Вы находитесь на странице: 1из 5

1156 Biochemical Society Transactions (2007) Volume 35, part 5

Resveratrol as an antioxidant and pro-oxidant


agent: mechanisms and clinical implications
C. Alarcón de la Lastra1 and I. Villegas
Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville, Spain

Abstract
Resveratrol (3,4 ,5-trihydroxystilbene) is found in various plants, including grapes, berries and peanuts. It
is also present in wines, especially red wines. During the last years, it has been the focus of numerous
in vitro and in vivo studies investigating its biological attributes, which include mainly antioxidant and anti-
inflammatory activities, anti-platelet aggregation effect, anti-atherogenic property, oestrogen-like growth-
promoting effect, growth-inhibiting activity, immunomodulation and chemoprevention. In fact, recently, it
has been demonstrated that the stilbene blocks the multistep process of carcinogenesis at various stages:
tumour initiation, promotion and progression. More recent results provide interesting insights into the effect
of this compound on the life span of yeasts and flies, implicating the potential of resveratrol as an anti-
aging agent in treating age-related human diseases. Nevertheless, depending on the concentration of the
phytoalexin and the cell type, it has also been shown that resveratrol can exhibit pro-oxidant properties,
leading to oxidative breakage of cellular DNA in the presence of transition metal ions such as copper. Re-
cently, it has been proposed that such a pro-oxidant action could be a common mechanism for anticancer and
chemopreventive properties of plant polyphenols. The present paper is intended to provide the reader up-
to-date information on the antioxidant and pro-oxidant properties of resveratrol and its clinical implications.

Introduction effects may be due in part to resveratrol being a phyto-


Resveratrol (3,4 ,5-trihydroxystilbene) is a phytoalexin oestrogen, i.e. a plant compound that has biologically similar
found in a wide variety of dietary sources including grapes, properties to those of oestrogens [8].
plums and peanuts. It is also present in wines, especially More recent results provide interesting insights into the
red wines and to a much lesser extent in white wines. effect of this compound on the lifespan of yeasts and flies,
Its stilbene structure is related to the synthetic oestrogen implicating its potential as an anti-aging agent in treating
diethylstilbestrol. Resveratrol exists as cis- and trans-isomers. age-related human diseases [9,10].
Trans-resveratrol is the preferred steric form and is relatively Additionally, some investigators have indicated a potential
stable if it is protected from high pH and light. The synthesis neuroprotective activity for resveratrol based on its
of trans-resveratrol in the plants can be induced by microbial beneficial effects in several brain damage models. Similarly,
infections, UV radiation and exposure to ozone [1–3]. several studies, including ours, have identified resveratrol as a
A primary impetus for research on resveratrol was initiated beneficial agent in the control of inflammatory disorders such
from the paradoxical observation that a low incidence of car- as arthritis and inflammatory bowel disease [11,12]. Potential
diovascular diseases may co-exist with a high-fat diet intake mechanisms implicated include: inhibition of synthesis and
and moderate consumption of red wine [4,5], a phenomenon release of pro-inflammatory mediators, modification of
known as the French paradox [1]. The possible mechanisms eicosanoid synthesis, inhibition of activate immune cells and
by which resveratrol exerts its cardio- and vascular-protection inflammatory enzymes such as iNOS [inducible NOS (nitric
involve inhibition of platelet aggregation, arterial vasodilation oxide synthase)] and COX-2 (cyclo-oxygenase-2) through
mediated by NO (nitric oxide) release, favourable changes its inhibitory effects on NF-κB (nuclear factor κB) or the
in lipid metabolism such as LDL (low-density lipoprotein)– AP-1 (activator protein-1) signalling pathways [9].
cholesterol oxidation, antioxidant effects, stimulation of One of the most striking biological activities of resveratrol
angiogenesis [6], induction of cardioprotective protein intensely investigated during the last years has been its cancer-
expression, and insulin sensitization. Indeed, it reduces chemopreventive or anticancer properties. These properties
the synthesis of certain lipids and eicosanoids that tend were first appreciated when Jang et al. [13] demonstrated that
to promote inflammation and atherosclerosis; likewise, it resveratrol possesses cancer-chemopreventive and cytostatic
suppresses certain cardiac arrhythmias [7]. Some of these properties via the three major stages of carcinogenesis, i.e.
initiation, promotion and progression [14]. Since then, there
Key words: antioxidant, copper, DNA damage, pro-oxidant, reactive oxygen species, resveratrol. has been a flurry of papers reporting the implication of res-
Abbreviations used: COX, cyclo-oxygenase; LP, lipid peroxidation; LPS, lipopolysaccharide; NF- veratrol in cancer chemoprevention through a wide range of
κB, nuclear factor κB; NOS, nitric oxide synthase; iNOS, inducible NOS; RNS, reactive nitrogen
species; ROS, reactive oxygen species; SOD, superoxide dismutase; XO, xanthine oxidase.
actions that are poorly understood. It appears to help detoxify
1
To whom correspondence should be addressed (email calarcon@us.es). carcinogens, to reduce the synthesis of various cancer-related


C The Authors Journal compilation 
C 2007 Biochemical Society
Inflammation 1157

compounds and to interfere with cell survival programmes; Figure 1 Resveratrol antioxidant potential
for instance, resveratrol has been shown to promote PUFA, polyunsaturated fatty acid.
apoptosis in cancer cells by blocking anti-apoptotic proteins
expression or by inhibiting signal transduction through
the PI3K (phosphoinositide 3-kinase), MAPK (mitogen-
activated protein kinase) or NF-κB pathways [3,10,15].
Most of the scientific evidence for resveratrol’s benefits
is based on in vitro studies in which the diastereomers
trans- or cis-resveratrol have been tested. However,
from animal studies and human trials, we know that the
predominant isomer that is orally ingested with foods is
trans-resveratrol glucoside (piceid), which is biotransformed
and rapidly eliminated. In addition, these derivatives might
be less biologically active due to their esterified hydroxy
groups. However, the chemopreventive activity of orally
administered trans-resveratrol has almost been demonstrated
in cancer-induced animal models [16]. Nonetheless, future
studies are needed to know the effective dose required to
achieve the health benefits evidenced in experimental models.

Resveratrol as free radical scavenger and


antioxidant
Over the last few years, a number of studies have provided
evidence of an important role of ROS (reactive oxygen spe-
cies) in mediating the development of oxidative stress. Excess-
ive ROS accumulation may induce the oxidative modification
of cellular macromolecules (lipid, proteins and nucleic acids)
with deleterious potential. In fact, DNA damage by ROS In order to protect tissues against the deleterious effects
has been implicated in mutagenesis, oncogenesis and aging. of ROS, all cells possess numerous defence mechanisms
Oxidative lesions in DNA include base modifications, sugar that include enzymes such as SOD (superoxide dismutase),
damage, strand breaks and abasic sites [17]. Since gene tran- catalase, glutathione reductase and glutathione peroxidase.
scription can be regulated by oxidants, antioxidants and other Resveratrol can maintain the concentration of intracellular
determinants of the intracellular redox state, ROS can also antioxidants found in biological systems. For instance, in a
produce protein damage, inducing other types of mutations. study by Losa [21], stilbene appeared to maintain the gluta-
One of the biological activities that have been ascribed thione content in peripheral blood mononuclear cells isolated
to resveratrol involves its antioxidant potential. Resveratrol ex vivo from a healthy human from oxidative damage caused
is both a free radical scavenger and a potent antioxidant by 2-deoxy-D-ribose. In a previous study, in human blood
because of its ability to promote the activities of a variety of platelets, resveratrol markedly decreased oxidation of thiol
antioxidant enzymes (Figure 1). The ability of the polyphen- groups of proteins in these cells [24]. Similarly, resveratrol
olic compounds to act as antioxidants depends on the redox induced an increase in glutathione levels in a concentration-
properties of their phenolic hydroxy groups and the potential dependent manner in human lymphocytes activated with
for electron delocalization across the chemical structure [18]. H2 O2 . In another study, resveratrol increased the amounts
The common recognition of resveratrol as a natural of several antioxidant enzymes, including glutathione peroxi-
antioxidant was clarified by Zini et al. [19], who suggested dase, glutathione S-transferase and glutathione reductase [25].
three different antioxidant mechanisms: (i) competition with
coenzyme Q and, to decrease the oxidative chain complex, Effects of resveratrol on RNS (reactive
the site of ROS generation, (ii) scavenging O2 − radicals

nitrogen species) generation
formed in the mitochondria and (iii) inhibition of LP (lipid It is now widely accepted that a moderate concentration of
peroxidation) induced by Fenton reaction products. In NO appears to play cardio- and neuro-protective effects,
fact, numerous studies have demonstrated the ability of and, along these lines, several reports have shown the role of
resveratrol to scavenge both O2 − and OH radicals [20–22].
䊉 䊉
resveratrol in the regulation of NO production from vascular
By contrast, in a study by Orallo et al. [23], using the endothelium in the ischaemic heart, brain or kidney [26,27].
enzymatic hypoxanthine oxidase–XO (xanthine oxidase) However, abnormally high concentrations of NO and its
system, resveratrol neither affected the XO activity nor derivatives RNS have been associated with tumour growth
scavenged O2 − radicals in rat macrophage extracts.

and vascular invasion. In a previous study [28], the effects of


C The Authors Journal compilation 
C 2007 Biochemical Society
1158 Biochemical Society Transactions (2007) Volume 35, part 5

resveratrol and oxyresveratrol on nitrosative and oxidative Figure 2 Inhibition of LP by resveratrol and its antioxidant
stress derived from microglial cells was investigated. mechanisms in carcinogenesis
Phytoalexin considerably diminished NO production upon
the inducible isoform of NOS (iNOS expression), and it also
induced an inhibitory effect on the iNOS enzyme activity.
Bacterial endotoxic LPS (lipopolysaccharide) is one of the
most important stimuli for iNOS induction, resulting in NO
production that has bactericidal effects. For example, in LPS-
activated RAW 264.7 macrophages, pre-incubation of cells
with resveratrol reduced inflammation by down-regulation
of the iNOS and mRNA [29,30]. The results obtained
demonstrate that resveratrol is a potent inhibitor of the
antipathogen responses of rat macrophages and thus suggest
that this agent may have applications in the treatment of
diseases involving macrophage hyper-responsiveness [31,32].

Antioxidant activity of resveratrol and


carcinogenesis
Resveratrol prevents the initial DNA damage by two
different pathways: (i) acting as an antimutagen through
the induction of Phase II enzymes, such as quinine
reductase, capable of metabolically detoxifying carcinogens
by inhibiting COX and cytochrome P450, and (ii) acting as
an antioxidant through inhibition of DNA damage by ROS
[33]. It has been proposed that ROS derived from LP may
compound, depending on the concentration of resveratrol
function as tumour initiators [20]. Leonard et al. [20] have
and the cell type, leading to oxidative breakage of cellular
shown that resveratrol exhibits a protective effect against LP
DNA. Lately, it has been proposed that such pro-oxidant
in cell membranes and DNA damage caused by ROS.
action could be an important action mechanism of its
The antipromotional properties of resveratrol can be partly
anticancer and apoptotic inducing properties. Furthermore,
attributed to its ability to enhance gap-junctional intercellular
it has been shown that there is an interesting correlation
communications in cells exposed to tumour promoters such
among the antioxidant and pro-oxidant activities and
as PMA [34]. The tumour-promoting activity mediated by
cytotoxicity of dietary polyphenols [36].
PMA has also been associated with oxidative stress by in-
Every antioxidant is in fact a redox (reduction–oxidation)
creased production of O2 − and H2 O2 , reduction of SOD

agent and thus might become a pro-oxidant to accelerate


activity and interference with glutathione metabolism. In a
LP and/or induce DNA damage under special conditions.
model of PMA application to mouse skin, resveratrol induced
Studies have revealed pro-oxidant effects of antioxidant
the restoration of H2 O2 and glutathione levels, and also my-
vitamins and several classes of plant-derived polyphenols
eloperoxidase, glutathione reductase and SOD activities [35].
such as flavonoids [37], tannins [38] and curcumin [39].
The development of skin cancer is related to accumulative
Ahmad et al. [40] observed that exposure of human
exposure to solar UVB as well as the nuclear transcription
leukaemia cells to low concentrations of resveratrol (4–
factor NF-κB, which plays a critical role in skin biology.
8 µM) inhibited caspase activation and DNA fragmentation
NF-κB is involved in the inflammatory and carcinogenic
induced by incubation with H2 O2 . At these concentrations,
signalling cascades, and resveratrol was able to block
resveratrol elicited pro-oxidant properties as evidenced by an
the damage caused by UVB exposure via its antioxidant
increase in intracellular O2 − concentration. Likewise, in rat

properties blocking UVB-mediated NF-κB activation.


hepatocytes exposed to ferrylmyoglobin-induced oxidative
Finally, resveratrol could inhibit tumour progression, partly
stress, physiological concentrations (100 pM–100 nM) of
by an inhibition of DNA polymerase and deoxyribo-
resveratrol exerted pro-oxidant activities [22]. It has also
nucleotide synthesis through its ability to scavenge the
been shown that resveratrol has a pro-oxidative effect on
essential tyrosine radical of the ribonucleotide reductase and
DNA damage during interaction with ADP-Fe3+ in the
partly by inducing cell cycle arrest [18] (Figure 2).
presence of H2 O2 in tumour cell line cultures [34].
Similarly, the pro-oxidant effects of resveratrol were shown
Effects of resveratrol on intracellular redox on rat liver microsomal systems. Resveratrol inhibited LP;
state however, resveratrol increased OH generation, indicating

Recent results have provided interesting insight into the that OH played a minor role in LP [41]. In addition, it is well

effect of resveratrol on intracellular redox state. These results known that haem (iron-protoporphyrin IX) is a pro-oxidant
seem to support both anti- and pro-oxidant activities of this and its rapid degradation by haem oxygenase is believed


C The Authors Journal compilation 
C 2007 Biochemical Society
Inflammation 1159

to be neuroprotective. Using primary neuronal cultures, Figure 3 Cytotoxic mechanism of resveratrol probably involves
resveratrol was able to significantly induce haem oxy- mobilization of endogenous copper ions
genase 1. This study indicated that the increase of haem
oxygenase activity by resveratrol is a unique pathway by
which this compound can exert its neuroprotective actions
[42]. Further corroborating the pro-oxidant activity of
resveratrol, there are data that demonstrate its inefficiency
in protecting proteins (BSA) from oxidative damage induced
by metal-catalysed reaction or alkylperoxyl radicals [43].
Fukuhara and Miyata [44] first reported the pro-oxidant
activity of resveratrol in a plasmid-based DNA cleavage
assay in the presence of transition metal ions such as copper.
DNA degradation by resveratrol in the presence of cop-
per (10–100 µM) or alone (200 µM) (in the absence of added
copper) has also been shown in a cellular system of peripheral
lymphocytes isolated from human blood [45,46].
Copper is one of the most redox-active metal ions present
in the nucleus, serum and tissues [47]. Approximately 20%
of copper is located in the nucleus and is closely associated
with DNA bases, in particular, guanine [48]. Furthermore,
it has been shown that the concentration of copper is greatly
increased in various malignancies [45]. Copper ions from
chromatin can be mobilized by metal-chelating agents, giving
rise to internucleosomal DNA fragmentation, a property
that is the hallmark of cells undergoing apoptosis [17].
The cytotoxic mechanism of resveratrol probably involves
mobilization of endogenous copper ions, possibly chromatin-
bound copper. First, resveratrol undergoes oxidation in the
presence of Cu(II). The oxidative product of resveratrol is a
dimer, which possibly might be formed by dimerization of
resveratrol phenoxyl radical as a result of the reductive activa-
tion of molecular oxygen. Indeed, this initial electron transfer
generates the reduction of Cu(II) to Cu(I). Interestingly,
DNA strand scission occurred at neutral pH, indicating that
resveratrol can induce DNA cleavage without the oxygena-
tion of the benzene nuclei to the catechol moiety. However,
the structural feature of the copper–peroxide complex as the
reactive species responsible for the DNA cleavage is still un-
known. Secondly, the Cu(II)–peroxide complex is capable of as a consequence of its pro-oxidant action, resveratrol can
binding DNA and forms a DNA–resveratrol–Cu(II) ternary sensitize cancer cells, which may result in synergistic anti-
complex. The high binding affinity of a 4-hydroxy group at tumour activities when resveratrol is combined with con-
the 4-position with both Cu(II) and DNA makes it possible ventional chemotherapeutic agents or cytotoxic compounds
and therefore cleaves DNA efficiently [49] (Figure 3). [15,50]. However, further insights into the signalling network
and interaction points modulated by resveratrol may provide
the basis for novel discovery programmes to exploit res-
Clinical implications veratrol for the prevention and treatment of human diseases.
The body of evidence presented here speaks volumes
about the clinical potential of resveratrol as an antioxidant and
pro-oxidant. Insufficient activation of apoptosis because of
References
defects in apoptosis programmes or because of the dominance 1 Ignatowicz, E. and Baer-Dubowska, W. (2001) Pol. J. Pharmacol. 53,
of survival signals may result in cancer cell resistance. Despite 557–569
aggressive therapies, resistance of many tumours to current 2 Soleas, G.J., Yan, J. and Goldberg, D.M. (2001) Methods Enzymol. 335,
130–145
treatment protocols still constitutes a major problem in cancer 3 Pervaiz, S. (2003) FASEB J. 17, 1975–1985
therapy. Poly-mechanistic phytochemicals such as resveratrol 4 Renaud, S.C. and De Lorgeril, M. (1992) Lancet 339, 1523–1526
may offer the advantage over targeted therapeutics and may 5 Soleas, G.J., Diamandis, E.P. and Goldberg, D.M.J. (1997) Clin. Lab. Anal.
11, 287–313
open new perspectives in cancer therapy. By blocking survival 6 Maulik, N. (2006) Antioxid. Redox Signaling 8, 2161–2168
and anti-apoptotic mechanisms or causing DNA degradation, 7 Providencia, R. (2006) Rev. Port. Cardiol. 25, 1043–1058


C The Authors Journal compilation 
C 2007 Biochemical Society
1160 Biochemical Society Transactions (2007) Volume 35, part 5

8 Delmas, D., Jannin, B. and Latruffe, N. (2005) Mol. Nutr. Food Res. 49, 31 Matsuda, H., Kageura, T., Morikawa, T., Toguchida, I., Harima, S. and
377–395 Yoshikawa, M. (2000) Bioorg. Med. Chem. Lett. 10, 323–327
9 Alarcón de la Lastra, C. and Villegas, I. (2005) Mol. Nutr. Food Res. 49, 32 Leiro, J., Alvarez, E., Garcia, D. and Orallo, F. (2002)
405–430 Int. Immunopharmacol. 2, 767–774
10 Holme, A.L. and Pervaiz, S. (2007) J. Bioenerg. Biomembr. 39, 59–63 33 Roemer, K. and Mahyar-Roemer, M. (2002) Drugs Today 38, 571–580
11 Martin, A.R., Villegas, I., La Casa, C. and de la Lastra, C.A. (2004) 34 Gusman, J., Malonne, H. and Atassi, G. (2001) Carcinogenesis 22,
Biochem. Pharmacol. 67, 1399–1410 1111–1117
12 Martin, A.R., Villegas, I., Sanchez-Hidalgo, M. and de la Lastra, C.A. 35 Jang, M. and Pezzuto, J.M. (1999) Drug. Exp. Clin. Res. 25, 65–77
(2006) Br. J. Pharmacol. 147, 873–885 36 Zheng, L.F., Wei, Q.Y., Cai, Y.J., Fang, J.G., Zhou, B., Yang, L. and Liu, Z.L.
13 Jang, M., Cai, L., Udeani, G.O., Slowing, K.V., Thomas, C.F., Beecher, C.W., (2006) Free Radical Biol. Med. 41, 1807–1816
Fong, H.H., Farnsworth, N.R., Kinghorn, A.D., Mehta, R.G. et al. (1997) 37 Rahman, A., Shahabuddin Hadi, S.M. and Parish, J.H. (1990)
Science 275, 218–220 Carcinogenesis 11, 2001–2003
14 Aziz, M.H., Kumar, R. and Ahmad, N. (2003) Int. J. Oncol. 23, 17–28 38 Singh, S., Asad, S.F., Ahmad, A., Khan, N.U. and Hadi, S.M. (2001)
15 Fulda, S. and Debatin, K.M. (2006) Cancer Detect. Prev. 30, 217–223 Cancer Lett. 169, 139–146
16 Somoza, V. (2005) Mol. Nutr. Food Res. 49, 373 39 Ahsan, H. and Hadi, S.M. (1998) Cancer Lett. 124, 23–30
17 Ahmad, A., Syed, F.A., Singh, S. and Hadi, S.M. (2005) Toxicol. Lett. 159, 40 Ahmad, K.A., Clement, M.V. and Pervaiz, S. (2003) Ann. N.Y. Acad. Sci.
1–12 1010, 365–373
18 Alarcón de la Lastra, C., Villegas, I. and Martı́n, A.R. (2006) in Resveratrol 41 Ozgová, S., Hermanek, J. and Gut, I. (2003) Biochem. Pharmacol. 66,
in Health and Disease (Aggarwald, B.B, and Shishodia, S., eds), 1127–1137
pp. 33–56, CRC Press, Boca Raton 42 Zhuang, H., Kim, Y.S., Koehler, R.C. and Dore, S. (2003) Ann. N.Y.
19 Zini, R., Morin, C., Bertelli, A., Bertelli, A.A. and Tillement, J.P. (1999) Acad. Sci. 993, 276–286
Drugs Exp. Clin. Res. 25, 87–97 43 Mayo, J.C., Tan, D.X., Sainz, R.M., Natarajan, M., Lopez-Burillo, S. and
20 Leonard, S., Xia, C., Jiang, B.H., Stinefelt, B., Klandorf, H., Harris, G.K. and Reiter, R.J. (2003) Biochim. Biophys. Acta 1620, 139–150
Shi, X. (2003) Biochem. Biophys. Res. Commun. 309, 1017–1026 44 Fukuhara, K. and Miyata, N. (1998) Bioorg. Med. Chem. Lett. 8,
21 Losa, G.A. (2003) Eur. J. Clin. Invest. 33, 818–823 3187–3192
22 Martı́nez, J. and Moreno, J.J. (2000) Biochem. Pharmacol. 59, 865–870 45 Azmi, A.S., Bhat, S.H. and Hadi, S.M. (2005) FEBS Lett. 579, 3131–3135
23 Orallo, F., Alvarez, E., Camina, M., Leiro, J.M., Gomez, E. and Fernandez, 46 Azmi, A.S., Bhat, S.H., Hanif, S. and Hadi, S.M. (2006) FEBS Lett. 580,
P. (2002) Mol. Pharmacol. 61, 294–302 533–538
24 Olas, B., Wachowicz, B., Bald, E. and Glowacki, R. (2004) J. Physiol. 47 Yoshida, Y., Furuta, S. and Niki, E. (1993) Biochim. Biophys. Acta 1210,
Pharmacol. 55, 467–476 81–88
25 Yen, G.C., Duh, P.D. and Lin, C.W. (2003) Free Radical Res. 37, 509–514 48 Agarwal, K., Sharma, A. and Talukder, G. (1989) Chem. Biol. Interact. 69,
26 Hung, L.M., Chen, J.K., Huang, S.S., Lee, R.S. and Su, M.J. (2000) 1–16
Cardiovasc. Res. 47, 549–555 49 Fukuhara, K., Nagakawa, M., Nakanishi, I., Ohkubo, K., Imai, K., Urano, S.,
27 Hattori, R., Otani, H., Maulik, N. and Das, D.K. (2002) Am. J. Physiol. Heart Fukuzumi, S., Ozawa, T., Ikota, N., Mochizuki, M. et al. (2006) Bioorg.
Circ. Physiol. 282, H1988–H1995 Med. Chem. 14, 1437–1443
28 Lorenz, P., Roychowdhury, S., Engelmann, M., Wolf, G. and Horn, T.F. 50 Cal, C., Garban, H., Jazirehi, A., Yeh, C., Mizutani, Y. and Bonavida, B.
(2003) Nitric Oxide 9, 64–76 (2003) Curr. Med. Chem. Anticancer Agents 3, 77–93
29 Tsai, S.H., Lin-Shiau, S.Y. and Lin, J.K. (1999) Br. J. Pharmacol. 126,
673–680
30 Wadsworth, T.L. and Koop, D.R. (1999) Biochem. Pharmacol. 57, Received 24 May 2007
941–949 doi:10.1042/BST0351156


C The Authors Journal compilation 
C 2007 Biochemical Society

Вам также может понравиться