Next Article in Journal
Effects of Drying Methods on Physicochemical and Immunomodulatory Properties of Polysaccharide-Protein Complexes from Litchi Pulp
Next Article in Special Issue
Antioxidant Activity and Mechanisms of Action of Natural Compounds Isolated from Lichens: A Systematic Review
Previous Article in Journal
Magnetite Nanostructures as Novel Strategies for Anti-Infectious Therapy
Previous Article in Special Issue
Allicin: Chemistry and Biological Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effect of Redox Modulating NRF2 Activators on Chronic Kidney Disease

1
College of Pharmacy, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Korea
2
Daewon Foreign Language High School, Kwangjin-gu, Seoul 143-713, Korea
*
Author to whom correspondence should be addressed.
Molecules 2014, 19(8), 12727-12759; https://doi.org/10.3390/molecules190812727
Submission received: 18 July 2014 / Revised: 6 August 2014 / Accepted: 12 August 2014 / Published: 20 August 2014

Abstract

:
Chronic kidney disease (CKD) is featured by a progressive decline of kidney function and is mainly caused by chronic diseases such as diabetes mellitus and hypertension. CKD is a complex disease due to cardiovascular complications and high morbidity; however, there is no single treatment to improve kidney function in CKD patients. Since biological markers representing oxidative stress are significantly elevated in CKD patients, oxidative stress is receiving attention as a contributing factor to CKD pathology. Nuclear factor erythroid-2 related factor 2 (NRF2) is a predominant transcription factor that regulates the expression of a wide array of genes encoding antioxidant proteins, thiol molecules and their generating enzymes, detoxifying enzymes, and stress response proteins, all of which can counteract inflammatory and oxidative damages. There is considerable experimental evidence suggesting that NRF2 signaling plays a protective role in renal injuries that are caused by various pathologic conditions. In addition, impaired NRF2 activity and consequent target gene repression have been observed in CKD animals. Therefore, a pharmacological intervention activating NRF2 signaling can be beneficial in protecting against kidney dysfunction in CKD. This review article provides an overview of the role of NRF2 in experimental CKD models and describes current findings on the renoprotective effects of naturally occurring NRF2 activators, including sulforaphane, resveratrol, curcumin, and cinnamic aldehyde. These experimental results, coupled with recent clinical experiences with a synthetic triterpenoid, bardoxolone methyl, have brought a light of hope for ameliorating CKD progression by preventing oxidative stress and maintaining cellular redox homeostasis.

Graphical Abstract

1. Chronic Kidney Disease (CKD)

An essential function of kidneys is the filtration of excess waste products that build up in the blood. As a result, waste will accumulate in the circulation system if the kidney fails to function properly, eventually progressing to end stage renal disease [1]. Renal failure occurs gradually, as opposed to showing overt, immediate symptoms. While chronic kidney disease (CKD) is being increasingly diagnosed worldwide, treatment methods such as dialysis or transplantation are either too costly or ineffective, so the condition is considered a public health problem that requires significant further research [2]. According to the Kidney Disease Quality Outcome Initiative (K/DOQI), CKD is defined as the atrophy of the kidney or of renal functions [3]. Renal atrophy can be identified in two different cases: when the glomerular filtration rate (GFR) does not reach above 60 mL/min/1.73 m2 for more than three months, and when albuminuria, defined as an albumin-to-creatinine ratio above 30 mg/g is present. However, when diagnosing the development and progression of CKD, the increase in urine albumin cannot be a direct marker—instead, it acts as a sign of renal deterioration as albuminuria indicates endothelial dysfunction of kidneys [2].
The progression of renal disease can be separated into five stages, the criterion of which is GFR. Patients in the first stage share similar GFR (≥90) to healthy counterparts (kidney damage with normal or increased GFR), making it difficult to identify the disease according to GFR alone. If kidney disease is suspected at this stage, it is imperative to delay its progression and minimize risk factors. Stage 2 is characterized by a 60–89 mL/min/1.73 m2 level of GFR (kidney damage with mild decreased GFR). As in Stage 1, it is difficult to distinguish CKD, but supplementary information such as albuminuria, proteinuria, and hematuria act as complementary indicators of CKD. The GFR of stage 3 is 30–59 mL/min/1.73 m2, and it is this stage in which early renal insufficiency is detected (moderately decreased GFR). Additional caution is required as complications like cardiovascular disease may accompany initial kidney damage. Stage 4, or pre-End-Stage Renal Disease (ESRD), has a GFR level of 15–29 mL/min/1.73 m2 (severely decreased GFR). Kidney replacement therapy must be considered when the condition worsens, since pharmacotherapy or lifestyle changes are not as effective in this stage. Finally, ESRD is declared upon the fifth stage when the kidney is completely deprived of its function. If transplantation is not available, it is crucial to undergo dialysis at this point [2].
Two main causes of CKD can be attributed to diabetes mellitus (DM) and hypertension [4]. Kidney damage that stems from diabetes is called diabetic nephropathy (DN). It occurs when high blood sugar resulting from diabetes impairs blood vessels in the kidney, an organ densely populated by myriad small vessels, which ultimately leads to severe renal degeneration. Gradual buildup of this damage eventually leads to renal failure, in which the kidney loses its function [5]. Major pathological changes instigated by DN are mesangial cell proliferation, glomerular hypertrophy and sclerosis, accumulation of extracellular matrix (ECM) in the glomerular basement membrane, and end-stage interstitial fibrosis [6,7]. Hypertension is the second leading cause of CKD, succeeding diabetes [8]. Hypertensive nephropathy patients are advised to maintain their blood pressure to 130/80 mm Hg to prevent CKD [9]. Currently, there is no single treatment to improve kidney function in CKD. Approaches to slow down the progression of CKD are limited to normalization of blood pressure blood glucose, and insulin. Therefore, the development of novel therapies to retard or reverse the decline of kidney function is highly needed.

2. Oxidative Stress and Inflammation in CKD

2.1. Oxidative Stress in CKD

Oxidative stress is produced when reactive oxygen species (ROS) production goes over the limit of the capacity of the antioxidant defense systems of the body [10]. This is a common phenomenon found in CKD lesions, and is considered to play a critical role in both the progression of CKD and related complications [10,11,12]. Over 90% of ROS are produced in the mitochondria during cellular respiration [13]. While a large majority of oxygen is converted into water, one electron reduction of O2 produces a “primary” ROS, superoxide (O2) [14]. In turn, O2 is transformed into hydrogen peroxide (H2O2) in mitochondria [15]. Yuan et al. [16] showed that mitochondrial dysfunction is involved in the pathogenesis of epithelial-mesenchymal transition (EMT) in renal proximal tubular epithelial cells, which is implicated for kidney fibrosis. The unilateral ureteral obstruction (UUO) model performed by Nishida et al. [17] demonstrated that mitochondrial proteins, especially in the electron transport complexes, decreased at an early stage of kidney fibrosis. Impaired mitochondrial respiration system and higher levels of oxidative stress markers were observed in CKD patients under hemodialysis treatment [18].
Additional cytosolic sources of ROS, including NADPH oxidase, xanthine oxidase, and lipoxygenases initiate and increase ROS production as well. Indeed, a chronic renal failure model, induced by renal mass reduction, showed up-regulated NAD(P)H oxidase in rats [19]. Fortuno et al. [20] reported that increased NADPH oxidase activity results in enhanced superoxide generation in patients with early CKD. Moreover, single nucleotide polymorphism in the coding region of p22phox, a key component of NADPH oxidase, was associated with elevated levels of oxidative stress in the kidney and consequent dialysis requirement [21]. Because the serum uric acid, an important source of oxidative stress in CKD, is produced by xanthine oxidase, blockade of xanthine oxidase led to renoprotective effects in 5/6 nephrectomized rats [22], diabetic mice [23] and UUO rats [24]. The 12/15-lipoxygenase was significantly up-regulated in the renal cortex of high-calorie/high-fat diet fed mice, which is a model of pre-diabetic neuropathy [25]. Another source of ROS production comes from activation of renin-angiotensin system (RAS). Activation of AT1 receptor by angiotensin II administration generated a higher level of superoxide in CKD animals than that of the normal animals [19,26]. A local increase of angiotensin-converting enzyme (ACE) in the kidney was associated with the progress of tubulointerstitial renal injury by hypokalemia, hypertension, and angiotensin II infusion in rats [27].
The level of ROS in the body is rigidly controlled by antioxidant enzymes, including superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and thiol molecules such as glutathione (GSH). High concentration glucose-treated cells produced more ROS than cells with normal glucose concentration. In this condition, overexpression of MnSOD effectively decreased ROS production, preventing related diabetic complications [28]. A study by Hinerfeld et al. [29] showed that mitochondrial sod null mice developed severe oxidative stress and diabetic complications. On the other hand, transgenic mice with CAT overexpression showed renoprotective effects following streptozotocin (STZ) treatment [30].

2.2. Inflammation in CKD

In pathophysiological conditions, oxidative stress and inflammation are inseparable from each other. Hasegawa et al. [31] found that levels of tumor necrosis factor-α (TNFα) and interleukin-1 (IL-1) were significantly increased in glomerular basement membranes of STZ-treated diabetic rats. Kidneys from diabetic rats showed higher IL-6 expression along with increased renal cortical IL-6 mRNA. The elevation of cytokines levels was related to the increase in kidney weight and urinary albumin excretion [32]. Additional reports verified that mRNA and protein levels of TNFα are elevated in glomerular cells of diabetic animals [33,34]. Particularly, TNFα increased ROS production in kidney mesangial cells [35], and activated NADPH oxidase causing glomerular injuries [36]. Besides, surface proteins of inflammatory cells, called adhesion molecules, play important roles in diabetic nephropathy. Coimbra et al. demonstrated that obese Zucker rats with T2DM developed early podocyte damage and interstitial macrophage infiltration, implying progressive renal disease, and particularly that an increase of intracellular adhesion molecule 1 (ICAM-1) was observed in glomerular and epithelial cells [37]. The level of vascular cell adhesion protein 1 (VCAM-1) is correlated to albuminuria in type 2 diabetic hypertensive patients [38]. Expression of pro-inflammatory cytokines and adhesion molecules, which are major mediators of inflammation, is regulated by transcription factor nuclear factor-κB (NF-κB) [39]. There are lines of evidence indicating that NF-κB activation is associated with oxidative stress-associated renal fibrosis [40,41]. Rats with 5/6 partial nephrectomy were shown to have increased levels of p65, a NF-κB subunit, in glomerular cells [42]. In STZ-induced diabetic rats, NF-κB activation was found in the renal cortex [43].

2.3. Biological Markers of Oxidative Stress and Inflammation in CKD

In the clinical setting, levels of oxidation markers of lipid, protein, and DNA are heightened in CKD patients (Table 1). Increased malondialdehyde (MDA), a lipid-associated oxidation marker, in CKD patients implies severe glomerulosclerosis [44]. Grone et al. [45] demonstrated that levels of hypochlorous acid (HCIO)-modified proteins increase in renal podocytes and glomeruli from CKD patients. Elevation of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG) was associated with ROS-related DNA damage in CKD patients [46]. Levels of plasma 8-isoprostanes and GFR showed an opposing relationship in CKD patients [47,48]. In addition, although there were some fluctuations depending on performed studies, levels of antioxidant genes such as SOD and CAT, and GSH contents were diminished in CKD patients [49,50,51]. Polymorphism of the MnSOD gene was found to be related to the incidence of DN in Chinese, Japanese and Koreans subjects with T2DM [52,53,54]. Moreover, SOD expression levels were positively correlated with GFR [55] and levels of GSH content were also positively associated with renal creatinine clearance [49].
Table 1. Biological markers of oxidative stress and inflammation in CKD patients.
Table 1. Biological markers of oxidative stress and inflammation in CKD patients.
Type of MarkersGroupSpecific MarkerRefs.
Oxidative markersLipidF2-isoprostanes[56,57,58]
Malondialdehyde (MDA)[59,60]
Thiobarbituric acid- reactive substance[61]
ProteinCarbonyls[62,63]
Advanced glycation end-products (AGEs)[64]
Advanced oxidation protein products (AOPP)[65,66]
Oxidized low density lipoproteins (OxLDL)[67]
DNA8-Oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG)[46,68]
DNA strand breaks[69,70]
Inflammatory markers C-reactive protein (CRP)[71,72]
IL-1[73]
IL-6[72]
TNFα[73,74]
In addition to the increase in oxidative markers, CKD patients are constantly exposed to transient infections, comorbidities, and intermittent stimulus of dialysis [75]. As a result, it is generally accepted that pro-inflammatory cytokines such as TNFα and IL-1β are amplified in hemodialysis patients [73,76]. According to Shlipak et al. [72], patients with renal insufficiency showed markedly higher levels of C-reactive protein (CRP) and IL-6 than those with normal kidney functions. The study by Oberg et al. reaffirmed this finding: inflammatory biomarkers such as CRP and IL-6 were amplified in CKD subjects as compared to healthy control groups [77].

3. Involvement of NRF2 Signaling in CKD Pathology

3.1. NRF2 as a Crucial Regulator of the Antioxidant Defense System

Nuclear factor-erythroid 2-related factor 2 (NRF2) is a redox-sensitive transcription factor with the basic leucine zipper (bZIP) motif. Belonging to the cap ‘n’ collar (CNC) family, NRF2 is crucial in regulating the basal and inducible expressions of various antioxidant and cytoprotective genes, which can counteract oxidative and electrophilic stress [78,79]. NRF2 activity is primarily governed by the Kelch-like ECH-associated protein 1 (KEAP1). Under normal conditions, cytosolic protein KEAP1 sequesters NRF2 in the cytoplasm and forwards ubiquitination and subsequent degradation by bridging NRF2 to a Cul3-based E3 ligase. The binding and regulation of KEAP1 and NRF2 have been explained by a “hinge and latch” model: the KEAP1 dimer formed through the BTB domain of this protein interacts with one molecule of NRF2 through conserved motifs DLG and ETGE (Figure 1). In particular, binding affinity of DLG motif of NRF2 is lower than that of ETGE motif; therefore a cysteine-rich protein KEAP1 can sensitively transduce a variety of redox signals to this binding motif. When KEAP1 cysteine residues are modified by ROS or electrophiles, KEAP1 undergoes conformational changes, leading to the dissociation of the DLG motif from KEAP1 and the blockade of ubiquitination/proteasomal degradation of this protein. Then NRF2 can translocate into the nucleus to activate its target gene transcription [80,81]. Within the nucleus, NRF2 heterodimerizes with small Maf proteins and transactivates the expression of genes with the antioxidant response element (ARE), which is also called as the electrophile response elements (EpRE).
Figure 1. Regulation of antioxidant and detoxifying genes by the NRF2-Keap1 pathway. Under normal conditions, NRF2 is continuously degraded by the KEAP1-Cul3-proteasome axis. When Cys residues of KEAP1 protein are modified by sulfhydryl reactive chemicals, conformational KEAP1 changes lead to NRF2 liberation and transcriptional activation of an array of ARE-bearing genes, encoding detoxifying enzymes, ROS scavenging enzymes, thiol molecules, and their generating enzymes.
Figure 1. Regulation of antioxidant and detoxifying genes by the NRF2-Keap1 pathway. Under normal conditions, NRF2 is continuously degraded by the KEAP1-Cul3-proteasome axis. When Cys residues of KEAP1 protein are modified by sulfhydryl reactive chemicals, conformational KEAP1 changes lead to NRF2 liberation and transcriptional activation of an array of ARE-bearing genes, encoding detoxifying enzymes, ROS scavenging enzymes, thiol molecules, and their generating enzymes.
Molecules 19 12727 g001
The first in vivo demonstration of the involvement of NRF2 in detoxifying gene regulation has been reported in 1997: The Yamamoto group showed that the induction of Nqo1 and glutathione S-transferase (Gst) by butylated hydroxyanisole (tBHA) treatment was largely attenuated in nrf2 deficient mice [82]. After this observation, numerous studies have identified a wide array of genes as NRF2-target genes [83,84,85,86]. These include genes encoding antioxidant enzymes, phase II detoxification enzymes, NADPH-generating enzymes, drug transporters, metabolism-related enzymes, and proteasome (Table 2). Among antioxidant proteins, NRF2 controls the expression of direct ROS scavenging enzymes such as glutathione peroxidase (GPx) and SOD; GSH generating enzymes such as the catalytic and modifier subunit of γ-glutamate cysteine ligase (GCLC and GCLM, respectively), and glutathione reductase (GSR); and thiol molecules such as thioredoxin. NRF2 is also involved in the expression of NADPH-generating system, which includes malic enzyme 1, glucose-6-phosphate 1-dehydrogenase, and 6-phosphogluconate dehydrogenase. Moreover, NAD(P)H:quinone oxido-reductase 1 (NQO1), heme oxygenase-1 (HO-1) and aldoketo-reductase family are also well-known NRF2 target genes.
Table 2. Genes regulated by NRF2 in mouse (m) and human (h).
Table 2. Genes regulated by NRF2 in mouse (m) and human (h).
Functional ClassificationGene NameDescriptionSpecies
Antioxidant proteinsGCLCγ-Glutamate-cysteine ligase, catalytic subunitmh
GCLMγ-Glutamate-cysteine ligase, modifier subunitmh
GSRGlutathione reductasemh
GPx1Glutathione peroxidase 1 (or 4)m
GPx2Glutathione peroxidase 2mh
TXNRDThioredoxin reductasemh
TXNThioredoxinmh
PRDX1 &6Peroxiredoxin 1 (or 6)mh
CATCatalasemh
SODSuperoxide dismutasemh
SRXN1Sulfiredoxin 1mh
GGT1γ-Glutamyltransferase 1 h
GLRXGlutaredoxin h
Phase I oxidation, reduction and hydrolysis enzymesALDH3A1Aldehyde dehydrogenase 3A1mh
ADH7Alcohol dehydrogenase 7m
AKR1B1Aldo-keto reductase 1B1mh
AKR1C1Aldo-keto reductase 1C1 h
CBR1Carbonyl reductase 1m
EPHX1Microsomal epoxide hydrolase 1mh
NQO1NAD(P)H:quinone oxidoreductase 1mh
CYP2B9Cytochrome p450, 2B9
Phase II detoxifying enzymesGSTM1Glutathione S-transferase class mu 1 (or 2,4,5,6)m
GSTM3Glutathione S-transferase class mu 3mh
GSTA1Glutathione S-transferase class alpha 1 (or 2,3,4)m
MGST1Microsomal glutathione S-transferase 2mh
MGST2Microsomal glutathione S-transferase 3m
UGT1A6UDP glucuronosyltransferase 1A6 h
UGT2B1UDP glucuronosyltransferase 2B1m
UGT2B5UDP glucuronosyltransferase 2B5m
NADPH-generating enzymesME1Malic enzyme 1mh
G6PDGlucose-6-phosphate 1-dehydrogenasemh
PGD6-Phosphogluconate dehydrogenasemh
Drug transportersABCB6ATP-binding cassette, subfamily B, member 6mh
ABCC1ATP-binding cassette, subfamily C, member 1m
ABCC2ATP-binding cassette, subfamily C, member 2mh
ABCC3ATP-binding cassette, subfamily C, member 3mh
ABCC4ATP-binding cassette, subfamily C, member 4m
ABCC5ATP-binding cassette, subfamily C, member 5m
Heme and metal metabolism (stress response protein)HO-1Heme oxygenase-1mh
FTH1Ferritin, heavy polypeptide 1mh
FTL1Ferritin, light polypeptide 1mh
MT1Metallothionein 1mh
MT2Metallothionein 2mh
Protein degradationUbCUbiquitin Cm
PSMB5Proteasome 26S PSMB5 subunitm
Lipid metabolismACOT7Acetyl-CoA thioesterase 7m
ACOX1Acetyl-CoA oxidase 1m
LIPHLipase, member Hm
CES1GCarboxylesterase 1Gm

3.2. NRF2 as a Multi-organ Protector against Oxidative Damages

Since NRF2 governs the expression of genes associated with xenobiotic metabolism and redox homeostasis, nrf2 null mice have been relatively more susceptible to acute damages of acetaminophen [87], cigarette smoke [88], and diesel exhaust [89] than wild-type mice. These mutant mice suffered from pulmonary inflammation following ovalbumin sensitization and showed a more severe asthmatic response than wild-type mice [90]. Cytotoxicity by 3-nitropropionic acid (3-NP), an inhibitor of mitochondrial complex II, was enhanced in nrf2-deficient primary neurons. In addition, nrf2-null mice showed increased lesion volume in the striatum along with impaired rotarod task performance following 3-NP injection [91].
The role of NRF2 in renoprotection has been suggested in multiple studies. Female mice with nrf2 gene ablation displayed a lupus-like autoimmune nephritis at over 60 weeks of age [92]. In a model of ischemia-reperfusion injury, renal function, vascular permeability, and survival of nrf2-knockout mice were significantly worse than wild-type mice [93]. Renal damage and interstitial fibrosis by cyclosporin A treatment were relatively higher in nrf2-knckout mice [94]. The STZ-induced diabetic nephropathy model revealed that nrf2-null mice developed a severe renal injury with greater oxidative DNA damage than wild-type mice [95].
Accordantly, there are a number of studies showing favorable effects of NRF2 inducers. Pharmacological intervention of NRF2 activators exerted protective roles against injuries from oxidative stress and inflammation in various in vitro and in vivo experimental models. Particularly, naturally occurring NRF2 activators have gained great attention: sulfur-containing compounds and phenolic antioxidant compounds are major dietary NRF2 activators [96,97]. In addition to these major groups, indoles, diterpenes, coumarins, and lactones are also regarded as NRF2 inducers [98]. Sulforaphane (SFN) and phenethyl isothiocyanate, which are abundant constituents of cruciferous plants, are well characterized NRF2 activators containing a sulfur motif, which can lead to KEAP1 sulfhydryl modification and ultimately NRF2 activation [97]. Cysteine residues of KEAP1 have an important role in activating NRF2 by SFN. Specifically, modification of Cys151 by SFN treatment is required for liberating NRF2 from KEAP1 [99]. A majority of evidence from in vitro and in vivo experiments elucidated that SFN increases the expression of phase II detoxifying enzymes and antioxidant proteins via NRF2 signaling [100,101]. In human hepatocarcinoma and prostate cancer cells, expression of NQO1 and GSTs were up-regulated by SFN treatment [102,103]. In an in vivo experiment, SFN treatment showed increased expression of NQO1 and GCL in intestines of wild-type mice but not in nrf2-null mice [104]. Incubation of phenethyl isothiocyanate also increased NQO1 and UDP-glucuronosyl transferase (UGT) expressions in Hepa1c1c7 cells [105]. Resveratrol, curcumin, cinnamic aldehyde (CA), and quercetin are phenolic antioxidant compounds. As a common chemical property, they have an electrophilic α, β-unsaturated carbonyl moiety called a Michael acceptor. This moiety is highly reactive to the Cys residues of KEAP1, which in turn induces conformational changes of KEAP1 [106,107]. In an early study by Dinkova-Kostova et al., dietary constituent curcumin increased phase II enzyme expression in murine hepatoma cells via arylhydrocarbon (Ah) receptor-independent manner [108]. There has been considerable evidence that people who consumed the foods containing quercetin had a lower risk of cancer than the control group [109]. In subsequent studies, the pharmacological benefits of quercetin were attributed to activation of NRF2 and its target gene expression [110,111].

3.3. NRF2 as Anti-inflammatory Modulator

There are notable reports showing reciprocal regulation of NRF2 and NFκB, suggesting an anti-inflammatory role of NRF2. For the activation of NF-κB, IκB, a cytosolic inhibitor protein of NF-κB, has to be phosphorylated by IκB kinase, and this leads to the degradation of IκB and nuclear translocation of NF-κB [112]. In this serial event, NRF2 is known to control NF-κB levels through suppression of IκB phosphorylation [113]. Mouse embryonic fibroblasts (MEF) from nrf2 null mice displayed a higher level of phosphorylated IκB than wild-type MEF [114], and these mutant cells possessed diminished levels of NF-κB subunit p50 and p65 [115]. In spinal cord injury mice, nrf2 knockout mice showed elevated NF-κB activity and TNFα expression [116]. Additionally, there have been reports on the role of NRF2 target proteins for NF-κB inhibition. Overexpression of HO-1 led to stabilization of IκB, resulting in NF-κB inhibition. Consistently, the inhibition of HO-1 activity suppressed p65 activity [117]. Overexpression of NRF2 or NQO1 repressed LPS-inducible expressions of TNFα and IL-1β in human monocytes [118]. Similarly, SFN treatment decreased DNA binding activity of NF-κB without affecting IκB levels in murine macrophages [119]. NRF2 inducer curcumin inhibited NF-κB activation through the down-regulation of IκB kinase [120]. A potent NRF2 inducer triterpenoid bardoxolone methyl directly blocked IκB kinase by interacting with Cys179 of the IκB kinase [121].

4. Role of the NRF2 System in CKD

Today increasing evidence supports the critical role of NRF2 in kidney pathology. In lupus nephritis, oxidative stress and inflammation have been suggested as core pathologic components: glomerular deposition of immune complex can bring about oxidative damage, leading to glomerular injuries [122]. Earlier, it was shown that female mice with nrf2 gene deletion developed lupus-like autoimmune nephritis [92]. A recent study by Jiang et al. [123] demonstrated that nrf2-deficient mice developed a greater degree of renal damages in a pristine-induced lupus nephritis model. In addition, administration of epigallocatechin-3-gallate prevented development of nephritis in spontaneously lupus-prone mice and this protection was mediated by NRF2 signaling [124]. Similarly, curcumin and bardoxolone methyl attenuated lupus nephritis in this lupus mouse model [125,126].
Experimental evidence showing the involvement of NRF2 in diabetic nephropathy was provided by multiple groups. Yoh et al. demonstrated a protective role of NRF2 in diabetic complications [127]. When diabetes was induced by a single injection of STZ, nrf2-null mice developed renal impairments earlier and suffered more severe mesangial injuries than wild-type mice. Similarly, in a study by Jiang et al. [95], when ROS production increased, oxidative DNA damage and renal injury worsened in STZ-treated nrf2 null mice compared to wild-type mice. In another study, these mutant mice showed exacerbated acute renal injuries following ischemia-reperfusion or cisplatin treatment. On the other hand, the administration of GSH or its precursor N-acetylcysteine could improve renal function in these mice [93]. Multiple in vitro studies also support the protective effects of NRF2 on renal pathology. TGFβ-stimulated ECM production was exacerbated in NRF2-silenced renal tubular epithelial cells, whereas TGFβ signaling and ECM expression were largely attenuated in KEAP1-silenced cells [128,129].
The study by Kim et al. [130], which used male Sprague-Dawley rats, was the first report to show the involvement of NRF2 signaling in an experimental model of 5/6 nephrectomy CKD. In this model, the remnant kidney exhibited GSH depletion, lipid peroxidation, NF-κB stimulation and NADPH oxidase activation. Notably, NRF2 activity was significantly diminished at 12 weeks in the remnant kidney, leading to a decrease in CAT, SOD, HO-1 and NQO1. In a study with Imai rats that is a model of spontaneous focal glomerulosclerosis, nuclear NRF2 levels and its target gene expressions were markedly reduced in the kidney in spite of the presence of a severe oxidative and inflammatory pathologic environment [26]. In a tubulointerstitial nephropathy model using adenine treatment, NRF2 activity was impaired and the expressions of CAT, HO-1 and GCL were repressed [131].
The above mentioned reports provide evidence for the necessity of NRF2 in animal models of CKD, and raises a possibility that dysregulation of NRF2 signaling may be involved in human CKD pathology. Indeed, lines of evidence show that the expression of several NRF2 target genes is diminished in kidneys from CKD patients. Activities of GPx and GSR were significantly decreased in T2DM patients with or without nephropathy. Moreover, repression of GPx and GSR was more severe in diabetic patients with nephropathy compared to patients without nephropathy [132]. According to Crawford et al. [133], CKD patients possessed lower levels of blood monocyte GPx and red blood cell CAT activities than controls. It was also notable that the single nucleotide polymorphism of GPx was more prevalent in CKD patients, suggesting the role of GPx in CKD pathology. A study by Puchades et al. [134] showed that levels of oxidized molecules such as protein carbonyls and 8-oxo-dG, and NRF2 target gene expression are increased in CKD patients with hemodialysis or peritoneal dialysis. Results showed that levels of oxidized molecules were significantly high, and enzyme activities of GPx, GSR, CAT and SOD were markedly diminished in these patients compared to the control group.

5. Naturally Occurring NRF2 Activators and CKD

Recently, many groups have demonstrated the beneficial effects of NRF2 on CKD. A study by Li et al. [135] elucidated how the increase of NRF2 target HO-1 and GCL is required in high glucose conditions to counteract high glucose-induced ROS increase and resultant oxidative damages in cultured mesangial cells. Due to protective effects, nrf2-null mice were vulnerable to STZ-induced nephropathy, characterized by severe renal dysfunction and oxidative injuries [95,127]. Therefore, experimental conditions with NRF2 activation in vitro and in vivo could provide protection against CKD. It was shown that incubation of human skin fibroblasts with proteasome inhibitor MG132 could induce nuclear translocation of NRF2 and increase target gene expression, suggesting that proteasome inhibitor treatment can activate NRF2 signaling [136]. When Sprague-Dawley rats were administered with low doses of MG132 for 12 weeks, renal mRNA levels for SOD1, CAT, and GPx increased and STZ-induced DN significantly improved [137]. A study by Cui et al. [138] used OVE26 mice as a model of transgenic type 1 diabetic mice. When a low dose of MG132 was administered to OVE26 for 3 months, there were significant improvements in renal structure alterations and kidney function.
In contrast, indoxyl sulfate (IS), one of the uremic toxins casing endothelial dysfunction and nephrotoxicity [139,140], was shown to repress renal expression of NRF2. A research by Bolati et al. [141] elucidated that IS administration in rats reduced the level of NRF2 and its target gene expression in the kidney, thereby increasing the renal level of 8-oxo-dG. However, treatment with AST-120, an oral sorbent lowering serum IS level, recovered NRF2 expression in CKD rat kidneys and consequently diminished ROS production.
On the basis of these reports, the positive relationship between NRF2 and CKD protection gained attention, leading researchers to make an effort to develop NRF2 activators as a novel therapeutic strategy for CKD protection/retardation. In particular, there have been extensive investigations on naturally occurring NRF2 activators for their potential efficacy in CKD protection. Here we review current findings of the role of NRF2 activating SFN, resveratrol, curcumin, and CA (chemical structures are shown in Table 3) in different animal CKD models.

5.1. SFN

SFN is abundant in cruciferous vegetables such as broccoli, brussels sprouts and cabbages [142]. It has long been recognized that SFN has potent chemopreventive efficacy in various animal carcinogenesis models and it had been accepted that the induction of phase II detoxifying enzymes is an underlying mechanism of SFN effect [143]. In addition to cancer prevention, accumulating evidence indicates that SFN effectively protects multiple organs from oxidative injuries. SFN treatment in cultured cardiomyocytes reduced ROS and DNA fragmentation [144] and the administration of SFN-containing broccoli in rats protected their hearts from ischemia-reperfusion injuries [145]. Animals fed SFN displayed reduced production of inflammation markers such as IL-6 and TNFα in response to LPS treatment in the brain [146]. Now, it has been firmly established that these multi-organ protective effects of SFN are attributed to activation of NRF2 signaling [147,148].
Table 3. Chemical structures of NRF2 activators.
Table 3. Chemical structures of NRF2 activators.
NRF2 ActivatorsChemical Structure
Sulforaphane (SFN) Molecules 19 12727 i001
Resveratrol Molecules 19 12727 i002
Curcumin Molecules 19 12727 i003
Cinnamic aldehyde (CA) Molecules 19 12727 i004
Bardoxolone methyl Molecules 19 12727 i005
The renoprotective effects of SFN have been evidenced in several in vivo studies. SFN administration protected kidneys from cisplatin and ischemia-reperfusion challenge and these protections were mediated through NRF2 activation [149,150,151]. A study by Zheng et al. [152] demonstrated that, in a STZ-induced diabetic mouse model, SFN treatment (started 2 weeks after STZ injection) noticeably improved metabolic dysfunction associated with diabetes, albuminuria and glomerular sclerosis. These beneficial effects of SFN were not examined in nrf2 deficient mice, indicating that NRF2 is a molecular target of SFN. This study further revealed that SFN attenuates high glucose-induced mesangial cell hypertrophy by NRF2-mediated TGFβ signaling repression. In a study by another group, SFN administration (0.5 mg/kg) for 3 months prevented STZ-induced DN: the albumin-to-creatinine ratio (ACR) and renal fibrosis were alleviated by SFN [153]. In addition, in vitro experiments using the human renal tubular KH11 cell line showed that oxidative damages by mannitol or high glucose plus palmitate were attenuated by SFN incubation; however protection was lost following NRF2 siRNA introduction. Beneficial effects of SFN were also demonstrated in the UUO model. In rats, UUO induced mitochondrial stress and subsequent apoptosis by decreasing nuclear NRF2 levels, whereas SFN treatment reversed NRF2 levels and modulated mitochondrial-stress damages and renal fibrosis [154]. An additional study with UUO showed that structural renal damages by UUO was remarkably improved after SFN treatment in rats [155].

5.2. Resveratrol

Resveratrol (3,5,4'-trihydroxystilbene) is a polyphenolic phytoalexin which is found in many plant species, including grapes and berries [156]. Initially, resveratrol gained attention for its cardioprotective effects when this compound, a key ingredient of red wine, was implicated in the French Paradox: the low incidence of cardiovascular diseases in the French population [157]. After this, numerous biological activities of resveratrol were identified. Resveratrol has been associated with multiple pharmacological health benefits that stem from its antioxidant [158], anti-inflammatory [159], anti-mutagenic [160], and anticancer properties [161]. This phytochemical exerts antioxidant effects by directly scavenging ROS or by increasing antioxidant enzyme expression through NRF2, AP-1 and forkhead box O (FOXO) [162,163,164]. In addition, resveratrol activates silent information regulator 1 (SIRT1), an NAD+-dependent deacetylase, via AMP-activated protein kinase (AMPK) activation [165,166]. Through these signaling alterations, resveratrol exhibits renoprotective effects in various animal models. In rat renal mesangial cells, resveratrol incubation decreased high concentration glucose-induced cell proliferation and fibronectin expression through inhibition of NF-κB/NADPH oxidase pathway [167]. Kitada et al. demonstrated the effect of resveratrol in a diabetic nephropathy model of db/db mice: resveratrol supplementation (400 mg/kg) attenuated albuminuria, urinary 8-oxo-dG excretion, and histological changes of kidney in db/db mice via AMPK-SIRT1-independent manner [162]. A study by Kim et al. confirmed beneficial effects of resveratrol in db/db mice [165]. Resveratrol treatment was started at the age of 8 weeks (maintained for 12 weeks) and it inhibited renal lipotoxicity and oxidative stress by enhancing AMPK-SIRT1-PGC1α signaling. Sharma et al. demonstrated a protective effect of resveratrol in a STZ diabetic model. The administration of resveratrol in STZ-treated rats improved renal dysfunction by reducing albuminuria and significantly diminishing oxidative stress markers such as MDA [168]. Resveratrol was also effective in preventing renal fibrosis in a UUO model: administration of resveratrol inhibited Smad3 acetylation, resulting in TGFβ signaling repression [169,170]. Together with these molecular targets, recent studies show that resveratrol affects NRF2 signaling in the kidney. Palsamy et al. reported that resveratrol treatment (5 mg/kg) for 30 days diminished creatinine clearance to the normal level in STZ/nicotinamide-induced diabetic rats [171]. Resveratrol also restrained lipid peroxidation and protein carbonyl levels in STZ rats. In particular, this study demonstrated that decreased expression of NRF2, GCS, GST and HO-1 in the STZ-diabetic kidney was normalized by resveratrol treatment. An in vitro experiment by Huang et al. showed that activation of SIRT1 by resveratrol leads to NRF2 increase in glomerular mesangial cells. Eventually, this event prevented AGE-induced TGFβ and fibronectin expressions [172].
Although there is no direct clinical evidence showing protective effects of resveratrol in CKD patients, several studies imply its beneficial effects on human subjects. Resveratrol supplementation in T2DM patients significantly reduced insulin resistance and urinary excretion of ortho-tyrosine levels, which is a marker of oxidative stress [173]. Moreover, dietary supplementation with red grape juice exerted antioxidative and anti-inflammatory efficacies in hemodialysis patients [174,175].

5.3. Curcumin

Curcumin, a phenolic compound derived from the hebaceous plant Curcuma longa, has been used as a traditional medicine for a long time [176]. Numerous studies have shown that curcumin has various biological and pharmacological functions; antioxidant [177], anti-inflammatory [178], anti-bacterial [179], anti-proliferative [180], hepatoprotective [181], cardioprotective [182] and neuroprotective activities [183]. In particular, curcumin was shown to improve diabetic symptoms and protect DN through multiple molecular targets [184]. In STZ diabetic rats, curcumin treatment (15 and 30 mg/kg) for 2 weeks after STZ injection ameliorated the increase of creatinine and urea clearance, proteinuria and oxidative stress markers [185]. In another similar experimental design, curcumin (100 mg/kg) treatment for 8 weeks significantly reversed all of abnormalities derived from STZ, including proteinuria, reduced creatinine clearance, and increased blood urea nitrogen [186]. Several signaling molecular targets have been suggested as underlying mechanisms of renoprotection of curcumin. In a study by Soetikno et al., normalization of renal dysfunction and lipid peroxidation in STZ rats was associated with inhibition of PKC-α and PKC-β1 by curcumin [187]. Chiu et al. demonstrated that renal oxidative stress, mesangial proliferation, and activation of histone acetyltransferase p300 and NF-κB by STZ were prevented by curcumin treatment [188]. Curcumin (100 mg/kg) administration in STZ diabetic animals markedly decreased infiltration of renal macrophages and renal production of proinflammatory cytokines such as TNF-α and IL-1β along with NFκB inhibition. In cultured glomerular mesangial cells, curcumin incubaton inhibited high concentration glucose-induced activation of activator protein-1 (AP-1) and sphingosine kinase 1 (SphK1) signaling, resulting in the supression of SphK1-regulated fibronectin and TGFβ overexpression [186,189]. In 5/6 nephrectomized rats, Ghosh et al. found that curcumin administration (75 mg/kg) for 8 weeks improved renal dysfunction and inhibited NF-κB activation and macrophage infiltration in the kidney [190].
Due to the poor bioavailability of curcumin, efforts have been made to improve the pharmacokinetic profile of this compound. A curcumin derivative B06 was developed for this reason and was proved to be effective in improving renal function and macrophage infiltration in diabetic rat kidneys [191]. B06 (0.2 mg/kg, 6 weeks) also inhibited pro-inflammatory cytokine production through the repression of c-Jun N-terminal kinase/NF-κB signaling. Another renoprotective curcumin derivative C66 was associated with the inactivation of mitogen-activated protein kinase (MAPK) pathway and the down-regulation of ACE/angiotensin II system [192].
Recent evidence supports the strong involvement of NRF2 signaling in renoprotective effects of curcumin. In a high-fat diet (HFD) mouse model, He et al. described that the short-term administration of curcumin (15 days) increases NRF2 signaling and diminishes lipid peroxide makers in the muscle, improving insulin tolerance, implying the beneficial effects of curcumin-NRF2 on diabetes [193]. A study by Soetikno et al. used a 5/6 nephrectomized rat model to demonstrate that curcumin treatment (75 mg/kg) prevents the decrease of NRF2 and HO-1 expressions in the remnant kidney along with the amelioration of renal dysfunction [194]. Similarly, other groups reported that NRF2 activation by curcumin is necessary for prevention of hemodynamic changes, glomerular hypertension, hyperfiltration, and antioxidant enzyme decrease in 5/6 nephrectomized rats [195,196].
Several clinical trials have evaluated the potential efficacy of curcumin in diabetes and DN. Usharani et al. showed that curcumin treatment (300 mg, twice a day for 8 weeks) in patients with T2DM significantly enhanced endothelial function and decreased oxidative stress and inflammatory markers [197]. A randomized, double-blind clinical trial by Chuengsamarn et al. reported that curcumin administration (1.5 g per day, 9 months) in pre-diabetic population was beneficial in preventing T2DM development, which was assessed by β-cell function, insulin resistance, and serum cytokine levels [198]. These studies allude to the potential benefit of curcumin in human DN and one clinical trial has been conducted to investigate this possibility. In a randomized, double-blind and placebo-controlled study by Khajehdehi et al. [199], patients with DN were received by turmeric (22.1 mg of curcumin, three times a day) for 2 months and the levels of TGFβ and pro-inflammatory cytokines were evaluated. Results indicate that serum levels of TGFβ and IL-8, and urinary levels of IL-8 were significantly decreased after curcumin supplementation when compared to the pre-supplementation group. In addition, proteinuria in DN patients was effectively improved by this supplementation without adverse effects.

5.4. CA

CA is a major compound isolated from Cinnamomum cassia [200]. Various lines of evidence indicate that CA has chemopreventive efficacy [201], anti-bacterial activities [200] and anti-inflammatory effects [202]. In addition, as a reactive Michael acceptor-containing chemical, CA is a potent NRF2 activator that induces the expression of its target genes [203]. There are a number of studies that evaluated renoprotective effects of CA. Zheng et al. elucidated that CA can improve diabetic-induced renal injuries [152]. CA treatment was able to ameliorate albuminuria and creatinine clearance in STZ-induced diabetic mice. While CA increased levels of NRF2 and its target genes NQO-1 and GCL, and suppressed TGF-β1 signaling in the kidneys of these diabetic mice, the treatment did not show any protective effect in nrf2-deficient mice, confirming the critical role of NRF2 signaling in DN protection. A study by Chao et al. [204] also showed that CA administration reduces high concentration glucose-induced cell proliferation and hypertrophy in renal interstitial fibroblast cells. Administration of cinnamon oil in alloxan-treated animals decreased fasting glucose levels and attenuated alloxan-induced DN [205].

6. Experience and Promise from Bardoxolone Methyl for CKD Management

Bardoxolone methyl (methyl-2-cyano-3,12-dioxooleano-1,9(11)dien-28-oate, CDDO-Me) is described as a synthetic triterpenoid, derived from the natural product oleanolic acid. This antioxidant inflammation modulator is known as one of most potent activators of NRF2 [206,207,208]. A study by Dinkova-Kostova et al. [206], used a series of synthetic triterpenoid analogs and demonstrated that phase II enzyme response and inflammation blocking effects are dependent on the presence of Michael reaction center. Further, a recent study provided direct biochemical evidence that bardoxolone methyl binds to the Cys151 of the BTB domain of KEAP1 forming an adduct which inhibits the association of Cul3 to KEAP1, resulting in NRF2 activation [209]. In addition, bardoxolone methyl binds to the Cys179 residue in IκB kinase IIKKβ protein, resulting in the inhibition of NFκB activation and pro-inflammatory cytokine production [121].
In animal experiments, treatment with bardoxolone methyl protected kidneys from acute injuries by elevating NRF2 target genes. Coordinated induction of NRF2 target genes in bardoxolone-treated kidneys ameliorated ischemia-reperfusion renal injuries [210]. Nephrotoxicity by iron nitrilotriacetate (FeNTA) was more severe in nrf2 null mice, and triterpenoid treatment protected FeNTA-induced renal injury through NRF2 activation [211]. Moreover, when STZ-induced diabetic mice were administered with a derivative of bardoxolone methyl, diabetes-associated atherosclerosis was reduced and diabetic kidney injury improved [212]. Another synthetic triterpenoid CDDO-dhTFEA could restore NRF2 decrease in rat CKD by 5/6 nephrectomy, and attenuated kidney inflammation and fibrosis [213].
Potential benefits of bardoxolone methyl in human kidney dysfunction have been recognized in cancer patients treated with this drug. The first Phase I clinical trial showed that bardoxolone treatment in patients with lymphomas and advanced solid tumors increased NQO1 mRNA in peripheral blood mononuclear cells and enhanced GFR [214]. After this intriguing observation, bardoxolone methyl was applied in Phase 2 clinical trial for patients with diabetes and CKD. An exploratory multi-center, opens-label, randomized clinical trial was performed to assess the renoprotective effect and safety of bardoxolone methyl [215]. Twenty patients with T2DM and CKD (stage 3 to 4) took 25 mg bardoxolone methyl for 28 days, then 75 mg for further 28 days. About 75% of patients took antihypertensive medicines, statins, and ACE inhibitors or ARBs, and most patients had significant albuminuria. Administration of bardoxolone showed a significant increase in GFR (increase of 2.8 mL/min/1.73 m2 at 4 weeks; increase of 7.2 mL/min/1.73 m2 at 8 weeks), a decrease in serum creatinine and blood urea nitrogen, and an increase of creatinine clearance. This study brought to light a therapeutic potential of bardoxolone methyl in CKD to light. However, there are some important limitations in this study such as a lack of a placebo control group and short term of treatment.
A large Phase 2b clinical trial (BEAM study) was prepared to confirm the effect of bardoxolone methyl in CKD patients [216]. In this double blind, randomized, placebo-controlled trial, 227 patients with diabetes and CKD participated. They received placebo or bardoxolone methyl with one of three dosage levels: 25, 75 or 150 mg. Bardoxolone methyl administration improved GFR within 4 weeks after treatment initiation of treatment and these improvements persisted for 52 weeks. In addition, adverse events were moderate and manageable, but muscle spasms and a mild increase in alanine aminotransferase levels were common. After this optimistic result, the BEACON trial, a multi-national, double-blind, placebo-controlled Phase 3 study began in 2011 [217]. A total of 2,185 patients with T2DM and stage 4 CKD were randomized and assigned a treatment of either 20 mg/day bardoxolone or placebo. This trial, however, was terminated prematurely in October 2012 by the Independent Data Monitoring Committee due to adverse events and mortality rates. Ninety six patients in the bardoxolone methyl group were hospitalized or dead due to heart failure, whereas in the placebo group, merely 55 cases were reported. In remaining patients, blood pressure, estimated GFR, and the urinary albumin-to-creatinine ratio increased significantly compared to those of the placebo group.

7. Conclusions

The failure of the bardoxolone methyl clinical study suggests the importance of animal experiments to identify precise modes of action of candidate drugs before applying them to a clinical setting; however, there still remains a possibility that NRF2 activators can be beneficial in managing CKD. As illustrated in Figure 2, there is increasing evidence that oxidative stress and inflammation are integrated contributing factors to CKD progression. Recent extensive studies have established the role of NRF2 signaling in renal protection against oxidative damage, and in modulation of inflammatory response. In addition, due to their traditional application for thousands years, plant-derived NRF2 activators such as resveratrol and curcumin have demonstrated their safety and health benefits in human subjects. Therefore, the utilization of naturally occurring NRF2 activators which share antioxidant and anti-inflammatory efficacies in CKD may be a promising strategy to ameliorate or retard kidney dysfunction.
Figure 2. Factors involved in the progression of kidney dysfunction in CKD and the role of NRF2. In CKD, the alteration of mitochondrial function and the activation of ROS-generating enzymes such as NADPH oxidase and xanthine oxidase participate in aggravated oxidative stress condition in the kidney. The activation of the renin-angiotensin system is another important contributing factor. In addition, oxidative stress triggers NF-κB activation and enhances inflammatory response, which is an important pathologic component of CKD. NRF2 provides renal cells with antioxidant potential by up-regulating an array of genes and consequently attenuates the production of pro-inflammatory cytokines and adhesion molecules.
Figure 2. Factors involved in the progression of kidney dysfunction in CKD and the role of NRF2. In CKD, the alteration of mitochondrial function and the activation of ROS-generating enzymes such as NADPH oxidase and xanthine oxidase participate in aggravated oxidative stress condition in the kidney. The activation of the renin-angiotensin system is another important contributing factor. In addition, oxidative stress triggers NF-κB activation and enhances inflammatory response, which is an important pathologic component of CKD. NRF2 provides renal cells with antioxidant potential by up-regulating an array of genes and consequently attenuates the production of pro-inflammatory cytokines and adhesion molecules.
Molecules 19 12727 g002

Abbreviation

CKD
chronic kidney disease
T2DM
type 2 diabetes mellitus
DN
diabetic nephropathy
GFR
glomerular filtration rate
ECM
extracellular matrix
UUO
unilateral ureteral obstruction
RAS
renin-angiotensin system
ACE
angiotensin-converting enzyme
STZ
streptozotocin
AGE
advanced glycation end products
ROS
reactive oxygen species
8-oxo-dG
8-oxo-7,8-dihydro-2'-deoxyguanosine
GSH
glutathione
GCL
γ-glutamate cysteine ligase
SOD
superoxide dismutase
GPx
glutathione peroxidase
NQO1
NAD(P)H quinone oxidoreductase-1
GST
glutathione S-transferase
HO-1
heme oxygenase-1
UGT
UDP-glucuronosyl transferase
TNFα
tumor necrosis factor-α
IL
interleukin
MDA
malondialdehyde
SFN
sulforaphane
CA
cinnamic aldehyde

Acknowledgments

This research was supported by the National Research Foundation (NRF) funded by the Ministry of Science, ICT & Future Planning (NRF-2013R1A2A2A01015497) and the Bio & Medical Technology Development Program of the NRF funded by the Ministry of Science, ICT & Future Planning (NRF-2013M3A9B5075839).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Small, D.M.; Coombes, J.S.; Bennett, N.; Johnson, D.W.; Gobe, G.C. Oxidative stress, anti-oxidant therapies and chronic kidney disease. Nephrology (Carlton) 2012, 17, 311–321. [Google Scholar] [CrossRef]
  2. Levey, A.S.; Coresh, J.; Balk, E.; Kausz, A.T.; Levin, A.; Steffes, M.W.; Hogg, R.J.; Perrone, R.D.; Lau, J.; Eknoyan, G. National kidney foundation practice guidelines for chronic kidney disease: Evaluation, classification, and stratification. Ann. Intern. Med. 2003, 139, 137–147. [Google Scholar] [CrossRef] [PubMed]
  3. Levey, A.S.; Eckardt, K.U.; Tsukamoto, Y.; Levin, A.; Coresh, J.; Rossert, J.; de Zeeuw, D.; Hostetter, T.H.; Lameire, N.; Eknoyan, G. Definition and classification of chronic kidney disease: A position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2005, 67, 2089–2100. [Google Scholar] [CrossRef] [PubMed]
  4. Laliberte, F.; Bookhart, B.K.; Vekeman, F.; Corral, M.; Duh, M.S.; Bailey, R.A.; Piech, C.T.; Lefebvre, P. Direct all-cause health care costs associated with chronic kidney disease in patients with diabetes and hypertension: A managed care perspective. J. Manag. Care Pharm. 2009, 15, 312–322. [Google Scholar] [PubMed]
  5. Roshan, B.; Stanton, R.C. A story of microalbuminuria and diabetic nephropathy. J. Nephropathol. 2013, 2, 234–240. [Google Scholar] [PubMed]
  6. Saito, H. Toxico-pharmacological perspective of the NRF2-KEAP1 defense system against oxidative stress in kidney diseases. Biochem. Pharmacol. 2013, 85, 865–872. [Google Scholar] [CrossRef] [PubMed]
  7. Iwano, M.; Neilson, E.G. Mechanisms of tubulointerstitial fibrosis. Curr. Opin. Nephrol. Hypertens. 2004, 13, 279–284. [Google Scholar] [CrossRef] [PubMed]
  8. Hart, P.D.; Bakris, G.L. Hypertensive nephropathy: Prevention and treatment recommendations. Expert Opin. Pharmacother. 2010, 11, 2675–2686. [Google Scholar] [CrossRef] [PubMed]
  9. Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am. J. Kidney Dis. 2004, 43, S1–S290. [Google Scholar]
  10. Vaziri, N.D. Oxidative stress in uremia: Nature, mechanisms, and potential consequences. Semin. Nephrol. 2004, 24, 469–473. [Google Scholar] [CrossRef] [PubMed]
  11. Himmelfarb, J.; Stenvinkel, P.; Ikizler, T.A.; Hakim, R.M. The elephant in uremia: Oxidant stress as a unifying concept of cardiovascular disease in uremia. Kidney Int. 2002, 62, 1524–1538. [Google Scholar] [CrossRef] [PubMed]
  12. Himmelfarb, J.; Hakim, R.M. Oxidative stress in uremia. Curr. Opin. Nephrol. Hypertens. 2003, 12, 593–598. [Google Scholar] [CrossRef] [PubMed]
  13. Pieczenik, S.R.; Neustadt, J. Mitochondrial dysfunction and molecular pathways of disease. Exp. Mol. Pathol. 2007, 83, 84–92. [Google Scholar] [CrossRef] [PubMed]
  14. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef] [PubMed]
  15. Wallace, D.C. A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: A dawn for evolutionary medicine. Annu. Rev. Genet. 2005, 39, 359–407. [Google Scholar] [CrossRef] [PubMed]
  16. Yuan, Y.; Chen, Y.; Zhang, P.; Huang, S.; Zhu, C.; Ding, G.; Liu, B.; Yang, T.; Zhang, A. Mitochondrial dysfunction accounts for aldosterone-induced epithelial-to-mesenchymal transition of renal proximal tubular epithelial cells. Free Radic. Biol. Med. 2012, 53, 30–43. [Google Scholar] [CrossRef] [PubMed]
  17. Nishida, H.; Kurahashi, T.; Saito, Y.; Otsuki, N.; Kwon, M.; Ohtake, H.; Yamakawa, M.; Yamada, K.I.; Miyata, S.; Tomita, Y.; et al. Kidney fibrosis is independent of the amount of ascorbic acid in mice with unilateral ureteral obstruction. Free Radic. Res. 2014, 48, 1115–1124. [Google Scholar] [CrossRef] [PubMed]
  18. Granata, S.; Zaza, G.; Simone, S.; Villani, G.; Latorre, D.; Pontrelli, P.; Carella, M.; Schena, F.P.; Grandaliano, G.; Pertosa, G. Mitochondrial dysregulation and oxidative stress in patients with chronic kidney disease. BMC Genomics 2009, 10, 388. [Google Scholar] [CrossRef] [PubMed]
  19. Vaziri, N.D.; Dicus, M.; Ho, N.D.; Boroujerdi-Rad, L.; Sindhu, R.K. Oxidative stress and dysregulation of superoxide dismutase and NADPH oxidase in renal insufficiency. Kidney Int. 2003, 63, 179–185. [Google Scholar] [CrossRef] [PubMed]
  20. Fortuno, A.; Beloqui, O.; San Jose, G.; Moreno, M.U.; Zalba, G.; Diez, J. Increased phagocytic nicotinamide adenine dinucleotide phosphate oxidase-dependent superoxide production in patients with early chronic kidney disease. Kidney Int. Suppl. 2005, 99, S71–S75. [Google Scholar] [CrossRef] [PubMed]
  21. Perianayagam, M.C.; Liangos, O.; Kolyada, A.Y.; Wald, R.; MacKinnon, R.W.; Li, L.; Rao, M.; Balakrishnan, V.S.; Bonventre, J.V.; Pereira, B.J.; et al. NADPH oxidase p22phox and catalase gene variants are associated with biomarkers of oxidative stress and adverse outcomes in acute renal failure. J. Am. Soc. Nephrol. 2007, 18, 255–263. [Google Scholar] [CrossRef] [PubMed]
  22. Sanchez-Lozada, L.G.; Tapia, E.; Soto, A.; Avila-Casado, C.; Franco, M.; Wessale, J.L.; Zhao, L.; Johnson, R.J. Effect of febuxostat on the progression of renal disease in 5/6 nephrectomy rats with and without hyperuricemia. Nephron. Physiol. 2008, 108, 69–78. [Google Scholar]
  23. Kosugi, T.; Nakayama, T.; Heinig, M.; Zhang, L.; Yuzawa, Y.; Sanchez-Lozada, L.G.; Roncal, C.; Johnson, R.J.; Nakagawa, T. Effect of lowering uric acid on renal disease in the type 2 diabetic db/db mice. Am. J. Physiol. Renal. Physiol. 2009, 297, F481–F488. [Google Scholar] [CrossRef] [PubMed]
  24. Omori, H.; Kawada, N.; Inoue, K.; Ueda, Y.; Yamamoto, R.; Matsui, I.; Kaimori, J.; Takabatake, Y.; Moriyama, T.; Isaka, Y.; et al. Use of xanthine oxidase inhibitor febuxostat inhibits renal interstitial inflammation and fibrosis in unilateral ureteral obstructive nephropathy. Clin. Exp. Nephrol. 2012, 16, 549–556. [Google Scholar] [CrossRef] [PubMed]
  25. Shevalye, H.; Lupachyk, S.; Watcho, P.; Stavniichuk, R.; Khazim, K.; Abboud, H.E.; Obrosova, I.G. Prediabetic nephropathy as an early consequence of the high-calorie/high-fat diet: Relation to oxidative stress. Endocrinology 2012, 153, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  26. Kim, H.J.; Sato, T.; Rodriguez-Iturbe, B.; Vaziri, N.D. Role of intrarenal angiotensin system activation, oxidative stress, inflammation, and impaired nuclear factor-erythroid-2-related factor 2 activity in the progression of focal glomerulosclerosis. J. Pharmacol. Exp. Ther. 2011, 337, 583–590. [Google Scholar] [CrossRef] [PubMed]
  27. Vio, C.P.; Jeanneret, V.A. Local induction of angiotensin-converting enzyme in the kidney as a mechanism of progressive renal diseases. Kidney Int. Suppl. 2003, 86, S57–S63. [Google Scholar] [CrossRef] [PubMed]
  28. Nishikawa, T.; Araki, E. Investigation of a novel mechanism of diabetic complications: Impacts of mitochondrial reactive oxygen species. Rinsho. Byori. 2008, 56, 712–719. [Google Scholar] [PubMed]
  29. Hinerfeld, D.; Traini, M.D.; Weinberger, R.P.; Cochran, B.; Doctrow, S.R.; Harry, J.; Melov, S. Endogenous mitochondrial oxidative stress: Neurodegeneration, proteomic analysis, specific respiratory chain defects, and efficacious antioxidant therapy in superoxide dismutase 2 null mice. J. Neurochem. 2004, 88, 657–667. [Google Scholar] [CrossRef] [PubMed]
  30. Brezniceanu, M.L.; Liu, F.; Wei, C.C.; Tran, S.; Sachetelli, S.; Zhang, S.L.; Guo, D.F.; Filep, J.G.; Ingelfinger, J.R.; Chan, J.S. Catalase overexpression attenuates angiotensinogen expression and apoptosis in diabetic mice. Kidney Int. 2007, 71, 912–923. [Google Scholar] [CrossRef] [PubMed]
  31. Hasegawa, G.; Nakano, K.; Sawada, M.; Uno, K.; Shibayama, Y.; Ienaga, K.; Kondo, M. Possible role of tumor necrosis factor and interleukin-1 in the development of diabetic nephropathy. Kidney Int. 1991, 40, 1007–1012. [Google Scholar] [CrossRef] [PubMed]
  32. Navarro, J.F.; Milena, F.J.; Mora, C.; Leon, C.; Garcia, J. Renal pro-inflammatory cytokine gene expression in diabetic nephropathy: Effect of angiotensin-converting enzyme inhibition and pentoxifylline administration. Am. J. Nephrol. 2006, 26, 562–570. [Google Scholar] [CrossRef] [PubMed]
  33. Nakamura, T.; Fukui, M.; Ebihara, I.; Osada, S.; Nagaoka, I.; Tomino, Y.; Koide, H. mRNA expression of growth factors in glomeruli from diabetic rats. Diabetes 1993, 42, 450–456. [Google Scholar] [CrossRef] [PubMed]
  34. Sugimoto, H.; Shikata, K.; Wada, J.; Horiuchi, S.; Makino, H. Advanced glycation end products-cytokine-nitric oxide sequence pathway in the development of diabetic nephropathy: Aminoguanidine ameliorates the overexpression of tumour necrosis factor-alpha and inducible nitric oxide synthase in diabetic rat glomeruli. Diabetologia 1999, 42, 878–886. [Google Scholar] [CrossRef] [PubMed]
  35. Radeke, H.H.; Meier, B.; Topley, N.; Floge, J.; Habermehl, G.G.; Resch, K. Interleukin 1-alpha and tumor necrosis factor-alpha induce oxygen radical production in mesangial cells. Kidney Int. 1990, 37, 767–775. [Google Scholar] [CrossRef] [PubMed]
  36. Koike, N.; Takamura, T.; Kaneko, S. Induction of reactive oxygen species from isolated rat glomeruli by protein kinase c activation and TNF-alpha stimulation, and effects of a phosphodiesterase inhibitor. Life Sci. 2007, 80, 1721–1728. [Google Scholar] [CrossRef] [PubMed]
  37. Coimbra, T.M.; Janssen, U.; Grone, H.J.; Ostendorf, T.; Kunter, U.; Schmidt, H.; Brabant, G.; Floege, J. Early events leading to renal injury in obese zucker (fatty) rats with type ii diabetes. Kidney Int. 2000, 57, 167–182. [Google Scholar] [CrossRef] [PubMed]
  38. Rubio-Guerra, A.F.; Vargas-Robles, H.; Lozano Nuevo, J.J.; Escalante-Acosta, B.A. Correlation between circulating adhesion molecule levels and albuminuria in type-2 diabetic hypertensive patients. Kidney Blood Press Res. 2009, 32, 106–109. [Google Scholar]
  39. Tak, P.P.; Firestein, G.S. NF-kappaB: A key role in inflammatory diseases. J. Clin. Investig. 2001, 107, 7–11. [Google Scholar] [CrossRef] [PubMed]
  40. Greiber, S.; Muller, B.; Daemisch, P.; Pavenstadt, H. Reactive oxygen species alter gene expression in podocytes: Induction of granulocyte macrophage-colony-stimulating factor. J. Am. Soc. Nephrol. 2002, 13, 86–95. [Google Scholar] [PubMed]
  41. Guijarro, C.; Egido, J. Transcription factor-kappa B (NF-kappa B) and renal disease. Kidney Int. 2001, 59, 415–424. [Google Scholar] [CrossRef] [PubMed]
  42. Fujihara, C.K.; Antunes, G.R.; Mattar, A.L.; Malheiros, D.M.; Vieira, J.M., Jr.; Zatz, R. Chronic inhibition of nuclear factor-kappaB attenuates renal injury in the 5/6 renal ablation model. Am. J. Physiol. Renal. Physiol. 2007, 292, F92–F99. [Google Scholar] [CrossRef] [PubMed]
  43. Iwamoto, M.; Mizuiri, S.; Arita, M.; Hemmi, H. Nuclear factor-kappab activation in diabetic rat kidney: Evidence for involvement of p-selectin in diabetic nephropathy. Tohoku J. Exp. Med. 2005, 206, 163–171. [Google Scholar] [CrossRef] [PubMed]
  44. Kuo, H.T.; Kuo, M.C.; Chiu, Y.W.; Chang, J.M.; Guh, J.Y.; Chen, H.C. Increased glomerular and extracellular malondialdehyde levels in patients and rats with focal segmental glomerulosclerosis. Eur. J. Clin. Investig. 2005, 35, 245–250. [Google Scholar] [CrossRef]
  45. Grone, H.J.; Grone, E.F.; Malle, E. Immunohistochemical detection of hypochlorite-modified proteins in glomeruli of human membranous glomerulonephritis. Lab. Investig. 2002, 82, 5–14. [Google Scholar] [CrossRef] [PubMed]
  46. Fujiwara, N.; Nakamura, T.; Sato, E.; Kawagoe, Y.; Hikichi, Y.; Ueda, Y.; Node, K. Renovascular protective effects of erythropoietin in patients with chronic kidney disease. Intern. Med. 2011, 50, 1929–1934. [Google Scholar] [CrossRef] [PubMed]
  47. Dounousi, E.; Papavasiliou, E.; Makedou, A.; Ioannou, K.; Katopodis, K.P.; Tselepis, A.; Siamopoulos, K.C.; Tsakiris, D. Oxidative stress is progressively enhanced with advancing stages of CKD. Am. J. Kidney Dis. 2006, 48, 752–760. [Google Scholar] [CrossRef] [PubMed]
  48. Terawaki, H.; Yoshimura, K.; Hasegawa, T.; Matsuyama, Y.; Negawa, T.; Yamada, K.; Matsushima, M.; Nakayama, M.; Hosoya, T.; Era, S. Oxidative stress is enhanced in correlation with renal dysfunction: Examination with the redox state of albumin. Kidney Int. 2004, 66, 1988–1993. [Google Scholar] [CrossRef] [PubMed]
  49. Ceballos-Picot, I.; Witko-Sarsat, V.; Merad-Boudia, M.; Nguyen, A.T.; Thevenin, M.; Jaudon, M.C.; Zingraff, J.; Verger, C.; Jungers, P.; Descamps-Latscha, B. Glutathione antioxidant system as a marker of oxidative stress in chronic renal failure. Free Radic. Biol. Med. 1996, 21, 845–853. [Google Scholar]
  50. Ongajooth, L.; Ongajyooth, S.; Likidlilid, A.; Chantachum, Y.; Shayakul, C.; Nilwarangkur, S. Role of lipid peroxidation, trace elements and anti-oxidant enzymes in chronic renal disease patients. J. Med. Assoc. Thai. 1996, 79, 791–800. [Google Scholar] [PubMed]
  51. Shurtz-Swirski, R.; Mashiach, E.; Kristal, B.; Shkolnik, T.; Shasha, S.M. Antioxidant enzymes activity in polymorphonuclear leukocytes in chronic renal failure. Nephron 1995, 71, 176–179. [Google Scholar]
  52. Lee, S.J.; Choi, M.G.; Kim, D.S.; Kim, T.W. Manganese superoxide dismutase gene polymorphism (V16A) is associated with stages of albuminuria in Korean type 2 diabetic patients. Metabolism 2006, 55, 1–7. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, L.; Zheng, T.; Wang, N.; Wang, F.; Li, M.; Jiang, J.; Zhao, R.; Li, L.; Zhao, W.; Zhu, Q.; et al. The manganese superoxide dismutase Val16Ala polymorphism is associated with decreased risk of diabetic nephropathy in Chinese patients with type 2 diabetes. Mol. Cell Biochem. 2009, 322, 87–91. [Google Scholar] [CrossRef] [PubMed]
  54. Nomiyama, T.; Tanaka, Y.; Piao, L.; Nagasaka, K.; Sakai, K.; Ogihara, T.; Nakajima, K.; Watada, H.; Kawamori, R. The polymorphism of manganese superoxide dismutase is associated with diabetic nephropathy in Japanese type 2 diabetic patients. J. Hum. Genet. 2003, 48, 138–141. [Google Scholar] [PubMed]
  55. Yilmaz, M.I.; Saglam, M.; Caglar, K.; Cakir, E.; Sonmez, A.; Ozgurtas, T.; Aydin, A.; Eyileten, T.; Ozcan, O.; Acikel, C.; et al. The determinants of endothelial dysfunction in CKD: Oxidative stress and asymmetric dimethylarginine. Am. J. Kidney Dis. 2006, 47, 42–50. [Google Scholar] [CrossRef] [PubMed]
  56. Ramos, L.F.; Shintani, A.; Ikizler, T.A.; Himmelfarb, J. Oxidative stress and inflammation are associated with adiposity in moderate to severe CKD. J. Am. Soc. Nephrol. 2008, 19, 593–599. [Google Scholar] [CrossRef] [PubMed]
  57. Ikizler, T.A.; Morrow, J.D.; Roberts, L.J.; Evanson, J.A.; Becker, B.; Hakim, R.M.; Shyr, Y.; Himmelfarb, J. Plasma F2-isoprostane levels are elevated in chronic hemodialysis patients. Clin. Nephrol. 2002, 58, 190–197. [Google Scholar] [CrossRef] [PubMed]
  58. Handelman, G.J.; Walter, M.F.; Adhikarla, R.; Gross, J.; Dallal, G.E.; Levin, N.W.; Blumberg, J.B. Elevated plasma F2-isoprostanes in patients on long-term hemodialysis. Kidney Int. 2001, 59, 1960–1966. [Google Scholar] [CrossRef] [PubMed]
  59. Atamer, A.; Kocyigit, Y.; Ecder, S.A.; Selek, S.; Ilhan, N.; Ecder, T.; Atamer, Y. Effect of oxidative stress on antioxidant enzyme activities, homocysteine and lipoproteins in chronic kidney disease. J. Nephrol. 2008, 21, 924–930. [Google Scholar] [PubMed]
  60. Apeland, T.; Mansoor, M.A.; Seljeflot, I.; Bronstad, I.; Goransson, L.; Strandjord, R.E. Homocysteine, malondialdehyde and endothelial markers in dialysis patients during low-dose folinic acid therapy. J. Intern. Med. 2002, 252, 456–464. [Google Scholar] [CrossRef] [PubMed]
  61. Caimi, G.; Carollo, C.; Montana, M.; Iatrino, R.; Bondi, B.; Lo Presti, R. Nitric oxide metabolites, leukocyte activation markers and oxidative status in dialyzed subjects. Blood Purif. 2009, 27, 194–198. [Google Scholar] [CrossRef] [PubMed]
  62. Pieniazek, A.; Brzeszczynska, J.; Kruszynska, I.; Gwozdzinski, K. Investigation of albumin properties in patients with chronic renal failure. Free Radic. Res. 2009, 43, 1008–1018. [Google Scholar] [CrossRef] [PubMed]
  63. Miyata, T.; Saito, A.; Kurokawa, K.; van Ypersele de Strihou, C. Advanced glycation and lipoxidation end products: Reactive carbonyl compounds-related uraemic toxicity. Nephrol. Dial. Transplant 2001, 16 (Suppl. 4), 8–11. [Google Scholar]
  64. Sakata, N.; Imanaga, Y.; Meng, J.; Tachikawa, Y.; Takebayashi, S.; Nagai, R.; Horiuchi, S. Increased advanced glycation end products in atherosclerotic lesions of patients with end-stage renal disease. Atherosclerosis 1999, 142, 67–77. [Google Scholar] [CrossRef] [PubMed]
  65. Witko-Sarsat, V.; Friedlander, M.; Capeillere-Blandin, C.; Nguyen, A.T.; Zingraff, J.; Jungers, P.; Descamps-Latscha, B. Advanced oxidation protein products as a novel marker of oxidative stress in uremia. Kidney Int. 1996, 49, 1304–1313. [Google Scholar]
  66. Valli, A.; Suliman, M.E.; Meert, N.; Vanholder, R.; Lindholm, B.; Stenvinkel, P.; Watanabe, M.; Barany, P.; Alvestrand, A.; Anderstam, B. Overestimation of advanced oxidation protein products in uremic plasma due to presence of triglycerides and other endogenous factors. Clin. Chim. Acta 2007, 379, 87–94. [Google Scholar] [CrossRef] [PubMed]
  67. Holvoet, P.; Donck, J.; Landeloos, M.; Brouwers, E.; Luijtens, K.; Arnout, J.; Lesaffre, E.; Vanrenterghem, Y.; Collen, D. Correlation between oxidized low density lipoproteins and von willebrand factor in chronic renal failure. Thromb. Haemost. 1996, 76, 663–669. [Google Scholar] [PubMed]
  68. Tarng, D.C.; Wen Chen, T.; Huang, T.P.; Chen, C.L.; Liu, T.Y.; Wei, Y.H. Increased oxidative damage to peripheral blood leukocyte DNA in chronic peritoneal dialysis patients. J. Am. Soc. Nephrol. 2002, 13, 1321–1330. [Google Scholar] [CrossRef] [PubMed]
  69. Ersson, C.; Thorman, R.; Rodhe, Y.; Moller, L.; Hylander, B. DNA damage in salivary gland tissue in patients with chronic kidney disease, measured by the comet assay. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2011, 112, 209–215. [Google Scholar] [CrossRef] [PubMed]
  70. Domenici, F.A.; Vannucchi, M.T.; Jordao, A.A., Jr.; Meirelles, M.S.; Vannucchi, H. DNA oxidative damage in patients with dialysis treatment. Ren. Fail. 2005, 27, 689–694. [Google Scholar] [CrossRef] [PubMed]
  71. Tonelli, M.; Sacks, F.; Pfeffer, M.; Jhangri, G.S.; Curhan, G. Biomarkers of inflammation and progression of chronic kidney disease. Kidney Int. 2005, 68, 237–245. [Google Scholar] [CrossRef] [PubMed]
  72. Shlipak, M.G.; Fried, L.F.; Crump, C.; Bleyer, A.J.; Manolio, T.A.; Tracy, R.P.; Furberg, C.D.; Psaty, B.M. Elevations of inflammatory and procoagulant biomarkers in elderly persons with renal insufficiency. Circulation 2003, 107, 87–92. [Google Scholar] [CrossRef] [PubMed]
  73. Pereira, B.J.; Shapiro, L.; King, A.J.; Falagas, M.E.; Strom, J.A.; Dinarello, C.A. Plasma levels of IL-1 beta, TNF alpha and their specific inhibitors in undialyzed chronic renal failure, CAPD and hemodialysis patients. Kidney Int. 1994, 45, 890–896. [Google Scholar] [CrossRef] [PubMed]
  74. Knight, E.L.; Rimm, E.B.; Pai, J.K.; Rexrode, K.M.; Cannuscio, C.C.; Manson, J.E.; Stampfer, M.J.; Curhan, G.C. Kidney dysfunction, inflammation, and coronary events: A prospective study. J. Am. Soc. Nephrol. 2004, 15, 1897–1903. [Google Scholar] [CrossRef] [PubMed]
  75. Meuwese, C.L.; Stenvinkel, P.; Dekker, F.W.; Carrero, J.J. Monitoring of inflammation in patients on dialysis: Forewarned is forearmed. Nat. Rev. Nephrol. 2011, 7, 166–176. [Google Scholar] [CrossRef] [PubMed]
  76. Herbelin, A.; Nguyen, A.T.; Zingraff, J.; Urena, P.; Descamps-Latscha, B. Influence of uremia and hemodialysis on circulating interleukin-1 and tumor necrosis factor alpha. Kidney Int. 1990, 37, 116–125. [Google Scholar] [CrossRef] [PubMed]
  77. Oberg, B.P.; McMenamin, E.; Lucas, F.L.; McMonagle, E.; Morrow, J.; Ikizler, T.A.; Himmelfarb, J. Increased prevalence of oxidant stress and inflammation in patients with moderate to severe chronic kidney disease. Kidney Int. 2004, 65, 1009–1016. [Google Scholar] [CrossRef] [PubMed]
  78. Ishii, T.; Itoh, K.; Takahashi, S.; Sato, H.; Yanagawa, T.; Katoh, Y.; Bannai, S.; Yamamoto, M. Transcription factor NRF2 coordinately regulates a group of oxidative stress-inducible genes in macrophages. J. Biol. Chem. 2000, 275, 16023–16029. [Google Scholar] [CrossRef] [PubMed]
  79. Itoh, K.; Wakabayashi, N.; Katoh, Y.; Ishii, T.; Igarashi, K.; Engel, J.D.; Yamamoto, M. KEAP1 represses nuclear activation of antioxidant responsive elements by NRF2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999, 13, 76–86. [Google Scholar] [CrossRef] [PubMed]
  80. Tong, K.I.; Katoh, Y.; Kusunoki, H.; Itoh, K.; Tanaka, T.; Yamamoto, M. KEAP1 recruits neh2 through binding to ETGE and DLG motifs: Characterization of the two-site molecular recognition model. Mol. Cell. Biol. 2006, 26, 2887–2900. [Google Scholar] [CrossRef] [PubMed]
  81. Tong, K.I.; Padmanabhan, B.; Kobayashi, A.; Shang, C.; Hirotsu, Y.; Yokoyama, S.; Yamamoto, M. Different electrostatic potentials define ETGE and DLG motifs as hinge and latch in oxidative stress response. Mol. Cell. Biol. 2007, 27, 7511–7521. [Google Scholar] [CrossRef] [PubMed]
  82. Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; et al. An NRF2/small Maf heterodimer mediates the induction of phase ii detoxifying enzyme genes through antioxidant response elements. Biochem. Biophys. Res. Commun. 1997, 236, 313–322. [Google Scholar] [CrossRef] [PubMed]
  83. Cho, H.Y.; Kleeberger, S.R. NRF2 protects against airway disorders. Toxicol. Appl. Pharmacol. 2010, 244, 43–56. [Google Scholar] [CrossRef] [PubMed]
  84. Hayes, J.D.; Dinkova-Kostova, A.T. The NRF2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 2014, 39, 199–218. [Google Scholar] [CrossRef] [PubMed]
  85. Hayes, J.D.; McMahon, M. NRF2 and KEAP1 mutations: Permanent activation of an adaptive response in cancer. Trends Biochem. Sci. 2009, 34, 176–188. [Google Scholar] [CrossRef] [PubMed]
  86. Jung, K.A.; Kwak, M.K. The NRF2 system as a potential target for the development of indirect antioxidants. Molecules 2010, 15, 7266–7291. [Google Scholar] [CrossRef] [PubMed]
  87. Enomoto, A.; Itoh, K.; Nagayoshi, E.; Haruta, J.; Kimura, T.; O’Connor, T.; Harada, T.; Yamamoto, M. High sensitivity of nrf2 knockout mice to acetaminophen hepatotoxicity associated with decreased expression of ARE-regulated drug metabolizing enzymes and antioxidant genes. Toxicol. Sci. 2001, 59, 169–177. [Google Scholar] [CrossRef] [PubMed]
  88. Rangasamy, T.; Cho, C.Y.; Thimmulappa, R.K.; Zhen, L.; Srisuma, S.S.; Kensler, T.W.; Yamamoto, M.; Petrache, I.; Tuder, R.M.; Biswal, S. Genetic ablation of NRF2 enhances susceptibility to cigarette smoke-induced emphysema in mice. J. Clin. Invest. 2004, 114, 1248–1259. [Google Scholar] [CrossRef] [PubMed]
  89. Aoki, Y.; Sato, H.; Nishimura, N.; Takahashi, S.; Itoh, K.; Yamamoto, M. Accelerated DNA adduct formation in the lung of the nrf2 knockout mouse exposed to diesel exhaust. Toxicol. Appl. Pharmacol. 2001, 173, 154–160. [Google Scholar] [CrossRef] [PubMed]
  90. Rangasamy, T.; Guo, J.; Mitzner, W.A.; Roman, J.; Singh, A.; Fryer, A.D.; Yamamoto, M.; Kensler, T.W.; Tuder, R.M.; Georas, S.N.; et al. Disruption of nrf2 enhances susceptibility to severe airway inflammation and asthma in mice. J. Exp. Med. 2005, 202, 47–59. [Google Scholar] [CrossRef] [PubMed]
  91. Calkins, M.J.; Johnson, D.A.; Townsend, J.A.; Vargas, M.R.; Dowell, J.A.; Williamson, T.P.; Kraft, A.D.; Lee, J.M.; Li, J.; Johnson, J.A. The NRF2/ARE pathway as a potential therapeutic target in neurodegenerative disease. Antioxid. Redox Signal. 2009, 11, 497–508. [Google Scholar] [CrossRef] [PubMed]
  92. Yoh, K.; Itoh, K.; Enomoto, A.; Hirayama, A.; Yamaguchi, N.; Kobayashi, M.; Morito, N.; Koyama, A.; Yamamoto, M.; Takahashi, S. Nrf2-deficient female mice develop lupus-like autoimmune nephritis. Kidney Int. 2001, 60, 1343–1353. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, M.; Grigoryev, D.N.; Crow, M.T.; Haas, M.; Yamamoto, M.; Reddy, S.P.; Rabb, H. Transcription factor NRF2 is protective during ischemic and nephrotoxic acute kidney injury in mice. Kidney Int. 2009, 76, 277–285. [Google Scholar] [CrossRef] [PubMed]
  94. Shin, D.H.; Park, H.M.; Jung, K.A.; Choi, H.G.; Kim, J.A.; Kim, D.D.; Kim, S.G.; Kang, K.W.; Ku, S.K.; Kensler, T.W.; et al. The NRF2-heme oxygenase-1 system modulates cyclosporin a-induced epithelial-mesenchymal transition and renal fibrosis. Free Radic. Biol. Med. 2010, 48, 1051–1063. [Google Scholar] [CrossRef] [PubMed]
  95. Jiang, T.; Huang, Z.; Lin, Y.; Zhang, Z.; Fang, D.; Zhang, D.D. The protective role of NRF2 in streptozotocin-induced diabetic nephropathy. Diabetes 2010, 59, 850–860. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, C.; Kong, A.N. Dietary cancer-chemopreventive compounds: From signaling and gene expression to pharmacological effects. Trends Pharmacol. Sci. 2005, 26, 318–326. [Google Scholar] [CrossRef] [PubMed]
  97. Zhao, C.R.; Gao, Z.H.; Qu, X.J. NRF2-ARE signaling pathway and natural products for cancer chemoprevention. Cancer Epidemiol. 2010, 34, 523–533. [Google Scholar] [CrossRef] [PubMed]
  98. Chen, C.; Kong, A.N. Dietary chemopreventive compounds and ARE/EpRE signaling. Free Radic. Biol. Med. 2004, 36, 1505–1516. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, D.D.; Hannink, M. Distinct cysteine residues in KEAP1 are required for KEAP1-dependent ubiquitination of NRF2 and for stabilization of NRF2 by chemopreventive agents and oxidative stress. Mol. Cell. Biol. 2003, 23, 8137–8151. [Google Scholar] [CrossRef] [PubMed]
  100. Brooks, J.D.; Paton, V. Potent induction of carcinogen defence enzymes with sulforaphane, a putative prostate cancer chemopreventive agent. Prostate Cancer Prostatic Dis. 1999, 2, S8. [Google Scholar] [CrossRef] [PubMed]
  101. Hu, R.; Hebbar, V.; Kim, B.R.; Chen, C.; Winnik, B.; Buckley, B.; Soteropoulos, P.; Tolias, P.; Hart, R.P.; Kong, A.N. In vivo pharmacokinetics and regulation of gene expression profiles by isothiocyanate sulforaphane in the rat. J. Pharmacol. Exp. Ther. 2004, 310, 263–271. [Google Scholar] [CrossRef] [PubMed]
  102. Bacon, J.R.; Williamson, G.; Garner, R.C.; Lappin, G.; Langouet, S.; Bao, Y. Sulforaphane and quercetin modulate PhIP-DNA adduct formation in human HepG2 cells and hepatocytes. Carcinogenesis 2003, 24, 1903–1911. [Google Scholar] [CrossRef] [PubMed]
  103. Basten, G.P.; Bao, Y.; Williamson, G. Sulforaphane and its glutathione conjugate but not sulforaphane nitrile induce UDP-glucuronosyl transferase (UGT1A1) and glutathione transferase (GSTA1) in cultured cells. Carcinogenesis 2002, 23, 1399–1404. [Google Scholar] [CrossRef] [PubMed]
  104. Thimmulappa, R.K.; Mai, K.H.; Srisuma, S.; Kensler, T.W.; Yamamoto, M.; Biswal, S. Identification of NRF2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray. Cancer Res. 2002, 62, 5196–5203. [Google Scholar] [PubMed]
  105. Rose, P.; Faulkner, K.; Williamson, G.; Mithen, R. 7-methylsulfinylheptyl and 8-methylsulfinyloctyl isothiocyanates from watercress are potent inducers of phase II enzymes. Carcinogenesis 2000, 21, 1983–1988. [Google Scholar] [CrossRef] [PubMed]
  106. Dinkova-Kostova, A.T.; Holtzclaw, W.D.; Cole, R.N.; Itoh, K.; Wakabayashi, N.; Katoh, Y.; Yamamoto, M.; Talalay, P. Direct evidence that sulfhydryl groups of KEAP1 are the sensors regulating induction of phase 2 enzymes that protect against carcinogens and oxidants. Proc. Natl. Acad. Sci. USA 2002, 99, 11908–11913. [Google Scholar]
  107. Dinkova-Kostova, A.T.; Massiah, M.A.; Bozak, R.E.; Hicks, R.J.; Talalay, P. Potency of michael reaction acceptors as inducers of enzymes that protect against carcinogenesis depends on their reactivity with sulfhydryl groups. Proc. Natl. Acad. Sci. USA 2001, 98, 3404–3409. [Google Scholar]
  108. Dinkova-Kostova, A.T.; Talalay, P. Relation of structure of curcumin analogs to their potencies as inducers of phase 2 detoxification enzymes. Carcinogenesis 1999, 20, 911–914. [Google Scholar]
  109. Murakami, A.; Ashida, H.; Terao, J. Multitargeted cancer prevention by quercetin. Cancer Lett. 2008, 269, 315–325. [Google Scholar] [CrossRef] [PubMed]
  110. Galati, G.; O’Brien, P.J. Potential toxicity of flavonoids and other dietary phenolics: Significance for their chemopreventive and anticancer properties. Free Radic. Biol. Med. 2004, 37, 287–303. [Google Scholar] [CrossRef] [PubMed]
  111. Tanigawa, S.; Fujii, M.; Hou, D.X. Action of NRF2 and KEAP1 in KEAP1-mediated NQO1 expression by quercetin. Free Radic. Biol. Med. 2007, 42, 1690–1703. [Google Scholar] [CrossRef] [PubMed]
  112. Salminen, A.; Huuskonen, J.; Ojala, J.; Kauppinen, A.; Kaarniranta, K.; Suuronen, T. Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res. Rev. 2008, 7, 83–105. [Google Scholar] [CrossRef] [PubMed]
  113. Pedruzzi, L.M.; Stockler-Pinto, M.B.; Leite, M., Jr.; Mafra, D. NRF2-KEAP1 system versus NF-kappaB: The good and the evil in chronic kidney disease? Biochimie 2012, 94, 2461–2466. [Google Scholar] [CrossRef] [PubMed]
  114. Thimmulappa, R.K.; Lee, H.; Rangasamy, T.; Reddy, S.P.; Yamamoto, M.; Kensler, T.W.; Biswal, S. NRF2 is a critical regulator of the innate immune response and survival during experimental sepsis. J. Clin. Invest. 2006, 116, 984–995. [Google Scholar] [CrossRef] [PubMed]
  115. Yang, H.; Magilnick, N.; Lee, C.; Kalmaz, D.; Ou, X.; Chan, J.Y.; Lu, S.C. NRF1 and NRF2 regulate rat glutamate-cysteine ligase catalytic subunit transcription indirectly via NF-kappaB and AP-1. Mol. Cell. Biol. 2005, 25, 5933–5946. [Google Scholar] [CrossRef] [PubMed]
  116. Mao, L.; Wang, H.; Qiao, L.; Wang, X. Disruption of nrf2 enhances the upregulation of nuclear factor-kappaB activity, tumor necrosis factor-alpha, and matrix metalloproteinase-9 after spinal cord injury in mice. Mediators Inflamm. 2010, 2010, 238321. [Google Scholar]
  117. Jun, C.D.; Kim, Y.; Choi, E.Y.; Kim, M.; Park, B.; Youn, B.; Yu, K.; Choi, K.S.; Yoon, K.H.; Choi, S.C.; et al. Gliotoxin reduces the severity of trinitrobenzene sulfonic acid-induced colitis in mice: Evidence of the connection between heme oxygenase-1 and the nuclear factor-kappaB pathway in vitro and in vivo. Inflamm. Bowel. Dis. 2006, 12, 619–629. [Google Scholar] [CrossRef] [PubMed]
  118. Rushworth, S.A.; MacEwan, D.J.; O'Connell, M.A. Lipopolysaccharide-induced expression of nad(p)h: Quinone oxidoreductase 1 and heme oxygenase-1 protects against excessive inflammatory responses in human monocytes. J. Immunol. 2008, 181, 6730–6737. [Google Scholar] [CrossRef] [PubMed]
  119. Heiss, E.; Herhaus, C.; Klimo, K.; Bartsch, H.; Gerhauser, C. Nuclear factor kappa B is a molecular target for sulforaphane-mediated anti-inflammatory mechanisms. J. Biol. Chem. 2001, 276, 32008–32015. [Google Scholar] [CrossRef] [PubMed]
  120. Pan, M.H.; Lin-Shiau, S.Y.; Lin, J.K. Comparative studies on the suppression of nitric oxide synthase by curcumin and its hydrogenated metabolites through down-regulation of IkappaB kinase and NFkappaB activation in macrophages. Biochem. Pharmacol. 2000, 60, 1665–1676. [Google Scholar] [CrossRef] [PubMed]
  121. Ahmad, R.; Raina, D.; Meyer, C.; Kharbanda, S.; Kufe, D. Triterpenoid CDDO-Me blocks the NF-kappaB pathway by direct inhibition of IKKbeta on Cys-179. J. Biol. Chem. 2006, 281, 35764–35769. [Google Scholar] [CrossRef] [PubMed]
  122. Kovacic, P.; Jacintho, J.D. Systemic lupus erythematosus and other autoimmune diseases from endogenous and exogenous agents: Unifying theme of oxidative stress. Mini Rev. Med. Chem. 2003, 3, 568–575. [Google Scholar] [CrossRef] [PubMed]
  123. Jiang, T.; Tian, F.; Zheng, H.; Whitman, S.A.; Lin, Y.; Zhang, Z.; Zhang, N.; Zhang, D.D. NRF2 suppresses lupus nephritis through inhibition of oxidative injury and the NF-kappaB-mediated inflammatory response. Kidney Int. 2014, 85, 333–343. [Google Scholar] [CrossRef] [PubMed]
  124. Tsai, P.Y.; Ka, S.M.; Chang, J.M.; Chen, H.C.; Shui, H.A.; Li, C.Y.; Hua, K.F.; Chang, W.L.; Huang, J.J.; Yang, S.S.; et al. Epigallocatechin-3-gallate prevents lupus nephritis development in mice via enhancing the NRF2 antioxidant pathway and inhibiting NLRP3 inflammasome activation. Free Radic. Biol. Med. 2011, 51, 744–754. [Google Scholar] [CrossRef] [PubMed]
  125. Lee, H.; Kim, H.; Lee, G.; Chung, H.S.; Bae, H. Curcumin attenuates lupus nephritis upon interaction with regulatory t cells in new zealand black/white mice. Br. J. Nutr. 2013, 110, 69–76. [Google Scholar] [CrossRef] [PubMed]
  126. Wu, T.; Ye, Y.; Min, S.Y.; Zhu, J.; Khobahy, E.; Zhou, J.; Yan, M.; Hemachandran, S.; Pathak, S.; Zhou, X.J.; et al. Targeting multiple signaling axes and oxidative stress using a synthetic triterpenoid prevents murine lupus nephritis. Arthritis Rheumatol. 2014. [Google Scholar] [CrossRef]
  127. Yoh, K.; Hirayama, A.; Ishizaki, K.; Yamada, A.; Takeuchi, M.; Yamagishi, S.; Morito, N.; Nakano, T.; Ojima, M.; Shimohata, H.; et al. Hyperglycemia induces oxidative and nitrosative stress and increases renal functional impairment in nrf2-deficient mice. Genes Cells 2008, 13, 1159–1170. [Google Scholar] [PubMed]
  128. Ryoo, I.G.; Ha, H.; Kwak, M.K. Inhibitory role of the KEAP1-NRF2 pathway in TGFbeta1-stimulated renal epithelial transition to fibroblastic cells: A modulatory effect on smad signaling. PLoS One 2014, 9, e93265. [Google Scholar] [CrossRef] [PubMed]
  129. Ryoo, I.G.; Shin, D.H.; Kang, K.S.; Kwak, M.K. Involvement of NRF2-GSH signaling in TGFbeta1-stimulated epithelial-to-mesenchymal transition changes in rat renal tubular cells. Arch. Pharm. Res. 2014. [Google Scholar] [CrossRef]
  130. Kim, H.J.; Vaziri, N.D. Contribution of impaired NRF2-KEAP1 pathway to oxidative stress and inflammation in chronic renal failure. Am. J. Physiol. Renal. Physiol. 2010, 298, F662–F671. [Google Scholar] [CrossRef] [PubMed]
  131. Aminzadeh, M.A.; Nicholas, S.B.; Norris, K.C.; Vaziri, N.D. Role of impaired NRF2 activation in the pathogenesis of oxidative stress and inflammation in chronic tubulo-interstitial nephropathy. Nephrol. Dial. Transplant. 2013, 28, 2038–2045. [Google Scholar] [CrossRef] [PubMed]
  132. Kumawat, M.; Sharma, T.K.; Singh, I.; Singh, N.; Ghalaut, V.S.; Vardey, S.K.; Shankar, V. Antioxidant enzymes and lipid peroxidation in type 2 diabetes mellitus patients with and without nephropathy. N. Am. J. Med. Sci. 2013, 5, 213–219. [Google Scholar] [CrossRef] [PubMed]
  133. Crawford, A.; Fassett, R.G.; Coombes, J.S.; Kunde, D.A.; Ahuja, K.D.; Robertson, I.K.; Ball, M.J.; Geraghty, D.P. Relationship between antioxidant enzyme genotype and activity and kidney function: A case-control study. Clin. Nephrol. 2012, 78, 135–144. [Google Scholar] [CrossRef] [PubMed]
  134. Puchades, M.J.; Saez, G.; Munoz, M.C.; Gonzalez, M.; Torregrosa, I.; Juan, I.; Miguel, A. Study of oxidative stress in patients with advanced renal disease and undergoing either hemodialysis or peritoneal dialysis. Clin. Nephrol. 2013, 80, 177–186. [Google Scholar] [CrossRef] [PubMed]
  135. Li, H.; Wang, F.; Zhang, L.; Cao, Y.; Liu, W.; Hao, J.; Liu, Q.; Duan, H. Modulation of NRF2 expression alters high glucose-induced oxidative stress and antioxidant gene expression in mouse mesangial cells. Cell Signal. 2011, 23, 1625–1632. [Google Scholar] [CrossRef] [PubMed]
  136. Kraft, D.C.; Deocaris, C.C.; Wadhwa, R.; Rattan, S.I. Preincubation with the proteasome inhibitor mg-132 enhances proteasome activity via the NRF2 transcription factor in aging human skin fibroblasts. Ann. New York Acad. Sci. 2006, 1067, 420–424. [Google Scholar]
  137. Luo, Z.F.; Qi, W.; Feng, B.; Mu, J.; Zeng, W.; Guo, Y.H.; Pang, Q.; Ye, Z.L.; Liu, L.; Yuan, F.H. Prevention of diabetic nephropathy in rats through enhanced renal antioxidative capacity by inhibition of the proteasome. Life Sci. 2011, 88, 512–520. [Google Scholar] [CrossRef] [PubMed]
  138. Cui, W.; Li, B.; Bai, Y.; Miao, X.; Chen, Q.; Sun, W.; Tan, Y.; Luo, P.; Zhang, C.; Zheng, S.; et al. Potential role for NRF2 activation in the therapeutic effect of MG132 on diabetic nephropathy in OVE26 diabetic mice. Am. J. Physiol. Endocrinol. Metab. 2013, 304, E87–E99. [Google Scholar] [CrossRef] [PubMed]
  139. Dou, L.; Jourde-Chiche, N.; Faure, V.; Cerini, C.; Berland, Y.; Dignat-George, F.; Brunet, P. The uremic solute indoxyl sulfate induces oxidative stress in endothelial cells. J. Thromb. Haemost. 2007, 5, 1302–1308. [Google Scholar] [CrossRef] [PubMed]
  140. Yu, M.; Kim, Y.J.; Kang, D.H. Indoxyl sulfate-induced endothelial dysfunction in patients with chronic kidney disease via an induction of oxidative stress. Clin. J. Am. Soc. Nephrol. 2011, 6, 30–39. [Google Scholar] [CrossRef] [PubMed]
  141. Bolati, D.; Shimizu, H.; Yisireyili, M.; Nishijima, F.; Niwa, T. Indoxyl sulfate, a uremic toxin, downregulates renal expression of NRF2 through activation of NF-kappaB. BMC Nephrol. 2013, 14, 56. [Google Scholar] [CrossRef] [PubMed]
  142. Fahey, J.W.; Talalay, P. Antioxidant functions of sulforaphane: A potent inducer of phase II detoxication enzymes. Food Chem. Toxicol. 1999, 37, 973–979. [Google Scholar] [CrossRef] [PubMed]
  143. Keum, Y.S.; Jeong, W.S.; Kong, A.N. Chemoprevention by isothiocyanates and their underlying molecular signaling mechanisms. Mutat. Res. 2004, 555, 191–202. [Google Scholar] [CrossRef] [PubMed]
  144. Angeloni, C.; Leoncini, E.; Malaguti, M.; Angelini, S.; Hrelia, P.; Hrelia, S. Modulation of phase ii enzymes by sulforaphane: Implications for its cardioprotective potential. J. Agric. Food Chem. 2009, 57, 5615–5622. [Google Scholar] [CrossRef] [PubMed]
  145. Mukherjee, S.; Gangopadhyay, H.; Das, D.K. Broccoli: A unique vegetable that protects mammalian hearts through the redox cycling of the thioredoxin superfamily. J. Agric. Food Chem. 2008, 56, 609–617. [Google Scholar] [CrossRef] [PubMed]
  146. Innamorato, N.G.; Rojo, A.I.; Garcia-Yague, A.J.; Yamamoto, M.; de Ceballos, M.L.; Cuadrado, A. The transcription factor NRF2 is a therapeutic target against brain inflammation. J. Immunol. 2008, 181, 680–689. [Google Scholar] [CrossRef] [PubMed]
  147. Juge, N.; Mithen, R.F.; Traka, M. Molecular basis for chemoprevention by sulforaphane: A comprehensive review. Cell. Mol. Life Sci. 2007, 64, 1105–1127. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, D.D. Mechanistic studies of the NRF2-KEAP1 signaling pathway. Drug Metab. Rev. 2006, 38, 769–789. [Google Scholar] [CrossRef] [PubMed]
  149. Guerrero-Beltran, C.E.; Calderon-Oliver, M.; Martinez-Abundis, E.; Tapia, E.; Zarco-Marquez, G.; Zazueta, C.; Pedraza-Chaverri, J. Protective effect of sulforaphane against cisplatin-induced mitochondrial alterations and impairment in the activity of NAD(P)H: Quinone oxidoreductase 1 and gamma glutamyl cysteine ligase: Studies in mitochondria isolated from rat kidney and in LLC-PK1 cells. Toxicol. Lett. 2010, 199, 80–92. [Google Scholar]
  150. Guerrero-Beltran, C.E.; Calderon-Oliver, M.; Tapia, E.; Medina-Campos, O.N.; Sanchez-Gonzalez, D.J.; Martinez-Martinez, C.M.; Ortiz-Vega, K.M.; Franco, M.; Pedraza-Chaverri, J. Sulforaphane protects against cisplatin-induced nephrotoxicity. Toxicol. Lett. 2010, 192, 278–285. [Google Scholar]
  151. Yoon, H.Y.; Kang, N.I.; Lee, H.K.; Jang, K.Y.; Park, J.W.; Park, B.H. Sulforaphane protects kidneys against ischemia-reperfusion injury through induction of the NRF2-dependent phase 2 enzyme. Biochem. Pharmacol. 2008, 75, 2214–2223. [Google Scholar] [CrossRef] [PubMed]
  152. Zheng, H.; Whitman, S.A.; Wu, W.; Wondrak, G.T.; Wong, P.K.; Fang, D.; Zhang, D.D. Therapeutic potential of NRF2 activators in streptozotocin-induced diabetic nephropathy. Diabetes 2011, 60, 3055–3066. [Google Scholar] [CrossRef] [PubMed]
  153. Cui, W.; Bai, Y.; Miao, X.; Luo, P.; Chen, Q.; Tan, Y.; Rane, M.J.; Miao, L.; Cai, L. Prevention of diabetic nephropathy by sulforaphane: Possible role of NRF2 upregulation and activation. Oxid. Med. Cell Longev. 2012, 2012, 821936. [Google Scholar]
  154. Chung, S.D.; Lai, T.Y.; Chien, C.T.; Yu, H.J. Activating NRF-2 signaling depresses unilateral ureteral obstruction-evoked mitochondrial stress-related autophagy, apoptosis and pyroptosis in kidney. PLoS One 2012, 7, e47299. [Google Scholar] [CrossRef] [PubMed]
  155. Noorafshan, A.; Karbalay-Doust, S.; Poorshahid, M. Stereological survey of the ameliorative effects of sulforaphane and quercetin on renal tissue in unilateral ureteral obstruction in rats. Acta Clin. Croat. 2012, 51, 555–562. [Google Scholar] [PubMed]
  156. Bertelli, A.A.; Das, D.K. Grapes, wines, resveratrol, and heart health. J. Cardiovasc. Pharmacol. 2009, 54, 468–476. [Google Scholar] [CrossRef] [PubMed]
  157. Liu, B.L.; Zhang, X.; Zhang, W.; Zhen, H.N. New enlightenment of french paradox: Resveratrol’s potential for cancer chemoprevention and anti-cancer therapy. Cancer Biol. Ther. 2007, 6, 1833–1836. [Google Scholar] [CrossRef] [PubMed]
  158. Leonard, S.S.; Xia, C.; Jiang, B.H.; Stinefelt, B.; Klandorf, H.; Harris, G.K.; Shi, X. Resveratrol scavenges reactive oxygen species and effects radical-induced cellular responses. Biochem. Biophys. Res. Commun. 2003, 309, 1017–1026. [Google Scholar] [CrossRef] [PubMed]
  159. Zhao, L.; Lee, J.Y.; Hwang, D.H. Inhibition of pattern recognition receptor-mediated inflammation by bioactive phytochemicals. Nutr. Rev. 2011, 69, 310–320. [Google Scholar] [CrossRef] [PubMed]
  160. Sgambato, A.; Ardito, R.; Faraglia, B.; Boninsegna, A.; Wolf, F.I.; Cittadini, A. Resveratrol, a natural phenolic compound, inhibits cell proliferation and prevents oxidative DNA damage. Mutat. Res. 2001, 496, 171–180. [Google Scholar] [CrossRef] [PubMed]
  161. Aggarwal, B.B.; Bhardwaj, A.; Aggarwal, R.S.; Seeram, N.P.; Shishodia, S.; Takada, Y. Role of resveratrol in prevention and therapy of cancer: Preclinical and clinical studies. Anticancer Res. 2004, 24, 2783–2840. [Google Scholar] [PubMed]
  162. Kitada, M.; Kume, S.; Imaizumi, N.; Koya, D. Resveratrol improves oxidative stress and protects against diabetic nephropathy through normalization of Mn-SOD dysfunction in AMPK/SIRT1-independent pathway. Diabetes 2011, 60, 634–643. [Google Scholar] [CrossRef] [PubMed]
  163. Mokni, M.; Elkahoui, S.; Limam, F.; Amri, M.; Aouani, E. Effect of resveratrol on antioxidant enzyme activities in the brain of healthy rat. Neurochem. Res. 2007, 32, 981–987. [Google Scholar] [CrossRef] [PubMed]
  164. Pervaiz, S.; Holme, A.L. Resveratrol: Its biologic targets and functional activity. Antioxid. Redox Signal. 2009, 11, 2851–2897. [Google Scholar] [CrossRef] [PubMed]
  165. Kim, M.Y.; Lim, J.H.; Youn, H.H.; Hong, Y.A.; Yang, K.S.; Park, H.S.; Chung, S.; Ko, S.H.; Shin, S.J.; Choi, B.S.; et al. Resveratrol prevents renal lipotoxicity and inhibits mesangial cell glucotoxicity in a manner dependent on the AMPK-SIRT1-PGC1alpha axis in db/db mice. Diabetologia 2013, 56, 204–217. [Google Scholar] [CrossRef] [PubMed]
  166. Lagouge, M.; Argmann, C.; Gerhart-Hines, Z.; Meziane, H.; Lerin, C.; Daussin, F.; Messadeq, N.; Milne, J.; Lambert, P.; Elliott, P.; et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 2006, 127, 1109–1122. [Google Scholar] [CrossRef] [PubMed]
  167. Zhang, L.; Pang, S.; Deng, B.; Qian, L.; Chen, J.; Zou, J.; Zheng, J.; Yang, L.; Zhang, C.; Chen, X.; et al. High glucose induces renal mesangial cell proliferation and fibronectin expression through JNK/NF-kappaB/NADPH oxidase/ROS pathway, which is inhibited by resveratrol. Int. J. Biochem. Cell Biol. 2012, 44, 629–638. [Google Scholar] [CrossRef] [PubMed]
  168. Sharma, S.; Anjaneyulu, M.; Kulkarni, S.K.; Chopra, K. Resveratrol, a polyphenolic phytoalexin, attenuates diabetic nephropathy in rats. Pharmacology 2006, 76, 69–75. [Google Scholar] [CrossRef] [PubMed]
  169. Li, J.; Qu, X.; Ricardo, S.D.; Bertram, J.F.; Nikolic-Paterson, D.J. Resveratrol inhibits renal fibrosis in the obstructed kidney: Potential role in deacetylation of Smad3. Am. J. Pathol. 2010, 177, 1065–1071. [Google Scholar] [CrossRef] [PubMed]
  170. Liang, J.; Tian, S.; Han, J.; Xiong, P. Resveratrol as a therapeutic agent for renal fibrosis induced by unilateral ureteral obstruction. Ren. Fail. 2014, 36, 285–291. [Google Scholar] [CrossRef] [PubMed]
  171. Palsamy, P.; Subramanian, S. Resveratrol protects diabetic kidney by attenuating hyperglycemia-mediated oxidative stress and renal inflammatory cytokines via NRF2-KEAP1 signaling. Biochim Biophys. Acta 2011, 1812, 719–731. [Google Scholar]
  172. Huang, K.; Huang, J.; Xie, X.; Wang, S.; Chen, C.; Shen, X.; Liu, P.; Huang, H. Sirt1 resists advanced glycation end products-induced expressions of fibronectin and TGF-beta1 by activating the NRF2/ARE pathway in glomerular mesangial cells. Free Radic. Biol. Med. 2013, 65, 528–540. [Google Scholar] [CrossRef] [PubMed]
  173. Brasnyo, P.; Molnar, G.A.; Mohas, M.; Marko, L.; Laczy, B.; Cseh, J.; Mikolas, E.; Szijarto, I.A.; Merei, A.; Halmai, R.; et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br. J. Nutr. 2011, 106, 383–389. [Google Scholar] [CrossRef] [PubMed]
  174. Castilla, P.; Davalos, A.; Teruel, J.L.; Cerrato, F.; Fernandez-Lucas, M.; Merino, J.L.; Sanchez-Martin, C.C.; Ortuno, J.; Lasuncion, M.A. Comparative effects of dietary supplementation with red grape juice and vitamin E on production of superoxide by circulating neutrophil NADPH oxidase in hemodialysis patients. Am. J. Clin. Nutr. 2008, 87, 1053–1061. [Google Scholar] [PubMed]
  175. Castilla, P.; Echarri, R.; Davalos, A.; Cerrato, F.; Ortega, H.; Teruel, J.L.; Lucas, M.F.; Gomez-Coronado, D.; Ortuno, J.; Lasuncion, M.A. Concentrated red grape juice exerts antioxidant, hypolipidemic, and antiinflammatory effects in both hemodialysis patients and healthy subjects. Am. J. Clin. Nutr. 2006, 84, 252–262. [Google Scholar] [PubMed]
  176. Gupta, S.C.; Patchva, S.; Koh, W.; Aggarwal, B.B. Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin. Exp. Pharmacol. Physiol. 2012, 39, 283–299. [Google Scholar] [CrossRef] [PubMed]
  177. Calabrese, V.; Bates, T.E.; Mancuso, C.; Cornelius, C.; Ventimiglia, B.; Cambria, M.T.; di Renzo, L.; de Lorenzo, A.; Dinkova-Kostova, A.T. Curcumin and the cellular stress response in free radical-related diseases. Mol. Nutr. Food Res. 2008, 52, 1062–1073. [Google Scholar] [CrossRef] [PubMed]
  178. Aggarwal, B.B.; Harikumar, K.B. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int. J. Biochem. Cell Biol. 2009, 41, 40–59. [Google Scholar] [CrossRef] [PubMed]
  179. Mun, S.H.; Joung, D.K.; Kim, Y.S.; Kang, O.H.; Kim, S.B.; Seo, Y.S.; Kim, Y.C.; Lee, D.S.; Shin, D.W.; Kweon, K.T.; et al. Synergistic antibacterial effect of curcumin against methicillin-resistant staphylococcus aureus. Phytomedicine 2013, 20, 714–718. [Google Scholar] [CrossRef] [PubMed]
  180. Bachmeier, B.E.; Killian, P.; Pfeffer, U.; Nerlich, A.G. Novel aspects for the application of curcumin in chemoprevention of various cancers. Front. Biosci. (Schol Ed.) 2010, 2, 697–717. [Google Scholar]
  181. Osawa, T. Nephroprotective and hepatoprotective effects of curcuminoids. Adv. Exp. Med. Biol. 2007, 595, 407–423. [Google Scholar] [PubMed]
  182. Gonzalez-Salazar, A.; Molina-Jijon, E.; Correa, F.; Zarco-Marquez, G.; Calderon-Oliver, M.; Tapia, E.; Zazueta, C.; Pedraza-Chaverri, J. Curcumin protects from cardiac reperfusion damage by attenuation of oxidant stress and mitochondrial dysfunction. Cardiovasc. Toxicol. 2011, 11, 357–364. [Google Scholar]
  183. Reyes-Fermin, L.M.; Gonzalez-Reyes, S.; Tarco-Alvarez, N.G.; Hernandez-Nava, M.; Orozco-Ibarra, M.; Pedraza-Chaverri, J. Neuroprotective effect of alpha-mangostin and curcumin against iodoacetate-induced cell death. Nutr. Neurosci. 2012, 15, 34–41. [Google Scholar]
  184. Soetikno, V.; Suzuki, K.; Veeraveedu, P.T.; Arumugam, S.; Lakshmanan, A.P.; Sone, H.; Watanabe, K. Molecular understanding of curcumin in diabetic nephropathy. Drug Discov. Today 2013, 18, 756–763. [Google Scholar] [CrossRef] [PubMed]
  185. Sharma, S.; Kulkarni, S.K.; Chopra, K. Curcumin, the active principle of turmeric (curcuma longa), ameliorates diabetic nephropathy in rats. Clin. Exp. Pharmacol. Physiol. 2006, 33, 940–945. [Google Scholar] [CrossRef] [PubMed]
  186. Soetikno, V.; Sari, F.R.; Veeraveedu, P.T.; Thandavarayan, R.A.; Harima, M.; Sukumaran, V.; Lakshmanan, A.P.; Suzuki, K.; Kawachi, H.; Watanabe, K. Curcumin ameliorates macrophage infiltration by inhibiting NF-kappaB activation and proinflammatory cytokines in streptozotocin induced-diabetic nephropathy. Nutr. Metab. (Lond) 2011, 8, 35. [Google Scholar] [CrossRef]
  187. Soetikno, V.; Watanabe, K.; Sari, F.R.; Harima, M.; Thandavarayan, R.A.; Veeraveedu, P.T.; Arozal, W.; Sukumaran, V.; Lakshmanan, A.P.; Arumugam, S.; et al. Curcumin attenuates diabetic nephropathy by inhibiting PKC-alpha and PKC-beta1 activity in streptozotocin-induced type I diabetic rats. Mol. Nutr. Food Res. 2011, 55, 1655–1665. [Google Scholar] [CrossRef] [PubMed]
  188. Chiu, J.; Khan, Z.A.; Farhangkhoee, H.; Chakrabarti, S. Curcumin prevents diabetes-associated abnormalities in the kidneys by inhibiting p300 and nuclear factor-kappaB. Nutrition 2009, 25, 964–972. [Google Scholar] [CrossRef] [PubMed]
  189. Huang, J.; Huang, K.; Lan, T.; Xie, X.; Shen, X.; Liu, P.; Huang, H. Curcumin ameliorates diabetic nephropathy by inhibiting the activation of the SphK1-S1P signaling pathway. Mol. Cell Endocrinol. 2013, 365, 231–240. [Google Scholar] [CrossRef] [PubMed]
  190. Ghosh, S.S.; Massey, H.D.; Krieg, R.; Fazelbhoy, Z.A.; Ghosh, S.; Sica, D.A.; Fakhry, I.; Gehr, T.W. Curcumin ameliorates renal failure in 5/6 nephrectomized rats: Role of inflammation. Am. J. Physiol. Renal. Physiol. 2009, 296, F1146–F1157. [Google Scholar] [CrossRef] [PubMed]
  191. Pan, Y.; Zhu, G.; Wang, Y.; Cai, L.; Cai, Y.; Hu, J.; Li, Y.; Yan, Y.; Wang, Z.; Li, X.; et al. Attenuation of high-glucose-induced inflammatory response by a novel curcumin derivative B06 contributes to its protection from diabetic pathogenic changes in rat kidney and heart. J. Nutr. Biochem. 2013, 24, 146–155. [Google Scholar] [CrossRef] [PubMed]
  192. Pan, Y.; Huang, Y.; Wang, Z.; Fang, Q.; Sun, Y.; Tong, C.; Peng, K.; Wang, Y.; Miao, L.; Cai, L.; et al. Inhibition of MAPK-mediated ACE expression by compound C66 prevents STZ-induced diabetic nephropathy. J. Cell Mol. Med. 2014, 18, 231–241. [Google Scholar] [CrossRef] [PubMed]
  193. He, H.J.; Wang, G.Y.; Gao, Y.; Ling, W.H.; Yu, Z.W.; Jin, T.R. Curcumin attenuates NRF2 signaling defect, oxidative stress in muscle and glucose intolerance in high fat diet-fed mice. World J. Diabetes 2012, 3, 94–104. [Google Scholar] [CrossRef] [PubMed]
  194. Soetikno, V.; Sari, F.R.; Lakshmanan, A.P.; Arumugam, S.; Harima, M.; Suzuki, K.; Kawachi, H.; Watanabe, K. Curcumin alleviates oxidative stress, inflammation, and renal fibrosis in remnant kidney through the NRF2-KEAP1 pathway. Mol. Nutr. Food Res. 2013, 57, 1649–1659. [Google Scholar] [CrossRef] [PubMed]
  195. Tapia, E.; Soto, V.; Ortiz-Vega, K.M.; Zarco-Marquez, G.; Molina-Jijon, E.; Cristobal-Garcia, M.; Santamaria, J.; Garcia-Nino, W.R.; Correa, F.; Zazueta, C.; et al. Curcumin induces NRF2 nuclear translocation and prevents glomerular hypertension, hyperfiltration, oxidant stress, and the decrease in antioxidant enzymes in 5/6 nephrectomized rats. Oxid. Med. Cell Longev. 2012, 2012, 269039. [Google Scholar]
  196. Tapia, E.; Zatarain-Barron, Z.L.; Hernandez-Pando, R.; Zarco-Marquez, G.; Molina-Jijon, E.; Cristobal-Garcia, M.; Santamaria, J.; Pedraza-Chaverri, J. Curcumin reverses glomerular hemodynamic alterations and oxidant stress in 5/6 nephrectomized rats. Phytomedicine 2013, 20, 359–366. [Google Scholar] [CrossRef] [PubMed]
  197. Usharani, P.; Mateen, A.A.; Naidu, M.U.; Raju, Y.S.; Chandra, N. Effect of NCB-02, atorvastatin and placebo on endothelial function, oxidative stress and inflammatory markers in patients with type 2 diabetes mellitus: A randomized, parallel-group, placebo-controlled, 8-week study. Drugs R D 2008, 9, 243–250. [Google Scholar] [CrossRef] [PubMed]
  198. Chuengsamarn, S.; Rattanamongkolgul, S.; Luechapudiporn, R.; Phisalaphong, C.; Jirawatnotai, S. Curcumin extract for prevention of type 2 diabetes. Diabetes Care 2012, 35, 2121–2127. [Google Scholar] [CrossRef] [PubMed]
  199. Khajehdehi, P.; Pakfetrat, M.; Javidnia, K.; Azad, F.; Malekmakan, L.; Nasab, M.H.; Dehghanzadeh, G. Oral supplementation of turmeric attenuates proteinuria, transforming growth factor-beta and interleukin-8 levels in patients with overt type 2 diabetic nephropathy: A randomized, double-blind and placebo-controlled study. Scand. J. Urol. Nephrol. 2011, 45, 365–370. [Google Scholar] [CrossRef] [PubMed]
  200. Chang, S.T.; Chen, P.F.; Chang, S.C. Antibacterial activity of leaf essential oils and their constituents from cinnamomum osmophloeum. J. Ethnopharmacol. 2001, 77, 123–127. [Google Scholar] [CrossRef] [PubMed]
  201. Huang, T.C.; Chung, Y.L.; Wu, M.L.; Chuang, S.M. Cinnamaldehyde enhances NRF2 nuclear translocation to upregulate phase II detoxifying enzyme expression in HepG2 cells. J. Agric. Food Chem. 2011, 59, 5164–5171. [Google Scholar] [CrossRef] [PubMed]
  202. Liao, J.C.; Deng, J.S.; Chiu, C.S.; Hou, W.C.; Huang, S.S.; Shie, P.H.; Huang, G.J. Anti-inflammatory activities of cinnamomum cassia constituents in vitro and in vivo. Evid. Based Complement. Alternat. Med. 2012, 2012, 429320. [Google Scholar]
  203. Wondrak, G.T.; Cabello, C.M.; Villeneuve, N.F.; Zhang, S.; Ley, S.; Li, Y.; Sun, Z.; Zhang, D.D. Cinnamoyl-based NRF2-activators targeting human skin cell photo-oxidative stress. Free Radic. Biol. Med. 2008, 45, 385–395. [Google Scholar] [CrossRef] [PubMed]
  204. Chao, L.K.; Chang, W.T.; Shih, Y.W.; Huang, J.S. Cinnamaldehyde impairs high glucose-induced hypertrophy in renal interstitial fibroblasts. Toxicol. Appl. Pharmacol. 2010, 244, 174–180. [Google Scholar] [CrossRef] [PubMed]
  205. Mishra, A.; Bhatti, R.; Singh, A.; Singh Ishar, M.P. Ameliorative effect of the cinnamon oil from cinnamomum zeylanicum upon early stage diabetic nephropathy. Planta Med. 2010, 76, 412–417. [Google Scholar] [CrossRef] [PubMed]
  206. Dinkova-Kostova, A.T.; Liby, K.T.; Stephenson, K.K.; Holtzclaw, W.D.; Gao, X.; Suh, N.; Williams, C.; Risingsong, R.; Honda, T.; Gribble, G.W.; et al. Extremely potent triterpenoid inducers of the phase 2 response: Correlations of protection against oxidant and inflammatory stress. Proc. Natl. Acad. Sci. USA 2005, 102, 4584–4589. [Google Scholar]
  207. Ruiz, S.; Pergola, P.E.; Zager, R.A.; Vaziri, N.D. Targeting the transcription factor NRF2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney Int. 2013, 83, 1029–1041. [Google Scholar] [CrossRef] [PubMed]
  208. Sporn, M.B.; Liby, K.T.; Yore, M.M.; Fu, L.; Lopchuk, J.M.; Gribble, G.W. New synthetic triterpenoids: Potent agents for prevention and treatment of tissue injury caused by inflammatory and oxidative stress. J. Nat. Prod. 2011, 74, 537–545. [Google Scholar] [CrossRef] [PubMed]
  209. Cleasby, A.; Yon, J.; Day, P.J.; Richardson, C.; Tickle, I.J.; Williams, P.A.; Callahan, J.F.; Carr, R.; Concha, N.; Kerns, J.K.; et al. Structure of the BTB domain of KEAP1 and its interaction with the triterpenoid antagonist CDDO. PLoS One 2014, 9, e98896. [Google Scholar] [CrossRef] [PubMed]
  210. Wu, Q.Q.; Wang, Y.; Senitko, M.; Meyer, C.; Wigley, W.C.; Ferguson, D.A.; Grossman, E.; Chen, J.; Zhou, X.J.; Hartono, J.; et al. Bardoxolone methyl (BARD) ameliorates ischemic AKI and increases expression of protective genes NRF2, PPARgamma, and HO-1. Am. J. Physiol. Renal. Physiol. 2011, 300, F1180–F1192. [Google Scholar] [CrossRef] [PubMed]
  211. Tanaka, Y.; Aleksunes, L.M.; Goedken, M.J.; Chen, C.; Reisman, S.A.; Manautou, J.E.; Klaassen, C.D. Coordinated induction of NRF2 target genes protects against iron nitrilotriacetate (FeNTA)-induced nephrotoxicity. Toxicol. Appl. Pharmacol. 2008, 231, 364–373. [Google Scholar] [CrossRef] [PubMed]
  212. Tan, S.M.; Sharma, A.; Stefanovic, N.; Yuen, D.Y.; Karagiannis, T.C.; Meyer, C.; Ward, K.W.; Cooper, M.E.; de Haan, J.B. A derivative of bardoxolone methyl, dh404, in an inverse dose-dependent manner, lessens diabetes-associated atherosclerosis and improves diabetic kidney disease. Diabetes 2014. [Google Scholar] [CrossRef]
  213. Aminzadeh, M.A.; Reisman, S.A.; Vaziri, N.D.; Khazaeli, M.; Yuan, J.; Meyer, C.J. The synthetic triterpenoid RTA dh404 (CDDO-dhTFEA) restores NRF2 activity and attenuates oxidative stress, inflammation, and fibrosis in rats with chronic kidney disease. Xenobiotica 2014, 44, 570–578. [Google Scholar] [CrossRef] [PubMed]
  214. Hong, D.S.; Kurzrock, R.; Supko, J.G.; He, X.; Naing, A.; Wheler, J.; Lawrence, D.; Eder, J.P.; Meyer, C.J.; Ferguson, D.A.; et al. A phase I first-in-human trial of bardoxolone methyl in patients with advanced solid tumors and lymphomas. Clin. Cancer Res. 2012, 18, 3396–3406. [Google Scholar] [CrossRef] [PubMed]
  215. Pergola, P.E.; Krauth, M.; Huff, J.W.; Ferguson, D.A.; Ruiz, S.; Meyer, C.J.; Warnock, D.G. Effect of bardoxolone methyl on kidney function in patients with T2D and stage 3b-4 CKD. Am. J. Nephrol. 2011, 33, 469–476. [Google Scholar] [CrossRef] [PubMed]
  216. Pergola, P.E.; Raskin, P.; Toto, R.D.; Meyer, C.J.; Huff, J.W.; Grossman, E.B.; Krauth, M.; Ruiz, S.; Audhya, P.; Christ-Schmidt, H.; et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N. Engl. J. Med. 2011, 365, 327–336. [Google Scholar] [CrossRef] [PubMed]
  217. De Zeeuw, D.; Akizawa, T.; Audhya, P.; Bakris, G.L.; Chin, M.; Christ-Schmidt, H.; Goldsberry, A.; Houser, M.; Krauth, M.; Lambers Heerspink, H.J.; et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N. Engl. J. Med. 2013, 369, 2492–2503. [Google Scholar]

Share and Cite

MDPI and ACS Style

Choi, B.-h.; Kang, K.-S.; Kwak, M.-K. Effect of Redox Modulating NRF2 Activators on Chronic Kidney Disease. Molecules 2014, 19, 12727-12759. https://doi.org/10.3390/molecules190812727

AMA Style

Choi B-h, Kang K-S, Kwak M-K. Effect of Redox Modulating NRF2 Activators on Chronic Kidney Disease. Molecules. 2014; 19(8):12727-12759. https://doi.org/10.3390/molecules190812727

Chicago/Turabian Style

Choi, Bo-hyun, Kyung-Shin Kang, and Mi-Kyoung Kwak. 2014. "Effect of Redox Modulating NRF2 Activators on Chronic Kidney Disease" Molecules 19, no. 8: 12727-12759. https://doi.org/10.3390/molecules190812727

Article Metrics

Back to TopTop