biochemical Journal. 2014;457(3):415-424. DOI: 10.1042/BJ20130863
  19. 19. Li S, Eguchi N, Lau H, Ichii H. The role of the Nrf2 signaling in obesity and insulin resistance. International Journal of Molecular Sciences. 2020;21(18):6973. DOI: 10.3390/ijms21186973
  20. 20. Fernández-Sánchez A, Madrigal-Santillán E, Bautista M, Esquivel-Soto J, Morales-González A, Esquivel-Chirino C, et al. Inflammation, oxidative stress, and obesity. International Journal of Molecular Sciences. 2011;12(5):3117-3132. DOI: 10.3390/ijms12053117
  21. 21. Kawai T, Autieri MV, Scalia R. Adipose tissue inflammation and metabolic dysfunction in obesity. American Journal of Physiology. Cell Physiology. 2021;320(3):C375-C391. DOI: 10.1152/ajpcell.00379.2020
  22. 22. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860-867. DOI: 10.1038/nature05485
  23. 23. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542(7640):177-185. DOI: 10.1038/nature21363
  24. 24. Vekic J, Stefanovic A, Zeljkovic A. Obesity and dyslipidemia: A review of current evidence. Current Obesity Reports. 2023;12(3):207-222. DOI: 10.1007/s13679-023-00518-z
  25. 25. Wang Y, Liu B, Wu P, Chu Y, Gui S, Zheng Y, et al. Dietary selenium alleviated mouse liver oxidative stress and NAFLD induced by obesity by regulating the KEAP1/NRF2 pathway. Antioxidants (Basel). 2022;11(2):349. DOI: 10.3390/antiox11020349
  26. 26. Ma Y, Yu K, Wang N, Xiao X, Leng Y, Fan J, et al. Sulfur dioxide-free wine with polyphenols promotes lipid metabolism via the Nrf2 pathway and gut microbiota modulation. Food Chemistry: X. 2023;21:101079. DOI: 10.1016/j.fochx.2023.101079
  27. 27. Zhou H, Liu Y, Su Y, Ji P, Kong L, Sun R, et al. Ginsenoside Rg1 attenuates lipopolysaccharide-induced chronic liver damage by activating Nrf2 signaling and inhibiting inflammasomes in hepatic cells. Journal of Ethnopharmacology. 2024;324:117794. DOI: 10.1016/j.jep.2024.117794
  28. 28. Dovinova I, Kvandová M, Balis P, Gresova L, Majzunova M, Horakova L, et al. The role of Nrf2 and PPARgamma in the improvement of oxidative stress in hypertension and cardiovascular diseases. Physiological Research. 2020;69(Suppl. 4):S541-S553. DOI: 10.33549/physiolres.934612
  29. 29. Liu QQ , Ren K, Liu SH, Li WM, Huang CJ, Yang XH. MicroRNA-140-5p aggravates hypertension and oxidative stress of atherosclerosis via targeting Nrf2 and Sirt2. International Journal of Molecular Medicine. 2019;43:839-849. DOI: 10.3892/ijmm.2018.3996
  30. 30. Lopes RA, Neves KB, Tostes RC, Montezano AC, Touyz RM. Downregulation of nuclear factor erythroid 2-related factor and associated antioxidant genes contributes to redox-sensitive vascular dysfunction in hypertension. Hypertension. 2015;66(6):1240-1250. DOI: 10.1161/hypertensionaha.115.06163
  31. 31. Masi S, Georgiopoulos G, Chiriacò M, Grassi G, Seravalle G, Savoia C, et al. The importance of endothelial dysfunction in resistance artery remodelling and cardiovascular risk. Cardiovascular Research. 2020;116(2):429-437. DOI: 10.1093/cvr/cvz096
  32. 32. Um HC, Jang JH, Kim DH, Lee C, Surh YJ. Nitric oxide activates Nrf2 through S-nitrosylation of Keap1 in PC12 cells. Nitric Oxide. 2011;25(2):161-168. DOI: 10.1016/j.niox.2011.06.001
  33. 33. Nowotny K, Jung T, Höhn A, Weber D, Grune T. Advanced glycation end products and oxidative stress in type 2 diabetes mellitus. Biomolecules. 2015;5(1):194-222. DOI: 10.3390/biom5010194
  34. 34. Henriksen EJ, Diamond-Stanic MK, Marchionne EM. Oxidative stress and the etiology of insulin resistance and type 2 diabetes. Free Radical Biology & Medicine. 2011;51(5):993-999. DOI: 10.1016/j.freeradbiomed.2010.12.005
  35. 35. David JA, Rifkin WJ, Rabbani PS, Ceradini DJ. The Nrf2/Keap1/ARE pathway and oxidative stress as a therapeutic target in type II diabetes mellitus. Journal Diabetes Research. 2017;2017:4826724. DOI: 10.1155/2017/4826724
  36. 36. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circulation Research. 2010;107(9):1058-1070. DOI: 10.1161/CIRCRESAHA.110.223545
  37. 37. Zhang C, Yang Y, Chen R, Wei Y, Feng Y, Zheng W, et al. Aberrant expression of oxidative stress related proteins affects the pregnancy outcome of gestational diabetes mellitus patients. American Journal of Translational Research. 2019;11(1):269-279
  38. 38. Cheng X, Chapple SJ, Patel B, Puszyk W, Sugden D, Yin X, et al. Gestational diabetes mellitus impairs Nrf2-mediated adaptive antioxidant defenses and redox signaling in fetal endothelial cells in utero. Diabetes. 2013;62(12):4088-4097. DOI: 10.2337/db13-0169
  39. 39. Lou Y, Kong M, Li L, Hu Y, Zhai W, Qi X, et al. Inhibition of the Keap1/Nrf2 signaling pathway significantly promotes the progression of type 1 diabetes mellitus. Oxidative Medicine and Cellular Longevity. 2021;2021:7866720. DOI: 10.1155/2021/7866720
  40. 40. Yagishita Y, Fukutomi T, Sugawara A, Kawamura H, Takahashi T, Pi J, et al. Nrf2 protects pancreatic β-cells from oxidative and nitrosative stress in diabetic model mice. Diabetes. 2014;63(2):605-618. DOI: 10.2337/db13-0909
  41. 41. Uruno A, Furusawa Y, Yagishita Y, Fukutomi T, Muramatsu H, Negishi T, et al. The Keap1-Nrf2 system prevents onset of diabetes mellitus. Molecular and Cellular Biology. 2013;33(15):2996-3010. DOI: 10.1128/MCB.00225-13
  42. 42. Dodson M, Shakya A, Anandhan A, Chen J, Garcia JGN, Zhang DD. NRF2 and diabetes: The good, the bad, and the complex. Diabetes. 2022;71(12):2463-2476. DOI: 10.2337/db22-0623
  43. 43. Artasensi A, Pedretti A, Vistoli G, Fumagalli L. Type 2 diabetes mellitus: A review of multi-target drugs. Molecules. 2020;25(8):1987. DOI: 10.3390/molecules25081987
  44. 44. Tan SM, de Haan JB. Combating oxidative stress in diabetic complications with Nrf2 activators: How much is too much? Redox Report. 2014;19(3):107-117. DOI: 10.1179/1351000214Y.0000000087
  45. 45. Zheng H, Whitman SA, Wu W, Wondrak GT, Wong PK, Fang D, et al. Therapeutic potential of Nrf2 activators in streptozotocin-induced diabetic nephropathy. Diabetes. 2011;60(11):3055-3066. DOI: 10.2337/db11-0807
  46. 46. Yang Y, Chen G, Cheng X, Teng Z, Cai X, Yang J, et al. Therapeutic potential of digitoflavone on diabetic nephropathy: Nuclear factor erythroid 2-related factor 2-dependent anti-oxidant and anti-inflammatory effect. Scientific Reports. 2015;5:12377. DOI: 10.1038/srep12377
  47. 47. Wu P, Yan Y, Ma LL, Hou BY, He YY, Zhang L, et al. Effects of the Nrf2 protein modulator salvianolic acid a alone or combined with metformin on diabetes-associated macrovascular and renal injury. The Journal of Biological Chemistry. 2016;291(42):22288-22301. DOI: 10.1074/jbc.M115.712703
  48. 48. Li S, Zheng L, Zhang J, Liu X, Wu Z. Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. Free Radical Biology & Medicine. 2021;162:435-449. DOI: 10.1016/j.freeradbiomed.2020.10.323
  49. 49. Dwivedi DK, Jena GB. Dimethyl fumarate-mediated Nrf2/ARE pathway activation and glibenclamide-mediated NLRP3 inflammasome cascade inhibition alleviate type II diabetes-associated fatty liver in rats by mitigating oxidative stress and inflammation. Journal of Biochemical and Molecular Toxicology. 2023;37(7):e23357. DOI: 10.1002/jbt.23357
  50. 50. Li R, Liu Y, Shan YG, Gao L, Wang F, Qiu CG. Bailcalin protects against diabetic cardiomyopathy through Keap1/Nrf2/AMPK-mediated antioxidative and lipid-lowering effects. Oxidative Medicine and Cellular Longevity. 2019;2019:3206542. DOI: 10.1155/2019/3206542
  51. 51. He X, Kan H, Cai L, Ma Q. Nrf2 is critical in defense against high glucose-induced oxidative damage in cardiomyocytes. Journal of Molecular and Cellular Cardiology. 2009;46(1):47-58. DOI: 10.1016/j.yjmcc.2008.10.007
  52. 52. Tan Y, Ichikawa T, Li J, Si Q , Yang H, Chen X, 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(2):625-633. DOI: 10.2337/db10-1164
  53. 53. Libby P. The changing landscape of atherosclerosis. Nature. 2021;592(7855):524-533. DOI: 10.1038/s41586-021-03392-8
  54. 54. Casper E. The crosstalk between Nrf2 and NF-κB pathways in coronary artery disease: Can it be regulated by SIRT6? Life Sciences. 2023;330:122007. DOI: 10.1016/j.lfs.2023.122007
  55. 55. Battino M, Giampieri F, Pistollato F, Sureda A, de Oliveira MR, Pittalà V, et al. Nrf2 as regulator of innate immunity: A molecular Swiss army knife! Biotechnology Advances. 2018;36(2):358-370. DOI: 10.1016/j.biotechadv.2017.12.012
  56. 56. Saha S, Buttari B, Panieri E, Profumo E, Saso L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules. 2020;25(22):5474. DOI: 10.3390/molecules25225474
  57. 57. Coto E, Reguero JR, Avanzas P, Pascual I, Martín M, Hevia S, et al. Gene variants in the NF-KB pathway (NFKB1, NFKBIA, NFKBIZ) and risk for early-onset coronary artery disease. Immunology Letters. 2019;208:39-43. DOI: 10.1016/j.imlet.2019.02.007
  58. 58. Tanriover C, Copur S, Mutlu A, Peltek IB, Galassi A, Ciceri P, et al. Early aging and premature vascular aging in chronic kidney disease. Clinical Kidney Journal. 2023;16(11):1751-1765. DOI: 10.1093/ckj/sfad076
  59. 59. Zhang J, Luo Z, Zhang Z, Zhao M, Tong C, Cong P, et al. Protective effect and mechanism of cannabidiol on myocardial injury in exhaustive exercise training mice. Chemico-Biological Interactions. 2022;365:110079. DOI: 10.1016/j.cbi.2022.110079
  60. 60. Javkhedkar AA, Quiroz Y, Rodriguez-Iturbe B, Vaziri ND, Lokhandwala MF, Banday AA. Resveratrol restored Nrf2 function, reduced renal inflammation, and mitigated hypertension in spontaneously hypertensive rats. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2015;308(10):R840-R846. DOI: 10.1152/ajpregu.00308.2014
  61. 61. Wang C, Luo Z, Carter G, Wellstein A, Jose PA, Tomlinson J, et al. NRF2 prevents hypertension, increased ADMA, microvascular oxidative stress, and dysfunction in mice with two weeks of ANG II infusion. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2018;314(3):R399-R406. DOI: 10.1152/ajpregu.00122.2017
  62. 62. Gao W, Xiao C, Hu J, Chen B, Wang C, Cui B, et al. Qing brick tea (QBT) aqueous extract protects monosodium glutamate-induced obese mice against metabolic syndrome and involves up-regulation transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) antioxidant pathway. Biomedicine & Pharmacotherapy. 2018;103:637-644. DOI: 10.1016/j.biopha.2018.04.043
  63. 63. Hüttl M, Markova I, Miklankova D, Makovicky P, Pelikanova T, Šeda O, et al. Adverse effects of methylglyoxal on transcriptome and metabolic changes in visceral adipose tissue in a Prediabetic rat model. Antioxidants (Basel). 2020;9(9):803. DOI: 10.3390/antiox9090803
  64. 64. Gu Y, Ma CT, Gu HL, Shi L, Tian XT, Xu WQ. Sitagliptin improves cardiac function after myocardial infarction through activation of autophagy in streptozotocin-induced diabetic mice. European Review for Medical and Pharmacological Sciences. 2018;22(24):8973-8983. DOI: 10.26355/eurrev_201812_16668
  65. 65. Yi M, Cruz Cisneros L, Cho EJ, Alexander M, Kimelman FA, Swentek L, et al. Nrf2 pathway and oxidative stress as a common target for treatment of diabetes and its comorbidities. International Journal of Molecular Sciences. 2024;25:821. DOI: 10.3390/ijms25020821
  66. 66. Wang G, Wang Y, Yang Q , Xu C, Zheng Y, Wang L, et al. Metformin prevents methylglyoxal-induced apoptosis by suppressing oxidative stress in vitro and in vivo. Cell Death & Disease. 2022;13(1):29. DOI: 10.1038/s41419-021-04478-x
  67. 67. Clarke JD, Hsu A, Williams DE, Dashwood RH, Stevens JF, Yamamoto M, et al. Metabolism and tissue distribution of sulforaphane in Nrf2 knockout and wild-type mice. Pharmaceutical Research. 2011;28(12):3171-3179. DOI: 10.1007/s11095-011-0500-z
  68. 68. Yoon HY, Kang NI, Lee HK, Jang KY, Park JW, Park BH. Sulforaphane protects kidneys against ischemia-reperfusion injury through induction of the Nrf2-dependent phase 2 enzyme. Biochemical Pharmacology. 2008;75(11):2214-2223. DOI: 10.1016/j.bcp.2008.02.029
  69. 69. Cui W, Bai Y, Miao X, Luo P, Chen Q , Tan Y, et al. Prevention of diabetic nephropathy by sulforaphane: Possible role of Nrf2 upregulation and activation. Oxidative Medicine and Cellular Longevity. 2012;2012:821936. DOI: 10.1155/2012/821936
  70. 70. Hang Y, Qin X, Ren T, Cao J. Baicalin reduces blood lipids and inflammation in patients with coronary artery disease and rheumatoid arthritis: A randomized, double-blind, placebo-controlled trial. Lipids in Health and Disease. 2018;17(1):146. DOI: 10.1186/s12944-018-0797-2
  71. 71. Wang G, Liang J, Gao LR, et al. Baicalin administration attenuates hyperglycemia-induced malformation of cardiovascular system. Cell Death & Disease. 2018;9(2):234. DOI: 10.1038/s41419-018-0318-2

Written By

Danyelly Bruneska Gondim Martins, Thaysa Walleria Aragão Santos, Maria Helena Menezes Estevam Alves and Rosângela Ferreira Frade de Araújo

Submitted: 06 March 2024 Reviewed: 11 March 2024 Published: 21 May 2024