Next Article in Journal
A Case of AML Characterized by a Novel t(4;15)(q31;q22) Translocation That Confers a Growth-Stimulatory Response to Retinoid-Based Therapy
Next Article in Special Issue
Correlation between Oxidative Stress, Nutrition, and Cancer Initiation
Previous Article in Journal
Inflammatory and Noninflammatory Itch: Implications in Pathophysiology-Directed Treatments
Previous Article in Special Issue
A Novel Therapeutic Approach in the Treatment of Pulmonary Arterial Hypertension: Allium ursinum Liophylisate Alleviates Symptoms Comparably to Sildenafil
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Naturally Occurring Compounds: New Potential Weapons against Oxidative Stress in Chronic Kidney Disease

Renal Unit, Department of Medicine, University-Hospital of Verona, Verona 37126, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2017, 18(7), 1481; https://doi.org/10.3390/ijms18071481
Submission received: 31 May 2017 / Revised: 22 June 2017 / Accepted: 8 July 2017 / Published: 10 July 2017
(This article belongs to the Special Issue Correlation between Nutrition, Oxidative Stress and Disease)

Abstract

:
Oxidative stress is a well-described imbalance between the production of reactive oxygen species (ROS) and the antioxidant defense system of cells and tissues. The overproduction of free radicals damages all components of the cell (proteins, lipids, nucleic acids) and modifies their physiological functions. As widely described, this condition is a biochemical hallmark of chronic kidney disease (CKD) and may dramatically influence the progression of renal impairment and the onset/development of major systemic comorbidities including cardiovascular diseases. This state is exacerbated by exposure of the body to uremic toxins and dialysis, a treatment that, although necessary to ensure patients’ survival, exposes cells to non-physiological contact with extracorporeal circuits and membranes with consequent mitochondrial and anti-redox cellular system alterations. Therefore, it is undeniable that counteracting oxidative stress machinery is a major pharmacological target in medicine/nephrology. As a consequence, in recent years several new naturally occurring compounds, administered alone or integrated with classical therapies and an appropriate lifestyle, have been proposed as therapeutic tools for CKD patients. In this paper, we reviewed the recent literature regarding the “pioneering” in vivo testing of these agents and their inclusion in small clinical trials performed in patients affected by CKD.

Graphical Abstract

1. Introduction

Oxidative stress, a biochemical imbalance between reactive oxygen species (ROS) production and antioxidant defenses, has been reported as an important biochemical hallmark of several human diseases.
In chronic kidney disease (CKD), this deregulated biochemical machinery has been associated with disease progression and with the onset/development of severe systemic complications (mainly atherosclerosis and other cardiovascular diseases), with a consequent remarkable impact on major clinical outcomes [1,2,3,4,5,6].
Although present from the early stages of renal impairment, it appears severely enhanced with advancing stages of CKD and the start of renal replacement therapy (hemodialysis or peritoneal dialysis) [3].
Several biological factors are responsible for oxidative stress in this large population of patients, but, as recently described, mitochondrial deregulation seems to have a primary role [7,8,9].
Mitochondria are organelles with a major role in generating energy for cellular metabolism by the oxidative phosphorylation system (OXPHOS), and they are involved in several physiological cellular functions (e.g., ion homeostasis, heme and steroid synthesis, calcium signaling, apoptosis [10,11,12,13]). During CKD mitochondria may undergo a profound deregulation that can induce functional alterations. CKD is associated with a decline in mitochondrial content at the early stage of the disease [14].
In particular, during renal damage, continuous mitochondrial insults may activate a vicious functional circle responsible for an overproduction of ROS which can induce mitochondrial DNA mutations, damage the mitochondrial respiratory chain, alter membrane permeability, and influence Ca2+ homeostasis and mitochondrial defense systems. Mitochondrial dysfunction and renal failure are strictly connected: it has been demonstrated that the role of mitochondria in podocyte injury leads to proteinuria [15,16] as well as epithelial to mesenchymal transition of tubular epithelial cells [17,18]. Moreover, uremic toxins impair OXPHOS in epithelial tubular cells [19].
Other biological factors contributing to oxidative stress in CKD are the enzymatic complex NADPH oxidase and the xanthine oxidase pathway. NADPH oxidase catalyzes the transfer of electrons to oxygen to produce superoxide anion (O2) that is immediately converted to H2O2. These, in turn, are precursors of other ROS that cause damages to proteins, lipids, and nucleic acids [20]. NADPH oxidase-derived ROS are involved in cell signaling, ion channel activity, gene expression, and in the direct killing of invading microbes in phagocytes. Its activity is upregulated in all stages of CKD and in dialysis [21,22,23].
Xanthine oxidase catalyzes the oxidation of hypoxanthine to xanthine and xanthine to uric acid together with ROS release. Xanthine oxidase activity is higher in CKD patients [24] and could be an independent predictor of cardiovascular events in CKD and hemodialysis patients [25]. In this context, allopurinol, the xanthine oxidase inhibitor acting as a competitive substrate for the enzyme, decreases serum uric acid levels and its toxic effects [26]. Several studies have shown that allopurinol treatment decreases C-reactive protein (CRP) levels, slows the progression of renal disease, decreases the number of hospitalizations, and reduces cardiovascular risk [27,28,29,30]. However, since it is excreted by urine, it requires individual dosage modification in CKD patients, and consequently may have poor control over the serum concentration of uric acid [31,32]. In addition, systematic reviews have highlighted that allopurinol could have only a partial therapeutic efficacy, and may also induce adverse effects in CKD patients [33,34].
Febuxostat, an orally administered nonpurine selective inhibitor of xanthine oxidase with two excretion pathways (urinary and fecal), was more effective in the reduction and maintenance of serum urate levels <6.0 mg/dL compared to allopurinol in patients with renal impairment [35,36,37].
At the same time, antioxidant systems are defective in CKD [38,39,40,41], but we recently described that nuclear factor erythroid 2-related factor 2 (NRF-2) and one of its target genes, superoxide dismutase (SOD) 2, are upregulated in dialysis-treated patients, defining a possible antioxidant system able to contrast ROS production.
Therefore, a worldwide interest is emerging in identifying and testing compounds able to significantly counteract oxidative damage in CKD. In particular, natural compounds targeting mitochondria, alone or combined with conventional therapies and lifestyle modifications, could represent valuable tools to prevent this condition and, because of the low adverse effects, they could be employed in patients undergoing both conservative and dialysis treatment [42].
These compounds are frequently included in the diet at biologically active concentrations and represent essential elements of traditional medicine from several countries. Although effective and relatively safety, they have been only partially investigated in nephrology and their efficacy in CKD is still debated.
To this purpose, a large number of studies have begun to address this objective by employing animal models and small clinical trials.

2. l-Carnitine

l-Carnitine (4-N-trimethylammonium-3-hydroxybutyric acid) is an amino acid-derived compound obtained mostly through the diet, particularly from animal-derived foods. It is endogenously synthesized in the kidney and liver from lysine and methionine, with a daily rate of 1.2 µmol/kg/day [43].
l-Carnitine acts as a transporter of long-chain fatty acids chains across the mitochondrial inner membrane, through a series of reversible transesterification reactions catalyzed by the group of carnitine acyltransferases. In these reactions, coenzyme A (CoA), bound to fatty acids, is substituted by the hydroxyl group of carnitine, forming acyl-carnitine. This molecule is transported from the cytosol to the inner mitochondrial matrix thanks to the combined activity of carnitine-acyl-carnitine translocase and carnitine palmitoyltransferase I and II. Here, acyl-carnitine is converted again into acyl-CoA, which can enter the β-oxidation and finally generate energy in the electron transport chain [44,45].
Carnitine is thus fundamental for mitochondrial and cellular physiological activity. It has been demonstrated that its deficiency leads to (1) the accumulation of free fatty acids into cells; (2) impaired export of the excess of organic acids, particularly in some of the secondary carnitine insufficiencies, including hemodialysis; and (3) increased mitochondrial dysfunction and ROS production, secondary to acetyl-CoA accumulation and multiple enzymatic inhibition [44,46].
Furthermore, carnitine acts as both a direct and indirect antioxidant by scavenging ROS, chelating iron [47], increasing the expression and activity of antioxidant enzymes [48], and by inhibiting lipid peroxidation and xanthine oxidase activity [49,50].
CKD is characterized by a reduced synthesis of l-carnitine, proportional to the decline of the glomerular filtration rate (GFR). Nevertheless, its level is higher in CKD patients under conservative therapy than in healthy individuals, probably through a compensatory effect of the liver [51].
On the other hand, CKD patients undergoing hemodialysis (HD) and peritoneal dialysis (PD) show a reduction of free l-carnitine levels in plasma and muscle. This is due to several factors, including loss during dialysis treatment, intestinal malabsorption, and the impaired synthetic capacity of the kidney. In particular, Di Liberato et al. suggested a potential role of PD modality (CAPD) on carnitine depletion and the possible advantage of carnitine-containing dialysis fluids [52].
Due to its antioxidant role, the oral administration of l-carnitine to HD patients resulted in an increased glutathione (GSH) level, increased glutathione peroxidase activity, and a decreased malondyaldeide (MDA) level [53,54]. Interestingly, several reports have suggested that its supplementation might have a positive effect on the response to erythropoietin (EPO) in long-term hemodialysis patients [55,56,57,58], but it did not modify EPO requirements in patients new to hemodialysis [59].
During the past few years, some federal agencies have suggested the use of l-carnitine in HD. In 2003, the National Kidney Foundation developed a practice recommendation for the use of l-carnitine in patients with dialysis-related carnitine disorders, most notably erythropoietin-resistant anemia, intradialytic hypotension, cardiomyopathy, and fatigability [60]. However, KDOQI guidelines state that there is not sufficient evidence to recommend the administration of L-carnitine to HD patients with anemia [61].
Two important meta-analyses of the l-carnitine administration in HD patients were recently published, with controversial results. Chen et al. concluded that l-carnitine administration decreases serum low density lipoprotein (LDL) and CRP, while it fails to ameliorate EPO responsiveness and anemia [62]. In contrast Yang et al. did not find any conclusive evidence of effectiveness against inflammation, oxidative stress, anemia, nutrition, dyslipidemia, hyperparathyroidism, or quality of life in HD patients undergoing l-carnitine administration. These contrasting results suggest that additional long-term controlled randomized clinical trials are necessary to definitively understand the clinical utility of carnitine administration in CKD patients [63].

3. Vitamin E

Vitamin E encompasses a group of eight chemically related molecules, comprising α, β, γ, δ tocopherol, and the derived tocotrienols [64]. The most biologically active form is α-tocopherol [65]. The main sources of vitamin E are seed oils (wheat germ oil: 150 mg/mL, almond oil: 95 mg/mL, olive oil: 15 mg/mL).
α-Tocopherol maintains the integrity of long-chain polyunsaturated fatty acids in the membranes of cells and thus preserves their bioactivity [66,67]. In particular, it protects lipid structures from peroxidation and increases LDL resistance to oxidative modification [68,69], suggesting a potential effect of vitamin E in the prevention of atherosclerosis [70,71].
Vitamin E is a powerful peroxyl radical scavenger that returns to its reduced state by reacting with vitamin C (or other hydrogen donors) [72].
Moreover, it reduces the mitochondrial generation of hydrogen peroxide and it regulates the expression of genes implicated in inflammation and fibrosis [73,74,75,76].
Although vitamin E therapy has been extensively studied in CKD patients, there is no consensus about the benefit obtained from its administration.
The SPACE study evaluated the cardiovascular protective effects of orally administered, high doses of vitamin E (800 IU/die) on HD patients with previous cardiovascular events, over a period of 519 days. The treatment resulted in a 40% decline in both composite cardiovascular events and myocardial infarction [77].
On the other hand, the administration of vitamin E (400 IU/day) to patients with mild to moderate renal failure failed to obtain beneficial effects on cardiovascular outcomes in the Heart Outcomes Prevention Evaluation (HOPE) study [78].
These contrasting results may be partially due to differences in the enrollment criteria (e.g., the SPACE trial included patients with higher cardiovascular risk, and participants were treated with a higher dose of vitamin E); moreover, most participants in the SPACE trial (43.3% of the vitamin E group) also consumed vitamin C. Finally, the SPACE study enrolled a smaller sample (196 HD patients) compared with other trials, which resulted in large confidence intervals based on a broad composite end-point.
More recent studies have shown the positive effect of vitamin E administration before coronary procedures in the prevention of contrast-induced acute kidney injury in patients with CKD undergoing elective coronary procedures [79,80].
Additionally, long-term use of vitamin E-coated HD filters improved oxidative stress, inflammatory markers, and hemoglobin levels, and reduced Erythropoiesis-Stimulating Agents (ESA) requirement without affecting dialysis adequacy [81,82,83,84,85,86,87].
Vitamin E combined with Pravastatin and homocysteine-lowering therapy was evaluated against vascular stiffness progression over 18 months in patients with mild to moderate CKD. This treatment resulted in significant improvement of vascular compliance and distensibility, decrement in common carotid intima-media thickness, and increase in brachial artery flow-mediated dilatation; however, the effect of other confounding variables was unclear [88,89].
It is noteworthy that vitamin E could have also a pro-oxidant action under special conditions that can be encountered in HD patients [90,91]. In fact, oral α-tocopherol administration (500 mg/day) for 1 year to HD patients caused reduced SOD activity and total antioxidant status [91]. This could be due to the low level of other antioxidants necessary to restore the reduced form of vitamin E (e.g., vitamin C) [92].
Based on research studies, vitamin E supplementation could be a potential valuable adjuvant therapy to contrast oxidative stress and lower lipid peroxidation in CKD and HD patients. Still, because of its adverse effects [93], its clinical employment in nephrology needs to be better defined.

4. Vitamin C

Vitamin C (ascorbic acid) is a water-soluble essential antioxidant obtained from citrus fruits and some green vegetables such as broccoli and spinach [94]. Its recommended daily dose is approximately 90 mg in adult men and 75 mg in adult women [95].
Vitamin C prevents oxidative damage by scavenging ROS and reactive nitrogen species [96]. Moreover, it shows an anti-apoptotic activity by maintaining the mitochondrial membrane potential and protecting mitochondrial DNA from oxidant insults [97,98,99,100].
It has been demonstrated that a reduction in both the total vitamin C concentration and the active form (ascorbate) is probably caused by the limited intake of potassium-rich foods in CKD patients in conservative treatment, and by the loss during HD treatment. Another possible explanation is an impairment of enzymatic or non-enzymatic recycling of ascorbate from dehydroascorbate (the oxidized form of vitamin C), since the recycling is largely GSH-dependent [101] and dialysis patients have a marked GSH deficiency [102,103]. The low vitamin C plasma levels in CKD patients has been, then, associated with an increased risk for fatal and nonfatal major adverse cardiovascular events [104,105].
To avoid this condition, currently oral ascorbate (1–1.5 g/week) or parenteral ascorbate (300 mg/dialysis session) are suggested to balance subclinical deficiency [103].
Poly-vitamin supplementation, including vitamin C (250 mg/day), for eight weeks to HD patients did not induce any change in inflammation, malnutrition, or oxidative stress markers [106].
Moreover, in a double-blind randomized clinical trial on CKD patients (stage IV–V and HD), vitamin C (250 mg, three times a week) failed to demonstrate a real clinical benefit when evaluated against uremic symptoms and cardiovascular stability [107].
Conversely, several studies showed advantages of vitamin C administration. In a cross-over study, patients treated for three months with vitamin C (200 mg/day) showed decreased CRP levels and increased serum prealbumin concentration as marker of malnutrition [108]. Interestingly, the use of vitamin E-coated dialysis membranes significantly reduced oxidative stress, avoided a reduction of erythrocyte reductases activity, and decreased the level of proinflammatory cytokines [81]. Moreover, other studies reported that this vitamin improved the response to ESA with a significant enhancement of hemoglobin levels and transferrin saturation [109,110]. At the moment, despite encouraging results from clinical research studies, the use of vitamin C is still debated in nephrology.

5. Coenzyme Q10

Coenzyme Q10 (CoQ10) is a high lipophilic molecule diffuse in eukaryotic cells that is mostly concentrated in mitochondria, as component of the mitochondrial respiratory chain where it transports electrons from complex I/II to complex III [111].
It is synthesized in the human body and is assumed with food (animal muscle and liver, blue fish, soy beans, and olive oil) with an estimated daily intake of 3–6 mg [112].
Due to its abundant distribution and intra-membranous localization, CoQ10 prevents membrane lipid peroxidation since hydroxyl and superoxide radicals generated in the membrane during electron transport chain would otherwise rapidly react with neighboring lipid and protein molecules [113,114].
CoQ10 regenerates vitamin E from the α-tocopheroxyl radical and prevents the oxidation of nucleic acids, particularly of mitochondrial DNA [113]. Additionally, it inhibits mitochondria-dependent apoptosis by preventing permeability transition pore opening and mitochondrial membrane potential depolarization [111,115]. Moreover, increased mitochondrial CoQ10 content results in a general improvement of bioenergetic parameters, such as oxygen consumption, ATP content, mitochondrial potential, and protein synthesis [116].
As a consequence, CoQ10 has been studied in clinical settings in order to prevent atherosclerosis, aging, and the progression of chronic diseases such as CKD.
In CKD patients, it an inverse relationship between CoQ10 and renal function has been reported [117,118].
An interesting correlation was also demonstrated between reduced CoQ10 levels and epicardial fat tissue (EFT) in HD patients [119]. EFT is the visceral adipose tissue surrounding the subepicardial coronary vessels. It is an active source of proinflammatory factors [120,121,122], and, in greater amounts, has recently been related to a higher risk for atherosclerosis and heart disease in PD and HD patients [123,124]. Thus, increased EFT in HD patients could cause the consumption of CoQ10 as an antioxidant molecule, and its supplementation was hypothesized able to correct this imbalance, reducing cardiovascular risk.
In a recent trial involving patients in maintenance HD, oral CoQ10 (200 mg/day) supplementation did not induce changes in exercise performance and in the blood level of oxidative system markers compared with a placebo [125].
As with other similar compounds, at present, there is no a clear evidence that justifies the “day by day” clinical employment of CoQ10 in CKD patients in both conservative and dialysis treatment.

6. α-Lipoic Acid

α-Lipoic acid (ALA) is a disulfide compound that acts as a coenzyme in pyruvate dehydrogenase and α-ketoglutarate dehydrogenase mitochondrial reactions [126]. Typical dietary sources of ALA are muscle meats, heart, kidney, and liver, and to a lesser degree, fruits and vegetables [127].
ALA and its reduced form, dihydrolipoic acid, have several antioxidant, anti-inflammatory, and metabolic activities.
ALA exerts direct antioxidant activity by chelating Cu2+, Zn2+, and Fe2+ [128,129], and it recycles other cellular antioxidants including CoQ10, vitamins C and E, and GSH. It also regulates the expression of numerous genes trough the inhibition of NFkB [130] and the induction of NRF2 [131].
As a modulator of peroxisome proliferator-activated receptor (PPAR)-α and -γ expression, ALA is implied in the regulation of glucose and lipid metabolism [131].
Moreover, it inhibits vascular calcification and vascular smooth muscle cell apoptosis by preserving mitochondrial functions and activating the PI3K/Akt pathway [132].
In a model of diabetic nephropathy, it reduced the progression of renal damage, glomerular mesangial matrix expansion, and glomerulosclerosis by restoring GSH, reducing MDA levels, and protecting mitochondrial function [133,134].
However, ALA supplementation in hemodialyzed patients showed uncertain results. A daily dose of 600 mg for eight weeks caused a reduction of CRP levels, without showing effects on MDA, total antioxidant status, total cholesterol, triglyceride, high-density lipoprotein cholesterol, or LDL levels [135]. Contrarily, in other studies, ALA combined with mixed tocopherols did not modify the level of several biomarkers of inflammation and oxidative stress [136,137,138,139].
Further studies, probably using mixed antioxidants and longer courses of treatment, could be useful to demonstrate the clinical utility of ALA supplementation in this patient population.

7. Selenium

Selenium is a non-metal element essential for human physiology, since it acts as a cofactor for several enzymes with an antioxidant role (glutathione peroxidase, thioredoxin reductase) [140]. It is obtained from meat, seafood, grains, cereals, fish (tuna and mackerel), and plants (garlic, onions, and broccoli), but the best source of selenium is Brazil nuts [141].
Several reports have demonstrated a selenium deficiency in CKD and dialyzed patients [142,143,144,145].
In patients with CKD, selenium supplementation increased the glutathione peroxidase activity [146], but unfortunately, this positive effect was detectable only if this agent was administered in the early stages of renal impairment [147].
Selenium supplementation (200 µg) for 12 weeks prevents the damage of DNA [148] and improved MDA levels [149], without showing any significant effects on inflammatory and thyroid functional biomarkers [150].
Moreover, 600 μg sodium selenide co-administered with 400 IU vitamin E before an HD session offsets the serum MDA increment induced by iron infusion [151].
At the moment, there is no indication to the use of selenium in CKD and additional, larger clinical studies are necessary to define its future potential employment in this clinical setting.

8. Green Tea

Green tea is composed of several polyphenolic compounds, but (−)-Epigallocatechin-3-gallate (EGCG) is the most abundant and effective as an antioxidant [152]. In fact, it is implicated in ROS scavenging, the inhibition of lipid peroxidation, and the chelation of metal ions such as copper (II) and iron (III) [153,154,155,156].
Recently these protective properties have become of interest specifically in kidney diseases, and EGCG has been tested in several animal models of glomerulonephritis [157]. Treatment with EGCG reduced proteinuria and serum creatinine, and it determined a marked improvement in several renal histological features. These effects seemed to be mediated by the direct and indirect effects of these agents on the redox cellular/biological system and the immune-inflammatory pathway [158,159,160]. In particular, EGCG increased renal NRF2 and glutathione peroxidase activity; reduced renal oxidative stress, NF-κB activation, and NLRP3 mRNA/protein expression as well as protein levels of mature caspase-1, IL-1β, and IL-18; and enhanced splenic regulatory T cell activity. EGCG-treated mice also showed a reduction in p-Akt, p-JNK, p-ERK1/2, and p-P38 as well as the restoration of peroxisome proliferator-activated receptor (PPAR)γ and sirtuin-1 (SIRT1) levels [158,159,160].
In streptozotocin-induced diabetic nephropathy, EGCG administration for 50 days improved renal function as well as reduced renal AGE accumulation, lipid peroxidation, and fibronectin levels [161].
In animal models of unilateral urethral obstruction, EGCG caused the upregulation and nuclear translocation of NRF2, with the consequent enhancement of antioxidant enzymes such as γ-glutamylcysteine synthetase [162].
EGCG significantly reduced uremic toxins, such as methylguanidine levels, in a dose-dependent manner in rats with adenine-induced renal failure [163].
In an ischemia reperfusion injury model, EGCG treatment, via monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF-β) downregulation and heme oxygenase 1 (HO-1) augmentation, protected the kidneys from a massive infiltration of macrophages [164] and the development of chronic renal alterations.
It must be noted, however, that all of the abovementioned studies are speculative and no large clinical trials have been published in nephrology.

9. Resveratrol

Resveratrol (3,5,4′-trihydroxystilbene) is a naturally occurring polyphenolic compound present in more than 70 species of plants, with the greatest amount found in grapes, berries, red wine, and peanuts [165].
Resveratrol can directly scavenge ROS and modulate the expression and activity of antioxidant enzymes such as SOD, glutathione peroxidase, and catalase, through transcriptional regulation via NRF-2, activator protein 1 (AP-1), forkhead box O (FOXO), and SP-1 [166,167]. Moreover, it is an activator of SIRT1, an NAD+-dependent deacetylase of histones, which results in reduced transcriptional activity. SIRT1 plays a key role in responding to nutritional and environmental perturbations such as fasting, calorie restriction, starvation, and nutrient deprivation, as well as in oxidative stress conditions [168,169]. SIRT1 activates peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α) and attenuates sterol-regulatory-element-binding protein (SREBP) activity, resulting in increased fatty acid oxidation, reduced lipogenesis and cholesterolgenesis, and improved glucose homeostasis and mitochondrial function [170,171]. Moreover, it deacetylates FOXO, p53, hypoxia-inducible factor (HIF)-1α, and NFkB, mediating antiapoptotic, antioxidative, and anti-inflammatory effects [172,173].
Due to its cytoprotective effects, resveratrol has been largely tested in animal models of chronic renal diseases. In streptozotocin-induced diabetic rats, resveratrol (5 or 10 mg/kg administered orally) improved renal dysfunction and oxidative stress, and attenuated cytokine levels [174,175].
Resveratrol, then, seemed to prevent in vitro high glucose-induced mesangial cell proliferation and fibronectin expression by inhibiting JNK and NF-κB, as well as NADPH oxidase activity and ROS production [176]. These results were in line with more recent studies that demonstrated a positive effect of this agent in podocytes of db/db mice through the activation of the autophagic pathway [177].
Resveratrol, through its antioxidant mechanisms and the deacetylation of Smad3, directly prevents EMT and renal fibrosis [176,178,179,180,181], and inhibits CKD-induced skeletal muscle atrophy mediated by NF-κB [182].
Moreover, by enhancing the AMPK/SIRT-1/PGC-1α axis [183] this agent could attenuate mitochondrial dysfunction and aldosterone-induced podocyte injury [184].
In other animal models of AKI (cisplatin nephropathy, ischemia-reperfusion, and sepsis-related acute renal damages), resveratrol demonstrated a considerable renal protective effect by normalizing Nrf2 renal expression, enhancing antioxidant factors expression (HO-1, GST), and reducing inflammatory mediators (TNF-α, IL-6) [185,186,187].
Unfortunately, although resveratrol has demonstrated low adverse effects, no study has been developed to evaluate its efficacy in CKD patients, probably because its low bioavailability [188]. In the future, the employment of this agent in combination with other compounds could probably permit its use to counteract the progression to end-stage renal disease.

10. Curcumin

Curcumin is the active element of curcuma longa (or turmeric), a perennial herbaceous plant member of the ginger family (Zingiberaceae), prevalent in India, China, and Southeast Asia [189].
In Ayurvedic and Chinese medicine, curcumin is used as anti-inflammatory, antioxidant, antibacterial, and antimicrobial reagent, as well as to treat chronic diseases [190,191,192,193,194].
Its antioxidant properties are due to both a direct scavenger activity and the upregulation of antioxidant and cytoprotective genes. Curcumin is able to directly scavenge superoxide anions, hydroxyl radicals, H2O2, singlet oxygen, nitric oxide, and peroxynitrite [195,196,197,198], probably by means of phenolic groups in its molecular structure. Curcumin also has an indirect antioxidant ability mediated by the induction of the expression of cytoprotective enzymes such as SOD, catalase [199], glutathione reductase, HO-1 [200], glutathione S-transferase, NAD(P)H:quinone oxidoreductase 1 [201], and γ-glutamylcysteine ligase [202].
Several studies have reported the reno-protective effects of curcumin in a mouse model of diabetic nephropathy. Administration of curcumin (15–150 mg/kg/day for two to eight weeks) ameliorated renal function through the upregulation of antioxidant enzymes, inhibition of NADPH oxidase and NFkB, and it reduced macrophages infiltration and proinflammatory cytokines together with antifibrotic activities [203,204,205,206,207].
Likewise, in an animal model of CKD, curcumin significantly reversed proteinuria, hypertension, interstitial fibrosis, fibrotic glomeruli, tubular atrophy, and mesangial expansion [208,209,210,211,212,213]. Interestingly, the preservation of mitochondrial dynamics, bioenergetics, and oxidative stress has been recently demonstrated in a rat model of CKD, which may be associated with ameliorated renal function [214].
Moreover, curcumin improved structural and functional manifestations of cardiac injury associated with renal failure, in part through the inhibition of NLRP3 inflammasome and the preservation of mitochondrial function [215,216,217].
Khajehdehi et al. showed that an oral supplementation of 500 mg turmeric (of which 22.1 mg was the active ingredient curcumin) for three months has strong renal protective effects in lupus nephritis and type 2 diabetic nephropathies [218,219].
However, although it is a promising agent for the treatment of chronic glomerulopathies and tubular renal dysfunctions, at the moment, the clinical evidences are not adequate to justify a large utilization of this compound in our nephrology patients.

11. Omega-3 Polyunsaturated Fatty Acids

Omega-3 polyunsaturated fatty acids (omega-3 PUFAs) are a class of essential long-chain fatty acids obtained primarily from dietary sources.
The two most bioactive and extensively studied omega-3 fatty acids are eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) [220].
Omega-3 PUFAs exert anti-inflammatory activity by modifying the expression of adhesion molecules, chemotactic factors, and proinflammatory cytokines (TNF-α, IL-1β, and IL-6) [221,222,223,224].
Another mechanism is the prevention of the conversion of arachidonic acid into proinflammatory eicosanoids such as prostaglandin (PG) and leukotriene (LT) [225], and serving as an alternative substrate to produce less potent 5-series LTs, 3-series PGs, and thromboxanes [226]. In addition, omega-3 PUFA-derived resolvins such as resolvin E1 (RvE1) and D-series resolvins and protectin D1 from DHA have potent anti-inflammatory actions [227,228].
These compounds maintain the structure and function of cell and organelle membranes [229], participating in membrane fluidity, ion channels transport (sodium, potassium, and calcium) [230], and mitochondrial biogenesis [231].
In addition to anti-inflammatory properties, omega-3 fatty acids also have antioxidant effects.
They enhance endogenous antioxidant defense systems such as GSH through the increased activity of γ-glutamyl-cysteinyl ligase, glutathione reductase, and glutathione S-transferase [232], and compete with arachidonic acid at COX2 and xanthine oxidase sites, reducing ROS synthesis [233].
In animal models of CKD, EPA and DHA supplementation reduce inflammation, fibrosis, and oxidative stress [234,235,236].
In CKD and dialysis patients, the administration of omega-3 PUFAs can reduce inflammation associated with CKD progression through the upregulation of E- and D-series resolvins [237], together with the reduced level of endothelial chemokines, RANTES, and MCP-1 [238].
At the same time, recent studies suggest a benefit of omega-3 PUFAs supplementation in ameliorating uremic symptoms (particularly pruritus) [239] and reducing hypertension [240,241].
In a randomized controlled clinical trial on 78 patients affected by IgA nephropathy, PUFAs associated with renin-angiotensin system blockers were more effective than renin-angiotensin system blockers alone in reducing proteinuria. Unfortunately, the small number of patients, the short time of observation (six months) and the single-center nature of the study limits the power of these conclusions [242].
Because of the few strong evidences of its benefit in CKD, additional studies are warranted to assess the real efficacy of these agents in slowing the progression of renal failure and to establish the formal intake recommendations and dosing in the CKD patient population (particularly in dialysis).

12. Conclusions

CKD is associated with enhanced oxidative stress that is a well-known risk factor for the onset/development of severe systemic complications and cardiovascular diseases. Several studies report a higher level of oxidative stress markers together with reduced antioxidants in pre-dialysis patients. This condition is exacerbated during the progression of renal failure and in renal replacement therapy. Several biological mechanisms contribute to oxidative stress, including mitochondrial activity, xanthine oxidase, and NADPH oxidase. Thus, in recent years the administration of antioxidants, both food-derived and through drugs with additional antioxidant effects, have demonstrated positive effects. Many of these compounds have direct ROS scavenger properties due to their molecular structure (l-carnitine, vitamin E, vitamin C, α-lipoic acid, green tea, resveratrol, curcumin), others also have indirect antioxidant effects mediated by the upregulation of antioxidant enzymes (l-carnitine, green tea, α-lipoic acid, resveratrol, curcumin) or by additional anti-inflammatory properties (vitamin E, resveratrol, curcumin, omega-3).
However, the great limit of most of these studies is the low sample size and short-term follow-up. As a consequence, none of these molecules have been introduced into usual clinical practice. Therefore, future prospective and comparative studies analyzing the co-administration of different antioxidants with long-term follow-up are warranted.

Author Contributions

Lorenzo Signorini, Simona Granata and Gianluigi Zaza searched the literature and wrote the manuscript; Antonio Lupo revised the manuscript; All authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cachofeiro, V.; Goicochea, M.; de Vinuesa, S.G.; Oubiña, P.; Lahera, V.; Luño, J. Oxidative stress and inflammation, a link between chronic kidney disease and cardiovascular disease. Kidney Int. Suppl. 2008, 111, S4–S9. [Google Scholar] [CrossRef] [PubMed]
  2. 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]
  3. Dursun, B.; Dursun, E.; Suleymanlar, G.; Ozben, B.; Capraz, I.; Apaydin, A.; Ozben, T. Carotid artery intima-media thickness correlates with oxidative stress in chronic haemodialysis patients with accelerated atherosclerosis. Nephrol. Dial. Transplant. 2008, 23, 1697–1703. [Google Scholar] [CrossRef] [PubMed]
  4. Kato, A.; Odamaki, M.; Hishida, A. Blood 8-hydroxy-2′-deoxyguanosine is associated with erythropoietin resistance in hemodialysis patients. Nephrol. Dial. Transplant. 2003, 18, 931–936. [Google Scholar] [CrossRef] [PubMed]
  5. Del Vecchio, L.; Locatelli, F.; Carini, M. What we know about oxidative stress in patients with chronic kidney disease on dialysis—Clinical effects, potential treatment, and prevention. Semin Dial. 2011, 24, 56–64. [Google Scholar] [CrossRef] [PubMed]
  6. Locatelli, F.; Canaud, B.; Eckardt, K.; Stenvinkel, P.; Wanner, C.; Zoccali, C. Oxidative stress in end-stage renal disease: An emerging treat to patient outcome. Nephrol. Dial. Transplant. 2003, 18, 1272–1280. [Google Scholar] [CrossRef]
  7. 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 Genom. 2009, 10, 388. [Google Scholar] [CrossRef] [PubMed]
  8. Zaza, G.; Granata, S.; Masola, V.; Rugiu, C.; Fantin, F.; Gesualdo, L.; Schena, F.P.; Lupo, A. Downregulation of nuclear-encoded genes of oxidative metabolism in dialyzed chronic kidney disease patients. PLoS ONE 2013, 8, e77847. [Google Scholar] [CrossRef]
  9. Granata, S.; Masola, V.; Zoratti, E.; Scupoli, M.T.; Baruzzi, A.; Messa, M.; Sallustio, F.; Gesualdo, L.; Lupo, A.; Zaza, G. NLRP3 inflammasome activation in dialyzed chronic kidney disease patients. PLoS ONE 2015, 10, e0122272. [Google Scholar] [CrossRef] [PubMed]
  10. Hajnóczky, G.; Csordás, G.; Das, S.; Garcia-Perez, C.; Saotome, M.; Sinha Roy, S. Mitochondrial calcium signalling and cell death: Approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium 2006, 40, 553–560. [Google Scholar] [CrossRef] [PubMed]
  11. Ajioka, R.S.; Phillips, J.D.; Kushner, J.P. Biosynthesis of heme in mammals. Biochim. Biophys. Acta 2006, 1763, 723–736. [Google Scholar] [CrossRef] [PubMed]
  12. Rossier, M.F. T channels and steroid biosynthesis: In search of a link with mitochondria. Cell Calcium 2006, 40, 155–164. [Google Scholar] [CrossRef] [PubMed]
  13. Green, D.R. Apoptotic pathways: The roads to ruin. Cell 1998, 94, 695–698. [Google Scholar] [CrossRef]
  14. Gamboa, J.L.; Billings, F.T.; Bojanowski, M.T.; Gilliam, L.A.; Yu, C.; Roshanravan, B.; Roberts, L.J.; Himmelfarb, J.; Ikizler, T.A.; Brown, N.J. Mitochondrial dysfunction and oxidative stress in patients with chronic kidney disease. Physiol. Rep. 2016, 4, e12780. [Google Scholar] [CrossRef] [PubMed]
  15. Su, M.; Dhoopun, A.R.; Yuan, Y.; Huang, S.; Zhu, C.; Ding, G.; Liu, B.; Yang, T.; Zhang, A. Mitochondrial dysfunction is an early event in aldosterone-induced podocyte injury. Am. J. Physiol. Ren. Physiol. 2013, 305, F520–F531. [Google Scholar] [CrossRef] [PubMed]
  16. Zhao, M.; Yuan, Y.; Bai, M.; Ding, G.; Jia, Z.; Huang, S.; Zhang, A. PGC-1α overexpression protects against aldosterone-induced podocyte depletion: Role of mitochondria. Oncotarget 2016, 7, 12150–12162. [Google Scholar] [PubMed]
  17. 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]
  18. Zhang, A.; Jia, Z.; Guo, X.; Yang, T. Aldosterone induces epithelial-mesenchymal transition via ROS of mitochondrial origin. Am. J. Physiol. Ren. Physiol. 2007, 293, F723–F731. [Google Scholar] [CrossRef] [PubMed]
  19. Mutsaers, H.A.; Wilmer, M.J.; Reijnders, D.; Jansen, J.; van den Broek, P.H.; Forkink, M.; Schepers, E.; Glorieux, G.; Vanholder, R.; van den Heuvel, L.P.; et al. Uremic toxins inhibit renal metabolic capacity through interference with glucuronidation and mitochondrial respiration. Biochim. Biophys. Acta 2013, 1832, 142–150. [Google Scholar] [CrossRef] [PubMed]
  20. Sedeek, M.; Nasrallah, R.; Touyz, R.M.; Hébert, R.L. NADPH oxidases, reactive oxygen species, and the kidney: Friend and foe. J. Am. Soc. Nephrol. 2013, 24, 1512–1518. [Google Scholar] [CrossRef] [PubMed]
  21. 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]
  22. Fortuño, A.; Beloqui, O.; San José, G.; Moreno, M.U.; Zalba, G.; Díez, 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]
  23. Morena, M.; Cristol, J.P.; Senécal, L.; Leray-Moragues, H.; Krieter, D.; Canaud, B. Oxidative stress in hemodialysis patients: Is NADPH oxidase complex the culprit? Kidney Int. Suppl. 2002, 80, 109–114. [Google Scholar] [CrossRef] [PubMed]
  24. Choi, J.Y.; Yoon, Y.J.; Choi, H.J.; Park, S.H.; Kim, C.D.; Kim, I.S.; Kwon, T.H.; Do, J.Y.; Kim, S.H.; Ryu, D.H.; et al. Dialysis modality-dependent changes in serum metabolites: Accumulation of inosine and hypoxanthine in patients on haemodialysis. Nephrol. Dial Transplant. 2011, 26, 1304–1313. [Google Scholar] [CrossRef] [PubMed]
  25. Gondouin, B.; Jourde-Chiche, N.; Sallee, M.; Dou, L.; Cerini, C.; Loundou, A.; Morange, S.; Berland, Y.; Burtey, S.; Brunet, P.; et al. Plasma Xanthine Oxidase Activity Is Predictive of Cardiovascular Disease in Patients with Chronic Kidney Disease, Independently of Uric Acid Levels. Nephron 2015, 131, 167–174. [Google Scholar] [CrossRef] [PubMed]
  26. Small, D.M.; Gobe, G.C. Oxidative Stress and Antioxidant Therapy in Chronic Kidney and Cardiovascular Disease. In Oxidative Stress and Chronic Degenerative Diseases—A Role for Antioxidants; In Tech: Vienna, Austria, 2013. [Google Scholar]
  27. Goicoechea, M.; de Vinuesa, S.G.; Verdalles, U.; Ruiz-Caro, C.; Ampuero, J.; Rincón, A.; Arroyo, D.; Luño, J. Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin. J. Am. Soc. Nephrol. 2010, 5, 1388–1393. [Google Scholar] [CrossRef] [PubMed]
  28. Siu, Y.P.; Leung, K.T.; Tong, M.K.; Kwan, T.H. Use of allopurinol in slowing the progression of renal disease through its ability to lower serum uric acid level. Am. J. Kidney Dis. 2006, 47, 51–59. [Google Scholar] [CrossRef] [PubMed]
  29. Kanbay, M.; Ozkara, A.; Selcoki, Y.; Isik, B.; Turgut, F.; Bavbek, N.; Uz, E.; Akcay, A.; Yigitoglu, R.; Covic, A. Effect of treatment of hyperuricemia with allopurinol on blood pressure, creatinine clearence, and proteinuria in patients with normal renal functions. Int. Urol. Nephrol. 2007, 39, 1227–1233. [Google Scholar] [CrossRef] [PubMed]
  30. Goicoechea, M.; Garcia de Vinuesa, S.; Verdalles, U.; Verde, E.; Macias, N.; Santos, A.; Pérez de Jose, A.; Cedeño, S.; Linares, T.; Luño, J. Allopurinol and progression of CKD and cardiovascular events: Long-term follow-up of a randomized clinical trial. Am. J. Kidney Dis. 2015, 65, 543–549. [Google Scholar] [CrossRef] [PubMed]
  31. Becker, M.A.; Schumacher, H.R., Jr.; Wortmann, R.L.; MacDonald, P.A.; Eustace, D.; Palo, W.A.; Streit, J.; Joseph-Ridge, N. Febuxostat compared with allopurinol in patients with hyperuricemia and gout. N. Engl. J. Med. 2005, 353, 2450–2561. [Google Scholar] [CrossRef] [PubMed]
  32. Dalbeth, N.; Kumar, S.; Stamp, L.; Gow, P. Dose adjustment of allopurinol according to creatinine clearance does not provide adequate control of hyperuricemia in patients with gout. J. Rheumatol. 2006, 33, 1646–1650. [Google Scholar] [PubMed]
  33. Thurston, M.M.; Phillips, B.B.; Bourg, C.A. Safety and efficacy of allopurinol in chronic kidney disease. Ann. Pharmacother. 2013, 47, 1507–1516. [Google Scholar] [CrossRef] [PubMed]
  34. Fleeman, N.; Pilkington, G.; Dundar, Y.; Dwan, K.; Boland, A.; Dickson, R.; Anijeet, H.; Kennedy, T.; Pyatt, J. Allopurinol for the treatment of chronic kidney disease: A systematic review. Health Technol. Assess. 2014, 18, 1–77. [Google Scholar] [CrossRef] [PubMed]
  35. Schumacher, H.R., Jr.; Becker, M.A.; Wortmann, R.L.; Macdonald, P.A.; Hunt, B.; Streit, J.; Lademacher, C.; Joseph-Ridge, N. Effects of febuxostat versus allopurinol and placebo in reducing serum urate in subjects with hyperuricemia and gout: A 28-week, phase, I.I.I.; randomized, double-blind, parallel-group trial. Arthritis Rheum. 2008, 59, 1540–1548. [Google Scholar] [CrossRef] [PubMed]
  36. Becker, M.A.; Schumacher, H.R.; Espinoza, L.R.; Wells, A.F.; MacDonald, P.; Lloyd, E.; Lademacher, C. The urate-lowering efficacy and safety of febuxostat in the treatment of the hyperuricemia of gout: The CONFIRMS trial. Arthritis Res. Ther. 2010, 12, R63. [Google Scholar] [CrossRef] [PubMed]
  37. Yamaguchi, A.; Harada, M.; Yamada, Y.; Hashimoto, K.; Kamijo, Y. Identification of chronic kidney disease patient characteristics influencing the renoprotective effects of febuxostat therapy: A retrospective follow-up study. BMC Nephrol. 2017, 18, 162. [Google Scholar] [CrossRef] [PubMed]
  38. Sindhu, R.K.; Ehdaie, A.; Farmand, F.; Dhaliwal, K.K.; Nguyen, T.; Zhan, C.D.; Roberts, C.K.; Vaziri, N.D. Expression of catalase and glutathione peroxidase in renal insufficiency. Biochim. Biophys. Acta 2005, 1743, 86–92. [Google Scholar] [CrossRef] [PubMed]
  39. Ross, E.A.; Koo, L.C.; Moberly, J.B. Low whole blood and erythrocyte levels of glutathione in hemodialysis and peritoneal dialysis patients. Am. J. Kidney Dis. 1997, 30, 489–494. [Google Scholar] [CrossRef]
  40. Ceballos-Picot, I.; Witko-Sarsat, V.; Merad-Boudia, M.; Nguyen, A.T.; Thévenin, 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] [CrossRef]
  41. Tbahriti, H.F.; Kaddous, A.; Bouchenak, M.; Mekki, K. Effect of different stages of chronic kidney disease and renal replacement therapies on oxidant-antioxidant balance in uremic patients. Biochem. Res. Int. 2013, 2013, 358985. [Google Scholar] [CrossRef] [PubMed]
  42. Granata, S.; Dalla Gassa, A.; Tomei, P.; Lupo, A.; Zaza, G. Mitochondria: A new therapeutic target in chronic kidney disease. Nutr. Metab. 2015, 12, 49. [Google Scholar] [CrossRef] [PubMed]
  43. Guarnieri, G.; Situlin, R.; Biolo, G. Carnitine metabolism in uremia. Am. J. Kidney Dis. 2001, 38, S63–S67. [Google Scholar] [CrossRef] [PubMed]
  44. Sharma, S.; Black, S.M. Carnitine homeostasis, mitochondrial function, and cardiovascular disease. Drug Discov. Today Dis. Mech. 2009, 6, e31–e39. [Google Scholar] [CrossRef] [PubMed]
  45. Kerner, J.; Hoppel, C. Fatty acid import into mitochondria. Biochim. Biophys. Acta 2000, 1486, 1–17. [Google Scholar] [CrossRef]
  46. Flanagan, J.L.; Simmons, P.A.; Vehige, J.; Willcox, M.D.; Garrett, Q. Role of carnitine in disease. Nutr. Metab. 2010, 7, 30. [Google Scholar] [CrossRef] [PubMed]
  47. Gülçin, I. Antioxidant and antiradical activities of l-carnitine. Life Sci. 2006, 78, 803–811. [Google Scholar] [CrossRef] [PubMed]
  48. Calò, L.A.; Pagnin, E.; Davis, P.A.; Semplicini, A.; Nicolai, R.; Calvani, M.; Pessina, A.C. Antioxidant effect of l-carnitine and its short chain esters: Relevance for the protection from oxidative stress related cardiovascular damage. Int. J. Cardiol. 2006, 107, 54–60. [Google Scholar] [CrossRef] [PubMed]
  49. Arockia Rani, P.J.; Panneerselvam, C. Carnitine as a free radical scavenger in aging. Exp. Gerontol. 2001, 36, 1713–1726. [Google Scholar] [CrossRef]
  50. Rani, P.J.; Panneerselvam, C. Effect of l-carnitine on brain lipid peroxidation and antioxidant enzymes in old rats. J. Gerontol. A Biol. Sci. Med. Sci. 2002, 57, B134–B137. [Google Scholar] [CrossRef] [PubMed]
  51. Guarnieri, G. Carnitine in maintenance hemodialysis patients. J. Ren. Nutr. 2015, 25, 169–175. [Google Scholar] [CrossRef] [PubMed]
  52. Di Liberato, L.; Arduini, A.; Rossi, C.; Di Castelnuovo, A.; Posari, C.; Sacchetta, P.; Urbani, A.; Bonomini, M. l-Carnitine status in end-stage renal disease patients on automated peritoneal dialysis. J. Nephrol. 2014, 27, 699–706. [Google Scholar] [CrossRef] [PubMed]
  53. Fatouros, I.G.; Douroudos, I.; Panagoutsos, S.; Pasadakis, P.; Nikolaidis, M.G.; Chatzinikolaou, A.; Sovatzidis, A.; Michailidis, Y.; Jamurtas, A.Z.; Mandalidis, D.; et al. Effects of l-carnitine on oxidative stress responses in patients with renal disease. Med. Sci. Sports Exerc. 2010, 42, 1809–1818. [Google Scholar] [CrossRef] [PubMed]
  54. Pertosa, G.; Grandaliano, G.; Simone, S.; Soccio, M.; Schena, F.P. Inflammation and carnitine in hemodialysis patients. J. Ren. Nutr. 2005, 15, 8–12. [Google Scholar] [CrossRef] [PubMed]
  55. Golper, T.A.; Goral, S.; Becker, B.N.; Langman, C.B. l-carnitine treatment of anemia. Am. J. Kidney Dis. 2003, 41, S27–S34. [Google Scholar] [CrossRef]
  56. Kletzmayr, J.; Mayer, G.; Legenstein, E.; Heinz-Peer, G.; Leitha, T.; Hörl, W.H.; Kovarik, J. Anemia and carnitine supplementation in hemodialyzed patients. Kidney Int. Suppl. 1999, 69, S93–S106. [Google Scholar] [CrossRef] [PubMed]
  57. Yee, J. l-carnitine for anemia in hemodialysis patients: A last resort. Clin. J. Am. Soc. Nephrol. 2012, 7, 1746–1748. [Google Scholar] [CrossRef] [PubMed]
  58. Hurot, J.M.; Cucherat, M.; Haugh, M.; Fouque, D. Effects of l-carnitine supplementation in maintenance hemodialysis patients: A systematic review. J. Am. Soc. Nephrol. 2002, 13, 708–714. [Google Scholar] [PubMed]
  59. Mercadal, L.; Coudert, M.; Vassault, A.; Pieroni, L.; Debure, A.; Ouziala, M.; Depreneuf, H.; Fumeron, C.; Servais, A.; Bassilios, N.; et al. l-carnitine treatment in incident hemodialysis patients: The multicenter, randomized, double-blinded, placebo-controlled CARNIDIAL trial. Clin. J. Am. Soc. Nephrol. 2012, 7, 1836–1842. [Google Scholar] [CrossRef] [PubMed]
  60. Eknoyan, G.; Latos, D.L.; Lindberg, J.; National Kidney Foundation Carnitine Consensus Conference. Practice recommendations for the use of l-carnitine in dialysis-related carnitine disorder. National Kidney Foundation Carnitine Consensus Conference. Am. J. Kidney Dis. 2003, 41, 868–876. [Google Scholar] [CrossRef]
  61. Kliger, A.S.; Foley, R.N.; Goldfarb, D.S.; Goldstein, S.L.; Johansen, K.; Singh, A.; Szczech, L. KDOQI US commentary on the 2012 KDIGO Clinical Practice Guideline for Anemia in CKD. Am. J. Kidney Dis. 2013, 62, 849–859. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, Y.; Abbate, M.; Tang, L.; Cai, G.; Gong, Z.; Wei, R.; Zhou, J.; Chen, X. l-Carnitine supplementation for adults with end-stage kidney disease requiring maintenance hemodialysis: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2014, 99, 408–422. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, S.K.; Xiao, L.; Song, P.A.; Xu, X.; Liu, F.Y.; Sun, L. Effect of l-carnitine therapy on patients in maintenance hemodialysis: A systematic review and meta-analysis. J. Nephrol. 2014, 27, 317–329. [Google Scholar] [CrossRef] [PubMed]
  64. IUPAC-IUB Joint Commission on Biochemical Nomenclature (JCBN). Nomenclature of tocopherols and related compounds. Recommendations 1981. Eur. J. Biochem. 1982, 123, 473–475. [Google Scholar]
  65. Brigelius-Flohe, R.; Traber, M.G. Vitamin E: Function and metabolism. FASEB J. 1999, 13, 1145–1155. [Google Scholar] [PubMed]
  66. Traber, M.G.; Atkinson, J. Vitamin E antioxidant and nothing more. Free Radic. Biol. Med. 2007, 43, 4–15. [Google Scholar] [CrossRef] [PubMed]
  67. Serbinova, E.; Kagan, V.; Han, D.; Packer, L. Free radical recycling and intramembrane mobility in the antioxidant properties of α-tocopherol and α-tocotrienol. Free Radic. Biol. Med. 1991, 10, 263–275. [Google Scholar] [CrossRef]
  68. Princen, H.M.; van Duyvenvoorde, W.; Buytenhek, R.; van der Laarse, A.; van Poppel, G.; Gevers Leuven, J.A.; van Hinsbergh, V.W. Supplementation with low doses of vitamin E protects LDL from lipid peroxidation in men and women. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 325–333. [Google Scholar] [CrossRef] [PubMed]
  69. Cachia, O.; Benna, J.E.; Pedruzzi, E.; Descomps, B.; Gougerot-Pocidalo, M.A.; Leger, C.L. α-tocopherol inhibits the respiratory burst in human monocytes. Attenuation of p47(phox) membrane translocation and phosphorylation. J. Biol. Chem. 1998, 273, 32801–32805. [Google Scholar] [CrossRef] [PubMed]
  70. Bozaykut, P.; Karademir, B.; Yazgan, B.; Sozen, E.; Siow, R.C.; Mann, G.E.; Ozer, N.K. Effects of vitamin E on peroxisome proliferator-activated receptor γ and nuclear factor-erythroid 2-related factor 2 in hypercholesterolemia-induced atherosclerosis. Free Radic. Biol. Med. 2014, 70, 174–181. [Google Scholar] [CrossRef] [PubMed]
  71. Stephens, N.G.; Parsons, A.; Schofield, P.M.; Kelly, F.; Cheeseman, K.; Mitchinson, M.J. Randomised controlled trial of vitamin E in patients with coronary disease: Cambridge Heart Antioxidant Study (CHAOS). Lancet 1996, 347, 781–786. [Google Scholar] [CrossRef]
  72. Traber, M.G.; Stevens, J.F. Vitamins C and E: Beneficial effects from a mechanistic perspective. Free Radic. Biol. Med. 2011, 51, 1000–1013. [Google Scholar] [CrossRef] [PubMed]
  73. Yoshikawa, T.; Yoshida, N.; Manabe, H.; Terasawa, Y.; Takemura, T.; Kondo, M. α-Tocopherol protects against expression of adhesion molecules on neutrophils and endothelial cells. Biofactors 1998, 7, 15–19. [Google Scholar] [CrossRef] [PubMed]
  74. Villacorta, L.; Graça-Souza, A.V.; Ricciarelli, R.; Zingg, J.M.; Azzi, A. α-Tocopherol induces expression of connective tissue growth factor and antagonizes tumor necrosis factor-α-mediated downregulation in human smooth muscle cells. Circ. Res. 2003, 92, 104–110. [Google Scholar] [CrossRef] [PubMed]
  75. Azzi, A.; Gysin, R.; Kempná, P.; Munteanu, A.; Negis, Y.; Villacorta, L.; Visarius, T.; Zingg, J.M. Vitamin E mediates cell signaling and regulation of gene expression. Ann. N. Y. Acad. Sci. 2004, 1031, 86–95. [Google Scholar] [CrossRef] [PubMed]
  76. Kuemerle, N.B.; Brandt, R.B.; Chan, W.; Krieg, R.J., Jr.; Chan, J.C. Inhibition of transforming growth factor β-1 induction by dietary vitamin E in unilateral obstruction in rats. Biochem. Mol. Med. 1997, 61, 82–96. [Google Scholar] [CrossRef]
  77. Boaz, M.; Smetana, S.; Weinstein, T.; Matas, Z.; Gafter, U.; Iaina, A.; Knecht, A.; Weissgarten, Y.; Brunner, D.; Fainaru, M.; et al. Secondary prevention with antioxidants of cardiovascular disease in end stage renal disease (SPACE): Randomised placebo-controlled trial. Lancet 2000, 356, 1213–1218. [Google Scholar] [CrossRef]
  78. Mann, J.F.; Lonn, E.M.; Yi, Q.; Gerstein, H.C.; Hoogwerf, B.J.; Pogue, J.; Bosch, J.; Dagenais, G.R.; Yusuf, S.; HOPE Investigators. Effects of vitamin E on cardiovascular outcomes in people with mild to moderate renal insufficiency: Results of the HOPE study. Kidney Int. 2004, 65, 1375–1380. [Google Scholar] [CrossRef] [PubMed]
  79. Tasanarong, A.; Vohakiat, A.; Hutayanon, P.; Piyayotai, D. New strategy of α- and γ-tocopherol to prevent contrast-induced acute kidney injury in chronic kidney disease patients undergoing elective coronary procedures. Nephrol. Dial. Transplant. 2013, 28, 337–344. [Google Scholar] [CrossRef] [PubMed]
  80. Rezaei, Y.; Khademvatani, K.; Rahimi, B.; Khoshfetrat, M.; Arjmand, N.; Seyyed-Mohammadzad, M.H. Short-Term High-Dose Vitamin E to Prevent Contrast Medium-Induced Acute Kidney Injury in Patients With Chronic Kidney Disease Undergoing Elective Coronary Angiography: A Randomized Placebo-Controlled Trial. J. Am. Heart. Assoc. 2016, 5, e002919. [Google Scholar] [CrossRef] [PubMed]
  81. Yang, C.C.; Hsu, S.P.; Wu, M.S.; Hsu, S.M.; Chien, C.T. Effects of vitamin C infusion and vitamin E-coated membrane on hemodialysis-induced oxidative stress. Kidney Int. 2006, 69, 706–714. [Google Scholar] [CrossRef] [PubMed]
  82. Takouli, L.; Hadjiyannakos, D.; Metaxaki, P.; Sideris, V.; Filiopoulos, V.; Anogiati, A.; Vlassopoulos, D. Vitamin E-coated cellulose acetate dialysis membrane: Long-term effect on inflammation and oxidative stress. Ren. Fail. 2010, 32, 287–293. [Google Scholar] [CrossRef] [PubMed]
  83. Kirmizis, D.; Papagianni, A.; Belechri, A.M.; Memmos, D. Effects of vitamin E-coated membrane dialyser on markers of oxidative stress and inflammation in patients on chronic haemodialysis. Nephrol. Dial. Transplant. 2011, 26, 2296–2301. [Google Scholar] [CrossRef] [PubMed]
  84. Panichi, V.; Rosati, A.; Paoletti, S.; Ferrandello, P.; Migliori, M.; Beati, S.; Bernabini, G.; Daini, R.; Casani, A.; Angelini, D.; et al. A vitamin E-coated polysulfone membrane reduces serum levels of inflammatory markers and resistance to erythropoietin-stimulating agents in hemodialysis patients: Results of a randomized cross-over multicenter trial. Blood Purif. 2011, 32, 7–14. [Google Scholar] [CrossRef] [PubMed]
  85. Kitamura, Y.; Kamimura, K.; Yoshioka, N.; Hosotani, Y.; Tsuchida, K.; Koremoto, M.; Minakuchi, J. The effect of vitamin E-bonded polysulfone membrane dialyzer on a new oxidative lipid marker. J. Artif. Organ. 2013, 16, 206–210. [Google Scholar] [CrossRef] [PubMed]
  86. Lines, S.W.; Carter, A.M.; Dunn, E.J.; Lindley, E.J.; Tattersall, J.E.; Wright, M.J. A randomized controlled trial evaluating the erythropoiesis stimulating agent sparing potential of a vitamin E-bonded polysulfone dialysis membrane. Nephrol. Dial. Transplant. 2014, 29, 649–656. [Google Scholar] [CrossRef] [PubMed]
  87. Yang, S.K.; Xiao, L.; Xu, B.; Xu, X.X.; Liu, F.Y.; Sun, L. Effects of vitamin E-coated dialyzer on oxidative stress and inflammation status in hemodialysis patients: A systematic review and meta-analysis. Ren. Fail. 2014, 36, 722–731. [Google Scholar] [CrossRef] [PubMed]
  88. Veringa, S.J.; Nanayakkara, P.W.; van Ittersum, F.J.; Vegting, I.L.; van Guldener, C.; Smulders, Y.M.; ter Wee, P.M.; Stehouwer, C.D. Effect of a treatment strategy consisting of pravastatin, vitamin, E.; and homocysteine lowering on arterial compliance and distensibility in patients with mild to moderate chronic kidney disease. Clin. Nephrol. 2012, 78, 263–272. [Google Scholar] [CrossRef] [PubMed]
  89. Nanayakkara, P.W.; van Guldener, C.; ter Wee, P.M.; Scheffer, P.G.; van Ittersum, F.J.; Twisk, J.W.; Teerlink, T.; van Dorp, W.; Stehouwer, C.D. Effect of a treatment strategy consisting of pravastatin, vitamin, E.; and homocysteine lowering on carotid intima-media thickness, endothelial function, and renal function in patients with mild to moderate chronic kidney disease: Results from the Antioxidant Therapy in Chronic Renal Insufficiency (ATIC) Study. Arch. Intern. Med. 2007, 167, 1262–1270. [Google Scholar] [PubMed]
  90. Pearson, P.; Lewis, S.A.; Britton, J.; Young, I.S.; Fogarty, A. The pro-oxidant activity of high-dose vitamin E supplements in vivo. BioDrugs 2006, 20, 271–273. [Google Scholar] [CrossRef] [PubMed]
  91. Antoniadi, G.; Eleftheriadis, T.; Liakopoulos, V.; Kakasi, E.; Kartsios, C.; Passadakis, P.; Vargemezis, V. Effect of one-year oral α-tocopherol administration on the antioxidant defense system in hemodialysis patients. Ther. Apher. Dial. 2008, 12, 237–242. [Google Scholar] [CrossRef] [PubMed]
  92. Kagan, V.E.; Serbinova, E.A.; Forte, T.; Scita, G.; Packer, L. Recycling of vitamin E in human low density lipoproteins. J. Lipid Res. 1992, 33, 385–397. [Google Scholar] [PubMed]
  93. Sesso, H.D.; Buring, J.E.; Christen, W.G.; Kurth, T.; Belanger, C.; MacFadyen, J.; Bubes, V.; Manson, J.E.; Glynn, R.J.; Gaziano, J.M. Vitamins E and C in the prevention of cardiovascular disease in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2008, 300, 2123–2133. [Google Scholar] [CrossRef] [PubMed]
  94. Clase, C.M.; Ki, V.; Holden, R.M. Water-soluble vitamins in people with low glomerular filtration rate or on dialysis: A review. Semin. Dial. 2013, 26, 546–567. [Google Scholar] [CrossRef] [PubMed]
  95. Monsen, E.R. Dietary reference intakes for the antioxidant nutrients: Vitamin, C.; vitamin, E.; selenium, and carotenoids. J. Am. Diet Assoc. 2000, 100, 637–640. [Google Scholar] [CrossRef]
  96. Descamps-Latscha, B.; Drüeke, T.; Witko-Sarsat, V. Dialysis-induced oxidative stress: Biological aspects, clinical consequences, and therapy. Semin. Dial. 2001, 14, 193–199. [Google Scholar] [CrossRef] [PubMed]
  97. Gruss-Fischer, T.; Fabian, I. Protection by ascorbic acid from denaturation and release of cytochrome c, alteration of mitochondrial membrane potential and activation of multiple caspases induced by H2O2, in human leukemia cells. Biochem. Pharmacol. 2002, 63, 1325–1335. [Google Scholar] [CrossRef]
  98. Perez-Cruz, I.; Carcamo, J.M.; Golde, D.W. Vitamin C inhibits FAS-induced apoptosis in monocytes and U937 cells. Blood 2003, 102, 336–343. [Google Scholar] [CrossRef] [PubMed]
  99. Dhar-Mascareño, M.; Cárcamo, J.M.; Golde, D.W. Hypoxia-reoxygenation-induced mitochondrial damage and apoptosis in human endothelial cells are inhibited by vitamin C. Free Radic. Biol. Med. 2005, 38, 1311–1322. [Google Scholar]
  100. KC, S.; Cárcamo, J.M.; Golde, D.W. Vitamin C enters mitochondria via facilitative glucose transporter 1 (Glut1) and confers mitochondrial protection against oxidative injury. FASEB J. 2005, 19, 1657–1667. [Google Scholar] [CrossRef] [PubMed]
  101. May, J.M.; Qu, Z.C.; Whitesell, R.R.; Cobb, C.E. Ascorbate recycling in human erythrocytes: Role of GSH in reducing dehydroascorbate. Free Radic Biol Med. 1996, 20, 543–551. [Google Scholar] [CrossRef]
  102. Nguyen-Khoa, T.; Massy, Z.A.; De Bandt, J.P.; Kebede, M.; Salama, L.; Lambrey, G.; Witko-Sarsat, V.; Drüeke, T.B.; Lacour, B.; Thévenin, M. Oxidative stress and haemodialysis: Role of inflammation and duration of dialysis treatment. Nephrol. Dial. Transplant. 2001, 16, 335–3540. [Google Scholar] [CrossRef] [PubMed]
  103. Deicher, R.; Horl, W.H. Vitamin C in chronic kidney disease and hemodialysis patients. Kidney Blood Press. Res. 2003, 26, 100–106. [Google Scholar] [CrossRef] [PubMed]
  104. Deicher, R.; Ziai, F.; Bieglmayer, C.; Schillinger, M.; Hörl, W.H. Low total vitamin C plasma level is a risk factor for cardiovascular morbidity and mortality in hemodialysis patients. J. Am. Soc. Nephrol. 2005, 16, 1811–1818. [Google Scholar] [CrossRef] [PubMed]
  105. Takahashi, N.; Morimoto, S.; Okigaki, M.; Seo, M.; Someya, K.; Morita, T.; Matsubara, H.; Sugiura, T.; Iwasaka, T. Decreased plasma level of vitamin C in chronic kidney disease: Comparison between diabetic and non-diabetic patients. Nephrol. Dial. Transplant. 2011, 26, 1252–1257. [Google Scholar] [CrossRef] [PubMed]
  106. Kamgar, M.; Zaldivar, F.; Vaziri, N.D.; Pahl, M.V. Antioxidant therapy does not ameliorate oxidative stress and inflammation in patients with end-stage renal disease. J. Natl. Med. Assoc. 2009, 101, 336–344. [Google Scholar] [CrossRef]
  107. Singer, R.F. Vitamin C supplementation in kidney failure: Effect on uraemic symptoms. Nephrol. Dial. Transplant. 2011, 26, 614–620. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, K.; Li, Y.; Cheng, X.; Liu, L.; Bai, W.; Guo, W.; Wu, L.; Zuo, L. Cross-over study of influence of oral vitamin C supplementation on inflammatory status in maintenance hemodialysis patients. BMC Nephrol. 2013, 14, 252. [Google Scholar] [CrossRef] [PubMed]
  109. Deved, V.; Poyah, P.; James, M.T.; Tonelli, M.; Manns, B.J.; Walsh, M.; Hemmelgarn, B.R. Alberta Kidney Disease Network. Ascorbic acid for anemia management in hemodialysis patients: A systematic review and meta-analysis. Am. J. Kidney Dis. 2009, 54, 1089–1097. [Google Scholar] [CrossRef] [PubMed]
  110. Kang, D.W.; Ahn, C.Y.; Ryu, B.K.; Shin, B.C.; Chung, J.H.; Kim, H.L. The effect of intravenous ascorbic acid in hemodialysis patients with normoferritinemic anemia. Kidney Res. Clin. Pract. 2012, 31, 48–53. [Google Scholar] [CrossRef] [PubMed]
  111. Ernster, L.; Dallner, G. Biochemical, physiological and medical aspects of ubiquinone function. Biochim. Biophys. Acta 1995, 1271, 195–204. [Google Scholar] [CrossRef]
  112. Pravst, I.; Zmitek, K.; Zmitek, J. Coenzyme Q10 contents in foods and fortification strategies. Crit. Rev. Food Sci. Nutr. 2010, 50, 269–280. [Google Scholar] [CrossRef] [PubMed]
  113. Bentinger, M.; Tekle, M.; Dallner, G. Coenzyme Q—Biosynthesis and functions. Biochem. Biophys. Res. Commun. 2010, 396, 74–79. [Google Scholar] [CrossRef] [PubMed]
  114. Bentinger, M.; Brismar, K.; Dallner, G. The antioxidant role of coenzyme Q. Mitochondrion 2007, 7, S41–S50. [Google Scholar] [CrossRef] [PubMed]
  115. Papucci, L.; Schiavone, N.; Witort, E.; Donnini, M.; Lapucci, A.; Tempestini, A.; Formigli, L.; Zecchi-Orlandini, S.; Orlandini, G.; et al. Coenzyme q10 prevents apoptosis by inhibiting mitochondrial depolarization independently of its free radical scavenging property. J. Biol. Chem. 2003, 278, 28220–28228. [Google Scholar] [CrossRef] [PubMed]
  116. Bergamini, C.; Moruzzi, N.; Sblendido, A.; Lenaz, G.; Fato, R. A water soluble CoQ10 formulation improves intracellular distribution and promotes mitochondrial respiration in cultured cells. PLoS ONE 2012, 7, e33712. [Google Scholar] [CrossRef] [PubMed]
  117. Gazdíková, K.; Gvozdjáková, A.; Kucharská, J.; Spustová, V.; Braunová, Z.; Dzúrik, R. Oxidative stress and plasma concentrations of coenzyme Q10, α-tocopherol, and beta-carotene in patients with a mild to moderate decrease of kidney function. Nephron 2001, 88, 285. [Google Scholar] [CrossRef] [PubMed]
  118. Lippa, S.; Colacicco, L.; Callà, C.; Sagliaschi, G.; Angelitti, A.G. Coenzyme Q10 levels, plasma lipids and peroxidation extent in renal failure and in hemodialytic patients. Mol. Aspects Med. 1994, 15, S213–S219. [Google Scholar] [CrossRef]
  119. Macunluoglu, B.; Atakan, A.; Ari, E.; Kaya, Y.; Kaspar, C.; Demir, H.; Alp, H.H. Epicardial fat tissue thickness is correlated with diminished levels of co-enzyme Q10, a major antioxidant molecule among hemodialysis patients. Clin. Biochem. 2014, 47, 1231–1234. [Google Scholar] [CrossRef] [PubMed]
  120. Mazurek, T.; Zhang, L.; Zalewski, A.; Mannion, J.D.; Diehl, J.T.; Arafat, H.; Sarov-Blat, L.; O’Brien, S.; Keiper, E.A.; Johnson, A.G.; et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation 2003, 108, 2460–2466. [Google Scholar] [CrossRef] [PubMed]
  121. Baker, A.R.; Silva, N.F.; Quinn, D.W.; Harte, A.L.; Pagano, D.; Bonser, R.S.; Kumar, S.; McTernan, P.G. Human epicardial adipose tissue expresses a pathogenic profile of adipocytokines in patients with cardiovascular disease. Cardiovasc. Diabetol. 2006, 5, 1. [Google Scholar] [CrossRef] [PubMed]
  122. Sacks, H.S.; Fain, J.N. Human epicardial adipose tissue: A review. Am. Heart J. 2007, 153, 907–917. [Google Scholar] [CrossRef] [PubMed]
  123. Turkmen, K.; Ozbek, O.; Kayikcioğlu, H.; Kayrak, M.; Solak, Y.; Nayman, A.; Anil, M.; Babur, H.; Tonbul, H.Z. The Relationship between Epicardial Adipose Tissue and Coronary Artery Calcification in Peritoneal Dialysis Patients. Cardiorenal. Med. 2012, 2, 43–51. [Google Scholar] [CrossRef] [PubMed]
  124. Atakan, A.; Macunluoglu, B.; Kaya, Y.; Ari, E.; Demir, H.; Asicioglu, E.; Kaspar, C. Epicardial fat thickness is associated with impaired coronary flow reserve in hemodialysis patients. Hemodial. Int. 2014, 18, 62–69. [Google Scholar] [CrossRef] [PubMed]
  125. Gokbel, H.; Turk, S.; Okudan, N.; Atalay, H.; Belviranli, M.; Gaipov, A.; Solak, Y. Effects of Coenzyme Q10 Supplementation on Exercise Performance and Markers of Oxidative Stress in Hemodialysis Patients: A Double-Blind Placebo-Controlled Crossover Trial. Am. J. Ther. 2016, 23, e1736–e1743. [Google Scholar] [CrossRef] [PubMed]
  126. Reed, L.J.; debusk, B.G.; Gunsalus, I.; Hornberger, C., Jr. Crystalline α-lipoic acid; A catalytic agent associated with pyruvate dehydrogenase. Science 1951, 114, 93–94. [Google Scholar] [CrossRef] [PubMed]
  127. Shay, K.P.; Moreau, R.F.; Smith, E.J.; Smith, A.R.; Hagen, T.M. α-lipoic acid as a dietary supplement: Molecular mechanisms and therapeutic potential. Biochim. Biophys. Acta 2009, 1790, 1149–1160. [Google Scholar] [CrossRef] [PubMed]
  128. Sigel, H.; Prijs, B.; McCormick, D.B.; Shih, J.C. Stability of binary and ternary complexes of a-lipoate and lipoate derivatives with Mn2+, Cu2+, and Zn2+ in solution. Arch. Biochem. Biophys. 1978, 187, 208–214. [Google Scholar] [CrossRef]
  129. Liu, J. The effects and mechanisms of mitochondrial nutrient α-lipoic acid on improving age-associated mitochondrial and cognitive dysfunction: An overview. Neurochem. Res. 2008, 33, 194–203. [Google Scholar] [CrossRef] [PubMed]
  130. Packer, L. α-Lipoic acid: A metabolic antioxidant which regulates NF-κB signal transduction and protects against oxidative injury. Drug Metab. Rev. 1998, 30, 245–275. [Google Scholar] [CrossRef] [PubMed]
  131. Petersen Shay, K.; Moreau, R.F.; Smith, E.J.; Hagen, T.M. Is α-lipoic acid a scavenger of reactive oxygen species in vivo? Evidence for its initiation of stress signaling pathways that promote endogenous antioxidant capacity. IUBMB Life 2008, 60, 362–367. [Google Scholar] [CrossRef] [PubMed]
  132. Kim, H.; Kim, H.J.; Lee, K.; Kim, J.M.; Kim, H.S.; Kim, J.R.; Ha, C.M.; Choi, Y.K.; Lee, S.J.; Kim, J.Y.; Harris, R.A.; Jeong, D.; Lee, I.K. α-Lipoic acid attenuates vascular calcification via reversal of mitochondrial function and restoration of Gas6/Axl/Akt survival pathway. J. Cell. Mol. Med. 2012, 16, 273–286. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, L.; Wu, C.G.; Fang, C.Q.; Gao, J.; Liu, Y.Z.; Chen, Y.; Chen, Y.N.; Xu, Z.G. The protective effect of α-Lipoic acid on mitochondria in the kidney of diabetic rats. Int. J. Clin. Exp. Med. 2013, 6, 90–97. [Google Scholar] [PubMed]
  134. Melhem, M.F.; Craven, P.A.; Liachenko, J.; De Rubertis, F.R. α-lipoic acid attenuates hyperglycemia and prevents glomerular mesangial matrix expansion in diabetes. J. Am. Soc. Nephrol. 2002, 13, 16. [Google Scholar]
  135. Khabbazi, T.; Mahdavi, R.; Safa, J.; Pour-Abdollahi, P. Effects of α-lipoic acid supplementation on inflammation, oxidative stress, and serum lipid profile levels in patients with end-stage renal disease on hemodialysis. J. Ren. Nutr. 2012, 22, 244–250. [Google Scholar] [CrossRef] [PubMed]
  136. Chang, J.W.; Lee, E.K.; Kim, T.H.; Min, W.K.; Chun, S.; Lee, K.U.; Kim, S.B.; Park, J.S. Effects of α lipoic acid on the plasma levels of asymmetric dimethylarginine in diabetic end-stage renal disease patients on hemodialysis: A pilot study. Am. J. Nephrol. 2007, 27, 70–74. [Google Scholar] [CrossRef] [PubMed]
  137. Safa, J.; Ardalan, M.R.; Rezazadehsaatlou, M.; Mesgari, M.; Mahdavi, R.; Jadid, M.P. Effects of α lipoic acid supplementation on serum levels of IL-8 and TNF-α in patient with ESRD undergoing hemodialysis. Int. Urol. Nephrol. 2014, 46, 1633–1638. [Google Scholar] [CrossRef] [PubMed]
  138. Ramos, L.F.; Kane, J.; McMonagle, E.; Le, P.; Wu, P.; Shintani, A. Effects of combination tocopherols and α lipoic acid therapy on oxidative stress and inflammatory biomarkers in chronic kidney disease. J. Ren. Nutr. 2011, 21, 211–218. [Google Scholar] [CrossRef] [PubMed]
  139. Himmelfarb, J.; Ikizler, T.A.; Ellis, C.; Wu, P.; Shintani, A.; Dalal, S.; Kaplan, M.; Chonchol, M.; Hakim, R.M. Provision of antioxidant therapy in hemodialysis (PATH): A randomized clinical trial. J. Am. Soc. Nephrol. 2014, 25, 623–633. [Google Scholar] [CrossRef]
  140. Tinggi, U. Selenium: Its role as antioxidant in human health. Environ. Health Prev. Med. 2008, 13, 102–108. [Google Scholar] [CrossRef]
  141. Rayman, M.P. Selenium and human health. Lancet 2012, 379, 1256. [Google Scholar] [CrossRef]
  142. Fujishima, Y.; Ohsawa, M.; Itai, K.; Kato, K.; Tanno, K.; Turin, T.C.; Onoda, T.; Endo, S.; Okayama, A.; Fujioka, T. Serum selenium levels are inversely associated with death risk among hemodialysis patients. Nephrol. Dial. Transplant. 2011, 26, 3331–3338. [Google Scholar] [CrossRef] [PubMed]
  143. Rucker, D.; Thadhani, R.; Tonelli, M. Trace element status in hemodialysis patients. Semin. Dial. 2010, 23, 389–395. [Google Scholar] [CrossRef] [PubMed]
  144. Tonelli, M.; Wiebe, N.; Hemmelgarn, B.; Klarenbach, S.; Field, C.; Manns, B.; Thadhani, R.; Gill, J. Alberta Kidney Disease Network. Trace elements in hemodialysis patients: A systematic review and meta-analysis. BMC Med. 2009, 7, 25. [Google Scholar] [CrossRef] [PubMed]
  145. Pakfetrat, M.; Malekmakan, L.; Hasheminasab, M. Diminished selenium levels in hemodialysis and continuous ambulatory peritoneal dialysis patients. Biol. Trace Elem. Res. 2010, 137, 335–339. [Google Scholar] [CrossRef] [PubMed]
  146. Sedighi, O.; Zargari, M.; Varshi, G. Effect of selenium supplementation on glutathione peroxidase enzyme activity in patients with chronic kidney disease: A randomized clinical trial. Nephrourol. Mon. 2014, 6, e17945. [Google Scholar] [CrossRef] [PubMed]
  147. Zachara, B.A.; Koterska, D.; Manitius, J.; Sadowski, L.; Dziedziczko, A.; Salak, A.; Wasowicz, W. Selenium supplementation on plasma glutathione peroxidase activity in patients with end-stage chronic renal failure. Biol. Trace Elem Res. 2004, 97, 15–30. [Google Scholar] [CrossRef]
  148. Zachara, B.A.; Gromadzinska, J.; Palus, J.; Zbrog, Z.; Swiech, R.; Twardowska, E.; Wasowicz, W. The effect of selenium supplementation in the prevention of DNA damage in white blood cells of hemodialyzed patients: A pilot study. Biol. Trace Elem. Res. 2011, 142, 274–283. [Google Scholar] [CrossRef] [PubMed]
  149. Salehi, M.; Sohrabi, Z.; Ekramzadeh, M.; Fallahzadeh, M.K.; Ayatollahi, M.; Geramizadeh, B.; Hassanzadeh, J.; Sagheb, M.M. Selenium supplementation improves the nutritional status of hemodialysis patients: A randomized, double-blind, placebo-controlled trial. Nephrol. Dial. Transplant. 2013, 28, 716–723. [Google Scholar] [CrossRef] [PubMed]
  150. Omrani, H.R.; Rahimi, M.; Nikseresht, K. The effect of selenium supplementation on acute phase reactants and thyroid function tests in hemodialysis patients. Nephrourol. Mon. 2015, 7, e24781. [Google Scholar] [CrossRef] [PubMed]
  151. Ardalan, M.R.; Tubbs, R.S.; Shoja, M.M. Vitamin E and selenium co-supplementation attenuates oxidative stress in haemodialysis patients receiving intra-dialysis iron infusion. Nephrol. Dial. Transplant. 2007, 22, 973–975. [Google Scholar] [CrossRef] [PubMed]
  152. Baptista, J.A.B.; Tavares, J.F.D.; Carvalho, R.C.B. Comparison of catechins and aromas among different green teas using HPLC/SPME-GC. Food Res. Int. 1998, 31, 729–736. [Google Scholar] [CrossRef]
  153. Yokozawa, T.; Dong, E.; Nakagawa Kashiwagi, H.; Nakagawa, H.; Takeuchi, S.; Chung, H.Y. In vitro and in vivo studies on the radical-scavenging activity of tea. J. Agric. Food Chem. 1998, 46, 2143–2150. [Google Scholar] [CrossRef]
  154. Nanjo, F.; Mori, M.; Goto, K.; Hara, Y. Radical scavenging activity of tea catechins and their related compounds. Biosci. Biotechnol. Biochem. 1999, 63, 1621–1623. [Google Scholar] [CrossRef] [PubMed]
  155. Zhao, B.; Guo, Q.; Xin, W. Free radical scavenging by green tea polyphenols. Methods Enzymolol. 2001, 335, 217–231. [Google Scholar]
  156. Higdon, J.V.; Frei, B. Tea catechins and polyphenols: Health effects, metabolism, and antioxidant functions. Crit. Rev. Food Sci. Nutr. 2003, 43, 89–143. [Google Scholar] [CrossRef] [PubMed]
  157. Bao, H.; Peng, A. The Green Tea Polyphenol(−)-epigallocatechin-3-gallate and its beneficial roles in chronic kidney disease. J. Transl. Int. Med. 2016, 4, 99–103. [Google Scholar] [CrossRef] [PubMed]
  158. Peng, A.; Ye, T.; Rakheja, D.; Tu, Y.; Wang, T.; Du, Y.; Zhou, J.K.; Vaziri, N.D.; Hu, Z.; Mohan, C.; Zhou, X.J. The green tea polyphenol (−)-epigallocatechin-3-gallate ameliorates experimental immune-mediated glomerulonephritis. Kidney Int. 2011, 80, 601–611. [Google Scholar] [CrossRef] [PubMed]
  159. Ye, T.; Zhen, J.; Du, Y.; Zhou, J.K.; Peng, A.; Vaziri, N.D.; Mohan, C.; Xu, Y.; Zhou, X.J. Green tea polyphenol (−)-epigallocatechin-3-gallate restores Nrf2 activity and ameliorates crescentic glomerulonephritis. PLoS ONE 2015, 10, e0119543. [Google Scholar] [CrossRef] [PubMed]
  160. 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]
  161. Yamabe, N.; Yokozawa, T.; Oya, T.; Kim, M. Therapeutic potential of (−)-epigallocatechin 3-O-gallate on renal damage in diabetic nephropathy model rats. J. Pharmacol. Exp. Ther. 2006, 319, 228–236. [Google Scholar] [CrossRef] [PubMed]
  162. Zhou, P.; Yu, J.F.; Zhao, C.G.; Sui, F.X.; Teng, X.; Wu, Y.B. Therapeutic potential of EGCG on acute renal damage in a rat model of obstructive nephropathy. Mol. Med. Rep. 2013, 7, 1096–1102. [Google Scholar] [PubMed]
  163. Nakagawa, T.; Yokozawa, T.; Sano, M.; Takeuchi, S.; Kim, M.; Minamoto, S. Activity of (−)-epigallocatechin 3-O-gallate against oxidative stress in rats with adenine-induced renal failure. J. Agric. Food Chem. 2004, 52, 2103–2107. [Google Scholar] [CrossRef] [PubMed]
  164. Kakuta, Y.; Okumi, M.; Isaka, Y.; Tsutahara, K.; Abe, T.; Yazawa, K.; Ichimaru, N.; Matsumura, K.; Hyon, S.H.; Takahara, S.; Nonomura, N. Epigallocatechin-3-gallate protects kidneys from ischemia reperfusion injury by HO-1 upregulation and inhibition of macrophage infiltration. Transpl. Int. 2011, 24, 514–522. [Google Scholar] [CrossRef] [PubMed]
  165. Bertelli, A.A.; Das, D.K. Grapes, wines, resveratrol, and heart health. J. Cardiovasc. Pharmacol. 2009, 54, 468–476. [Google Scholar] [CrossRef] [PubMed]
  166. 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]
  167. Spanier, G.; Xu, H.; Xia, N.; Tobias, S.; Deng, S.; Wojnowski, L.; Forstermann, U.; Li, H. Resveratrol reduces endothelial oxidative stress by modulating the gene expression of superoxide dismutase 1 (SOD1), glutathione peroxidase 1 (GPx1) and NADPH oxidase subunit (Nox4). J. Physiol. Pharmacol. 2009, 60, 111–116. [Google Scholar] [PubMed]
  168. Hao, C.M.; Haase, V.H. Sirtuins and their relevance to the kidney. J. Am. Soc. Nephrol. 2010, 21, 1620–1627. [Google Scholar] [CrossRef] [PubMed]
  169. Imai, S.; Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014, 24, 464–471. [Google Scholar] [CrossRef] [PubMed]
  170. Rodgers, J.T.; Lerin, C.; Haas, W.; Gygi, S.P.; Spiegelman, B.M.; Puigserver, P. Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Nature 2005, 434, 113–118. [Google Scholar] [CrossRef] [PubMed]
  171. Walker, A.K.; Yang, F.; Jiang, K.; Ji, J.Y.; Watts, J.L.; Purushotham, A.; Boss, O.; Hirsch, M.L.; Ribich, S.; Smith, J.J.; et al. Conserved role of SIRT1 orthologs in fasting-dependent inhibition of the lipid/cholesterol regulator SREBP. Genes Dev. 2010, 24, 1403–1417. [Google Scholar] [CrossRef] [PubMed]
  172. Kitada, M.; Kume, S.; Takeda-Watanabe, A.; Kanasaki, K.; Koya, D. Sirtuins and renal diseases: Relationship with aging and diabetic nephropathy. Clin. Sci. 2013, 124, 153–164. [Google Scholar] [CrossRef] [PubMed]
  173. Kitada, M.; Koya, D. Renal protective effects of resveratrol. Oxid. Med. Cell. Longev. 2013, 2013, 568093. [Google Scholar] [CrossRef] [PubMed]
  174. 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]
  175. 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] [CrossRef] [PubMed]
  176. 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-κB/NADPH oxidase/ROS pathway, which is inhibited by resveratrol. Int. J. Biochem. Cell Biol. 2012, 44, 629–638. [Google Scholar] [CrossRef] [PubMed]
  177. Huang, S.S.; Ding, D.F.; Chen, S.; Dong, C.L.; Ye, X.L.; Yuan, Y.G.; Feng, Y.M.; You, N.; Xu, J.R.; Miao, H.; et al. Resveratrol protects podocytes against apoptosis via stimulation of autophagy in a mouse model of diabetic nephropathy. Sci. Rep. 2017, 7, 45692. [Google Scholar] [CrossRef] [PubMed]
  178. 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]
  179. 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]
  180. He, T.; Guan, X.; Wang, S.; Xiao, T.; Yang, K.; Xu, X.; Wang, J.; Zhao, J. Resveratrol prevents high glucose-induced epithelial-mesenchymal transition in renal tubular epithelial cells by inhibiting NADPH oxidase/ROS/ERK pathway. Mol. Cell Endocrinol. 2015, 402, 13–20. [Google Scholar] [CrossRef] [PubMed]
  181. Huang, X.Z.; Wen, D.; Zhang, M.; Xie, Q.; Ma, L.; Guan, Y.; Ren, Y.; Chen, J.; Hao, C.M. Sirt1 activation ameliorates renal fibrosis by inhibiting the TGF-β/Smad3 pathway. J. Cell. Biochem. 2014, 115, 996–1005. [Google Scholar] [CrossRef] [PubMed]
  182. Sun, L.J.; Sun, Y.N.; Chen, S.J.; Liu, S.; Jiang, G.R. Resveratrol attenuates skeletal muscle atrophy induced by chronic kidney disease via MuRF1 signaling pathway. Biochem. Biophys. Res. Commun. 2017, 487, 83–89. [Google Scholar] [CrossRef] [PubMed]
  183. Um, J.H.; Park, S.J.; Kang, H.; Yang, S.; Foretz, M.; McBurney, M.W.; Kim, M.K.; Viollet, B.; Chung, J.H. AMP-activated protein kinase-deficient mice are resistant to the metabolic effects of resveratrol. Diabetes 2010, 59, 554–563. [Google Scholar] [CrossRef] [PubMed]
  184. Yuan, Y.; Huang, S.; Wang, W.; Wang, Y.; Zhang, P.; Zhu, C. Activation of peroxisome proliferator-activated receptor-γ coactivator 1α ameliorates mitochondrial dysfunction and protects podocytes from aldosterone-induced injury. Kidney Int. 2012, 82, 771–789. [Google Scholar] [CrossRef] [PubMed]
  185. Do Amaral, C.L.; Francescato, H.D.; Coimbra, T.M.; Costa, R.S.; Darin, J.D.; Antunes, L.M.; Bianchi, M.L. Resveratrol attenuates cisplatin-induced nephrotoxicity in rats. Arch. Toxicol. 2008, 82, 363–370. [Google Scholar] [CrossRef] [PubMed]
  186. Bertelli, A.A.; Migliori, M.; Panichi, V.; Origlia, N.; Filippi, C.; Das, D.K.; Giovannini, L. Resveratrol, a component of wine and grapes, in the prevention of kidney disease. Ann. N. Y. Acad. Sci. 2002, 957, 230–238. [Google Scholar] [CrossRef] [PubMed]
  187. Chen, L.; Yang, S.; Zumbrun, E.E.; Guan, H.; Nagarkatti, P.S.; Nagarkatti, M. Resveratrol attenuates lipopolysaccharide-induced acute kidney injury by suppressing inflammation driven by macrophages. Mol. Nutr. Food Res. 2015, 59, 853–864. [Google Scholar] [CrossRef] [PubMed]
  188. Smoliga, J.M.; Blanchard, O. Enhancing the delivery of resveratrol in humans: If low bioavailability is the problem, what is the solution? Molecules 2014, 19, 17154–17172. [Google Scholar] [CrossRef] [PubMed]
  189. Trujillo, J.; Chirino, Y.I.; Molina-Jijón, E.; Andérica-Romero, A.C.; Tapia, E.; Pedraza-Chaverrí, J. Renoprotective effect of the antioxidant curcumin: Recent findings. Redox Biol. 2013, 1, 448–456. [Google Scholar] [CrossRef] [PubMed]
  190. Singh, S. From exotic spice to modern drug? Cell 2007, 130, 765–768. [Google Scholar] [CrossRef] [PubMed]
  191. Calabrese, V.; Bates, T.E.; Mancuso, C.; Cornelius, C.; Ventimiglia, B.; Cambria, M.T. Curcumin and the cellular stress response in free radical-related diseases. Mol. Nutr. Food Res. 2008, 52, 1062–1073. [Google Scholar] [CrossRef] [PubMed]
  192. 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]
  193. Fujisawa, S.; Atsumi, T.; Ishihara, M.; Kadoma, Y. Cytotoxicity, ROS-generation activity and radical-scavenging activity of curcumin and related compounds. Anticancer Res. 2004, 24, 563–569. [Google Scholar] [PubMed]
  194. 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.; Kwon, D.Y. Synergistic antibacterial effect of curcumin against methicillin-resistant Staphylococcus aureus. Phytomedicine 2013, 20, 714–718. [Google Scholar] [CrossRef] [PubMed]
  195. Ak, T.; Gülçin, I. Antioxidant and radical scavenging properties of curcumin. Chem. Biol. Interact. 2008, 174, 27–37. [Google Scholar] [CrossRef] [PubMed]
  196. Barzegar, A.; Moosavi-Movahedi, A.A. Intracellular ROS protection efficiency and free radical-scavenging activity of curcumin. PLoS ONE 2011, 6, e26012. [Google Scholar] [CrossRef] [PubMed]
  197. Das, K.C.; Das, C.K. Curcumin (diferuloylmethane), a singlet oxygen ((1)O(2)) quencher. Biochem. Biophys. Res. Commun. 2002, 295, 62–66. [Google Scholar] [CrossRef]
  198. Kim, J.E.; Kim, A.R.; Chung, H.Y.; Han, S.Y.; Kim, B.S.; Choi, J.S. In vitro peroxynitrite scavenging activity of diarylheptanoids from Curcuma longa. Phytother. Res. 2003, 17, 481–484. [Google Scholar] [CrossRef] [PubMed]
  199. Aggarwal, B.B.; Sung, B. Pharmacological basis for the role of curcumin in chronic diseases: An age-old spice with modern targets. Trends Pharmacol. Sci. 2009, 30, 85–94. [Google Scholar] [CrossRef] [PubMed]
  200. Jeong, G.S.; Oh, G.S.; Pae, H.O.; Jeong, S.O.; Kim, Y.C.; Shin, M.K.; Seo, B.Y.; Han, S.Y.; Lee, H.S.; Jeong, J.G.; et al. Comparative effects of curcuminoids on endothelial heme oxygenase-1 expression: Ortho-methoxy groups are essential to enhance heme oxygenase activity and protection. Exp. Mol. Med. 2006, 38, 393–400. [Google Scholar] [CrossRef] [PubMed]
  201. Ye, S.F.; Hou, Z.Q.; Zhong, L.M.; Zhang, Q.Q. Effect of curcumin on the induction of glutathione S-transferases and NADP(H):quinone oxidoreductase and its possible mechanism of action. Yao Xue Xue Bao 2007, 42, 376–380. [Google Scholar] [PubMed]
  202. Rushworth, S.A.; Ogborne, R.M.; Charalambos, C.A.; O’Connell, M.A. Role of protein kinase C delta in curcumin-induced antioxidant response element-mediated gene expression in human monocytes. Biochem. Biophys. Res. Commun. 2006, 341, 1007–1016. [Google Scholar] [CrossRef] [PubMed]
  203. 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]
  204. Soetikno, V.; Watanabe, K.; Sari, F.R.; Harima, M.; Thandavarayan, R.A.; Veeraveedu, P.T.; Arozal, W.; Sukumaran, V.; Lakshmanan, A.P.; Arumugam, S.; Suzuki, K. Curcumin attenuates diabetic nephropathy by inhibiting PKC-α and PKC-β1 activity in streptozotocin-induced type I diabetic rats. Mol. Nutr. Food Res. 2011, 55, 1655–1665. [Google Scholar] [CrossRef] [PubMed]
  205. 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-κB activation and proinflammatory cytokines in streptozotocin induced-diabetic nephropathy. Nutr. Metab. 2011, 8, 35. [Google Scholar] [CrossRef] [PubMed]
  206. Chiu, J.; Khan, Z.A.; Farhangkhoee, H.; Chakrabarti, S. Curcumin prevents diabetes-associated abnormalities in the kidneys by inhibiting p300 and nuclear factor-κB. Nutrition 2009, 25, 964–972. [Google Scholar] [CrossRef] [PubMed]
  207. Lu, M.; Yin, N.; Liu, W.; Cui, X.; Chen, S.; Wang, E. Curcumin Ameliorates Diabetic Nephropathy by Suppressing NLRP3 Inflammasome Signaling. BioMed Res. Int. 2017, 2017, 1516985. [Google Scholar] [CrossRef] [PubMed]
  208. 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. Ren. Physiol. 2009, 296, F1146–F1157. [Google Scholar] [CrossRef] [PubMed]
  209. Ghosh, S.S.; Gehr, T.W.; Ghosh, S. Curcumin and chronic kidney disease (CKD): Major mode of action through stimulating endogenous intestinal alkaline phosphatase. Molecules 2014, 19, 20139–20156. [Google Scholar] [CrossRef] [PubMed]
  210. Jacob, A.; Chaves, L.; Eadon, M.T.; Chang, A.; Quigg, R.J.; Alexander, J.J. Curcumin alleviates immune-complex-mediated glomerulonephritis in factor-H-deficient mice. Immunology 2013, 139, 328–337. [Google Scholar] [CrossRef] [PubMed]
  211. Tapia, E.; Soto, V.; Ortiz-Vega, K.M.; Zarco-Márquez, G.; Molina-Jijón, E.; Cristóbal-García, M.; Santamaría, J.; García-Niño, 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] [CrossRef] [PubMed]
  212. 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]
  213. Tapia, E.; Zatarain-Barrón, Z.L.; Hernández-Pando, R.; Zarco-Márquez, G.; Molina-Jijón, E.; Cristóbal-García, M.; Santamaría, 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]
  214. Aparicio-Trejo, O.E.; Tapia, E.; Molina-Jijón, E.; Medina-Campos, O.N.; Macías-Ruvalcaba, N.A.; León-Contreras, J.C.; Hernández-Pando, R.; García-Arroyo, F.E.; Cristóbal, M.; Sánchez-Lozada, L.G.; et al. Curcumin prevents mitochondrial dynamics disturbances in early 5/6 nephrectomy: Relation to oxidative stress and mitochondrial bioenergetics. Biofactors 2017, 43, 293–310. [Google Scholar] [CrossRef] [PubMed]
  215. Bugyei-Twum, A.; Abadeh, A.; Thai, K.; Zhang, Y.; Mitchell, M.; Kabir, G.; Connelly, K.A. Suppression of NLRP3 Inflammasome Activation Ameliorates Chronic Kidney Disease-Induced Cardiac Fibrosis and Diastolic Dysfunction. Sci. Rep. 2016, 6, 39551. [Google Scholar] [CrossRef] [PubMed]
  216. Correa, F.; Buelna-Chontal, M.; Hernández-Reséndiz, S.; García-Niño, W.R.; Roldán, F.J.; Soto, V.; Silva-Palacios, A.; Amador, A.; Pedraza-Chaverrí, J.; Tapia, E.; et al. Curcumin maintains cardiac and mitochondrial function in chronic kidney disease. Free Radic. Biol. Med. 2013, 61, 119–129. [Google Scholar] [CrossRef] [PubMed]
  217. Hernández-Reséndiz, S.; Correa, F.; García-Niño, W.R.; Buelna-Chontal, M.; Roldán, F.J.; Ramírez-Camacho, I.; Delgado-Toral, C.; Carbó, R.; Pedraza-Chaverrí, J.; Tapia, E.; Zazueta, C. Cardioprotection by curcumin post-treatment in rats with established chronic kidney disease. Cardiovasc. Drugs Ther. 2015, 29, 111–120. [Google Scholar] [CrossRef] [PubMed]
  218. 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]
  219. Khajehdehi, P.; Zanjaninejad, B.; Aflaki, E.; Nazarinia, M.; Azad, F.; Malekmakan, L. Oral supplementation of turmeric decreases proteinuria, hematuria, and systolic blood pressure in patients suffering from relapsing or refractory lupus nephritis: A randomized and placebo-controlled study. J. Ren. Nutr. 2012, 22, 50–57. [Google Scholar] [CrossRef] [PubMed]
  220. Sprecher, H. The roles of anabolic and catabolic reactions in the synthesis and recycling of polyunsaturated fatty acids. Prostaglandins Leukot. Essent. Fat. Acids 2002, 67, 79–83. [Google Scholar] [CrossRef] [PubMed]
  221. Hassan, K.S.; Hassan, S.K.; Hijazi, E.G.; Khazim, K.O. Effects of omega-3 on lipid profile and inflammation markers in peritoneal dialysis patients. Ren. Fail. 2010, 32, 1031–1035. [Google Scholar] [CrossRef] [PubMed]
  222. Shing, C.M.; Adams, M.J.; Fassett, R.G.; Coombes, J.S. Nutritional compounds influence tissue factor expression and inflammation of chronic kidney disease patients in vitro. Nutrition 2011, 27, 967–972. [Google Scholar] [CrossRef] [PubMed]
  223. Novak, T.E.; Babcock, T.A.; Jho, D.H.; Helton, W.S.; Espat, N.J. NF-κB inhibition by omega-3 fatty acids modulates LPS-stimulated macrophage TNF-α transcription. Am. J. Physiol. Lung Cell Mol. Physiol. 2003, 284, L84–L89. [Google Scholar] [CrossRef] [PubMed]
  224. De Caterina, R.; Massaro, M. Omega-3 fatty acids and the regulation of expression of endothelial pro-atherogenic and pro-inflammatory genes. J. Membr. Biol. 2005, 206, 103–116. [Google Scholar] [CrossRef] [PubMed]
  225. Corey, E.J.; Shih, C.; Cashman, J.R. Docosahexaenoic acid is a strong inhibitor of prostaglandin but not leukotriene biosynthesis. Proc. Natl. Acad. Sci. USA 1983, 80, 3581–3584. [Google Scholar] [CrossRef] [PubMed]
  226. Needleman, P.; Raz, A.; Minkes, M.S.; Ferrendelli, J.A.; Sprecher, H. Triene prostaglandins: Prostacyclin and thromboxane biosynthesis and unique biological properties. Proc. Natl. Acad. Sci. USA 1979, 76, 944–948. [Google Scholar] [CrossRef] [PubMed]
  227. Seki, H.; Tani, Y.; Arita, M. Omega-3 PUFA derived anti-inflammatory lipid mediator resolvin E1. Prostaglandins Other Lipid Mediat. 2009, 89, 126–130. [Google Scholar] [CrossRef] [PubMed]
  228. Wiktorowska-Owczarek, A.; Berezińska, M.; Nowak, J.Z. PUFAs: Structures, Metabolism and Functions. Adv. Clin. Exp. Med. 2015, 24, 931–941. [Google Scholar] [CrossRef] [PubMed]
  229. Surette, M.E. The science behind dietary omega-3 fatty acids. CMA J. 2008, 178, 177–180. [Google Scholar] [CrossRef] [PubMed]
  230. Calder, P.C.; Yaqoob, P.; Thies, F.; Wallace, F.A.; Miles, E.A. Fatty acids and lymphocyte functions. Br. J. Nutr. 2002, 87, S31–S48. [Google Scholar] [CrossRef] [PubMed]
  231. Vaughan, R.A.; Garcia-Smith, R.; Bisoffi, M.; Conn, C.A.; Trujillo, K.A. Conjugated linoleic acid or omega 3 fatty acids increase mitochondrial biosynthesis and metabolism in skeletal muscle cells. Lipids Health Dis. 2012, 11, 142. [Google Scholar] [CrossRef] [PubMed]
  232. Arab, K.; Rossary, A.; Flourié, F.; Tourneur, Y.; Steghens, J.P. Docosahexaenoic acid enhances the antioxidant response of human fibroblasts by upregulating gamma-glutamyl-cysteinyl ligase and glutathione reductase. Br. J. Nutr. 2006, 95, 18–26. [Google Scholar] [CrossRef] [PubMed]
  233. Kim, Y.J.; Chung, H.Y. Antioxidative and anti-inflammatory actions of docosahexaenoic acid and eicosapentaenoic acid in renal epithelial cells and macrophages. J. Med. Food 2007, 10, 225–231. [Google Scholar] [CrossRef] [PubMed]
  234. Peake, J.M.; Gobe, G.C.; Fassett, R.G.; Coombes, J.S. The effects of dietary fish oil on inflammation, fibrosis and oxidative stress associated with obstructive renal injury in rats. Mol. Nutr. Food Res. 2011, 55, 400–410. [Google Scholar] [CrossRef] [PubMed]
  235. An, W.S.; Kim, H.J.; Cho, K.H.; Vaziri, N.D. Omega-3 fatty acid supplementation attenuates oxidative stress, inflammation, and tubulointerstitial fibrosis in the remnant kidney. Am. J. Physiol. Ren. Physiol. 2009, 297, F895–F903. [Google Scholar] [CrossRef] [PubMed]
  236. de Mattos, A.M.; da Costa, J.A.C.; Jordão Júnior, A.A.; Chiarello, P.G. Omega-3 Fatty Acid Supplementation is Associated with Oxidative Stress and Dyslipidemia, but Does not Contribute to Better Lipid and Oxidative Status on Hemodialysis Patients. J. Ren. Nutr. 2017. [Google Scholar] [CrossRef] [PubMed]
  237. Mas, E.; Barden, A.; Burke, V.; Beilin, L.J.; Watts, G.F.; Huang, R.C.; Puddey, I.B.; Irish, A.B.; Mori, T.A. A randomized controlled trial of the effects of n-3 fatty acids on resolvins in chronic kidney disease. Clin. Nutr. 2016, 35, 331–336. [Google Scholar] [CrossRef] [PubMed]
  238. Hung, A.M.; Booker, C.; Ellis, C.D.; Siew, E.D.; Graves, A.J.; Shintani, A.; Abumrad, N.N.; Himmelfarb, J.; Ikizler, T.A. Omega-3 fatty acids inhibit the up-regulation of endothelial chemokines in maintenance hemodialysis patients. Nephrol. Dial. Transplant. 2015, 30, 266–274. [Google Scholar] [CrossRef] [PubMed]
  239. Panahi, Y.; Dashti-Khavidaki, S.; Farnood, F.; Noshad, H.; Lotfi, M.; Gharekhani, A. Therapeutic Effects of Omega-3 Fatty Acids on Chronic Kidney Disease-Associated Pruritus: A Literature Review. Adv. Pharm. Bull. 2016, 6, 509–514. [Google Scholar] [CrossRef] [PubMed]
  240. Mori, T.A.; Burke, V.; Puddey, I.; Irish, A.; Cowpland, C.A.; Beilin, L.; Dogra, G.; Watts, G.F. The effects of omega-3 fatty acids and coenzyme Q10 on blood pressure and heart rate in chronic kidney disease: A randomized controlled trial. J. Hypertens. 2009, 27, 1863–1872. [Google Scholar] [CrossRef] [PubMed]
  241. Barden, A.E.; Burke, V.; Mas, E.; Beilin, L.J.; Puddey, I.B.; Watts, G.F.; Irish, A.B.; Mori, T.A. n-3 fatty acids reduce plasma 20-hydroxyeicosatetraenoic acid and blood pressure in patients with chronic kidney disease. J. Hypertens. 2015, 33, 1947–1953. [Google Scholar] [CrossRef] [PubMed]
  242. Ferraro, P.M.; Ferraccioli, G.F.; Gambaro, G.; Fulignati, P.; Costanzi, S. Combined treatment with renin-angiotensin system blockers and polyunsaturated fatty acids in proteinuric IgA nephropathy: A randomized controlled trial. Nephrol. Dial. Transplant. 2009, 24, 156–160. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Signorini, L.; Granata, S.; Lupo, A.; Zaza, G. Naturally Occurring Compounds: New Potential Weapons against Oxidative Stress in Chronic Kidney Disease. Int. J. Mol. Sci. 2017, 18, 1481. https://doi.org/10.3390/ijms18071481

AMA Style

Signorini L, Granata S, Lupo A, Zaza G. Naturally Occurring Compounds: New Potential Weapons against Oxidative Stress in Chronic Kidney Disease. International Journal of Molecular Sciences. 2017; 18(7):1481. https://doi.org/10.3390/ijms18071481

Chicago/Turabian Style

Signorini, Lorenzo, Simona Granata, Antonio Lupo, and Gianluigi Zaza. 2017. "Naturally Occurring Compounds: New Potential Weapons against Oxidative Stress in Chronic Kidney Disease" International Journal of Molecular Sciences 18, no. 7: 1481. https://doi.org/10.3390/ijms18071481

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop