Skip to main content
Oxidative Medicine and Cellular Longevity logoLink to Oxidative Medicine and Cellular Longevity
. 2010 Mar-Apr;3(2):86–100. doi: 10.4161/oxim.3.2.3

Multifaceted approach to resveratrol bioactivity

Focus on antioxidant action, cell signaling and safety

Peter Kovacic 1,, Ratnasamy Somanathan 2
PMCID: PMC2952093  PMID: 20716933

Abstract

Resveratrol (RVT) is a naturally occurring trihydroxy stilbene that displays a wide spectrum of physiological activity. Its ability to behave therapeutically as a component of red wine has attracted wide attention. The phenol acts as a protective agent involving various body constituents. Most attention has been given to beneficial effects in insults involving cancer, aging, cardiovascular system, inflammation and the central nervous system. One of the principal modes of action appears to be as antioxidant. Other mechanistic pathways entail cell signaling, apoptosis and gene expression. There is an intriguing dichotomy in relation to pro-oxidant property. Also discussed are metabolism, receptor binding, rationale for safety and suggestions for future work. This is the first comprehensive review of RVT based on a broad, unifying mechanism.

Key words: resveratrol, bioactivity, mechanism, antioxidant, metabolism, cell signaling, safety

Introduction

Resveratrol 1 (Fig. 1) (RVT) is a naturally occurring stilbene derivative that possesses three phenolic groups, two of which are part of a resorcinol structure. Most work involves the naturally occurring trans isomer. It is present in various dietary sources, including grapes, peanuts, plums and many plants. Much recent attention was paid to the “French paradox” (see below) which provides evidence for the beneficial effects of red wine. Investigations pointed to RVT as the principal therapeutic agent. The literature contains voluminous reports on the wide variety of properties by the phenol. Most attention has been devoted to anticancer, anti-aging, anti-inflammatory, cardioprotection and countering of insults to the central nervous system. Articles document a large number of mechanisms involved in the biological activity. One of the most important is the antioxidant (AO) attribute. Considerable literature correlates the presence of reactive oxygen species (ROS) to oxidative stress (OS) leading to large numbers of adverse effects suffered by body constituents. Hence, it is reasonable to assign a widespread protective role to RVT as an AO. Other mechanisms in which the drug is involved include cell signaling, apoptosis and gene expression. The dichotomy of the AO RVT also exhibiting pro-oxidant action is addressed and rationalized. Another important aspect is the finding that RVT has been shown to produce no adverse effects, even when consumed in high concentrations.1 Hence, it exhibits good protection for use in therapy for various diseases.

Figure 1.

Figure 1

Resveratrol structure. The compound is a stilbene derivative containing three phenolic groups. Hence, it can exert AO action which evidently plays an important role in the protective effects observed against oxidative insult involving various body constituents. Metabolic studies show major formation of water-soluble conjugates. Many investigations deal with cell signaling pathways in the bioactivity. Particular focus has been devoted to antiaging, anti-cancer actions, cardio protection and prevention of CNS damage.

Reviews demonstrate the widespread involvement of ROS-OS in cancer and toxicity associated with many illnesses. The preponderance of bioactive substances and their metabolites incorporate ET functionalities, which, we believe, play an important role in physiological responses. The main groups include quinones (or phenolic precursors), metal complexes (or complexors), aromatic nitro compounds (or reduced hydroxylamine and nitroso derivatives) and conjugated imines or iminium species. In vivo redox cycling with oxygen can occur giving rise to OS through generation of ROS, such as hydrogen peroxide, hydroperoxides, alkyl peroxides, and diverse radicals (hydroxyl, alkoxyl, hydroperoxyl), and superoxide. In some cases, ET results in interference with normal electrical effects, e.g., in respiration or neurochemistry. Generally, active entities possessing ET groups display reduction potentials in the physiologically responsive range, i.e., more positive than −0.5 V ET, ROS and OS have been increasingly implicated in the mode of action of drugs and toxins (toxicants) e.g., antiinfective agents,2 anticancer drugs,3 carcinogens,4 reproductive toxins,5 nephrotoxins,6 hepatotoxins,7 cardiovascular toxins,8 nerve toxins,9 mitochondrial toxins,10 abused drugs,11 ototoxins,12 pulmonary toxins,13 immune system toxins,14 and various other categories of drugs and toxins, including human illnesses.15

There is a plethora of experimental evidence supporting the OS theoretical framework, including generation of the common ROS, lipid peroxidation, degradation products of oxidation, depletion of AOs, and DNA oxidation and cleavage products, as well as electrochemical data. Of particular relevance in the present case is the prevalent beneficial effect of AOs,16 in connection with RVT. This comprehensive, unifying mechanism is in keeping with the frequent observations that many ET substances display a variety of activities, e.g., multiple drug properties, as well as toxic effects. Knowledge of events at the molecular level can result in practical application in medicine.

The protective effects of RVT are demonstrated for cancer and insults to many body constituents, including the cardiovascular system, central nervous system (CNS), liver, kidney, DNA and others. There is a beneficial effect in connection with aging, arthritis and inflammation. The drug is a versatile agent that also functions through cell signaling and gene expression. Other aspects treated are metabolism, receptor interaction, electrochemistry, safety and pro-oxidant behavior. Suggestions for future work are offered.

This review demonstrates that the ET-ROS-OS-AO unifying theme, which has been successful for many other classes of drugs and toxins, can also be applied to RVT, and this comprises the first comprehensive, mechanistic review devoted to that agent. However, it should be emphasized that physiological activity of endogenous and exogenous substances is often complex and multifaceted. Our objective does not encompass extensive treatment of all other modes of action. The citations are usually representative, rather than exhaustive. A number of original references may be found in the reviews and articles cited.

Metabolism

A common metabolic route for phenols involves oxidation to catechols or hydroquinones with subsequent conversion to o- or p-quinones. An ensuing result is often redox cycling entailing oxygen with production of ROS. It is quite significant that finding comparable literature for phenolic RVT is difficult. A rationale is provided by various metabolic reports. Rapid metabolism leads to about 75% excretion in urine and feces in the form of RVT glucuronides and sulfates.17 The following conjugates are reported: 3-O-and 4′-O-glucuronides;18 most abundant, 3-O-glucuronide and 3-sulfate;19 main metabolites, monoglucuronide, dihydro monosulfate, monosulfate and dihydro RVT;20 3-sulfate, 4′-sulfate, 3,5-disulfate, 3,4′-disulfate, 3,4′,5-trisulfate, 3-glucuronide and aglycone.21 In a study with two cell lines, RVT-3sulfate was identified.22 The absence of toxic effects apparently results from preferential conjugation, rather than appreciable oxidation to ET quinones.

The parent RVT could act as an AO, similar to the familiar phenolic AO vitamin E. Also, the conjugates possessing free phenolic groups might exert AO effects provided contact is made with free radical species. However, conjugation with highly hydrophilic groups results in increased water solubility. An AO analog would be water soluble vitamin C.

There is scant literature on oxidative metabolism of RVT. In a metabolic study, mention is made of hydroxylation, in addition to glucuronidation, sulfation and hydrogenation.23 Oxidation catalyzed by lipoxygenase yields a complex mixture of decomposition products, similar to those obtained with hydrogen peroxide.24 Quinones, apparently not yet identified, may be formed in low yield, difficult to isolate due to reaction with protein nucleophiles. However, only small amounts are required to carry out catalytic ET with generation of large quantities of ROS.

Antioxidant

The AO property is apparently an important aspect of the physiological activity of RVT, particularly in relation to protection from oxidative injury. The literature is extensive and is also cited in other parts of this review.

The AO effect of RVT was manifested in blood mononuclear cells by a significant reduction in malonaldehyde content, an indication of oxidant injury.25 The cells acquired AO capacity. Grape RVT may be a useful dietary supplement for minimizing oxidative injury in immune-perturbed states and human chronic degenerative diseases. The effect of RVT on oxidative/nitrative stress by peroxynitrite, which is a strong physiological oxidant and inflammatory mediator, was determined in human blood platelets.26 Protein oxidation was significantly inhibited. Oxidation of thiol groups in protein and GSH was markedly reduced. There was distinct reduction in platelets lipid peroxidation. Various protective effects against peroxynitrite induced oxidative/nitrative damages to human platelet proteins and lipids were observed. RVT reduces OS induced by cisplatin and Se-Pt in human blood platelets, lymphocytes and plasma.27 The AO decreased lipid peroxidation and reduced activities of anti-oxidative enzymes, such as SOD, catalase and GSH peroxidase. A significant decrease in DNA damage was observed. Another study dealt with the antioxidation and free radical scavenging activities in the protective effects on ischemia-reperfusion induced injuries of rat hearts.28 Scavenging of the stable free radical DPPH was also observed. Astringinin, a more water soluble catechol-type analog, has a superoxide scavenging ability about 160 times more potent than that of RVT, and, hence could potentially be used as an AO and cardioprotective agent in biological systems. The efficiency of RVT for protecting polyunsaturated fatty acids was higher than that of flavonoids during oxidation.29 A related study deals with inhibition of human LDL oxidation by RVT.30 The phenol exhibits potent AO properties related to anti-inflammatory and anti-catabolic effects.31 The findings suggest possible application of RVT in therapy of human and animal osteoarthritis.

RVT could be a useful drug for the protection of liver cells from OS induced damage.32 The phenol is of interest for its beneficial properties in a variety of pathologies, including neurodegeneration.33 Many of the beneficial effects have been attributed to the ability to reduce OS. Bioactive phenols, such as RVT, can quench ROS and thus avoid pro-oxidative damage.34 This highly effective protection against OS damage suggests that this AO property constitutes the major part of preventing tumor induction. An investigation was made of the mechanism of cardioprotection by RVT in ischemia-reperfusion.35 The protection of cardiomyocytes from injury occurs partly by suppression of superoxide levels via AO action.

The mechanism of phenolic AO action has been addressed in a recent book.16 A key step is stoppage of radical propagation by formation of a resonance-stabilized peroxy radical, shown in Figure 2 for RVT. A report deals with mechanism and efficiency of AO action involving RVT and analogs.36 The comparison of the radical-scavenging effects of RVT and its analogues trans-4-hydroxystilbene and trans-3,5-dihydroxystilbene revealed that the two analogs showed almost the same effect and were more efficient than trans-3,5-dihydroxystilbene. These findings indicate greater radical-scavenging activity of the trans-resveratrol para-hydroxy group than its metahydroxy groups. Other data showed great similarity between RVT and trans-4-hydroxystilbene which seems to confirm that the para-hydroxy group of trans-resveratrol scavenges free radicals more effectively than its meta-hydroxy groups. The results can be rationalized by the greater delocalization possible for the radical from the para hydroxy group vs. the meta analog. Another important aspect entails termination of radical propagation by the phenoxy radical. This favorable aspect involves conversion to non-radical product by C-C coupling.16 This process is reported to occur with the radical derived from RVT.37 Trapping of the radical occurs by dimerization yielding trans-delta-viniferin which is unable to chain propagate.

Figure 2.

Figure 2

Resveratrol radical. The illustrated phenoxy radical is generated during the widespread AO action of RVT. The radical formation is facile due to the high degree of resonance stabilization afforded by this species. Hence, energetically, this radical is favored over the ones that would be formed from the meta-hydroxy resorcinol group in the other ring. Radical propagation is terminated by C-C radical coupling leading to a more stable dimer.

Cell Signaling

An investigation was made of the regulatory effect of RVT on signal transduction pathways in leukemia.38 Data indicate reduction in activation of JAK1/STAT3 tyrosine phosphorylation, as well as downregulation of expression. Evidence indicates activation of the Raf/Ras cascade and reversal of the sustained phosphorylation of JNK/SAPK.39 The study deals with the effect of RVT on signal transduction pathways involved in paclitaxel-induced apoptosis. The phenol inhibits phorbol ester activation of JNK and PKC.40 The inhibition may have a therapeutic potential, perhaps providing a novel means of controlling growth and invasiveness of tumors. A report deals with RVT modulation of phorbol ester-induced signal transduction pathways leading to elevated COX-2 expression.41 Other signals involved are NFkappaB, MAP kinases, AP-1 and protein kinase (ERK). An overview summarizes RVT modulation of signal transduction in apoptosis and cell survival.42 There is interference with many intracellular signaling pathways which regulate cell survival and apoptosis. Further insight is provided into the signaling network and interaction points. A review discusses the diverse molecular targets with focus on those involved in intracellular transduction.43 RVT impacts many components of intracellular signaling pathways including regulation of apoptosis and cell survival, tumor switches involved with kinases, transcription factors and their regulators. Evidence indicates that the stilbene derivative targets PTPIB to inhibit PDGFR mitogenic signaling.44 RVT mediates its effects through modulation of many different pathways by binding to numerous cell-signaling molecules.45 The compound activates various transcription factors, e.g., NFκB, STAT3, HIF-1α, β-catenin and PPAR-γ, suppresses the expression of antiapoptotic gene products, inhibits protein kinases, e.g., src, P13K, JNK and AKT, induces AO enzymes, and modulates cell cycle regulatory genes. The polyphenol holds promise against numerous age-associated diseases. An investigation was made of signaling pathways influenced by RVT involved with apoptosis and growth control in leukemia.46 Apoptosis is induced by modulating three different signaling pathways that regulate cell death and survival. The survival signaling pathway Notch is inhibited. The phenol inhibits P13K/Aket and activates Gsk/3β. Pro-apoptotic proteins p53 are also influenced. RVT reduces paclitaxel-induced apoptosis by modulating cell signaling pathways.39

Since the literature on cell signaling involving RVT is voluminous, additional, representative material is provided in abbreviated form. The organ or action is indicated along with the cell signaling involved in Tables 15.

Table 1.

Apoptosis

Cell signaling agent Organ or action Reference
(1) MAP kinases Human breast cancer cell MCF-11 47
(2) Fas redistribution in the rafts Colon cancer cells 48
(3) Ceramide Breast cancer cells 49
(4) STAT3 and nuclear factor-κB-regulation Human multiple myceloma cells 50
(5) Protein kinases and p38 kinase Antitumor activity 51
(6) Src and Stat3 Breast cancer 52
(7) TRAIL-induced Mitochondria 53
(8) Phosphorylated Akt and Caspace-9 MCF7 human breast cancer cells 54
(9) Cdc42 activation of ASK1/JNK Human leukemia cancer cells 55
(10) Phosphatidylinositol 3′-kinase Akt Human prostate cancer cells 56
(11) WAF-1/p21-mediated G1 phase Human epidermoid Carcinoma A431 cells 57
(12) AMPK Colon cancer cells 58
(13) JNK and c-JUN/AP-1 Prevented HNE-induced JNK 59
(14) c-Myc Downregulation in human medulloblastoma 60
(15) ERK Initiates p53 dependent apoptosis via αVβ3 61
(16) Caspase-3 Human breast cancer cell 62
(17) CD95 Human tumor cells 63
(18) TNBS Inhibiting Notch pathway 64
(19) p53 and P13K/Akt Human T-cell acute lymphoblastic leukemia MOLT-4 cells 65

Table 5.

Miscellaneous

Cell signaling agent Organ or action Reference
(1) NF-E2 HO-1 gene expression 91
(2) Kinase Reduces oxidation of human retinal pigment epithelial cells 92
(3) AMPK Antidiabetic 93
(4) Nrf2 Cigarette smoke-mediated OS 94
(5) SIRT1 and AMP kinase Human alcoholic liver disease 95
(6) Ras Stimultes sirtuins and extends life ras 96
(7) Caspase-3 Inhibits IL-1β-induced stimulation 97
(8) 3-Kinase/Akt Inhibition of phosphor-inositide-dependent kinase-1 activity 98
(9) NFκB Stimulates cytokines IL-6-1β and NFκB 99
(10) P13K Inhibition 100
(11) AMP kinase Stimulate AMPK in neurons 101
(12) NFκB and AP-1 Activate and regulate gene expression 67
(13) MAPK Activate through estrogen receptors 102
(14) NFκB, STAT3, HIF-1α Activate transcription factors 45

In relation to mechanism at the basic level, recent proposals focus on ROS and electrochemistry. Cell signaling is known to be importantly involved in various aspects of biological function, including normal processes, therapeutic drug action and toxicology. More than 10 years ago, ROS attracted attention in relation to cell signaling. Since then several books103,104 and a book chapter105 have addressed this aspect. A recent review has further insight.106 Evidence has accumulated that ROS, such as hydrogen peroxide, superoxide, and the hydroxyl radical, are important chemical mediators that regulate the transduction of signals by modulating protein activity via redox chemistry. Authors have proposed that ROS have been conserved throughout evolution as universal second messengers. Nearly every step in signal transfer is sensitive to ROS, which can function as second messengers in the activation of transcription factors. Various types of radiation, which are generators of ROS, also influence cell communications. Since the messengers must possess appreciable lifetime in order to migrate, a certain degree of stability is required. For example, the hydroxyl radical would not be a messenger due to its extremely high reactivity with its resultant very short time existence, although it would generate messenger radicals. Likely candidates include superoxide and resonance stabilized peroxyl radicals. Others would be stable ROS arising from AOs, such as vitamin C, vitamin E and flavonoids. Members can be envisioned from the reactive nitrogen species (RNS) category. NO is a well known radical that plays an important role in cell signaling. In relation to theory, a puzzling aspect is its relatively short life time. Perhaps more stable complexes are involved. Other nitrogen radicals can play a role. Important candidates comprise small proteins possessing redox groups.

In effect, cell signaling can be regarded as proceeding via a long redox chain in which the standard parameters of initiation, propagation and termination pertain, involving omnipresent conduit species with unshared electrons. A series of relay stations may be operative. Based on the redox chain framework, the second messenger might be superoxide formed by redox process involving oxygen and a second messenger electron from an ET functionality in the receptor site. At the termination of the initial journey, radical character would be transmitted to a site, mobile or stationary, e.g., a redox amino acid (AA) side chain acting as a relay (transfer station), that could then pass on (initiate) radical character to a third messenger. These types of interactions, widespread in AA chemistry, usually involving electron and/or hydrogen abstraction to generate radical species, are treated in a review.107 The numerous redox moieties in anchored proteins might fall in the relay category. There has been dramatic increase in attention devoted to free radical species in cell signaling, although the bulk of the signal transduction literature pays no attention to this aspect.

Electrochemistry also appears to play a role in cell signaling, including electron transfer and electrostatics. Discussion is present in recent reviews on receptor-ligand activity,108 phosphates and sulfates109 and metal cations.110

Receptors

The literature on the RVT receptor is very limited. RVT is a phytoestrogen which binds to and activates estrogen receptors that regulate the transcription of estrogen-responsive target genes.111 The effect on gene expression appears to correlate with chemoprevention. The drug regulates mRNA expression of several genes involved in cell cycle control, apoptosis, metastasis, cell-cell adhesion and receptor signaling pathways. A report shows RVT binds to the sulfonyl urea receptor.112 Electrophysiological measurements revealed that the bound ligand is a blocker of pancreatic SUR channels and enhances apoptosis. RVT is known to influence the androgen receptor (AR). AR pathways are involved in the development and progression of prostate cancer.113 The ability to modulate AR function may contribute to the chemopreventive activity of RVT. Also, the drug regulates AR target gene expression, at least in part, by repressing AR transcriptional activity.114,115 RVT represses different classes of AR. AR upregulates genes at the protein or mRNA level, and may be a useful preventive or therapeutic agent for prostate cancer.116 Aryl hydrocarbon receptor ligands, such as dioxin and polynuclear aromatic hydrocarbons, are environmental contaminants with many adverse effects.117 RVT, a competitive antagonist of these ligands, promotes receptor translocation to the nucleus and binding to DNA. The phenol inhibits the transactivation of several dioxin-inducible genes. Clinical testing as a prophylactic against the insults is warranted.

Electrochemistry

Glucose-induced depolarization was counteracted by RVT.118 There is inhibition of electrical activity and insulin release from insulinoma cells by blockage of voltage-gated Ca channels and chloride currents, with inhibition of K (ATP) currents. RVT inhibits the electrical activity of paraventricular nucleus neurons and exerts neuroprotective effects on central neurons.119 The effects of RVT on neuron discharges in rat subfornical organ were examined.120 The inhibition of electrical activity may be related to blockade of voltage-gated Ca channels and NO promotion. A study demonstrated RVT-induced depression of electrical activity in the rat heart.121 The shortened action potential in the left atrium is likely due to activation of K (ATP) channels. The importance of electrochemistry in living systems has been reviewed recently.122

Aging

The anti-aging property of RVT has enjoyed much attention. This aspect is also addressed in many other portions of this review. Considerable prior literature identifies ROS with the aging process.

An article deals with prevention and treatment of common clinical conditions of aging.123 Aging is associated with a variety of common conditions, such as cancer, diabetes, cardiovascular disease and Alzheimer's disease. Properties of the phenol associated with the beneficial aspects include AO, regulation of the cell cycle, activation of intracellular pathways, vascular tone, oncogene suppression and mitochondrial energy production. There is a striking transcriptional overlap of RVT and calorie restriction in the heart, skeletal muscle and brain.124 Both interventions inhibit gene expression associated with cardiac and skeletal muscle aging, as well as prevention of cardiac dysfunction. RVT fulfills the definition of a dietary compound that mimics some aspect of calorie restriction. A review summarizes the anti-aging properties of RVT, including cardiovascular benefits via increased NO production, downregulation of vasoactive peptides, lowered levels of oxidized low-density lipoprotein, cyclooxygenase inhibition, effects on neural tissues, phytohormonal actions, anticancer properties via modulation of signal transduction (anti-initiation, antipromotion and anti-progression effects), antimicrobial effects, sirtuin activation, possible benefits in Alzheimer's disease and prevention of photoaging.125 Comparison was made with other AOs used in skin care products. RVT consistently retards aging in organisms as diverse as yeast, worm, fly and fish.126 It prolonged lifespan and delayed the onset of age-related dysfunctions in fish. A review focuses on the role of OS and inflammation in cardiovascular dysfunction in aging, and on emerging anti-aging therapeutic strategies offered by RVT and other polyphenols.127 In a study of RVT influence on aging, the drug mitigated the metabolic dysfunction of mice fed high-fat diets.128 The effects may be mediated partly by activation of a deacetylase enzyme that regulates several transcription factors and enzymes responsive to nutrient availability.

Cancer

Antioxidant.

OS arising from ROS appears to be associated with many aspects of carcinogenesis.4 For example, there is involvement in three major stages of the process. Literature cited129134 indicates that RVT as an AO provides a beneficial effect in alleviation of harmful OS. In some cases, there is evidence for significant modulation of oxidative imbalance and effect on levels of other AOs.

Apoptosis.

Apoptosis plays a part in many aspect of biological chemistry. There are many articles which deal with it in connection with cancer. The reports addressed herein do so in connection with presence of RVT.62,135149

Cell signaling.

This aspect has been addressed in a general approach (see above). Cell signaling has played a role in many aspects of biochemistry. It is not surprising that considerable attention has been devoted to its relationship with cancer. Mechanistic aspects of signaling are summarized in an above section. Representative articles are provided that involve RVT.43,58,150158

Nitric oxide.

This fascinating gas possesses a wide variety of bioactivities, both beneficial and deleterious. Many modes of action have been implicated. One review puts focus on electron transfer.159 Peroxynitrite formed by reaction with superoxide, plays a role, particularly in toxicity. A sampling of literature is presented for the effects of NO along with RVT.135,160,161

Miscellaneous aspects.

There are many other approaches that deal with the cancer problem, along with involvement of RVT.162174 Examples involve hormones, polyamines, autophagy, cathepsin D, DNA synthesis and mitochondria.

Cardiovascular System

RVT displays many bioactivities, such as protection from or reduction of the incidence of coronary heart disease, including counteraction to ischemia-reperfusion injury.175 It might be that there is inhibition of signaling pathways and gene expression which involve the disruption of the ERK pathway via attenuation of ROS. A review focuses on the role of the phenol on OS and inflammation in cardiovascular disease.176 Intake may contribute to the “French paradox” involving the unexpectedly low cardiovascular morbidity in the Mediterranean population, which may reflect the AO and anti-inflammatory effects. Upregulation of NO production might also be involved.177 The amount of malondialdehyde, indicative of lipid peroxidation, was decreased in the postischemic myocardium indicating a reduction of OS.178 There was prevention of superoxide-dependent inflammation response induced by ischemia-reperfusion and oxidants.179 AO properties may be involved. RVT exhibits multifaceted properties in preventing various harmful vascular alterations, in addition to aging, including cell signaling, enzymatic pathways, apoptosis, gene expression and AO action.180 The review discusses the relationship with ROS and regulation of pro-inflammatory genes. Administration of RVT exerts cardioprotection against ROS-mediated menadione toxicity.181 Results indicate that the drug interferes with the release of inflammatory mediators, thus providing biological plausibility to the protective effect of moderate red wine consumption against coronary heart disease.182 In a related study a protective AO effect of RVT in red wine was demonstrated against oxidative injury on red blood cells.183 Authors proposed that the AO and antiapoptotic effects, together with the anti-inflammatory actions, are responsible, at least in part, for the cardioprotective effects.184 Upregulation of endogenous AOs and certain enzymes by red wine RVT in aortic muscle cells leads to protection against oxidative and electrophilic stress.185 Various investigations dealt with the effects on blood platelets, such as, action as AO for reduction of OS,186 inhibition of superoxide generation, ROS production and lipid peroxidation.187 The drug reduces ROS levels, blunts the inflammatory pathways and stimulates NO generation.188 There is inhibition of ROS production, e.g., superoxide, hydrogen peroxide, singlet oxygen and organic radicals,189 and a variety of actions including anti-inflammatory, AO, ROS, scavenger and reduction of lipid peroxidation.190

A prior comprehensive review on prevention of cardiotoxicity is based on the unifying theme of ET-ROS-OS.8

Central Nervous System (CNS)

Much research supports the thesis that ROS play a role in insults to the CNS.9 Since the brain is deficient in defenses by AOs, supplementation should be investigated as a potential clinical approach. There are many reports demonstrating the beneficial effect of RVT in preventing toxic attacks in relation to brain neuronal injury in conditions such as Alzheimer's disease and Parkinson's disease.191199 In many cases, attribution is given to the AO property. In the treatment of traumatic brain injury, the drug provided neuroprotection by reducing OS.200 There is a related report.201 Increased OS has been implicated in the mechanism of neuronal cell death following cerebral ischemic insult.202,203 RVT exerted a protective action, apparently as an AO. The phenol is a potent neuroprotective agent against diabetic oxidative damage.204 There was reduction in lipid oxidation product and NO production, as well as OS and DNA fragmentation.205 A neuroprotective effect via free radical scavenging was observed with induced Parkinsonism.206,207 Intervention by AOs can be a potential beneficial approach in the treatment of epilepsy.208 The protective effect of RVT against kainic acid-induced convulsions and the attenuation of lipid oxidation product suggest the potential use of AOs as adjunct therapy in epilepsy. The neuroprotective ability of RVT against NO-related toxicity in hippocampal neurons is attributed to AO involvement.209 Also, a neuroprotective effect was observed in cerebral ischemia.210 There was decrease in product from lipid oxidation and induction of an important role for NO.

Inflammation

An appreciable amount of research is reported on the anti-inflammatory effects of RVT. Evidence shows that the condition is associated with the presence of ROS.15 Hence, it is not surprising that the powerful phenolic AO exerts a beneficial effect. Literature in the area is also discussed elsewhere in this review, such as in cell signaling.

Some research in this area involves the lungs.211 With human airway epithelial cells, RVT inhibits NFκB-protein dependent transcription and cytokine-stimulated inducible NO synthase cells. The salutary effects of the drug on attenuation of lung injury following trauma hemorrhage are likely due to reduction of pro-inflammatory mediators.212 Plausible mechanisms of anti-inflammatory activity are discussed.213 This property may have relevant clinical implications. The drug could alleviate the severity of pancreatitis through its anti-inflammatory effects, regulation of inflammatory mediators and inhibition of NFκB expression.214 Also, there is prevention of superoxide-dependent inflammatory response induced by ischemia/reperfusion platelet- activating factor or oxidants.215 The phenol decreases the degree of inflammation associated with colitis.64 The effect may result from the countering of OS and proinflammatory cytokines. Attenuation of ischemia/reperfusion injury in rats is due to the ant-inflammatory action of RVT, apparently through a NO-dependent mechanism.216

Arthritis

Much evidence supports a role for ROS-OS in arthritis.15 This includes oxidative damage to DNA, protein and lipids. In accord with involvement of ROS, there is depletion of GSH, ascorbate and α-tocopherol. Hence, it is not surprising that the AO RVT possesses the potential for arthritis treatment.217 Inflammation, which is commonly associated with arthritis, appears to have a ROS link.15 Involvement of cell signaling pathways is also treated.217 The inflammatory process plays a pivotal role during the pathogenesis of osteroarthritis, dominated by catabolic processes initiated by pro-inflammatory cytokines, such as IL-1 beta.218 RVT appears to be an effective anti-inflammatory agent that has a chondroprotective capacity through suppression of IL-1β, ROS and tumor suppression protein p53 production. NFκB is a pivotal transcription factor involved in the activation of the TNFα and IL-β genes.219 Activation of NFκB is a feature seen in arthritis patients. RVT is a potent and specific inhibitor of TNFα and IL-1β. Injection of the drug may protect cartilage against the development of inflammatory arthritis.

Liver

Pyrogallol causes hepatotoxicity in experimental animals.220 RVT reduces the increase in lipid peroxidation due to OS by the toxicant. The accompanying decrease in GSH, GSH peroxidase and GSH reductase activities was significantly attenuated by the phenol. In a study of the liver damage with a high-fat diet, RVT reduces oxidized LDL and hepatic OS.221 RVT-mediated chemoprevention of hepatocarcinogenesis occurred with nitrosamine-initiated carcinogenesis.222 The favorable effect was attributed to inhibition of cell proliferation and induction of apoptosis. A review documents evidence for the favorable effect entailing destruction of cancer-initiating ROS by AO action (see Introduction). Liver damage caused by chronic alcohol consumption was reduced by RVT.223 Inhibition of oxidation of polyunsaturated fatty acids is proposed as a basis of the hepatoprotective effect. The involvement of OS in the pathogenesis of alcoholic diseases in the liver has been repeatedly confirmed.224 Dietary supplementation with RVT during ethanol treatment inhibited hepatic lipid peroxidation and ameliorated the reduction in activity of SOD, catalase and GSH peroxidase.

Kidney

The protective effect of the AO resveratrol and others was investigated in the kidney of rats treated with the carcinogen KBrO3.225 The 100% increase in 8-OH-dG from oxidation in the renal genome DNA was completely abolished by treatment with RVT. ROS have been implicated in cell injury that occurs after ischemia.226 Products of lipid peroxidation are generated on reperfusion. This oxidation can be prevented by AOs. Resveratrol was found to exert a favorable effect in reducing such injury. A related report is available.227 An hypothesis was advanced concerning the beneficial effects of red wine which contains resveratrol.228 Evidence suggests a protective role of moderate wine consumption against the onset and progression of renal diseases, based on the concept of kidney injury mediated by OS. A similar study was reported.229 ROS are observed in gentamicin-induced nephrotoxicity.230 Renal lipid peroxidation increases with the toxicant alone, which was prevented by the administration of RVT.230 Addition of the phenol resulted in an increase in the levels of AOs. At least a part of the favorable effects was attributed to the AO activity.

DNA

RVT reduces nuclear DNA fragmentation.231 Results indicate that the drug can act as an antimutagenic/anticarcinogenic agent by preventing DNA damage which plays a pivotal role in the carcinogenic activity of many genotoxic agents. The phenol reduced DNA damage induced by Cr (III) based on reduced 8-OH-dG formation.232 The protective effect against Cr-induced carcinogenesis may relate to the free radical-scavenging ability. The genoprotective effects of the drug were investigated under conditions of OS induced by hydrogen peroxide in glioma cells.233 Due to attenuation of oxidative DNA damage, RVT may be important in protecting against DNA fragmentation and oxidation arising from OS. In a study of Alzheimer's disease, red wine micronutrients were found protective. The phenols reduced ROS production, prevented DNA fragmentation and protected the cellular membrane from oxidative damage.

Genes

RVT significantly blocked the expression of genes related to the NFκB family.234 The phenol has a significant modulatory effect on the NFκB signaling pathway and an important AO role that may help explain the cardioprotective effects attributed to long-term moderate red wine consumption. Through its phytoestrogenic properties, RVT regulates the expression of hormone- dependent genes in breast cells and provides a protective effect against several types of cancer, notably breast cancer.111 The drug modulates the expression of genes in a pattern dependent on the state of estrogen receptors. Gene expression is regulated via the estrogen receptor pathway and also an undetermined pathway. The effects of RVT on circadian clocks of Rat-1 cells were analyzed.235 A dose, which did not exhibit toxicity, regulated the expression of various clock genes. Relevant material is also present elsewhere in this review.

Other Body Constituents

In addition to the extensive studies presented in the above sections, there are many reports of the beneficial effects of RVT on other body constituents under stress. Much of the salutary responses can be attributed to AO action under OS. RVT limited dysfunction of rat brain mitochondria in an anoxia-reoxygenation model.236 At least three mechanisms were proposed, including AO properties. Data indicate that the drug may have application in the treatment of bronchial asthma, accompanied by inhibition of increases in cytokinases.237 A review provides evidence for AO protection of various pulmonary disorders.13 The phenol exerted beneficial activity against alcohol injury, e.g., peroxidation of lipids.238 The adverse effects, countered by AO action involve ROS and OS. Oxidative stress entailing rapid degeneration of endothelial cell function, is deeply involved in systemic sclerosis pathogenesis.239 There is potential for RVT as an AO for reverting endothelial dysfunction, scavenging lipid peroxides and reducing hypoxia-reperfusion injury. Examination revealed a significant improvement in ovarian morphology in RVT-treated rats, compared with the ischemia-reperfusion group.240 Drug administration reduced lipid peroxide products and countered the reduction in GSH levels. The phenol exerted a favorable influence against lipid peroxidation in cell membranes, including a decrease in DNA damage.241 An important AO role was assigned. Also, an inhibitory effect was observed on the NFκB signaling pathway after exposure to metal-induced radicals. Obesity in the US has become a serious problem, leading to a sharp increase in diabetes. In a controlled study, RVT treatment caused the greatest and most consistent loss of fat content in animal subjects.242 The underlying target protein likely involves sirtuin family members. A study was carried out involving RVT and Cd-induced oxidative damage in mice.243 The phenol and other AOs effectively protect against lipid peroxidation generated by the metal, and were able to counter the inhibition of catalase activity. Metals are well-known generators of ROS and OS.215

Skin cancer is a common illness among humans, due to solar radiation.243 A way to reduce the occurrence is by use of photochemopreventive agent, often in the AO category.244 Results suggest that RVT may afford substantial protection against damage caused by UVB exposure, which may be mediated via its AO properties. RVT reduces colitis, alleviates oxidative events and stimulates apoptosis.245 Much attention has recently been paid to nanoparticles. RVT-loaded nanoparticles protected cells from β-amyloid peptide toxicity by attenuating intracellular OS and caspase-3-activity.246 A review addresses various approaches to immunomodulation.247 Disruption of the accompanying proinflammatory cascade is by various therapies, including RVT, involving various mechanisms, including AO effects and alterations in cell signaling.

Pro-Oxidant

The literature contains extensive documentation for AO action by RVT as presented above. However, there is an apparent dichotomy based on an appreciable number of reports providing evidence for pro-oxidant action. The following material addresses these references, followed by rationale for the dichotomy.

Compounds, such as RVT, acting as AOs to lipids often have a pro-oxidant effect on DNA or protein.248 Free radicals derived from the phenol appear to mediate between anti-and pro-oxidative actions. Dietary polyphenols with phenol rings are metabolized by peroxidase to form pro-oxidant phenoxy radicals which are sufficiently reactive to co-oxidize GSH or NADH accompanied by extensive oxygen uptake and ROS formation.249 The experimental conditions are important for the pro-oxidant activity, causing oxidative DNA damage that may lead to cell cycle arrest or apoptosis.250 At certain concentrations, RVT elicits pro-oxidant properties as evidenced by an increase in intracellular superoxide.251 The pro-oxidant effect is further supported by other observations. An unusual study reveals opposite effects on rat tissue lipid peroxidation.252 With a dark-light cycle, RVT behaved as an AO during the dark span, and as a pro-oxidant during the light span, comprising further support for the importance of conditions.

Discussion of the AO-pro-oxidant aspect is available for other well-known AOs, such as vitamins E and C, flavonoids and thiols.16 For the phenols, a pro-oxidant appears to involve conversion to a quinone capable of ET-ROS-OS. A similar route would apply to RVT in relation to oxidation to the stilbene quinone “3” (Fig. 3). However, we have not found such a report (see Metabolism section). It may be that the quinone has not yet been isolated due to formation in small quantities with much being tied to protein by nucleophilic attack (see Future Work).

Figure 3.

Figure 3

Resveratrol quinone. This stilbene quinone would be expected from facile oxidation of RVT. However, we are unable to find any report of its formation. It may be that it is present only in very small amounts making for greater difficulty in isolation. Nevertheless, only minor quantities will suffice for the catalytic generation of large amounts of ROS. Quinone formation is one rationale for the observed pro-oxidant effects of RVT involving redox cycling with oxygen leading to ROS.

A superficial approach might lead to assignment of quinone “3” (Fig. 3) as the product from oxidation of “1.” However, application of basic principles of organic chemistry could lead to different conclusion. Compound “3” (Fig. 3) is a conjugated dienone incorporating two enol groups. Generally, at equilibrium, the keto state is energetically favored over the enol tautomer. An exception is cyclohexane-1,2-dione for which enol “4” (Fig. 4) makes a substantial contribution due to enol stabilization by hydrogen bonding with the carbonyl.253 It is reasonable to apply this reasoning to “3” (Fig. 3) as shown in structure “5” (Fig. 5). On the other hand, such interaction makes similar H-bonding by the other enol less likely. As a result, the keto form shown in “6” (Fig. 6) could prevail. Therefore, the quinonoid structure capable of redox cycling with generation of toxic ROS would not pertain. This scenario is in line with the very low toxicity exhibited by RVT.1 Another possibility is H-bonding involving both enol groups, as illustrated in “7” (Fig. 7), or conversion to an ET o-quinone via oxidation of adjacent hydroxyl groups in the tetrahydroxy precursor of “3” (Fig. 3).

Figure 4.

Figure 4

Enol tautomer of cyclohexane-1,2-dione. Although the keto form is generally favored over the enol tautomer, in the case of “4,” hydrogen bonding of enol with the carbonyl results in increased contribution of enol to the equilibrium state.

Figure 5.

Figure 5

Mono-hydrogen bonded quinone. This structure is analogous to that in Figure 4. Hence, the enol moiety is stabilized.

Figure 6.

Figure 6

Diketo-enol tautermer of “3” (Fig. 3). Structure “5” (Fig. 5) shows the stabilization of an enol group by H-bonding, similar to that in “4” (Fig. 4). Since this is now less likely for the other enol group. It may tautomerize mostly to the keto form as illustrated in “6” (Fig. 6). The ability to redox cycle would be substancially decreased due to absence of the quinone functionality. The conjugated α-dicarbonyl structure in “6” (Fig. 6) should be capable of electron uptake, but not be a good generator of ROS.

Figure 7.

Figure 7

Di-hydrogen bonded quinone. Perhaps both enols participate in H-bonding with the carbonyl. Therefore, the quinone structure is maintained.

It should be recognized that “6” (Fig. 6) could also be capable of electron uptake, but less efficiently than the related quinone. Compound “6” (Fig. 6) is a conjugated analog of diacetyl which is electron affinic, but exhibits negligible ability to generate significant OS via ROS.253

The placement of the hydroxyl groups apparently is beneficial due to phenolic AO action, but largely avoids the damaging redox cycling that often results from quinone generation via oxidation. This represents another example of a clever strategy employed in the biochemical domain.

Analogy can be made to metabolism of PCBs, in which oxidation proceeds via mono- and di-hydroxyl derivatives to the quinone “8” (Fig. 8) stage. Compound “8” (Fig. 8) is known to redox cycle with formation of ROS which appear responsible for some of the toxicity.254

Figure 8.

Figure 8

Quinone metabolite of PCB. This metabolite route to “8” is quite similar to that proposed for RVT, which provides support for the proposed oxidative metabolism. The monohydroxy metabolite of PCB is analogous to RVT.

An alternate interpretation exists for the pro-oxidant behavior. Phenols, including RVT, form complexes with metals. In the case of heavier metals, the favorable reduction potential can lead in vivo to redox cycling involving oxygen with formation of ROS leading to OS.215 In the presence of Cu ions, DNA damage by RVT occurs which is attributed to oxidative intermediates formed by redox cycles involving the metal complex.255 Similar studies were made.256261

Future Work

A principal aspect of metabolism (see above) involves formation of conjugates. It would be helpful to ascertain whether or not the mono- and di-derivatives with at least one free phenolic group possess AO properties. A careful analysis of metabolic products from oxidation should be performed with the aim of detecting the proposed quinone. Authentic material would be synthesized for comparison. Also, it could be illuminating to oxidize 1 at the carbon between the resorcinol hydroxyl groups and determine the structure in relation to the hypothetical quinone. Computational studies should cast light on the structure of “3” (Fig. 3).

Other Aspects

Related articles are available, including ones dealing with antioxidants.262271

Table 2.

Cancer

Cell signaling Organ or action Reference
(1) NFκB and Ap-1 Genes 66
(2) p38, MAPK, p53 and p21 Cancer cells 67
(3) NFα mediated MMP-9 expression Hepatocellular carcinoma cells 68
(4) TRAIL through gene expression Human melanomas 69
(5) STAT3 Medulloblastoma cell lines 70
(6) MAPK Breast cancer cells 71
(7) c-Jun-NH2-terminal-kinase Waldenström's macroglobulinemia 72
(8) NFκB Cardiovascular, neurological and mitochondrial dysfunction 73
(9) ATM/ATR-Chk1/2-Cdc42 Ovarian cancer cells 74
(10) Rac and Cdc42 Breast cancer cells 75
(11) P13K Cultured muscle cells 76

Table 3.

Inflammation

Cell signaling agent Organ or action Reference
(1) NFκB Chromatin structure Glutathione biosynthesis 77
(2) IL-6-induced ICAM-1 gene expression in endothelial cells 78
(3) P13KK/AKt/ERK dependent interleukin IL-17 Mouse cardiac fibroblast 79
(4) TNFα Endothelial cells 80
(5) Cytokine 1L-1β Human chondrocytes 81
(6) NFκB and AP-1 Mouse skin cell 82
(7) TLR-derived Mouse embryonic fibroblast 83

Table 4.

Heart

Cell signaling agent Organ or action Reference
(1) AMPK H9c2 cardiac muscle cells 84
(2) NFκB Endothelial cells of coronary artery 85
(3) p38 and ERK/1/2 Inhibits EMMMPRIN THP-1 cells 86
(4) p38 Mitogen-activated protein kinase Triggers an MAPK path involving erK ½ and P38 MAPK 87
(5) Bcl-2 Adenosine A3 receptor Activation 88
(6) p38 map kinase and P1-3-kinase HO-1 mediated mechanism 89
(7) Akt/protein Kinase B Suppresses angiotensin II-induced protein 90

Acknowledgements

Editorial assistance by Thelma Chavez is acknowledged.

Abbreviations

RVT

resveratrol

ET

electron transfer

ROS

reactive oxygen species

OS

oxidative stress

AO

antioxidant

CNS

central nervous system

ATP

adenosine triphosphate

GSH

glutathione

NO

nitric oxide

8-OH-dG

8-hydroxy-d-guanosine

PCB

polychlorinated biphenyl

AR

androgen receptor

SOD

superoxide dismutase

AA

amino acid

Footnotes

References

  • 1.Udenigwe CC, Ramprasth VR, Aluko RE, Jones PJ. Potential of resveratrol in anticancer and anti-inflammatory therapy. Nutr Rev. 2008;66:445–454. doi: 10.1111/j.1753-4887.2008.00076.x. [DOI] [PubMed] [Google Scholar]
  • 2.Kovacic P, Becvar LE. Mode of action of anti-infective agents: emphasis on oxidative stress and electron transfer. Curr Pharm Des. 2000;6:143–167. doi: 10.2174/1381612810006020143. [DOI] [PubMed] [Google Scholar]
  • 3.Kovacic P, Osuna JA. Mechanisms of anticancer agents: emphasis on oxidative stress and electron transfer. Curr Pharm Des. 2000;6:277–309. doi: 10.2174/1381612003401046. [DOI] [PubMed] [Google Scholar]
  • 4.Kovacic P, Jacintho JD. Mechanism of carcinogenesis: focus on oxidative stress and electron transfer. Curr Med Chem. 2001;8:773–796. doi: 10.2174/0929867013373084. [DOI] [PubMed] [Google Scholar]
  • 5.Kovacic P, Jacintho JD. Reproductive toxins: pervasive theme of oxidative stress and electron transfer. Curr Med Chem. 2001;8:863–892. doi: 10.2174/0929867013372878. [DOI] [PubMed] [Google Scholar]
  • 6.Kovacic P, Sacman A, Wu-Weis M. Nephrotoxins: widespread role of oxidative stress and electron transfer. Curr Med Chem. 2002;9:823–847. doi: 10.2174/0929867024606803. [DOI] [PubMed] [Google Scholar]
  • 7.Poli G, Cheeseman GH, Dianzani MU, Slater TF. Free Radicals in the Pathogenesis of Liver Injury. Oxford: Pergamon Press; 1989. pp. 1–330. [Google Scholar]
  • 8.Kovacic P, Thurn LA. Cardiovascular toxins from the perspective of oxidative stress and electron transfer. Curr Vasc Pharmacol. 2005;3:107–117. doi: 10.2174/1570161053586912. [DOI] [PubMed] [Google Scholar]
  • 9.Kovacic P, Somanathan R. Neurotoxicity: the broad framework of electron transfer, oxidative stress and protection by antioxidants. Curr Med Chem-CNS Agents. 2005;5:249–258. [Google Scholar]
  • 10.Kovacic P, Pozos RS, Somanathan R, Shangari N, O'Brien PG. Mechanism of mitochondrial uncouplers, inhibitors and toxins: focus on electron transfer, free radicals and structure-activity relationships. Curr Med Chem. 2005;5:2601–2623. doi: 10.2174/092986705774370646. [DOI] [PubMed] [Google Scholar]
  • 11.Kovacic P, Cooksy AL. Unifying mechanism for toxicity and addiction by abused drugs: electron transfer and reactive oxygen species. Med Hypotheses. 2005;64:357–366. doi: 10.1016/j.mehy.2004.07.021. [DOI] [PubMed] [Google Scholar]
  • 12.Kovacic P, Somanathan R. Ototoxicity and noise trauma: electron transfer, reactive oxygen species, cell signaling, electrical effects and protection by antioxidants: practical medical aspect. Med Hypotheses. 2008;70:914–923. doi: 10.1016/j.mehy.2007.06.045. [DOI] [PubMed] [Google Scholar]
  • 13.Kovacic P, Somanathan R. Pulmonary toxicity and environmental contamination: radicals, electron transfer, and protection by antioxidants. In: Whitacre WD, editor. Reviews of Environmental Contamination and Toxicology. Vol. 201. New York: Springer; 2009. pp. 41–69. [DOI] [PubMed] [Google Scholar]
  • 14.Kovacic P, Somanathan R. Intergrated approach to immunotoxicity: electron transfer, reactive oxygen species, antioxidants, cell signaling and receptors. J Recept Signal Transduct. 2008;28:323–346. doi: 10.1080/10799890802305217. [DOI] [PubMed] [Google Scholar]
  • 15.Halliwell B, Gutteridge JMC. Free Radicals in Biology and Medicine. New York: Oxford University Press; 1999. pp. 1–936. [Google Scholar]
  • 16.Kovacic R, Somanathan R. Beneficial effects of antioxidants in relation to carcinogens, toxins and various illnesses. In: Panglossi HV, editor. Antioxidants: New Research. NY: Nova Science Publishers Inc.; 2006. p. 1. [Google Scholar]
  • 17.Wenzel E, Somoza V. Metabolism and bioavailability of trans-resveratrol. Mol Nutr Food Res. 2005;49:472–481. doi: 10.1002/mnfr.200500010. [DOI] [PubMed] [Google Scholar]
  • 18.Wang LX, Heredia A, Song H, Zhang Z, Yu B, Davis C, et al. Resveratrol glucuronides as the metabolites of resveratrol in humans: characterization, synthesis and anti-HIV activity. J Pharm Sci. 2004;93:2448–2457. doi: 10.1002/jps.20156. [DOI] [PubMed] [Google Scholar]
  • 19.Yu C, Shin YG, Chow A, Li Y, Kosmedar JW, Lee YS, et al. Human, rat and mouse metabolism of resveratrol. Pharm Res. 2002;19:1907–1914. doi: 10.1023/a:1021414129280. [DOI] [PubMed] [Google Scholar]
  • 20.Wang D, Hang T, Wu C, Liu W. Identification of the major metabolites of resveratrol in rat urine by HPLCMS/MS. J Chromatogr B Analyt Technol Biomed Life Sci. 2005;829:97–106. doi: 10.1016/j.jchromb.2005.09.040. [DOI] [PubMed] [Google Scholar]
  • 21.Wenzel E, Soldo T, Erbersdobler H, Somoza V. Bioactivity and metabolism of trans-resveratrol orally administered to Wister rats. Mol Nutr Food Res. 2005;49:482–494. doi: 10.1002/mnfr.200500003. [DOI] [PubMed] [Google Scholar]
  • 22.Muris M, Miksits M, Aust S, Spatzenegger M, Thalhammer T, Szekeres T, et al. Metabolism of resveratrol in breast cancer cell lines: impact of sulfotransferase 1A1 expression on cell growth inhibition. Cancer Lett. 2008;261:172–182. doi: 10.1016/j.canlet.2007.11.008. [DOI] [PubMed] [Google Scholar]
  • 23.Shanker S, Singh G, Srivastava RK. Chemoprevention by resveratrol: molecular mechanisms and therapeutic potential. Front Biosci. 2007;12:4839–4854. doi: 10.2741/2432. [DOI] [PubMed] [Google Scholar]
  • 24.Pinto MC, García-Barrardo JA, Macías P. Oxidation of resveratrol catalyzed by soybean lipoxygenase. J Agric Food Chem. 2003;51:1653–1657. doi: 10.1021/jf025818d. [DOI] [PubMed] [Google Scholar]
  • 25.Losa GA. Resveratrol modulates apoptosis and oxidation in human blood mononuclear cells. Eur J Clin Invest. 2003;33:818–823. doi: 10.1046/j.1365-2362.2003.01219.x. [DOI] [PubMed] [Google Scholar]
  • 26.Olas B, Wachowicz B, Nowak P, Stochmal A, Oleszek W, Glowacki R, et al. Comparative studies of the antioxidant effects of naturally occurring resveratrol analogue trans-3,3′,5,5′-tetrahydroxy-4′-methoxystilbene and resveratrol against oxidation and nitration of biomolecules in blood platelets. Cell Biol Toxicol. 2007;24:331–340. doi: 10.1007/s10565-007-9045-7. [DOI] [PubMed] [Google Scholar]
  • 27.Olas B, Wachowicz B, Majsterek I, Blasiak J. Resveratrol may reduce oxidative stress induced by platinum compounds in human plasma, blood platelets and lymphocytes. Anticancer Drugs. 2005;16:659–665. doi: 10.1097/00001813-200507000-00011. [DOI] [PubMed] [Google Scholar]
  • 28.Hung L-M, Su M-J, Chu W-K, Chiao RC-W, Chan RW-F, Chen RJ-K. The protective effect of resveratrol on ischaemia-reperfusion injuries of rat hearts in correlated with antioxidant efficacy. Brit J Pharmacol. 2002;135:1627–1633. doi: 10.1038/sj.bjp.0704637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Frémont L, Belguendouz L, Delpal S. Antioxidant activity of resveratrol and alcohol-free wine polyphenols related to LDL oxidation and polyunsaturated fatty acids. Life Sci. 1999;64:2511–2521. doi: 10.1016/s0024-3205(99)00209-x. [DOI] [PubMed] [Google Scholar]
  • 30.Frankel EN, Waterhouse AL, Kinsella JE. Inhibition of human LDL by resveratrol. Lancet. 1993;341:1103–1104. doi: 10.1016/0140-6736(93)92472-6. [DOI] [PubMed] [Google Scholar]
  • 31.Shakibaei M, John T, Seifarth C, Mobasheri A. Resveratrol inhibited IL-1 beta-induced stimulation of caspase-3 and cleavage of PARP in human articular chondrocytes in vitro. Ann NY Acad Sci. 2007;1095:554–563. doi: 10.1196/annals.1397.060. [DOI] [PubMed] [Google Scholar]
  • 32.Rubiolo JA, Mithieux G, Vega FV. Resveratrol protects primary rat hepatocytes against oxidative stress damage: activation of the Nrf2 transcription factor and augmented activities of antioxidant enzymes. Eur J Pharmacol. 2008;591:66–72. doi: 10.1016/j.ejphar.2008.06.067. [DOI] [PubMed] [Google Scholar]
  • 33.Robb EL, Winkelmolen L, Visanji N, Brotchie J, Stuart JA. Dietary resveratrol administration increases MnSOD expression and activity in mouse brain. Biochem Biophys Res Commun. 2008;372:254–259. doi: 10.1016/j.bbrc.2008.05.028. [DOI] [PubMed] [Google Scholar]
  • 34.Kaindl U, Eyberg I, Rohr-Udilov N, Heinzle C, Marian B. The dietary antioxidants resveratrol and quercetin protect cells from exogenous pro-oxidative damage. Food Chem Toxicol. 2008;46:1320–1326. doi: 10.1016/j.fct.2007.09.002. [DOI] [PubMed] [Google Scholar]
  • 35.Goh SSC, Woodman OL, Pepe S, Cao AH, Qin C, Ritchie RH. The red wine antioxidant resveratrol prevents cardiomyocyte injury following ischemia-reperfusion via multiple sites and mechanisms. Antioxid Redox Signal. 2007;9:101–113. doi: 10.1089/ars.2007.9.101. [DOI] [PubMed] [Google Scholar]
  • 36.Stojanovic S, Sprinz H, Brede O. Efficiency and mechanism of the antioxidant action of trans-resveratrol and its analogues in the radical liposome oxidation. Arch Biochem Biophys. 2001;391:79–89. doi: 10.1006/abbi.2001.2388. [DOI] [PubMed] [Google Scholar]
  • 37.Szewczuk LM, Lee SH, Blair IA, Penning TM. Viniferin formation by COX-1; evidence for radical intermediates during co-oxidation of resveratrol. J Nat Prod. 2005;68:36–42. doi: 10.1021/np049702i. [DOI] [PubMed] [Google Scholar]
  • 38.Li T, Wang W, Li T. Effect of resveratrol on JAK/STAT3 signal transduction pathway in leukemia. Zhongguo Shi Yan Xue Ye Za Zhi. 2008;16:772–776. [PubMed] [Google Scholar]
  • 39.Nicolini G, Rigolio R, Scuteri A, Miloso M, Saccomanno D, Cavaletti G, et al. Effect of trans-resveratrol on signal transduction pathways in paclitaxel-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neurochem Int. 2003;42:419–429. doi: 10.1016/s0197-0186(02)00132-8. [DOI] [PubMed] [Google Scholar]
  • 40.Woo JH, Lim JH, Kim YH, Suh SI, Min DS, Chang JS, et al. Resveratrol inhibits phorbol myristate acetate-induced matrix metalloproteinase-9 expression by inhibiting JNK and PKCdelta signal transduction. Oncogene. 2004;23:1845–1853. doi: 10.1038/sj.onc.1207307. [DOI] [PubMed] [Google Scholar]
  • 41.Kundu Jk, Shin YK, Surh YJ. Resveratrol modulates phorbol ester-induced pro-inflammatory signal transduction pathways in mouse skin in vivo: NFkappaB and AP-1 as prime targets. Biochem Pharmacol. 2006;72:1506–1515. doi: 10.1016/j.bcp.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 42.Fulda S, Debatin KM. Resveratrol modulation of signal transduction in apoptosis and cell survival: mini-review. Cancer Detec Prev. 2006;30:217–223. doi: 10.1016/j.cdp.2006.03.007. [DOI] [PubMed] [Google Scholar]
  • 43.Kundu JK, Surh YJ. Cancer chemopreventive and therapeutic potential of resveratrol: mechanistic perspectives. Cancer Lett. 2008;269:243–261. doi: 10.1016/j.canlet.2008.03.057. [DOI] [PubMed] [Google Scholar]
  • 44.Venkatesan B, Ghosh-Choudhury N, Das F, Mahimainathan L, Kamat A, Kasinath BS, et al. Resveratrol inhibits PDGE receptor mitogenic signaling in mesangial cells: role of PTP1B. FASEB J. 2008;22:3469–3482. doi: 10.1096/fj.08-109488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Harikumar KB, Aggarwal BB. Resveratrol: a multitargeted agent for age-associated chronic diseases. Cell Cycle. 2008;7:1020–1035. doi: 10.4161/cc.7.8.5740. [DOI] [PubMed] [Google Scholar]
  • 46.Cecchinato V, Chiaramonte R, Nizzardo M, Cristofaro B, Basile A, Sherbet GV, et al. Resveratrol-induced apoptosis in human T-cell acute lymphoblastic leukaemia MOLT-4 cells. Biochem Pharmacol. 2007;74:1568–1574. doi: 10.1016/j.bcp.2007.08.001. [DOI] [PubMed] [Google Scholar]
  • 47.Filomeni G, Graziani I, Rotilio G. Trans-Resveratrol induces apoptosis in human breast cancer cells MCF-7 by the activation of MAP kinases pathways. Genes Nutr. 2007;2:295–305. doi: 10.1007/s12263-007-0059-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Delmas D, Rébé C, Lacour S, Filomenko R, Athias A, Gambert P, et al. Resveratrol-induced apoptosis is associated with Fas redistribution in the Rafts and the formation of a death-inducing signal complex in colon cancer cells. J Biol Chem. 2003;278:41482–41490. doi: 10.1074/jbc.M304896200. [DOI] [PubMed] [Google Scholar]
  • 49.Scarlatti F, Sala G, Somenzi G, Signorelli P, Sacchi N, Ghidoni R. Resveratrol induces growth inhibition and apoptosis in metastatic breast cancer cells via de novo ceramide signaling. FASEB J. 2003;17:2339–2341. doi: 10.1096/fj.03-0292fje. [DOI] [PubMed] [Google Scholar]
  • 50.Bhardwaj A, Sethi G, Vardhan-raj S, Bueso-Ramos C, Takada Y, Gaur U, et al. Resveratrol inhibits proliferation, induces apoptosis, and overcomes chemoresistance through downregulation of STAT3 and nuclear factor-kB-regulated antiapoptotic and cell survival gene products in human multiple myeloma cells. Blood. 2007;109:2293–2302. doi: 10.1182/blood-2006-02-003988. [DOI] [PubMed] [Google Scholar]
  • 51.She Q-B, Bode AM, Ma W-Y, Chen N-Y, Dong Z. Resveratrol-induced activation of p53 and apoptosis is mediated by extracellular-signal-regulated protein kinases and p38 kinase. Cancer Res. 2001;61:1604–1610. [PubMed] [Google Scholar]
  • 52.Kotha A, Sekharam M, Cilenti L, Siddiquee K, Khaled A, Zervos AS, et al. Resveratrol inhibits Src and Atat3 signaling and induces the apoptosis of malignant cells containing activated Stat3 protein. Mol Cancer Ther. 2006;5:621–629. doi: 10.1158/1535-7163.MCT-05-0268. [DOI] [PubMed] [Google Scholar]
  • 53.Fulda S, Debatin K-M. Resveratrol-mediated sensitization to TRAIL-induced apoptosis depends on death receptor and mitochondrial signaling. Eur J Cancer. 2005;41:786–798. doi: 10.1016/j.ejca.2004.12.020. [DOI] [PubMed] [Google Scholar]
  • 54.Li Y, Liu J, Liu X, Xing K, Wang Y, Li F, et al. Resveratrol-induced cell inhibition of growth and apoptosis in MCF7 human breast cancer cells are associated with modulation of phosphorylated Akt and Caspase-9. Appl Biochem Biotech. 2006;135:181–192. doi: 10.1385/abab:135:3:181. [DOI] [PubMed] [Google Scholar]
  • 55.Su J-L, lin M-T, Hong C-C, Chang A-C, Shiah S-G, Wu C-W, et al. Resveratrol induces FasL-related apoptosis through Cdc42 activation of ASK1/JNK-dependent signaling pathway in human leukemia HL-60 cells. Carcinogenesis. 2005;26:1–10. doi: 10.1093/carcin/bgh220. [DOI] [PubMed] [Google Scholar]
  • 56.Aziz MH, Nihal M, Fu VX, Jarrard DF, Ahmad N. Resveratrol-caused apoptosis of human prostate carcinoma LNCaP cells is mediated via modulation of phosphatidylinositol 3′-kinase/Akt pathway and Bcl-2 family proteins. Mol Cancer Ther. 2006;5:1335–1341. doi: 10.1158/1535-7163.MCT-05-0526. [DOI] [PubMed] [Google Scholar]
  • 57.Ahmad N, Adhami VM, Afaq F, Feyes DK, Mukhtar H. Resveratrol causes WAF-1/p21-mediated G1-phase arrest of cell cycle and induction of apoptosis in human epidermoid carcinoma A431 cells. Clin Cancer Res. 2001;7:1466–1473. [PubMed] [Google Scholar]
  • 58.Hwang J-T, Kwak DW, lin SK, Kim HM, Kim YM, Park OJ. Resveratrol induces apoptosis in chemoresistant cancer cells via modulation of AMPK signaling pathway. Ann NY Acad Sci. 2007;1095:441–448. doi: 10.1196/annals.1397.047. [DOI] [PubMed] [Google Scholar]
  • 59.Kutuk O, Poli G, Basaga H. Resveratrol protects against 4-hydroxynonenal-induced apoptosis by blocking JNK and c-JUN/AP-1 signaling. Toxicol Sci. 2006;90:120–132. doi: 10.1093/toxsci/kfj055. [DOI] [PubMed] [Google Scholar]
  • 60.Zhang P, Li H, Wu M-L, Chen X-Y, Kong Q-Y, Wang X-W, et al. c-Myc downregulation: a critical molecular event in resveratrol-induced cell cycle arrest and apoptosis of human medulloblastoma cells. J Neurooncol. 2006;80:123–131. doi: 10.1007/s11060-006-9172-7. [DOI] [PubMed] [Google Scholar]
  • 61.Lin H-Y, Tang H-Y, Keating T, Wu Y-H, Shih A, Hammond D, et al. Resveratrol is pro-apoptotic and thyroid hormone is anti-apoptotic in glioma cells: both actions are intergrin and ERK mediated. Carcinogenesis. 2008;29:62–69. doi: 10.1093/carcin/bgm239. [DOI] [PubMed] [Google Scholar]
  • 62.Alkhalf M, El-Mowafy A, Rachid O, Ali A, Al-Attyiah R. Resveratrol-induced apoptosis in human breast cancer cells is mediated primarily through the Caspase-3-dependent pathway. Arch Med Res. 2008;39:162–168. doi: 10.1016/j.arcmed.2007.09.003. [DOI] [PubMed] [Google Scholar]
  • 63.Clément M-V, Hirpara JL, Chawdhury S-H, Pervaiz S. Chemopreventive agent resveratrol, a natural product derived from grapes, triggers CD95 signaling-dependent apoptosis in human tumor cells. Blood. 1998;92:996–1002. [PubMed] [Google Scholar]
  • 64.Martín AR, Villegas I, Sánchez-Hidalgo M, de la Lastra CA. The effects of resveratrol, a phytoalexin derived from red wines, on chronic inflammation induced in an experimentally induced colitis model. Brit J Pharmacol. 2006;147:873–885. doi: 10.1038/sj.bjp.0706469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Cecchinato V, Chiaramonte R, Nizzardo M, Cristofaro B, Basile A, Sherbet GV, et al. Resveratrol-induced apoptosis in human T-cell acute lymphoblastic leukaemia MOLT-4 cells. Biochem Pharmacol. 2007;74:1568–1574. doi: 10.1016/j.bcp.2007.08.001. [DOI] [PubMed] [Google Scholar]
  • 66.Kundu JK, Surh Y-J. Molecular basis of chemoprevention by resveratrol: NFκB and AP-1 as potential targets. Mutat Res. 2004;555:65–80. doi: 10.1016/j.mrfmmm.2004.05.019. [DOI] [PubMed] [Google Scholar]
  • 67.Heiss EH, Schilder YDC, Dirsch VM. Chronic treatment with resveratrol induces redox stress- and ataxia telangiectasia-mutated (ATM)-dependent senescence in p53-positive cancer cells. J Biol Chem. 2007;282:26759–26766. doi: 10.1074/jbc.M703229200. [DOI] [PubMed] [Google Scholar]
  • 68.Yu H, Pan C, Zhao S, Wang Z, Zhang H, Wu W. Resveratrol inhibits tumor necrosis factor-α-mediated matrix metalloproteinase-9 expression and invasion of human hepatocellular carcinoma cells. Biomed Pharmacother. 2008;62:366–372. doi: 10.1016/j.biopha.2007.09.006. [DOI] [PubMed] [Google Scholar]
  • 69.Ivanov V, Partridge MA, Johnson GE, Huang SXL, Zhou H, Hei TK. Resveratrol sensitizes melanomas to TRAIL through modulation of antiapoptotic gene expression. Expt Cell Res. 2008;314:1163–1176. doi: 10.1016/j.yexcr.2007.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Yu L-J, Wu M-L, Li H, Chen X-Y, Wang Q, Sun Y, et al. Inhibition of STAT3 expression and signaling in resveratrol-differentiated medulloblastoma cells. Neoplasia. 2008;10:736–744. doi: 10.1593/neo.08304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Alkhalaf M. Resveratrol-induced growth inhibition in MDA-MB-231 breast cancer cells is associated with mitogen-activated protein kinase signaling and protein translation. Eur J Cancer Prevent. 2007;16:334–341. doi: 10.1097/01.cej.0000228413.06471.4c. [DOI] [PubMed] [Google Scholar]
  • 72.Roccaro AM, Leleu X, Sacco A, Moreau A-S, Hatjiharissi E, Jia X, et al. Resveratrol exerts antiproliferative activity and induces apoptosis in Waldenström's macroglobulinemia. Clin Cancer Res. 2008;14:1849–1858. doi: 10.1158/1078-0432.CCR-07-1750. [DOI] [PubMed] [Google Scholar]
  • 73.Saiko P, Szakmary A, Jaeger W, Szekeres T. Resveratrol and its analogs: defense against cancer, coronary disease and neurodegenerative maladies or just a fad? Mutat Res. 2008;658:68–94. doi: 10.1016/j.mrrev.2007.08.004. [DOI] [PubMed] [Google Scholar]
  • 74.Tyagi A, Singh RP, Agarwal C, Siriwardane S, Sclafani RA, Agarwal R. Resveratrol causes Cdc2-tyr15 phosphorylation via ATM/ATR-Chk1/2-Cdc25C carcinoma ovcar-3 cells. Carcinogenesis. 2005;26:1978–1987. doi: 10.1093/carcin/bgi165. [DOI] [PubMed] [Google Scholar]
  • 75.Azios NG, Krishnamoorthy L, Harris M, Cubano LA, Cammer M, Dharmawardane SF. Estrogen and resveratrol regulate Rac and Cdc42 signaling to the actin cytoskeleton of metastatic breast cancer cells. Neoplasia. 2007;9:147–158. doi: 10.1593/neo.06778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Fröjdö S, Cozzone D, Vidal H, Pirola L. Resveratrol is a class IA phosphoinositide 3-kinase inhibitor. Biochem J. 2007;406:511–518. doi: 10.1042/BJ20070236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rahman I, Biswas SK, Kirkham PA. Regulation of inflammation and redox signaling by dietary polyphenols. Biochem Pharmacol. 2006;72:1439–1452. doi: 10.1016/j.bcp.2006.07.004. [DOI] [PubMed] [Google Scholar]
  • 78.Wung BS, Hsu MC, Hsieh CW. Resveratrol suppresses IL-6-induced ICAM-1 gene expression in endothelial cells: effects on the inhibition of STAT3 phosphorylation. Life Sci. 2005;78:389–397. doi: 10.1016/j.lfs.2005.04.052. [DOI] [PubMed] [Google Scholar]
  • 79.Venkatachalem K, Mummidi S, Cortez DM, Prabhu SD, Valente AJ, Chandrasekar B. Resveratrol inhibits high glucose-induced P13/Akt? ERK-dependent interleukin-17 expression in primary mouse cardiac fibroblast. Am J Physiol Heart Circ Physiol. 2008;294:2078–2087. doi: 10.1152/ajpheart.01363.2007. [DOI] [PubMed] [Google Scholar]
  • 80.Moon S-O, Kim W, Sung MJ, Lee S, Kang KP, Kim DH, et al. Resveratrol suppresses tumor necrosis factor-α-induced fractalkine expression in endothelial cells. Mol Pharmacol. 2006;70:112–119. doi: 10.1124/mol.106.022392. [DOI] [PubMed] [Google Scholar]
  • 81.Csaki C, Keshishzadeh N, Fischer K, Shakibaei M. Regulation of inflammation signaling by resveratrol in human chondrocytes in vitro. Biochem Pharmacol. 2008;75:677–687. doi: 10.1016/j.bcp.2007.09.014. [DOI] [PubMed] [Google Scholar]
  • 82.Kundu JK, Shin YK, Surh Y-J. Resveratrol modulates phorbol ester induced pro-inflammatory signal transduction pathways in mouse skin in vivo: NFκB and AP1 as prime targets. Biochem Pharmacol. 2006;72:1506–1515. doi: 10.1016/j.bcp.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 83.Youn HS, Lee JY, Fitzgerald KA, Young HA, Akira S. Specific inhibition of MyD88-independent signaling pathways of TLR3 and TLR4 by resveratrol: molecular targets are TBK1 and RIP1 in TRIF complex. J Immunol. 2005;175:3339–3346. doi: 10.4049/jimmunol.175.5.3339. [DOI] [PubMed] [Google Scholar]
  • 84.Hwang J-T, Kwon DY, Park OJ, Kim MS. Resveratrol protects ROS-induced cell death by activating AMPK in H9c2 cardiac muscle cells. Genes Nutr. 2008;2:323–326. doi: 10.1007/s12263-007-0069-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Csiszar A, Smith K, Labinskyy N, Orosz Z, Rivera A, Ungvsri Z. Resveratrol attenuates TNFα-induced activation of coronary arterial endothelial cells: role of NFκB inhibition. Am J Physiol Heart Circ Physiol. 2006;291:1694–1699. doi: 10.1152/ajpheart.00340.2006. [DOI] [PubMed] [Google Scholar]
  • 86.Huang Z, Wang C, Wei L, Wang J, Fan Y, Wang L, et al. Resveratrol inhibits EMMMPRIN expression via P38 and ERK1/2 pathways in PMA-induced THP-1 cells. Biochem Biophys Res Commun. 2008;374:517–521. doi: 10.1016/j.bbrc.2008.07.058. [DOI] [PubMed] [Google Scholar]
  • 87.Das S, Tosaki A, Bagchi D, Maulik N, Das DK. Potentiation of a survival signal in the ischemic heart by resveratrol through p38 mitogen-activated protein kinase/mitogen-and stress-activated protein kinase 1/cAMP response element-binding protein signaling. J Pharmacol Expt Therapeut. 2006;317:980–988. doi: 10.1124/jpet.105.095133. [DOI] [PubMed] [Google Scholar]
  • 88.Das S, Cordis GA, Maulik N, Das DK. Pharmacological preconditioning with resveratrol: role of CREB-dependent Bcl-2 signaling via adenosine A3 receptor activation. Am J Physiol Heart Circ Physiol. 2005;288:328–335. doi: 10.1152/ajpheart.00453.2004. [DOI] [PubMed] [Google Scholar]
  • 89.Das S, Fraga CG, Das DK. Cardioprotective effect of resveratrol via HO-1 expression involves p38 map kinase and P13-kinase signaling, but does not involve NFκB. Free Rad Res. 2006;40:1066–1075. doi: 10.1080/10715760600833085. [DOI] [PubMed] [Google Scholar]
  • 90.Haider UGB, Sorescu D, Griendling KK, Vollmar AM, Dirsch VM. Resveratrol suppresses angiotensin II-induced Akt/proten kinase B and p70 S6 kinase phosphorylation and subsequent hypertrophy in rat aortic smooth muscle cells. Mol Pharmacol. 2002;62:772–777. doi: 10.1124/mol.62.4.772. [DOI] [PubMed] [Google Scholar]
  • 91.Chen C-Y, Jang J-H, Li M-H, Surh Y-J. Resveratrol upregulates heme oxygenase-1 expression via activation of NF-E2-related factor 2 in PC12 cells. Biochem Biophys Res Commun. 2005;331:993–1000. doi: 10.1016/j.bbrc.2005.03.237. [DOI] [PubMed] [Google Scholar]
  • 92.King RE, Kent KD, Bomser JA. Resveratrol reduces oxidation and proliferation of human epithelial cells via extracellular signal-regulated kinase inhibition. Chem Biol Interact. 2005;151:243–249. doi: 10.1016/j.cbi.2004.11.003. [DOI] [PubMed] [Google Scholar]
  • 93.Breen DM, Sanil T, Giacca A, Tsiani E. Stimulation of muscle cell glucose uptake by resveratrol through sirtuins and AMPK. Biochem Biophys Res Commun. 2008;374:117–122. doi: 10.1016/j.bbrc.2008.06.104. [DOI] [PubMed] [Google Scholar]
  • 94.Kode A, Rajendrasozhan S, Caito S, Yang S-R, Megson IL, Rahman I. Resveratrol induces glutathione synthesis by activation of Nrf2 and protects against cigarette smoke-mediated oxidative stress in human lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2008;294:478–488. doi: 10.1152/ajplung.00361.2007. [DOI] [PubMed] [Google Scholar]
  • 95.Ajmo JM, Liang X, Rogers CQ, Pennock B, You M. Resveratrol alleviates alcoholic fatty liver in mice. Am J Gastroint Liver Physiol. 2008;295:833–842. doi: 10.1152/ajpgi.90358.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Stefani M, Markus MA, Lin RCY, Pinese M, Dawes IW, Morris BJ. The effect of resveratrol on cell model of human aging. Ann NY Acad Sci. 2007;1114:407–418. doi: 10.1196/annals.1396.001. [DOI] [PubMed] [Google Scholar]
  • 97.Shakibaei M, John T, Seifarth C, Mobasheri A. Resveratrol inhibits IL-1β-induced stimulation of caspase-3 and cleavage of PARP in human articular chondrocytes in vitro. Ann NY Acad Sci. 2007;1095:554–563. doi: 10.1196/annals.1397.060. [DOI] [PubMed] [Google Scholar]
  • 98.Godichaud S, Si-Tayeb K, Augé N, Desmouliére A, Balabaud C, Payrastre B, et al. The grape-derived polyphenol resveratrol differently affects epidermal and platelet-derived growth factor signaling in human liver myofibroblasts. Int J Biochem Cell Biol. 2006;38:629–637. doi: 10.1016/j.biocel.2005.11.001. [DOI] [PubMed] [Google Scholar]
  • 99.Leiro J, Arranz JA, Fraiz N, Sanmartin ML, Quezada E, Orallo F. Effect of cis-resveratrol on genes involved in nuclear factor kappaB signaling. Int Immunopharmacol. 2005;5:393–406. doi: 10.1016/j.intimp.2004.10.006. [DOI] [PubMed] [Google Scholar]
  • 100.Poolman TM, Ng LL, Farmer PB, Manson MM. Inhibition of the respiratory burst by resveratrol in human monocytes: correlation with inhibition of P13K signaling. Free Radic Biol Med. 2005;39:118–132. doi: 10.1016/j.freeradbiomed.2005.02.036. [DOI] [PubMed] [Google Scholar]
  • 101.Dasgupta B, Milbrandt J. Resveratrol stimulates AMP kinase activity in neurons. PNAS. 2007;104:7217–7222. doi: 10.1073/pnas.0610068104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Klinge CM, Blankenship KA, Risingert KE, Bhatnagar S, Noisin EL, Sumanasekara WK, et al. Resveratrol and estradiol rapidly activate MAPK signaling through estrogen receptors α and β in endothelial cells. J Biol Chem. 2005;280:7460–7468. doi: 10.1074/jbc.M411565200. [DOI] [PubMed] [Google Scholar]
  • 103.Forman HG, Cadenas E. Oxidative Stress and, Signal Transduction. New York: Chapman and Hall; 1997. pp. 1–475. [Google Scholar]
  • 104.Hancock GT. Cell Signaling. New York: Oxford University Press; 2005. pp. 1–296. [Google Scholar]
  • 105.Demple B. Oxidative stress and signal transduction. In: Bradshaw RA, Dennis EA, editors. Handbook of Cell Signaling. Vol. 78. Boston: Academic Press; 2004. pp. 293–307. [Google Scholar]
  • 106.Kovacic P, Pozos RS. Cell signaling (mechanism and reproductive toxicity): redox chains, electrons, relays, conduit, electrochemistry and other medical implications. Birth Defects Res Part C. 2006;28:333–344. doi: 10.1002/bdrc.20083. [DOI] [PubMed] [Google Scholar]
  • 107.Kovacic P. Protein electron transfer (mechanism and reproductive toxicity): iminium, hydrogen bonding, homoconjugation, amino acid side chains and cell signaling. Birth Defects Res. 2007;81:51–64. doi: 10.1002/bdrc.20086. [DOI] [PubMed] [Google Scholar]
  • 108.Kovacic P, Pozos RS, Draskovich CD. Unifying electrostatic mechanism for receptor-ligand activity. J Recept Signal Transduct. 2007;27:411–432. doi: 10.1080/10799890701699686. [DOI] [PubMed] [Google Scholar]
  • 109.Kovacic P, Draskovich CD, Pozos RS. Unifying electrostatic mechanism for phosphates and sulfates in cell signaling. J Recept Signal Transduct. 2007;27:433–443. doi: 10.1080/10799890701699702. [DOI] [PubMed] [Google Scholar]
  • 110.Kovacic P. Unifying electrostatic mechanism for metal cations in receptors and cell signaling. J Recept Signal Transduct. 2008;28:153–162. doi: 10.1080/10799890802084234. [DOI] [PubMed] [Google Scholar]
  • 111.Le Corre L, Fustier P, Chalabi N, Bignon YJ, Bernard-Gallon D. Effects of resveratrol on expression of panel of genes interacting with the BRCA1 oncosuppressor in human breast cancer cell lines. Clin Chim Acta. 2004;344:115–121. doi: 10.1016/j.cccn.2004.02.024. [DOI] [PubMed] [Google Scholar]
  • 112.Hambrock A, Franz CBDO, Hiller S, Grenz A, Ackerman S, Schulze DU, et al. Resveratrol binds to the sulfonylurea receptor (SUR) and induces apoptosis in a SUR subtype-specific manner. J Biol Chem. 2007;282:3347–3356. doi: 10.1074/jbc.M608216200. [DOI] [PubMed] [Google Scholar]
  • 113.Gao S, Liu GZ, Wang Z. Modulation of androgen receptor-dependent transcription by resveratrol and genistein in prostate cancer cells. Prostate. 2004;59:214–225. doi: 10.1002/pros.10375. [DOI] [PubMed] [Google Scholar]
  • 114.Shi WF, Leong M, Cho E, Farrell J, Chen HC, Tian J, Zhang D. Repressive effects of resveratrol on androgen receptor transcriptional activity. PLOS One. 2009;4:7398. doi: 10.1371/journal.pone.0007398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Harada N, Murata Y, Yamji R, Miura T, Inui H, Nakano Y. Resveratrol downregulates the androgen receptor at the post-translational level in prostate cancer cells. J Nutr Sci Vitaminol. 2007;53:556–560. doi: 10.3177/jnsv.53.556. [DOI] [PubMed] [Google Scholar]
  • 116.Mitchell SH, Zhu, Young CY. Resveratrol inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells. Cancer Res. 1999;59:5892–5895. [PubMed] [Google Scholar]
  • 117.Casper RF, Quesne M, Rogers IM, Shirota T, Jolivet A, Milgrom E, Savouret J-F. Resveratrol has antagonist activity on the aryl hydrocarbon receptor: implications for prevention of dioxin toxicity. Mol Pharmacol. 1999;56:784–790. [PubMed] [Google Scholar]
  • 118.Jakab M, Lach S, Bacová Z, Langelüddecke C, Strbák V, Schmidt S, et al. Resveratrol inhibits electrical activity and insulin release from insulinoma cells by block of voltage-gated Ca+ channels and swelling-dependent Cl− currents. Cell Physiol Biochem. 2008;22:567–578. doi: 10.1159/000185541. [DOI] [PubMed] [Google Scholar]
  • 119.Wang R, Xiao L, Ma HJ, Zhang LH, He RR, Wu YM, et al. Resveratrol inhibits electrical activity of paraventricular nucleus neurons in rat hypothalamic slices. Acta Physologica Sinica. 2008;60:279–283. [PubMed] [Google Scholar]
  • 120.Li M, Wang QS, Chen Y, Wang ZM, Liu Z, Guo SM, et al. Resveratrol inhibits the electrical activity of subfornical organ neurons in rat. Acta Physiologica Sinica. 2005;57:523–528. [PubMed] [Google Scholar]
  • 121.Buluc M, Ayaz M, Turan B, Demirel-Yilmaz E. Resveratrol-induced depression of the mechanical and electrical activities of the rat heart is reversed by glyburide: evidence for possible K(ATO) channels activation. Arch Pharm Res. 2007;30:603–607. doi: 10.1007/BF02977655. [DOI] [PubMed] [Google Scholar]
  • 122.Kovacic P, Hall ME. Bioelectrochemistry, reactive oxygen species, receptor and cell signaling: How interrelated? J Recept Signal Tranduct. 2010;30:1–9. doi: 10.3109/10799890903517939. [DOI] [PubMed] [Google Scholar]
  • 123.Markus MA, Morris BJ. Resveratrol in prevention and treatment of common conditions of aging. Clin Interv Aging. 2008;3:331–339. [PMC free article] [PubMed] [Google Scholar]
  • 124.Barger JL, Kayo T, Vann JM, Arias EB, Wang J, Hacker TA, et al. A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS One. 2008;3:2264. doi: 10.1371/journal.pone.0002264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Baxter RA. Anti-aging properties of resveratrol: review and report of potent new antioxidant skin care formulation. J Cosmet Dermatol. 2008;7:2–7. doi: 10.1111/j.1473-2165.2008.00354.x. [DOI] [PubMed] [Google Scholar]
  • 126.Valenzano DR, Cellerino A. Resveratrol and the pharmacology of aging: a new vertebtrate model to validate an old molecule. Cell Cycle. 2006;5:1027–1032. doi: 10.4161/cc.5.10.2739. [DOI] [PubMed] [Google Scholar]
  • 127.Labinskyy N, Csiszar A, Veress G, Stef G, Pacher P, Oroszi G, et al. Vascular dysfunction in aging: potential effects of resveratrol, an anti-inflammatory phytoestrogen. Curr Med Chem. 2006;13:989–996. doi: 10.2174/092986706776360987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Jnutson MD, Leeuwenburgh C. Resveratrol and novel potent activators of SIRT1: effects on aging and age-related diseases. Nutr Rev. 2008;66:591–596. doi: 10.1111/j.1753-4887.2008.00109.x. [DOI] [PubMed] [Google Scholar]
  • 129.Sengottuvelan M, Viswanathan P, Nalini N. Chemopreventive effect of trans-resveratrol-1 phytoalexin against colonic aberrant crypt foci and cell proliferation in 1,2-dimethylhydrazine induced colon carcinogenesis. Carcinogenesis. 2006;27:1038–1046. doi: 10.1093/carcin/bgi286. [DOI] [PubMed] [Google Scholar]
  • 130.Athar M, Back JH, Tang X, Kim KH, Kopelovich L, Bickers DR, et al. Resveratrol: a review of preclinical studies for human cancer prevention. Toxicol Appl Pharmacol. 2007;224:274–283. doi: 10.1016/j.taap.2006.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Chan W-H. Effect of resveratrol on high glucose-induced stress in human leukemia K562 cells. J Cellular Biochem. 2005;94:1267–1279. doi: 10.1002/jcb.20408. [DOI] [PubMed] [Google Scholar]
  • 132.Jang M, Pezzuto JM. Effect of resveratrol on 12-O-tetradecanolphenol-13-acetate-induced oxidative events and gene expression in mouse skin. Cancer Lett. 1998;134:81–89. doi: 10.1016/s0304-3835(98)00250-x. [DOI] [PubMed] [Google Scholar]
  • 133.Kozuki Y, Miura Y, Yagasaki K. Resveratrol suppresses hepatoma cell invasion independently of its anti-proliferation action. Cancer Lett. 2001;167:151–156. doi: 10.1016/s0304-3835(01)00476-1. [DOI] [PubMed] [Google Scholar]
  • 134.Le Corre L, Chalabi N, Delort L, Bignon Y-J, Bernard-Gallon DJ. Resveratrol and breast cancer chemoprevention: molecular mechanisms. Mol Nutr Food Res. 2005;49:462–471. doi: 10.1002/mnfr.200400094. [DOI] [PubMed] [Google Scholar]
  • 135.Bhat KPL, Pezzuto JM. Cancer chemopreventive activity of resveratrol. Ann NY Acad Sci. 2002;957:210–229. doi: 10.1111/j.1749-6632.2002.tb02918.x. [DOI] [PubMed] [Google Scholar]
  • 136.Ratan HL, Steward WP, Gescher AJ, Mellon JK. Resveratrol-a prostate cancer chemopreventive agent? Urologic Oncolo. 2002;7:223–227. doi: 10.1016/s1078-1439(02)00194-1. [DOI] [PubMed] [Google Scholar]
  • 137.Sun W, Wang W, Kim J, Keng P, Yang S, Zhang H, et al. Anti-cancer effect of resveratrol is associated with induction of apoptosis via a mitochondrial pathway alignment. Adv Exp Med Biol. 2008;614:179–186. doi: 10.1007/978-0-387-74911-2_21. [DOI] [PubMed] [Google Scholar]
  • 138.Signorelli P, Ghidoni R. Resveratrol as an anticancer nutrient: molecular basis, open questions and promises. J Nutr Biochem. 2005;16:449–466. doi: 10.1016/j.jnutbio.2005.01.017. [DOI] [PubMed] [Google Scholar]
  • 139.Joe AK, Liu H, Suzui M, Vural ME, Xiao D, Weinstein B. Resveratrol induces growth inhibition, S-phase arrest, apoptosis, and cell changes in biomarker expression in several human cancer cell lines. Clin Cancer Res. 2002;8:893–903. [PubMed] [Google Scholar]
  • 140.Shanker S, Siddiqui I, Srivastava RK. Molecular mechanisms of resveratrol (3,4,5-trihydroxy-trans-stilbene) and its interaction with TNF-related apoptosis inducing ligand (TRAIL) in androgen-insensitive prostate cancer cells. Mol Cell Biochem. 2007;304:273–285. doi: 10.1007/s11010-007-9510-x. [DOI] [PubMed] [Google Scholar]
  • 141.Puissant A, Grosso S, Jacquel A, Belhacene N, Colosetti P, Cassuto J-P, et al. Imatinib mesylate-resistant human chronic myelogenous leukemia cell line exhibit high sensitivity to the phytoalexin resveratrol. FASEB J. 2008;22:1894–1904. doi: 10.1096/fj.07-101394. [DOI] [PubMed] [Google Scholar]
  • 142.Filomeni G, Graziani I, Rorilio G, Ciriolo MR. trans-Resveratrol induces apoptosis in human breast cancer cells MCF-7 by the activation of MAP kinases pathways. Genes Nutr. 2007;2:295–305. doi: 10.1007/s12263-007-0059-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Tang H-Y, Shih A, Cao J, Davis FB, Lin H-Y. Resveratrol-induced cyclooxygenase-2-facilitates p53-dependent apoptosis in human breast cancer cells. Mol Cancer Ther. 2006;5:2034–2042. doi: 10.1158/1535-7163.MCT-06-0216. [DOI] [PubMed] [Google Scholar]
  • 144.Mohan J, Gandhi AA, Bhavya AV, Rashmi R, Karunagaran D, Indu R, et al. Caspase-2 triggers Bax-Bak-dependent and -independent cell death in colon cancer cells treated with resveratrol. J Biol Chem. 2006;281:17599–17611. doi: 10.1074/jbc.M602641200. [DOI] [PubMed] [Google Scholar]
  • 145.Atten MJ, Godoy-Romero E, Attar BM, Milson T, Zopel M, Holian O. Resveratrol regulates cellular PKCa and d to inhibit growth and induces apoptosis in gastric cancer cells. Invest New Drugs. 2005;23:111–119. doi: 10.1007/s10637-005-5855-8. [DOI] [PubMed] [Google Scholar]
  • 146.Shih A, Davis FB, Lin H-Y, Davis PJ. Resveratrol induces apoptosis in thyroid cancer cell lines via a MAPK-and p53-dependent mechanism. J Clin Endocrinol Metabol. 2002;87:1223–1232. doi: 10.1210/jcem.87.3.8345. [DOI] [PubMed] [Google Scholar]
  • 147.Ahmad KA, Clement M-V, Hanif IM, Pervez S. Resveratrol inhibits drug-induced apoptosis in human leukemia cells by creating an intracellular milieu non-permissive for death execution. Cancer Res. 2004;64:1452–1459. doi: 10.1158/0008-5472.can-03-2414. [DOI] [PubMed] [Google Scholar]
  • 148.Johnson GE, Ivonov VN, Hei TK. Radiosensitization of melanoma cells through combined inhibition of protein regulators of cell survival. Int J Programmed Cell Death. 2008;13:790–802. doi: 10.1007/s10495-008-0212-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Shao J, Li X, Lu X, Jiang C, Hu Y, Li Q, et al. Enhanced growth inhibition effect resveratrol incorporated into biodegradable nanoparticles against glioma cells is mediated by the induction of intracellualr reactive oxygen species levels. Colloids Surf B; Biointerfaces. 2009;72:40–47. doi: 10.1016/j.colsurfb.2009.03.010. [DOI] [PubMed] [Google Scholar]
  • 150.Stewart JR, O'Brian CA. Resveratrol antagonized EGFR-dependent Erk1/2 activation in human androgen-independent prostate cancer cells with associated isozyme-selective PKCα inhibition. Invest New Drugs. 2004;22:107–117. doi: 10.1023/B:DRUG.0000011787.75522.ec. [DOI] [PubMed] [Google Scholar]
  • 151.Golkar L, Ding X-Z, Ujiki MB, Salabat MR, Kelly DL, Scholtens D, et al. Resveratrol inhibits pancreatic cancer cell proliferation through transcriptional induction of macrophage inhibitory cytokine-1. J Surg Res. 2007;138:163–169. doi: 10.1016/j.jss.2006.05.037. [DOI] [PubMed] [Google Scholar]
  • 152.Harper CE, Patel BB, Wang J, Arabshahi A, Eltoum IA, Lamartiniere A. Resveratrol suppresses prostate cancer progression in transgenic mice. Carcinogenesis. 2007;28:1946–1953. doi: 10.1093/carcin/bgm144. [DOI] [PubMed] [Google Scholar]
  • 153.Hudson TS, Hartle DK, Hursting SD, Nunez NP, Wang TTY, Young HA, et al. Inhibition of prostate cancer growth by muscadine grape skin extract and resveratrol through distinct mechanisms. Cancer Res. 2007;67:8396–8405. doi: 10.1158/0008-5472.CAN-06-4069. [DOI] [PubMed] [Google Scholar]
  • 154.Benitez DA, Pozo-Guisado E, Clementi M, Castellón E, Fernandez-Salguero PM. Non-genomic action of resveratrol on androgen and oestrogen receptors in prostate cancer: modulation of the phosphoinosotide 3-kinase pathway. Brit J Cancer. 2007;96:1595–1604. doi: 10.1038/sj.bjc.6603755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Aggarwal BB, Bhardwaj A, Aggarwal RS, Seeram NP, Shishodia S, Takada Y. Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies. Anticancer Res. 2004;24:2783–2840. [PubMed] [Google Scholar]
  • 156.Lin HY, Sun M, Tang HY, Simone TM, Wu YH, Grandis JR, et al. Resveratrol causes COX-2-and p53-dependent apoptosis in head and neck squamous cell cancer cells. J Cell Biochem. 2008;104:2131–2142. doi: 10.1002/jcb.21772. [DOI] [PubMed] [Google Scholar]
  • 157.Hope C, Planutis K, Planutiene M, Moyer MP, Johal KS, Woo J, et al. Low concentration of resveratrol inhibit Wnt signal throughput in colon-derived cells: implications for colon cancer prevention. Mol Nutr Food Res. 2008;52:52–61. doi: 10.1002/mnfr.200700448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Gill C, Walsh SE, Morrissey C, Fitzpatrick JM, Watson RW. Resveratrol sensitizes androgen independent prostate cancer cells to death-receptor mediated apoptosis through multiple mechanisms. Prostate. 2007;67:1641–1653. doi: 10.1002/pros.20653. [DOI] [PubMed] [Google Scholar]
  • 159.Kovacic P. Electron transfer mechanism for regulatory action of nitic oxide. Bioelectrochem Bioenerg. 1996;39:155–159. [Google Scholar]
  • 160.Holian O, Wahid S, Atten MJ, Attar BM. Inhibition of gastric cancer cell proliferation by resveratrol; role of nitric oxide. Am J Physiol Gastrointest Liver Physiol. 2002;282:809–816. doi: 10.1152/ajpgi.00193.2001. [DOI] [PubMed] [Google Scholar]
  • 161.Gusman J, Malonne H, Atassi G. A reappraisal of the potential chemopreventive and chemotherapeutic properties of resveratrol. Carcinogenesis. 2001;22:1111–1117. doi: 10.1093/carcin/22.8.1111. [DOI] [PubMed] [Google Scholar]
  • 162.Stewart JR, Artime MC, O'Brian CA. Resveratrol: a candidate nutritional substance for prostate cancer prevention. J Nutr. 2003;133:2440–2443. doi: 10.1093/jn/133.7.2440S. [DOI] [PubMed] [Google Scholar]
  • 163.Zhang Q, Tang X, Lu QY, Zhang ZF, Brown J, Le AD. Resveratrol inhibits hypoxia-induced accumulation of hypoxia-inducible factor-1α and VEGF expression in human tongue squamous cell carcinoma and hepatoma cells. Mol Cancer Ther. 2005;4:1465–1474. doi: 10.1158/1535-7163.MCT-05-0198. [DOI] [PubMed] [Google Scholar]
  • 164.Wolter F, Ulrich S, Stein J. Molecular mechanisms of the chemopreventive effects of resveratrol and its analogs in colorectal cancer: key role of polyamines? J Nutr. 2004;134:3219–3222. doi: 10.1093/jn/134.12.3219. [DOI] [PubMed] [Google Scholar]
  • 165.Wang TTY, Hudson TS, Wang T-C, Remsberg CM, Davis NM, Takahashi Y, et al. Differential effects of resveratrol on androgen-responsive LNCaP human protate cancer cells in vitro and in vivo. Carcinogenesis. 2008;29:2001–2010. doi: 10.1093/carcin/bgn131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Tang F-Y, Chiang E-PI, Sun Y-C. Resveratrol inhibits heregulin-β1-mediated matrix metalloproteinase-9 expression and cell invasion in human breast cancer cells. J Nutr Biochem. 2008;19:287–294. doi: 10.1016/j.jnutbio.2007.03.003. [DOI] [PubMed] [Google Scholar]
  • 167.Scarlatti F, Maffei R, Beau I, Codogno P, Ghidoni R. Role of non-canonical Beclin 1-independent autophagy in cell death induced by resveratrol in human breast cancer cells. Cell Death Differentiation. 2008;15:1318–1329. doi: 10.1038/cdd.2008.51. [DOI] [PubMed] [Google Scholar]
  • 168.Nakagawa H, Kiyozuka Y, Uemura Y, Senzaki H, Shikata N, Hioki K, et al. Resveratrol inhibits human breast cancer cell growth and may mitigate the effect of linoleic acid, a potent breast cancer cell stimulator. J Cancer Res Clin Oncol. 2001;127:258–264. doi: 10.1007/s004320000190. [DOI] [PubMed] [Google Scholar]
  • 169.Melilo G. Targeting hypoxia cell signaling for cancer therapy. Cancer Metastasis Rev. 2007;26:341–352. doi: 10.1007/s10555-007-9059-x. [DOI] [PubMed] [Google Scholar]
  • 170.Trincheri NF, Nicotra G, Follo C, Castino R, Isidoro C. Resveratrol induces cell death in colorectal cancer cells by novel pathway involving lysosomal cathepsin D. Carcinogenesis. 2007;28:922–931. doi: 10.1093/carcin/bgl223. [DOI] [PubMed] [Google Scholar]
  • 171.Kuwajerwala N, Cifuentes E, Gautam S, Menon M, Barrack ER, Reddy GPV. Resveratrol induces prostate cancer cell entry into S phase and inhibits DNA synthesis. Cancer Res. 2002;62:2488–2492. [PubMed] [Google Scholar]
  • 172.van Ginkel PR, Darjatmoko SR, Sareen D, Subramanian L, Bhattacharya S, Lindstrom MJ, et al. Resveratrol inhibits uveal melanoma tumor growth via early mitochondrial dysfunction. Invest Ophthalmol Vis Sci. 2008;49:1299–1306. doi: 10.1167/iovs.07-1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Wolter F, Akoglu B, Clausnitzer A, Stein J. Downregulation of the cyclin D1/Cdk4 complex occurs during resveratrol-induced cell cycle arrest in colon cancer cell lines. J Nutr. 2001;131:2197–2203. doi: 10.1093/jn/131.8.2197. [DOI] [PubMed] [Google Scholar]
  • 174.Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nature Rev Drug Disc. 2006;5:493–506. doi: 10.1038/nrd2060. [DOI] [PubMed] [Google Scholar]
  • 175.Chao H-H, Juan S-H, Liu J-C, Yang H-Y, Yang E, Cheng T-H, et al. Resveratrol inhibits angiotensin II-induced endothelin-1 gene expression and subsequent proliferation in rat aortic smooth muscle cells. Eur J Pharmacol. 2005;515:1–9. doi: 10.1016/j.ejphar.2005.03.035. [DOI] [PubMed] [Google Scholar]
  • 176.Labinskyy N, Csiszar A, Veress G, Stef G, Pacher P, Oroszi G, et al. Vascular dysfunction in aging: potential effects of resveratrol, an anti-inflammatory phytoestrogen. Curr Med Chem. 2006;13:989–996. doi: 10.2174/092986706776360987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Hung L-M, Chen J-K, Huang S-S, Lee R-S, Su M-J. Cardioprotective effect of resveratrol, a natural anti-oxidant derived from grapes. Cardiovascular Res. 2000;47:549–555. doi: 10.1016/s0008-6363(00)00102-4. [DOI] [PubMed] [Google Scholar]
  • 178.Sato M, Ray PS, Maulik G, Maulik N, Engelman RM, Bertelli AAE, et al. Myocardial protection with red wine extract. J Cardiovascular Pharmacol. 2000;35:263–268. doi: 10.1097/00005344-200002000-00013. [DOI] [PubMed] [Google Scholar]
  • 179.Shigematsu S, Ishida S, Hara M, Takahashi N, Yoshimatsu H, Sakata T, et al. Resveratrol, a red wine constituent polyphenol, prevents superoxide-dependent imflammatory response induced by ischemia/reperfusion, platelet-activating factor, or oxidants. Free Radic Biol Med. 2003;34:810–817. doi: 10.1016/s0891-5849(02)01430-2. [DOI] [PubMed] [Google Scholar]
  • 180.Delms D, Jannin B, Latruffe N. Resveratrol: preventing properties against vascular alterations and aging. Mol Nutr Food Res. 2005;49:377–395. doi: 10.1002/mnfr.200400098. [DOI] [PubMed] [Google Scholar]
  • 181.Floreani M, Napoli E, Quintieri L, Palatini P. Oral administration of trans-resveratrol to guinea pigs increases cardiac DT-diaphorase and catalase activities, and protects isolated atria from menadione toxicity. Life Sci. 2003;72:2741–2751. doi: 10.1016/s0024-3205(03)00179-6. [DOI] [PubMed] [Google Scholar]
  • 182.Rotonado S, Rajtar G, Manarini S, Celardo A, Rotillo D, de Gaetano G, et al. Effect of trans-resveratrol, a natural polyphenolic compound, on human polymorphonuclear leukocyte function. Brit J Pharmacol. 1998;123:1691–1699. doi: 10.1038/sj.bjp.0701784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Tedesco I, Russo M, Russo P, Iacomino G, Russo GL, Carraturo A, et al. Antioxidant effect of red wine polyphenols on red blood cells. J Nutr Biochem. 2000;11:114–119. doi: 10.1016/s0955-2863(99)00080-7. [DOI] [PubMed] [Google Scholar]
  • 184.Ungvari Z, Orosz Z, Rivera A, Labinskyy N, Xiangmin Z, Olson S, et al. Resveratrol increases vascular oxidative stress resistance. Am J Physiol Heart Circ Physiol. 2007;292:2417–2424. doi: 10.1152/ajpheart.01258.2006. [DOI] [PubMed] [Google Scholar]
  • 185.Li Y, Cao Z, Zhu H. Upregulation of endogenous antioxidants and phase 2 enzymes by the red wine poly- phenol, resveratrol in cultured aortic smooth muscle cells leads to cytoprotection against oxidative and electrophilic stress. Pharmacol Res. 2006;53:6–15. doi: 10.1016/j.phrs.2005.08.002. [DOI] [PubMed] [Google Scholar]
  • 186.Olas B, Wachowicz B. Resveratrol and vitamin C as antioxidants in blood platelets. Thrombosis Res. 2002;106:143–144. doi: 10.1016/s0049-3848(02)00101-9. [DOI] [PubMed] [Google Scholar]
  • 187.Olas B, Wachowicz B, Saluk-Juszczak J, Zielinki T, Kaca W, Buczynski A. Antioxidant activity of resveratrol in endotoxin-stimulated blood platelets. Cell Biol Toxicol. 2001;17:117–125. doi: 10.1023/a:1010962222305. [DOI] [PubMed] [Google Scholar]
  • 188.Fulda S, Debatin KM. Resveratrol modulation of signal transduction in apoptosis and cell survival: mini-review. Cancer Detec Prev. 2006;30:217–223. doi: 10.1016/j.cdp.2006.03.007. [DOI] [PubMed] [Google Scholar]
  • 189.Olas B, Zbikowska HM, Wachowicz B, Krajewski T, Buczynski A, Magnuszewska A. Inhibitory effect of resveratrol on free radical generation in blood platelets. Acta Biochem Pol. 1999;46:961–966. [PubMed] [Google Scholar]
  • 190.Olas B, Wachowicz B. Resveratrol, a phenolic antioxidant with effects on blood platelet function. Platelets. 2005;16:251–260. doi: 10.1080/09537100400020591. [DOI] [PubMed] [Google Scholar]
  • 191.Zhuang H, Kim Y-S, Koehler RC, Doré S. Potential mechanism by which resveratrol, a red wine constituent, protects neurons. Ann NY Acad Sci. 2003;993:276–286. doi: 10.1111/j.1749-6632.2003.tb07534.x. [DOI] [PubMed] [Google Scholar]
  • 192.Bastianetto S, Quirion R. Natural extracts as possible protective agents of brain aging. Neurobiol Aging. 2002;23:891–897. doi: 10.1016/s0197-4580(02)00024-6. [DOI] [PubMed] [Google Scholar]
  • 193.Mokni M, Elkahoui S, Limam F, Amri M, Aouani E. Effect of resveratrol on antioxidant enzyme activities in the brain of healthy rat. Neurochem Res. 2007;32:981–987. doi: 10.1007/s11064-006-9255-z. [DOI] [PubMed] [Google Scholar]
  • 194.Anekonda TS. Resveratrol-a boon for treating Alzheimer's disease? Brain Res Rev. 2006;52:316–326. doi: 10.1016/j.brainresrev.2006.04.004. [DOI] [PubMed] [Google Scholar]
  • 195.Jang J-H, Surh Y-J. Protective effect of resveratrol on b-amyloid-induced oxidative PC12 cell death. Free Radic Biol Med. 2003;34:1100–1110. doi: 10.1016/s0891-5849(03)00062-5. [DOI] [PubMed] [Google Scholar]
  • 196.Rossi L, Mazzitelli S, Arcello M, Capo CR, Rotillo G. Benefits from dietary polyphenols for brain aging and Alzheimer's disease. Neurochem Res. 2008;33:2390–2400. doi: 10.1007/s11064-008-9696-7. [DOI] [PubMed] [Google Scholar]
  • 197.Sun AY, Simonyi A, Sun GY. The “French paradox” and beyond: neuroprotective effects of polyphenols. Free Radic Biol Med. 2002;32:314–318. doi: 10.1016/s0891-5849(01)00803-6. [DOI] [PubMed] [Google Scholar]
  • 198.Pallás M, Casadesús G, Smith MA, Coto-Montes A, Pelegri C, Vilaplana J, et al. Resveratrol and neurode-generative diseases: activation of SIRT1 as the potential pathway towards neroprotection. Curr Neurovasc Res. 2009;6:70–81. doi: 10.2174/156720209787466019. [DOI] [PubMed] [Google Scholar]
  • 199.Rocha-González HI, Ambriz-Tututi M, Granados-Soto V. Resveratrol: a natural compound with pharmacological potential in neurodegenerative diseases. CNS Neurosci Ther. 2008;14:234–247. doi: 10.1111/j.1755-5949.2008.00045.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Ates O, Cayli S, Altinoz E, Gurses I, Yucel N, Sener M, et al. Neuroprotection by resveratrol against traumatic brain injury in rats. Mol Cell Biochem. 2007;294:137–144. doi: 10.1007/s11010-006-9253-0. [DOI] [PubMed] [Google Scholar]
  • 201.Sönmez U, Sönmez A, Erbil G, Tekmen I, Baykara B. Neuroprotective effects of resveratrol against traumatic brain injury in immature rats. Neurosci Lett. 2007;420:133–137. doi: 10.1016/j.neulet.2007.04.070. [DOI] [PubMed] [Google Scholar]
  • 202.Wang Q, Xu J, Rottinghaus GE, Simonyi A, Lubahn D, Sun GY, et al. Resveratrol protects against global cerebral ischemic injury in gerbils. Brain Res. 2002;958:439–447. doi: 10.1016/s0006-8993(02)03543-6. [DOI] [PubMed] [Google Scholar]
  • 203.Yousuf S, Atif F, Ahmad M, Hoda N, Ishrat T, Khan B, et al. Resveratrol exerts its neuroprotective effect by modulating mitiochondrial dysfunction and associated cell death during cerebral ischemia. Brain Res. 2009;1250:242–253. doi: 10.1016/j.brainres.2008.10.068. [DOI] [PubMed] [Google Scholar]
  • 204.Ates O, Cayli SR, Yucel N, Altinoz E, Kocak A, Durak MA, et al. Central nervous system protection by resveratrol in streptozotocin-induced diabetic rats. J Clin Nerurosci. 2007;14:256–260. doi: 10.1016/j.jocn.2005.12.010. [DOI] [PubMed] [Google Scholar]
  • 205.Kumar A, Kaundal RK, Iyer S, Sharma SS. Effects of resveratrol on nerve functions, oxidative stress and DNA fragmentation in experimental diabetic neuropathy. Life Sci. 2007;80:1236–1244. doi: 10.1016/j.lfs.2006.12.036. [DOI] [PubMed] [Google Scholar]
  • 206.Lu KT, Ko MC, Chen BY, Huang JC, Hsieh JC, Lee MC, et al. Neuroprotective effects of resveratrol on MPTP-induced neuron loss mediated by free radical scavenging. J Agric Food Chem. 2008;56:6910–6913. doi: 10.1021/jf8007212. [DOI] [PubMed] [Google Scholar]
  • 207.Blanchet J, Longpré F, Bureau G, Morissette M, DiPaolo T, Bronchti G, et al. Resveratrol, a red wine polyphenol, protects dopaminergic neurons in MPTP-treated mice. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1243–1250. doi: 10.1016/j.pnpbp.2008.03.024. [DOI] [PubMed] [Google Scholar]
  • 208.Gupta YK, Briyal S, Chaudhary G. Protective effect of trans-resveratrol against kainic acid-induced seizures and oxidative stress in rats. Pharmacol Biochem Behav. 2002;71:245–249. doi: 10.1016/s0091-3057(01)00663-3. [DOI] [PubMed] [Google Scholar]
  • 209.Bastianetto S, Zheng W-H, Quirion R. Neuroprotective abilities of resveratrol and other red wine constituents against nitric oxide-related toxicity in cultured hippocampal neurons. Brit J Pharmacol. 2000;131:711–720. doi: 10.1038/sj.bjp.0703626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Tsai SK, Hung LM, Fu YT, Cheng H, Nien MW, Liu HY, et al. Resveratrol neuroprotective effects during focal cerebral ischemia injury via nitric oxide mechanism in rats. J Vasc Surg. 2007;46:346–353. doi: 10.1016/j.jvs.2007.04.044. [DOI] [PubMed] [Google Scholar]
  • 211.Donnelly LE, Newton R, Kennedy GE, Fenwick PS, Leung RH, Ito K, et al. Anti-inflammatory effects of resveratrol in lung epithelial cells: molecular mechanism. Am J Physiol Lung Cell Mol Physiol. 2004;287:774–783. doi: 10.1152/ajplung.00110.2004. [DOI] [PubMed] [Google Scholar]
  • 212.Wu CT, Yu HP, Chung CY, Lau YT, Liao SK. Attenuation of lung inflammation and pro-inflammatory cytokine production by resveratrol following trauma-hemorrhage. Chin J Physiol. 2008;51:363–368. [PubMed] [Google Scholar]
  • 213.de la Lastra CA, Villegas I. Resveratrol as an anti-inflammatory and anti-aging agent: mechanisms and clinical implications. Mol Nutr Food Res. 2005;49:405–430. doi: 10.1002/mnfr.200500022. [DOI] [PubMed] [Google Scholar]
  • 214.Meng Y, Ma QY, Kou XP, Xu J. Effect of resveratrol on activation of nuclear factor kappaB and inflammatory factors in rat model of acute pancreatitis. World J Gastroenterol. 2005;11:525–528. doi: 10.3748/wjg.v11.i4.525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Shiggematsu S, Ishida S, Hara M, Takahashi N, Yoshimatsu H, Sakata T, et al. Resveratrol, a red wine constituent polyphenol, prevents superoxide-dependent inflammatory response induced by ischemia/reperfusion, platelet-activating factor, or oxidants. Free Radic Biol Med. 2003;34:810–817. doi: 10.1016/s0891-5849(02)01430-2. [DOI] [PubMed] [Google Scholar]
  • 216.Das S, Falchi M, Bertelli A, Maulik N, Das DK. Attenuation of ischemia/reperfusion injury in rats by the anti-inflammatory action of resveratrol. Arzneimittel Forchung. 2006;56:700–706. doi: 10.1055/s-0031-1296776. [DOI] [PubMed] [Google Scholar]
  • 217.Khanna D, Sethi G, Ahn KS, Pandey MK, Kunnumakkara AB, Sung B, et al. Natural products as gold mine for the arthritis treatment. Curr Opin Pharmacol. 2007;7:344–351. doi: 10.1016/j.coph.2007.03.002. [DOI] [PubMed] [Google Scholar]
  • 218.Csaki C, Keshishzaden N, Fischer K, Shakibaei M. Regulation of inflammation signaling by resveratrol in human chondrocytes in vitro. Biochem Pharmacol. 2008;75:677–687. doi: 10.1016/j.bcp.2007.09.014. [DOI] [PubMed] [Google Scholar]
  • 219.Elmali N, Baysal O, Esenkaya I, Mizrak B. Effects of resveratrol in inflammatory arthritis. Inflammation. 2007;30:1–6. doi: 10.1007/s10753-006-9012-0. [DOI] [PubMed] [Google Scholar]
  • 220.Upadhyay G, Singh AK, Kumar A, Prakash O, Singh MP. Resveratrol modulates pyrogallol-induced changes in hepatic toxicity markers, xenobiotic metabolizing enzymes and oxidative stress. Eur J Pharmacol. 2008;596:146–152. doi: 10.1016/j.ejphar.2008.08.019. [DOI] [PubMed] [Google Scholar]
  • 221.Rocha KK, Souza GA, Ebaid GX, Seiva FR, Cataneo AC, Novelli EL. Resveratrol toxicity: effects on risk factors for atherosclerosis and hepatic oxidative stress in standard and high-fat diets. Food Chem Toxicol. 2009;47:1362–1367. doi: 10.1016/j.fct.2009.03.010. [DOI] [PubMed] [Google Scholar]
  • 222.Bishayee A, Dhir N. Resveratrol-mediated chemoprevention of diethynitrosamine-initiated hepatocarcinogenesis: inhibition of cell proliferation and induction of apoptosis. Chem Biol Interact. 2009;179:131–144. doi: 10.1016/j.cbi.2008.11.015. [DOI] [PubMed] [Google Scholar]
  • 223.Raal A, Pokk P, Arend A, Aunapuu M, Jõgi J, Okva K, et al. Trans-resveratrol alone and hydroxystilbenes of rhubarb (Rhenum rhaponticum L.) root reduce liver damage induced by chronic ethanol administration: a comparative study in mice. Phytother Res. 2009;23:525–532. doi: 10.1002/ptr.2665. [DOI] [PubMed] [Google Scholar]
  • 224.Kasdallah-Grissa A, Mornagui B, Aouani E, Hammami M, May ME, Gharbi N, et al. Resveratrol, a red wine polyphenol, attenuates ethanol-induced oxidative stress in rat liver. Life Sci. 2007;80:1033–1039. doi: 10.1016/j.lfs.2006.11.044. [DOI] [PubMed] [Google Scholar]
  • 225.Cadenas S, Barja G. Resveratrol, melatonin, vitamin E and PBN protect against renal oxidative DNA damage induced by the kidney carcinogen KBrO3. Free Radic Biol Med. 1999;26:1531–1537. doi: 10.1016/s0891-5849(99)00019-2. [DOI] [PubMed] [Google Scholar]
  • 226.Giovannini L, Migliori M, Longoni BM, Das DK, Bertelli AAE, Panichi V, et al. Resveratrol, a polyphenol found in wine, reduces ischemia reperfusion injury in rat kidneys. J Cardiovascul Pharmacol. 2001;37:262–270. doi: 10.1097/00005344-200103000-00004. [DOI] [PubMed] [Google Scholar]
  • 227.Sener G, Tugrepe H, Yüksel M, Çetinel S, Gedik N, Yegen C. Resveratrol improves ischemia/reperfusioninduced oxidative renal injury in rats. Arch Med Res. 2006;37:822–829. doi: 10.1016/j.arcmed.2006.04.003. [DOI] [PubMed] [Google Scholar]
  • 228.Rodrigo R, Rivera G. Renal damage mediated by oxidative stress: a hypothesis of protective effects of red wine. Free Radic Biol Med. 2002;33:409–422. doi: 10.1016/s0891-5849(02)00908-5. [DOI] [PubMed] [Google Scholar]
  • 229.Rodrigo R, Bosco C. Oxidative stress and protective effects of polyphenols: comparative studies in human and rodent kidney. A review. Comparat Biochem Physiol Part C. 2006;142:317–327. doi: 10.1016/j.cbpc.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • 230.Sgambato A, Ardito R, Faraglia B, Boninsegna A, Wolf FI, Cittadini A. Resveratrol, a natural phenolic compound, inhibits cell proliferation and prevents oxidative DNA damage. Mutat Res. 2001;496:171–180. doi: 10.1016/s1383-5718(01)00232-7. [DOI] [PubMed] [Google Scholar]
  • 231.Burkhardt S, Reiter RJ, Tan D-X, Hardeland R, Cabrera J, Karbownik M. DNA oxidatively damaged by chromium (III) and H2O2 is protected by the antioxidants melatonin, N1-acetyl-N2-formyl-5-methoxyynuramine, resveratrol and uric acid. Int J Biochem Cell Biol. 2001;33:775–783. doi: 10.1016/s1357-2725(01)00052-8. [DOI] [PubMed] [Google Scholar]
  • 232.Quincozes-Santos A, Andreazza AC, Nardin P, Funchal C, Gonçalves C-A, Gottfried C. Resveratrol attenuates oxidative-induced DNA damage in C6 Glioma cells. Neuro Toxicol. 2007;28:886–891. doi: 10.1016/j.neuro.2007.03.008. [DOI] [PubMed] [Google Scholar]
  • 233.Russo A, Palumbo M, Aliano C, Lempereur L, Scoto G, Renis M. Red wine micronutrients as protective agents in Alzheimer-like induced insult. Life Sci. 2003;72:2369–2379. doi: 10.1016/s0024-3205(03)00123-1. [DOI] [PubMed] [Google Scholar]
  • 234.Leiro J, Arranz JA, Fraiz N, Sanmartin ML, Quezada E, Orallo F. Effect of cis-resveratrol on genes involved in nuclear factor kappaB signaling. Int Immunopharmacol. 2005;5:393–406. doi: 10.1016/j.intimp.2004.10.006. [DOI] [PubMed] [Google Scholar]
  • 235.Oike H, Kobori M. Resveratrol regulates circadian clock genes in Rat-1 fibroblast cells. Biosci Biotechnol Biochem. 2008;72:3038–3040. doi: 10.1271/bbb.80426. [DOI] [PubMed] [Google Scholar]
  • 236.Zini R, Morin C, Bertelli A, Bertelli AAE, Tillement J-P. Resveratrol-induced limitation of dysfunction of mitochondria isolated from rat brain in an anoxia-reoxygenation model. Life Sci. 2002;71:3091–3108. doi: 10.1016/s0024-3205(02)02161-6. [DOI] [PubMed] [Google Scholar]
  • 237.Lee M, Kim S, Kwon OK, Oh SR, Lee HK, Ahn K. Anti-inflammatory and anti-asthmatic effects of resveratrol, a polyphenolic stilbene, in mouse model of allergic asthma. Int Immunopharmacol. 2009;9:418–424. doi: 10.1016/j.intimp.2009.01.005. [DOI] [PubMed] [Google Scholar]
  • 238.Kasdallah-Grissa A, Mornagui B, Aouani E, Hammami M, Gharbi N, Kamoun A, et al. Protective effect of resveratrol on ethanol-induced lipid peroxidation in rats. Alcohol Alcoholism. 2006;41:236–239. doi: 10.1093/alcalc/agh256. [DOI] [PubMed] [Google Scholar]
  • 239.Simonni G, Pignone A, Generini S, Falcini F, Cerinic MM. Emerging potentials for an antioxidant therapy as a new approach to the treatment of systemic sclerosis. Toxicology. 2000;155:1015. doi: 10.1016/s0300-483x(00)00272-9. [DOI] [PubMed] [Google Scholar]
  • 240.Hascalik S, Celik O, Turkoz Y, Hascalik M, Cigremis Y, Mizrak B, et al. Resveratrol, a red wine constituent polyphenol, protects from ischemia-reperfusion damage of the ovaries. Gynecol Obstetric Invest. 2004;57:218–223. doi: 10.1159/000076760. [DOI] [PubMed] [Google Scholar]
  • 241.Leonard SS, Xia C, Jiang B-H, Stinefelt B, Landorf H, Harris GH, et al. Resveratrol scavenges reactive oxygen species and effects radical-induced cellular responses. Biochem Biophys Res Commun. 2003;309:1017–1026. doi: 10.1016/j.bbrc.2003.08.105. [DOI] [PubMed] [Google Scholar]
  • 242.Jone KS, Alimov AP, Rilo HL, Jandacek RJ, Woollett LA, Penberthy WT. A high throughput live transparent animal bioassay to identify non-toxic small molecules or genes that regulate vertebrate fat metabolism for obesity drug development. Nutr Metab. 2008;6:23. doi: 10.1186/1743-7075-5-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Eybl V, Kotyzova D, Koutensky J. Comparative study of natural antioxidants-curcumin, resveratrol and melatonin in cadmium-induced oxidative damage in mice. Toxicology. 2006;225:150–156. doi: 10.1016/j.tox.2006.05.011. [DOI] [PubMed] [Google Scholar]
  • 244.Afaq F, Adhami VM, Ahamd N. Prevention of short- term ultraviolet B radiation-mediated damages by resveratrol in SKH-1 hairless mice. Toxicol Appl Pharmacol. 2003;186:28–37. doi: 10.1016/s0041-008x(02)00014-5. [DOI] [PubMed] [Google Scholar]
  • 245.Martín AR, Villegas I, La Casa C, de la Lastra CA. Resveratrol, a polyphenol found in grapes, suppresses oxidative damage and stimulates apoptosis during early colonic inflammation in rats. Biochem Pharmacol. 2004;67:1399–1410. doi: 10.1016/j.bcp.2003.12.024. [DOI] [PubMed] [Google Scholar]
  • 246.Lu X, Ji C, Xu H, Li X, Ding H, Ye M, et al. Resveratrol-loaded polymeric micelles protect cells from Aβ-induced oxidative stress. Int J Pharmaceut. 2009;375:89–96. doi: 10.1016/j.ijpharm.2009.03.021. [DOI] [PubMed] [Google Scholar]
  • 247.Clarke JO, Mullin GE. A review of complemetary and alternative approaches to immunomodulation. Nutr Clin Pract. 2008;23:49–62. doi: 10.1177/011542650802300149. [DOI] [PubMed] [Google Scholar]
  • 248.Miura T, Muraoka S, Fujimoto Y. Inactivation of creatine kinase induced by stilbene derivatives. Pharmacol Toxicol. 2002;90:66–72. doi: 10.1034/j.1600-0773.2002.900203.x. [DOI] [PubMed] [Google Scholar]
  • 249.Galati G, Sabzevari O, Wilson JX, O'Brien PJ. Prooxidant activity and cellular effects of the phenoxy radicals of dietary flavonoids and other polyphenolics. Toxicology. 2002;177:91–104. doi: 10.1016/s0300-483x(02)00198-1. [DOI] [PubMed] [Google Scholar]
  • 250.Athar M, Back JH, Kopelovich L, Bickers DR, Kim AL. Multiple molecular targets of resveratrol: anti-carcinogenic mechanism. Arch Biochem Biophys. 2009;486:95–1002. doi: 10.1016/j.abb.2009.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Ahmad KA, Clement MV, Pervaiz S. Pro-oxidant activity of low doses of resveratrol inhibits hydrogen peroxide-induced apoptosis. Ann NY Acad Sci. 2003;1010:365–373. doi: 10.1196/annals.1299.067. [DOI] [PubMed] [Google Scholar]
  • 252.Gadacha W, ben-Attia M, Bonnfont-Rousseelot D, Aouani E, Ghanem-Boughanmi N, Touitou Y. Resveratrol opposite effects on rat tissue lipid peroxidation: pro-oxidant during day-time and antioxidant at night. Free Radic Res. 2009;14:154–158. doi: 10.1179/135100009X466131. [DOI] [PubMed] [Google Scholar]
  • 253.Niufar NN, Haycock FL, Wesemann JL, MacStay JA, Heasley VL, Kovacic P. Reduction potentials of conjugated aliphatic ketones, oximes and imines. Rev Soc Quim Mex (J Mex Chem Soc) 2002;46:307–312. [Google Scholar]
  • 254.Ames JR, Kovacic P. A mechanism for the toxicity of polychlorinated biphenyls (PCBs), dioxins (TCDD) and dibenzofurans. J Biopharmaceut Sci. 1991;2:329–338. [Google Scholar]
  • 255.Zheng LF, Wei QY, Cai YJ, Fang JG, Zhou B, Yang L, et al. DNA damage induced by resveratrol and its synthetic analogues in the presence of Cu(II) ions: mechanism and structure-activity relationship. Free Radic Biol Med. 2006;41:1807–1816. doi: 10.1016/j.freeradbiomed.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 256.Azami AS, Bhat SH, Hanif S, Hadi SM. Plant polyphenols mobilize endogenous copper in human peripheral lymphocytes leading to oxidative DNA breakage: a putative mechanism for anticancer properties. FEBS Lett. 2006;580:533–538. doi: 10.1016/j.febslet.2005.12.059. [DOI] [PubMed] [Google Scholar]
  • 257.Ahamed A, Syed FA, Singh S, Hadi SM. Prooxidant activity of resveratrol in the presence of copper ions: mutagenicity in plasmid DNA. Toxicol Lett. 2005;159:1–12. doi: 10.1016/j.toxlet.2005.04.001. [DOI] [PubMed] [Google Scholar]
  • 258.Azami AS, Bhat SH, Hanif S, Hadi SM. Resveratrol-Cu(II) induced DNA breakage in human peripheral lymphocytes; implications for anticancer properties. FEBS Lett. 2005;579:3131–3135. doi: 10.1016/j.febslet.2005.04.077. [DOI] [PubMed] [Google Scholar]
  • 259.Hadi SM, Asad SF, Singh S, Ahmad A. Putative mechanism for anticancer and apoptosis-inducing properties of plant-derived polyphenolic compounds. IUBMB Life. 2000;50:167–171. doi: 10.1080/152165400300001471. [DOI] [PubMed] [Google Scholar]
  • 260.de la Lastra CA, Villegas I. Resveratrol as an antioxidant and pro-oxidant agent: mechanisms and clinical implications. Biochem Soc Trans. 2007;35:1156–1160. doi: 10.1042/BST0351156. [DOI] [PubMed] [Google Scholar]
  • 261.Rüweler M, Gülden M, Maser E, Murias M, Seibert H. Cytotoxic cytoprotective and antioxidant activities of resveratrol and analogues in C6 astroglioma cells in vitro. Chem Biol Interact. 2009;182:128–135. doi: 10.1016/j.cbi.2009.09.003. [DOI] [PubMed] [Google Scholar]
  • 262.Kovacic P, Somanathan R. Clinical physiology and mechanism of dizocilpine (MK-801): electron transfer, radicals, redox metabolites and bioactivity. Oxid Med Cell Longev. 2010;3:13–22. doi: 10.4161/oxim.3.1.10028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Pristov JB, Spasojevic I, Mikovic Z, Mandic, Cerovic N, Spasic M. Antioxidative defense enzymes in placenta protect placenta and fetus in inherited thrombophilia from hydrogen peroxide. Oxid Med Cell Longev. 2009;2:14–18. doi: 10.4161/oxim.2.1.7705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Fischer-Wellman K, Bell HK, Bloomer RJ. Oxidative stress and antioxidant defense mechanisms linked to exercise during cardiopulmonary and metabolic disorders. Oxid Med Cell Longev. 2009;2:43–51. doi: 10.4161/oxim.2.1.7732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Gupta A, Bhatt MLB, Misra MK. Lipid peroxidation and antioxidant status in head and neck squamous cell carcinoma patients. Oxid Med Cell Longev. 2009;2:66–72. doi: 10.4161/oxim.2.2.8160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Bharti VK, Srivatava RS. Pineal proteins upregulate specific antioxidant defense systems in the brain. Oxid Med Cell Longev. 2009;2:88–92. doi: 10.4161/oxim.2.2.8361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Ozsoy N, Candoken E, Akev N. Implications for degenerative disorders: antioxidative activity, total phenols, flavonoids, ascorbic acid, β-carotene, α-tocopherol in Aloe vera. Oxid Med Cell Longev. 2009;2:98–99. doi: 10.4161/oxim.2.2.8493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Gomes P, Simão S, Silva E, Pinto V, Amaral JS, Alfonso J, et al. Aging increases oxidative stress and renal expression of oxidant and antioxidant enzymes that are associated with an increased trend in systolic blood pressure. Oxid Med Cell Longev. 2009;2:130–137. doi: 10.4161/oxim.2.3.8819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Kovacic P, Somanathan R. Novel, unifying mechanism for mescaline in the central nervous system: electrochemistry, catechol redox metabolite, receptor, cell signaling and structure activity relationships. Oxid Med Cell Longev. 2009;2:181–190. doi: 10.4161/oxim.2.4.9380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Santos IMS, Tomé ADR, Saldanha GB, Ferreira PM, Militão GCG, de Freitas M. Oxidative stress in the hippocampus during experimental seizures can be ameliorated with the antioxidant ascorbic acid. Oxid Med Cell Longev. 2009;2:214–221. doi: 10.4161/oxim.2.4.8876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Kovacic P, Somanthan R. Zolpidem a clinical hypnotic that affects electronic transfer, alters synaptic activity through potential GABA receptors in the nervous system without significant free radical generation. Oxid Med Cell Longev. 2009;2:52–57. doi: 10.4161/oxim.2.1.7859. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Oxidative Medicine and Cellular Longevity are provided here courtesy of Wiley

RESOURCES