Hostname: page-component-7c8c6479df-8mjnm Total loading time: 0 Render date: 2024-03-28T12:27:04.073Z Has data issue: false hasContentIssue false

Use of conventional and -omics based methods for health claims of dietary antioxidants: a critical overview

Published online by Cambridge University Press:  01 May 2008

Siegfried Knasmüller*
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Armen Nersesyan
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Miroslav Mišík
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Christopher Gerner
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Wolfgang Mikulits
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Veronika Ehrlich
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Christine Hoelzl
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Akos Szakmary
Affiliation:
Institute of Cancer Research, Inner Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090Vienna, Austria
Karl-Heinz Wagner
Affiliation:
Department of Nutritional Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
*
*Corresponding author: Professor Siegfried Knasmüller, fax +431 4277 9651, email siegfried.knasmueller@mediuniwien.ac.at
Rights & Permissions [Opens in a new window]

Abstract

This article describes the principles and limitations of methods used to investigate reactive oxygen species (ROS) protective properties of dietary constituents and is aimed at providing a better understanding of the requirements for science based health claims of antioxidant (AO) effects of foods. A number of currently used biochemical measurements aimed of determining the total antioxidant capacity and oxidised lipids and proteins are carried out under unphysiologcial conditions and are prone to artefact formation. Probably the most reliable approaches are measurements of isoprostanes as a parameter of lipid peroxidation and determination of oxidative DNA damage. Also the design of the experimental models has a strong impact on the reliability of AO studies: the common strategy is the identification of AO by in vitro screening with cell lines. This approach is based on the assumption that protection towards ROS is due to scavenging, but recent findings indicate that activation of transcription factors which regulate genes involved in antioxidant defence plays a key role in the mode of action of AO. These processes are not adequately represented in cell lines. Another shortcoming of in vitro experiments is that AO are metabolised in vivo and that most cell lines are lacking enzymes which catalyse these reactions. Compounds with large molecular configurations (chlorophylls, anthocyans and polyphenolics) are potent AO in vitro, but weak or no effects were observed in animal/human studies with realistic doses as they are poorly absorbed. The development of -omics approaches will improve the scientific basis for health claims. The evaluation of results from microarray and proteomics studies shows that it is not possible to establish a general signature of alterations of transcription and protein patterns by AO. However, it was shown that alterations of gene expression and protein levels caused by experimentally induced oxidative stress and ROS related diseases can be normalised by dietary AO.

Type
Full Papers
Copyright
Copyright © The Authors 2008

It is well documented that oxidative stress, defined by Blomhoff(Reference Blomhoff1) as a “condition that is characterised by the accumulation of non-enzymatic oxidative damage to molecules that threaten the normal functions of a cell or the organism” is involved in the aetiology of a large number of human diseases. Typical examples are various forms of cancer (breast, colon and liver), neuropathological disorders such as Parkinson's and Altzheimer's disease, inflammations including hepatitis and inflammatory bowel diseases, different types of dermatitis as well as bacterial and viral infections (HBV, sepsis), diabetes, and rheumatoid arthritis (for review see(Reference Elahi and Matata2)). Also coronary heart diseases which are the major cause of death in industrialised countries(Reference Elahi and Matata2, Reference Lefer and Granger3) and idiopathic infertility(Reference Agarwal, Sharma, Nallella, Thomas, Alvarez and Sikka4, Reference Pasqualotto, Sharma, Kobayashi, Nelson, Thomas and Agarwal5), which has increased over the last decades in Western countries seem to be causally related to reactive oxygen species (ROS) meditated damage and it has been stressed that oxidative damage is also involved in several diseases of ageing, e.g. Werner's syndrome(Reference Von Kobbe, May, Grandori and Bohr6), progeria(Reference Yan, Li, Jiang and Oberley7), amyotrophic lateral sclerosis, cataract formation and decreased immune functions(Reference Kohen and Nyska8Reference Calabrese, Guagliano, Sapienza, Mancuso, Butterfield and Stella10).

Already half a century ago it was found that the acute toxic, DNA-damaging and carcinogenic effects of ionising radiation which are predominantly caused by the formation of ROS can be reduced by antioxidant vitamins such as C, E, and A(Reference Zimmermann and Kimmig11Reference Gebhart13). In the following decades, it became apparent that plant derived foods as well as beverages contain a large number of compounds which protect against oxidative damage and its consequences. Typical examples for such antioxidants which have been defined as “redox-active compounds that reduce pro-oxidative stress by reacting non-enzymatically with a reactive oxidant”(Reference Blomhoff1) are flavonoids and phenolic acids contained in fruits and vegetables(Reference Li, Tsao, Yang, Liu, Zhu and Young14Reference Erdman, Balentine, Arab, Beecher, Dwyer, Folts, Harnly, Hollman, Keen, Mazza, Messina, Scalbert, Vita, Williamson and Burrowes17), allyslulfides in Allium species(Reference Kim, Chang, Kim, Chang and Chun18), hydroxycinnamic acids in coffee(Reference Iwai, Kishimoto, Kakino, Mochida and Fujita19, Reference Fujioka and Shibamoto20), phenolic compounds in wines(Reference Lopez-Velez, Martinez-Martinez and Del Valle-Ribes21) and vegetable oils(Reference Hidalgo, Leon and Zamora22), catechins in teas(Reference Luximon-Ramma, Neergheen, Bahorun, Crozier, Zbarsky, Datla, Dexter and Aruoma23), specific ingredients of common spices such as capsaicin in chillies(Reference Antonious, Kochhar, Jarret and Snyder24), gingerol(Reference Masuda, Kikuzaki, Hisamoto and Nakatani25) and curcumin(Reference Somparn, Phisalaphong, Nakornchai, Unchern and Morales26), chlorophylls(Reference Hsu, Yang, Chen, Chao and Hu27), anthocyanins in berries(Reference Xiong, Melton, Easteal and Siew28) as well as carotenoids(Reference Semba, Lauretani and Ferrucci29) to name only a few.

The increasing evidence of the strong impact of the redox status on human health has stimulated intense research activities in this field. It has been estimated that around 10 papers dealing with oxidative stress and/or antioxidants are published daily(Reference Blomhoff1) and many of them concern the identification of dietary compounds in the diet and investigations concerning their mode of action. The results of these efforts have a strong impact on the development of nutritional recommendations and led to the development of supplements which contain high levels of food derived antioxidants(Reference Aruoma, Sun, Fujii, Neergheen, Bahorun, Kang and Sung30) and to the production of functional foods. A broad variety of different methods are currently used to study antioxidants in human foods and to identify and characterise their active principles. The models include chemical–analytical and physical measurements, experiments with subcellular fractions and intact cells, animal studies as well as human intervention trails. In the last decade, new biomarkers have been developed and validated which can be used in human studies and the rapid development of -omics techniques (in particular the use of microarrays and two dimensional gel electrophoresis) offers the possibility to explore the effects of antioxidants on gene expression and protein levels and to study alterations of disease related patterns(Reference Narayanan31Reference Scandalios33).

The aim of the present article is it to give a critical overview on the advantages and limitations of the different approaches which are currently used with particular emphasis on the newly developed methods. We anticipate that it will help in the interpretation of existing data and lead to the development of improved strategies concerning the detection of antioxidants.

The formation of ROS as well as their physical and chemical properties, their reactions with organic molecules and their inactivation by antioxidants have been extensively described in the scientific literature(Reference Choe and Min34Reference Karihtala and Soini36). Therefore, these topics are confined in the present article to short descriptions which are essential to understand the subsequent chapters.

Formation of reactive oxygen species (ROS)

Pro-oxidants (often termed as reactive oxygen species) can be classified in two groups, namely radicals and non-radicals. Radicals (O2, , OH, ROO, RO, and NO) contain unpaired electrons in the shells around the nucleus which causes the high reactivity of these species (except O2), due to their ability to donate or receive other atoms to obtain stability. Important non-radicals comprise hyperchlorous acid (HOCl), hydrogen peroxide (H2O2), organic peroxides, aldehydes and ozone (O3). The most relevant ROS as well as some of their main reactions are shown in Fig. 1.

Fig. 1 Different form of reactive oxygen species and their interaction with organic molecules.

Superoxide , which is formed for example during respiration in mitochondria (as a consequence of reduction of oxygen required for APT production) forms at low pH hydroperoxyl (HO2 which penetrates the cell membranes more easily than the charged form(Reference Fridovich37, Reference Halliwell and Gutteridge38). Enzymatic as well as non-enzymatic dismutation leads to formation of hydrogen peroxide (H2O2) which can be detoxified enzymatically (catalase, glutathion peroxidase). H2O2 molecules can damage cells at low concentrations and degrade haem proteins and oxidise DNA, enzymes, –SH groups and keto acids and are also the source of more deleterious species such as HOCl and OH. The latter radical is short lived and reacts at a high rate with most organic molecules (DNA, amino acids, sugars, proteins, lipids). Transition metals (first row of the D block of the periodic table) contain unpaired electrons (except Zn) and can therefore be considered as radicals. In particular copper and iron are contained at relatively high concentrations in many organisms. At physiological pH, most of the metals are present in oxidised forms (Fe3+∙, Fe2+), but after reduction (e.g. by ascorbic acid or via the Haber-Weiss reaction ), they can undergo “Fenton type” reactions (e.g. Fe2+ + H2O2 → Fe3+ + OH + OH). These two processes explain the formation of OHin vivo. However, it is notable that in organisms, metals are always bound to proteins and membranes and it has been shown that they can undergo in this state the aforementioned reactions and produce OH at a single site and convert non-reactive radicals to highly reactive species.

Nitric oxide (NO) is produced by oxydation of the terminal guanidine-nitrogen atoms of arginine(Reference Halliwell and Gutteridge38Reference Czapski and Goldstein40). This reaction is catalysed by nitric oxide synthetases (NOS, i.e. neuronal NOS, endothelial NOS and inducible NOS). NO can react with different radicals, the most important reaction under physiological conditions is the formation of peroxynitirite (ONOO− ) in which is involved(Reference Halliwell and Gutteridge38, Reference Czapski and Goldstein40). ONOO−  can cause damage similar to that induced by OH∙(Reference Beckman and Koppenol39).

The biological significance of the different ROS species has been discussed quite controversially. Some authors suggested that and NO are the most relevant ones, while other stressed that peroxyl radicals may be even more important(Reference Kohen and Nyska8).

Several exogenous factors contribute to oxidative stress. Ionising radiation causes toxic effects in organisms primarily via ionisation of intracellular water(Reference Shadyro, Yurkova and Kisel41). Also non-ionising radiation (UV light) can indirectly produce a variety of ROS species including O3 (Reference Pentland42). Other major sources of exposure are air pollutants such as cigarette smoke(Reference Koren43) and car exhausts(Reference Victorin44), drugs (bleomycin, doxorubicine)(Reference Yu and Anderson45) as well as pesticides and herbicides(Reference Wiseman, Ridgway, Goldfarb and Woods46) and industrial chemicals(Reference Parke and Sapota47). Also pathogenic microorganisms may produce oxygen species, but the most relevant external source is nutrition as most of the foods we consume are oxidised and contain oxidants such as peroxides, aldehydes, fatty acids and transition metals(Reference Ames48, Reference Kanner and Lapidot49).

The most important endogenous processes of ROS production are respiration processes in the mitochondria and the massive continuous production of radicals is even increased in ageing cells(Reference Richter, Gogvadze, Laffranchi, Schlapbach, Schweizer, Suter, Walter and Yaffee50, Reference Fleury, Mignotte and Vayssiere51). Another source are white blood cells involved in immune responses which can undergo a respiratory bust that is characterised by an up to 20-fold increase of oxygen production(Reference Dhalla, Temsah and Netticadan52, Reference Forman and Torres53). During this reaction, NAPDH serves as a donor of electrons which results in the production of from oxygen. The enzyme myeloperoxidase catalyses the production of HOCl by interaction between H2O2 peroxides and chlorides(Reference Hawkins, Brown and Davies54, Reference Rodrigues, Rodriguez, Henrique Catalani, Russo and Campa55).

Targets of oxidative damage

The continuous exposure to ROS from exogenous and endogenous sources results in oxidative damage of many cell components and alterations of cellular functions; some of these changes can be used as markers of oxidative stress and to investigate putative protective effects of phytochemicals.

Proteins

Radicals react in particular with nucleophilic amino acids for example with tryptophane, histidine and cysteine(Reference Davies56, Reference Grune, Reinheckel and Davies57). Apart from direct oxidation of SH-groups by H2O2 and , organic radicals may bind covalently to cellular proteins which are part of cell membranes or have enzymatic functions. One of the major adducts which can be easily detected is 3-nitrotyrosine which is produced by interactions between ONOO and other nitrogen reactive radicals with the amino acid tyrosine(Reference Levine and Stadtman58). Also relatively resistant amino acids such as lysine and proline can be hydroxylated non-enzymatically by OH∙(Reference Stadtman59).

It is also known that ROS can destroy peptide bonds and cause drastic alterations of their structures resulting in changes of their cellular functions (for review see(Reference Karihtala and Soini36)). NO reacts in particular with Fe–S centres of proteins which transport electrons and this affects the functions of mitochondria(Reference Kushnareva, Murphy and Andreyev60). Another important feature is their reaction with thiol groups of proteins; a typical example is the S-nitrosylation of caspases which are part of cell signalling processes(Reference Squadrito and Pryor61).

Lipids

All cellular membranes are vulnerable to oxidation due to their high concentrations of unsaturated fatty acids. Damage of lipids by ROS (lipid peroxidation, LP) occurs in three stages. In the first (“initiation phase”), double bonds of fatty acids are attacked by radicals which leads to formation of fatty acids radicals. During the “propagation”, a chain reaction takes place which leads to continuous formation of these radicals. The last stage (chain determination) occurs following interactions ROO or with other radical types and/or antioxidants (for a detailed description of LP see(Reference Halliwell and Gutteridge38, Reference Younes and Siegers62, Reference Horton and Fairhurst63)). Important marker molecules formed during LP are aldeydes and ketones, for example malonedialdehyde (MDA) and 4-hydroxynonenal (4-HNE) as well as the family of isoprostanes which are excreted in the urine. Oxidation of fatty acids can be measured with the relative change in the fatty acid pattern and the formation of conjugated dienes.

Blood cholesterols can be oxidised by ROS to form oxidised low density lipoproteins (LDL). The oxidative modification hypothesis of atherosclerosis assumes that circulating LDL particles are modified by oxidation and that these particles are then taken up by macrophages inside the arterial wall and form the start of atherosclerotic plaques(Reference Zock and Katan64, Reference Forrester and Shah65).

DNA damage

ROS can damage DNA either directly or indirectly (via LP). The major part of damage is attributable to OH radicals which interact either with the sugar phosphate chain or oxidise bases and form reaction products such as thymine glycol, 5-hydroxy-uracil and 8-hydoxy-desoxyguanosine (8-OH-dG)(Reference Moller and Wallin66, Reference Barzilai and Yamamoto67). Also NO can damage DNA indirectly via formation of peroxynitrite, another pathway of damage is the formation of covalent bonds between radicals and nucleobases(Reference Halliwell and Gutteridge38, Reference Niles, Wishnok and Tannenbaum68, Reference Tsutsui, Ide and Kinugawa69).

The most important indirect form of DNA damage is caused by LP (for review see(Reference Vaca, Wilhelm and Harms-Ringdahl70)). Lipid radicals formed during the chain reaction cause adduct formation, strand breaks and DNA-protein crosslinks. The former lesions are also induced by specific end products of the reaction such as alkenes and alkanes(Reference Ehrenberg, Osterman-Golkar, Segerbck, Svensson and Calleman71, Reference Segerback72) and numerous studies have been published which concern the genotoxic effects of reaction products such a MDA and 4-HNE(Reference Vaca, Wilhelm and Harms-Ringdahl70). Nuclelotide- as well as base excision repair mechanisms (BER and NER) prevent the persistence of oxidative lesions. Oxidised guanosine is removed by the action of glycosylases such as 8-oxoguanine DNA glycosylase 1 and 2 (OGG1 and OGG2); mispaired adenines by MYH and MTH1. NEIL glycosylases containing β/δ-elimination activities excise a broad range of oxidatively damaged bases, including 5-hydroxyuracil (5-OHU), thymine glycol (Tg), uracil, 8-oxoguanine (8-oxoG) and ring-fragmented purines(Reference Doublie, Bandaru, Bond and Wallace73). Also pyrimidine derived lesions are a substrate of this latter enzyme, important uracil specific enzymes are for example UNG and SMUG1. A detailed description of repair of oxidised DNA can be found in the articles of Sanderson et al. (Reference Sanderson, Bennett, Sung and Mosbaugh74), Krokan et al. (Reference Krokan, Drablos and Slupphaug75) and Cooke et al. (Reference Cooke, Evans, Dizdaroglu and Lunec76).

Oxidative stress, cell signalling and activation of transcription factors

The exposure of organisms to ROS causes dramatic changes in gene regulation patterns and protein synthesis. In lower eucaryotes, e.g. in yeast the expression of up to 1/3 of the genes is affected by oxidative stress(Reference Kim, Chang, Chung, Park and Seo77), mammalian cells are somewhat less flexible but still hundreds of genes have been identified which react towards oxidative damage. During the last decades, complex signalling pathways have been discovered which activate transcription factors involved in gene regulation. The networks involved in ROS mediated cellular responses have been described in a number of reviews(Reference Valko, Rhodes, Moncol, Izakovic and Mazur78Reference Valko, Leibfritz, Moncol, Cronin, Mazur and Telser84). In the present article we will give a short overview on the most important processes since the mode of action of dietary antioxidants may involve changes of signalling pathways which cause activation of cellular defence systems(Reference Ma, Battelli and Hubbs85).

Alterations of cell signalling pathways

Oxidants can impinge signalling either by influencing redox dependent protein–protein interactions or via altering enzyme activities, i.e. oxidations leading to inhibition of phosophor/serine/threonine-, phosphothyrosine- and phospholipid-phosphatases(Reference Mathers, Fraser, McMahon, Saunders, Hayes and McLellan82). The key reactions which have been identified are interactions with sulphydryl groups on their cysteine residues(Reference Poli, Leonarduzzi, Biasi and Chiarpotto86).

The intracellular responses can be grouped in two categories namely receptor-mediated ones and non-receptor effects. Fig. 2 gives a schematic overview on the different processes.

Fig. 2 Impact of ROS on cell signalling and activation of transcription factors.

Growth factors and cytokines (e.g. TNF-α, IL-1) cause ROS production in non-pathogenic cells and activate intracellular receptor mediated signalling which affect mitogen activated protein kinases (MAPKs). Growth factor receptors are tyrosine kinases (RTLs), apart from these, also non-receptor protein kinases have been identified which are also activated by ROS and belong for example to the Src family(Reference Abe and Berk87, Reference Esposito, Chirico, Montesano Gesualdi, Posadas, Ammendola, Russo, Cirino and Cimino88). Activated Src binds to membranes and initiates MAPKs, NfκB and PI3K signalling pathways(Reference Valko, Rhodes, Moncol, Izakovic and Mazur78). Other important targets involved in signalling are Ras (membrane bound G proteins involved in the regulation of cell growth), protein tyrosine phosophatases (PTP) and seronine/threonine kinases. The most important representative of the latter group is protein kinase C (PKC), its catalytic site is a zinc finger domain containing several cysteine rich regions which can be modified by various oxidants(Reference Gopalakrishna and Jaken89).

MAPKs (for a detailed description see(Reference Kyriakis and Avruch90)) relay signals generated by exogenous or endogenous stimuli to intracellular space via phosphorylation of proteins. During this process, the kinases interact also with downstream mediators including transcription factors(Reference Lopez-Ilasaca, Crespo, Pellici, Gutkind and Wetzker91). Studies on the upregulation of MAPKs have shown that these processes are type and stimuli specific. For example, it was found that endogenous H2O2 production by respiratory burst induces ERK but not p38 kinase(Reference Iles and Forman92) while exogenous peroxide treatment activates the latter enzyme(Reference Torres and Forman93).

Activation of transcription factors by ROS

The most significant effects of ROS on MAPKs concern the activation of transcription factors which control the expression of protective genes, arrest division of damaged cells and induce apoptosis (programmed cell death).

AP-1 is a collection of dimeric basic region-leucine zipper proteins which are for example induced by metals and H2O2 (Reference Rao, Luo and Hogan94, Reference Pinkus, Weiner and Daniel95) and regulate cell growth, differentiation and apoptosis.

NFκB is a DNA binding protein which is sequestered in the cytoplasm because of an interaction with a member of the inhibitory IκB family. Activation via ROS causes dissociation and allows NFκB to enter the nucleus and activate genes involved in inflammatory responses, transformation and angiogenesis(Reference Amiri and Richmond96). A number of investigations showed that that activation by different stimuli can be blocked by antioxidants including N-acetlylcysteine, cysteine, vitamin E, thiols and green tea polyphenolics(Reference Valko, Rhodes, Moncol, Izakovic and Mazur78).

Another important factor which plays a key role in protecting cells from malignant transformation is p53, also termed a “tumour suppressor” since it arrests cell cycle and induces apoptosis(Reference Hofseth, Hussain and Harris97). p53 is directly activated by oxidants and its overexpression leads to increase of intracellular ROS levels. One of the important functions of p53 is the up regulation of proteins that play a role in ROS mediated apoptosis namely ferrodoxin reductase (FDXR) and a novel stress-response gene Redd1/HIF-1 originally isolated as an HIF-1-response gene(Reference Jin, An, Lee, Woo, Seo, Choe, Yoo, Lee, Um, Lee, Park, Kim, Hong, Rhee and Park98). It was shown in a number of in vitro studies that antioxidants reduce apoptosis rates due to interaction with p53(Reference Li, Jang and Surh99, Reference Sablina, Budanov, Ilyinskaya, Agapova, Kravchenko and Chumakov100). In a recent human intervention study we observed a drastic reduction of the apoptosis frequencies in lymphocytes after consumption of wheat sprouts which is probably due to antioxidant effects(Reference Ehrlich, Hoelzl, Nersesyan, Winter, Koller, Ferk, Fenech, Dusinska, Knasmüller, Ďuračková, Slámenova and Micksche101).

Two other transcription factors affected by ROS are nuclear factor of activated T cells (NFAT) and HIF. The former family regulates muscle growth and differentiation as well as cytokine formation and angiogenesis(Reference Rao, Luo and Hogan94, Reference Jauliac, Lopez-Rodriguez, Shaw, Brown, Rao and Toker102); while the latter is a heterodimer controlling genes encoding for vascular endothelial growth factor (VEGF), aldolase, enolase and lactate dehydrogenase(Reference Semenza103).

Probably the most important contribution to cell defence against oxidative stress is mediated through transcriptional activation of genes via a cis-acting enhancer known as antioxidant responsive element (ARE) which was discovered by Pickett and co-workers(Reference Huang, Nguyen and Pickett104Reference Favreau and Pickett106) and identified in the 5′flanking regions of many genes. A number of studies showed that the transcription factor Nrf2 which belongs to the CNC (cap‘N'collar) basic leucine zipper family and is represented in many tissues is the key mediator of ARE dependent activation(Reference Gupta, Dobashi, Greene, Orak and Singh107, Reference Klaunig and Kamendulis108). Comparative investigations with genetically altered rodents (Nrf2+/+ and Nrf2 − / − ) showed that numerous genes are regulated by the element including those which encode for protection against ROS such as glutamate cystein ligase (GCL) which catalyses the rate limiting step of glutathione synthesis, NADPH quinone oxidoreductase (NQO), glutathione S-transferase (GST), aldehyde dehydrogenase (ADH), glutathione peroxidase, glutathione reductase, peroxiredoxin I (PrxI), superoxide dismutase (SOD), catalase, and thioredoxin(Reference Michiels, Raes, Toussaint and Remacle109, Reference Fridovich110). Also enzymes which are involved in the supply of reducing equivalents (e.g. glucose-6-phosphate dedydrogenase) as well as xenobiotic drug metabolising enzymes (e.g. CYPs), chaperones, and stress response proteins are regulated by Nrf2(Reference Kwak, Wakabayashi, Itoh, Motohashi, Yamamoto and Kensler111, Reference Thimmulappa, Mai, Srisuma, Kensler, Yamamoto and Biswal112).

Recent investigations showed that the actin binding protein Kelch-like Ech-associated protein (Keap1) regulates transcription factor Nrf2 by controlling its stability and subcellular localisation(Reference McMahon, Itoh, Yamamoto and Hayes113Reference Kwak and Kensler115). The disruption of the Keap-Nrf2 complex by oxidative stress leads to Nrf2 accumulation in the nucleus where it is associated with small MAF transcription factors and mediates ARE dependent gene expression (see Fig. 2).

It is well documented that chemicals which release ROS such as metals(Reference He, Lin, Chen, Zhang and Ma116, Reference He, Chen, Lin and Ma117) and vascular diseases (arteriosclerosis, diabetes, chronic renal failure, preeclampsia) both cause induction of the different transcription factors described above(Reference Mann, Niehueser-Saran, Watson, Gao, Ishii, de Winter and Siow118).

The interaction of phytochemicals with these processes has been reviewed in several articles(Reference Mann, Niehueser-Saran, Watson, Gao, Ishii, de Winter and Siow118Reference Surh, Kundu, Na and Lee121). It was shown that phenolics such as EGCG and resveratrol and spice ingredients (e.g. capsaicin, curcumin) inhibit the transcription factors NFkB, AP-1 and β-catenin-TcF signalling via interaction with upstream signalling pathways (IKK phosphorylation, MAPK phosphorylation and PI3K/Akt phosphorylation), in parallel proinflammatory mediators (TNF-α, IL, PGE2 and NO) and the activities of proinflammatory enzymes (COX 2, iNOS) were reduced(Reference Kundu and Surh119). In the case of COX 2, it is known that inhibition by synthetic compounds such as non-steroidal anti-inflammatory drugs (NSAIDS) is paralleled by decreased rates of colon and colorectal cancers in humans(Reference Stange122, Reference Chia, Newcomb, Bigler, Morimoto, Thibodeau and Potter123). On the contrary, the induction of detoxifying enzymes (including those which inactivate ROS) is due to activation of the transcription factor Nrf2. Typical example for dietary antioxidants which cause an induction of this transcription factor are synthetic and tea specific phenolics, isothicoyanates and sulforaphane(Reference Kong, Owuor, Yu, Hebbar, Chen, Hu and Mandlekar124), curcumin(Reference Nishinaka, Ichijo, Ito, Kimura, Katsuyama, Iwata, Miura, Terada and Yabe-Nishimura125) and flavonoids(Reference Tanigawa, Fujii and Hou126, Reference Gonzalez-Gallego, Sanchez-Campos and Tunon127).

The molecular mechanisms by which phytochemicals interact with signal transmission cascades are not precisely known. It is supposed that the downregulation of transcription factors may be due to the direct scavenging of ROS. In the case of Nrf2 it was shown that the activation by sulforaphane and synthetic alkylating compounds is due to modifications of cysteine residues of Keap1, a sensor protein which regulates Nrf2(Reference Hong, Freeman and Liebler128Reference Eggler, Liu, Pezzuto, van Breemen and Mesecar131). However it cannot be excluded that Nrf2 activation seen with certain phytochemicals may be due to release of ROS; it is known that antioxidants (in particular phenolics) can act under certain conditions as pro-oxidants.

Conventional and new methods for the detection of dietary antioxidants

A large number of different techniques have been developed to monitor oxidative damage and its consequences; these approaches can be also used to identify dietary antioxidants and their mode of action. These methods are often applied in experimental systems in which oxidative stress is induced by specific treatments or diseases. The most frequently employed models are described in the next chapter; the following sections describe physics–based and biochemical methods, techniques for the detection of oxidative DNA-damage, approaches used to investigate alterations of signalling pathways as well as the advantages and disadvantages of -omics techniques (Fig. 3).

Fig. 3 Overview of different methods used for the detection of dietary antioxidants.

Induction of oxidative stress in biological systems

In experiments with subcellular fractions and in in vitro experiments with cells, ROS are in most cases generated by chemical reactions, for example with the xanthine/xanthine oxidase system by hydroquinone oxidation(Reference Abdelwahed, Bouhlel, Skandrani, Valenti, Kadri, Guiraud, Steiman, Mariotte, Ghedira, Laporte, Dijoux-Franca and Chekir-Ghedira32, Reference Le, Hailer, Buhrow, Wang, Flatten, Pediaditakis, Bible, Lewis, Sausville, Pang, Ames, Lemasters, Holmuhamedov and Kaufmann132, Reference Wiseman133) which generate . Transition metals such as copper and iron play a major role in Fenton type reactions thereby forming mainly OH radicals. Another frequently used approach is the use of chemicals such as H2O2, t-butyl-hydrogenperoxide or bleomycin which release and OH or of compounds such as menadione(Reference Criddle, Gillies, Baumgartner-Wilson, Jaffar, Chinje, Passmore, Chvanov, Barrow, Gerasimenko, Tepikin, Sutton and Petersen134), paraquat(Reference McCarthy, Somayajulu, Sikorska, Borowy-Borowski and Pandey135), and plumbagin(Reference Srinivas, Gopinath, Banerji, Dinakar and Srinivas136) which form O2.

Activated phagocytic cells produce oxygen radicals as part of their defence system and a burst of ROS can be induced by exposing such cells to bacteria, particles or certain chemicals; one of the most powerful responses can be evoked with the tumour promotor phorbol myristate acetate(Reference Phillips, Anderson and Gangolli137).

Chemicals which generate ROS are rarely used in animal studies due to their high reactivity. A more convenient way to cause oxidative damage which has been also used in numerous in vitro experiments is ionising radiation. Indirect approaches are feeding of vitamin E deficient diets(Reference Minamiyama, Takemura, Hai, Suehiro and Okada138), iron overload(Reference Isomura, Fujie, Shibata, Inoue, Iizuka, Takebe, Takahashi, Nishihira, Izumi and Sakamoto139) or inhalation of oxygen(Reference Gregorevic, Lynch and Williams140).

Organ specific inflammations can be induced with certain chemicals; for example liver cirrhosis with CCl4(Reference He, Luo, Wang, Wang, Fu, Xu, Zhao and Liu141) or thioacetamide(Reference Bruck, Ashkenazi, Weiss, Goldiner, Shapiro, Aeed, Genina, Helpern and Pines142). 2,4,6-Trinitrobenzene sulfonic acid, oxazolone and dextran sodium sulfate are used in models for inflammatory bowel diseases(Reference Wirtz, Neufert, Weigmann and Neurath143); diabetes can be caused by the antibiotic streptozotocin.

In the last years, a variety of genetically altered mice and rat strains have been developed as models for ROS-related diseases for example animals which are deficient in specific SOD forms(Reference Fujimoto, Arakawa, Shibaya, Miida, Ando, Yasumo, Hara, Uchiyama, Iwabuchi, Takasaki, Manabe and Yamoto144) and GST isozymes(Reference Fujimoto, Arakawa, Shibaya, Miida, Ando, Yasumo, Hara, Uchiyama, Iwabuchi, Takasaki, Manabe and Yamoto144).

Disease related models include those of ataxia-telangiectasia(Reference Erker, Schubert, Elchuri, Huang, Tarin, Mueller, Zielen, Epstein and Wynshaw-Boris145), Alzheimer's(Reference Ashe146) and Parkinson's(Reference Hamaue, Ogata, Terado, Ohno, Kikuchi, Sasaki, Tashiro, Hirafuji and Minami147) disease, ageing models(Reference Zha, Le, Higami, Shimokawa, Taguchi and Razzaque148). In addition, also rodent species have been developed which are deficient in specific genes encoding for repair of oxidative DNA damage such as Ogg1 (Reference Larsen, Kwon, Coin, Egly and Klungland149) and Myh (Reference Xie, Yang, Cunanan, Okamoto, Shibata, Pan, Barnes, Lindahl, McIlhatton, Fishel and Miller150).

Most human studies on dietary antioxidants are carried out with healthy volunteers. In this context it is notable that a number of parameters such as age, sex, body mass index, and seasonal variations were found to affect the redox status and should be taken into consideration in dietary studies(Reference Glasziou and Sanders151). In the last years, a number of antioxidant studies has been conducted in which oxidative stress was induced by physical exercise(Reference Hartmann, Niess, Grunert-Fuchs, Poch and Speit152, Reference Sacheck, Milbury, Cannon, Roubenoff and Blumberg153) or hyperbaric treatment(Reference Dennog, Radermacher, Barnett and Speit154) and also with patients with ROS related diseases such as diabetes(Reference Astley, Langrish-Smith, Southon and Sampson155Reference Sampson, Astley, Richardson, Willis, Davies, Hughes and Southon157), HIV(Reference Jaruga, Jaruga, Gackowski, Olczak, Halota, Pawlowska and Olinski158), atherosclerosis and coronary artery diseases(Reference Soccio, Toniato, Evangelista, Carluccio and De Caterina159), cancer(Reference Hercberg, Czernichow and Galan160, Reference Flora161), uremia(Reference Kan, Undeger, Bali and Basaran162) or systemic lupus erythematosus(Reference Evans, Cooke, Akil, Samanta and Lunec163).

Conventional methods used for the detection of oxidative stress or for the identification of antioxidative dietary components

This part of the review will focus on conventional methods used to describe oxidative stress, and will be divided into five parts namely (1) physics based approaches, (2) methods used for the determination of the antioxidant compounds; (3) biochemical methods used to monitor the oxidation of macromolecules and their oxidation products, (4) approaches for the detection of ROS induced DNA-damage and (5) methods used to measure antioxidant enzymes and transcriptional factors.

Trapping of reactive species

The only technique that can measure free radicals directly and specify them is electron spin resonance (ESR), because it detects the presence of unpaired electrons. However, ESR can only be used to monitor only fairly unreactive radicals, since reactive ones do not accumulate at high-enough levels. One solution to this problem is to add ‘traps’ or ‘probes’, i.e. agents that intercept reactive radicals reacting with them to form a stable radical that can be detected by ESR. Whole-body ESR techniques are being used with rodents(Reference Berliner, Khramtsov, Fujii and Clanton164) but are currently not applicable to humans due to the lack of human safety data on the probes. A wide range of traps is available for use in animals and cell culture systems, not only N-tertbutyl-p-phenylnitrone (PBN) and 5,5-dimethyl-1-pyrroline-N-oxide (DMPO)(Reference Khan, Wilmot, Rosen, Demidenko, Sun, Joseph, O'Hara, Kalyanaraman and Swartz165) which were frequently used, but also “newcomers” as 1,1,3-trimethyl-isoindole N-oxide (TMINO)(Reference Bottle, Hanson and Micallef166), 5,5-diethylcarbonyl-1-pyrroline N-oxide (DECPO)(Reference Karoui, Clément, Rockenbauer, Siri and Tordo167), N-2-(2-ethoxycarbonyl-propyl)-a-phenyl-nitrone (EPPN)(Reference Stolze, Udilova, Rosenau, Hofinger and Nohl168), 5-diethoxyphosphoryl-5-methyl-1-pyrroline N-oxide (DEPMPO)(Reference Khan, Wilmot, Rosen, Demidenko, Sun, Joseph, O'Hara, Kalyanaraman and Swartz165) and 5-tert-butoxycarbonyl-5-methyl-1-pyrroline-N-oxide (BMPO)(Reference Zhao, Joseph, Zhang, Karoui and Kalyanaraman169). A generally underestimated problem is, that the reaction products giving an ESR signal, can be rapidly removed in vivo and in cultured cells, both by enzymic metabolism and by direct reduction by agents such as ascorbate(Reference Halliwell and Whiteman170).

Approaches to determine the total antioxidant capacity

Two main approaches have been developed to evaluate the antioxidant capacity in foods and human material (in particular plasma and LDL). The first measures the ability of a substance to transfer one electron to reduce compounds like radicals, carbonyls or metals. The most popular tests which belong to this category are the ferric iron reducing antioxidant parameter (FRAP), the Trolox equivalent antioxidant capacity (TEAC), and the diphenyl-1-picrylhydrazyl test (DPPH). Methods which fall into the second category are based on their ability to quench free radicals by hydrogen donation. Some scientists believe that these reactions are similar to the reaction mechanisms of antioxidants(Reference Aruoma171). The most popular methods are the oxygen radical absorbance capacity test (ORAC), the total radical trapping antioxidant parameter (TRAP), the total oxidant scavenging capacity (TOSC) method (all measuring effects in the hydrophilic compartment of the plasma) and the inhibition of linoleic acid and LDL oxidation.

Free radical quenching methods

The ORAC assay, which is based on the work of Ghiselli et al. (Reference Ghiselli, Serafini, Maiani, Azzini and Ferro-Luzzi172), Glazer(Reference Glazer173) and Cao et al. (Reference Cao, Alessio and Cutler174) measures the antioxidant inhibition of ROO induced oxidations. Therefore, it reflects the classical H donating ability of antioxidants in the hydrophilic compartment. The peroxyl-radical reacts with a fluorescent probe thereby forming a non-fluorescent product which can be quantitated by following the fluorescence over time. In earlier studies, β-phycoerythrin was used as the fluorescent agent emitting in the visible region (Exc 495 nm, Em 595 nm), but due to shortcomings and inconsistencies of the results, fluorescein or dichlorofluorescein are currently used, since they are less reactive and more stable(Reference Prior, Wu and Schaich175). The antioxidative activity can be expressed as the lag time or the net integrated area under the fluorescence curve (AUC). ORAC values are reported as Trolox equivalents. Originally, the ORAC assay was limited to the measurement of hydrophilic chain breaking antioxidant capacity. A newer protocol, in which lipophilic and hydrophilic compounds are selectively separated by extraction, allows now also the quantification of lipophilic antioxidants using a mixture of acetone and water(Reference Huang, Ou, Hampsch-Woodill, Flanagan and Deemer176). The advantage of the ORAC assay is that it can be automated. Convincing results have been obtained with 48 or 96 well plates coupled with a microplate reader(Reference Huang, Ou, Hampsch-Woodill, Flanagan and Prior177). One important parameter is the temperature control (37°C), as small temperature differences decrease the reproducibility of the test(Reference Lussignoli, Fraccaroli, Andrioli, Brocco and Bellavite178). A principal drawback of the test is, that the effect of oxidation of the photoreceptor of the protein used does not necessarily reflect protection against oxidative damage of the protein itself(Reference Frankel and Meyer179).

The TRAP assay, proposed by Wayner et al. (Reference Wayner, Burton, Ingold and Locke180) is based on the use of 2,2′-azobis(2,4-amidinopropane)dihydrochloride (AAPH), a hydrophilic azo-compound which generates peroxyl-radicals. AAPH decomposes at 37°C spontaneously with a known rate. Various substances like 2,2-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), R-phycoerythrin or 2′-7′-dichlorofluorescin (DCFH)(Reference DeLange and Glazer181Reference Rota, Chignell and Mason183) have been used as oxidizable agents. A comprehensive review on the different modifications has been published by Ghiselli et al. (Reference Ghiselli, Serafini, Natella and Scaccini184). The basic reactions of the procedure are similar to those of the ORAC assay. The probe reacts with ROO radicals at low concentrations with a significant spectroscopic change in between the native and the oxidized sample and no radical chain reaction beyond sample oxidation should occur. The antioxidant capacity is determined as the time required to consume all antioxidants by extension of the lag time for appearance of the oxidized probe when antioxidants are present. TRAP values are usually expressed as the lag time of the sample compared to the corresponding times for Trolox. The test is relatively complex to perform, requires experience and is time-consuming. The use of the lag phase is based on the assumption that all antioxidants show a lag phase and that the lag phase corresponds to the antioxidative capacity.

One drawback of TRAP and ORAC is the interference of proteins which contribute by ⩾80 % to the total antioxidant capacity(Reference Prior, Wu and Schaich175, Reference Ghiselli, Serafini, Natella and Scaccini184). Therefore, Trolox can be used as an internal standard or the samples must be deproteinized prior to the measurements.

The TOSC assay was initially used for environment related studies on marine organisms(Reference Winston, Regoli, Dugas, Fong and Blanchard185, Reference Regoli and Winston186). It is based on the inhibition of the radical-dependent formation of ethylene from ketomethiolbutyric acid by antioxidants. This procedure permits testing against three different ROS species (i.e. peroxyl-, hydroxyl-radicals as well as peroxynitrite) with physiological relevance and different reactivities. It can be conducted at physiological temperature and pH; non-linear concentration-dependent activity variations can be examined easily and different types of antioxidant reactions (retardant or fast-acting) can be distinguished. However, high throughput analyses are not possible and multiple injections of each sample are required in order to observe ethylene formation. Further limitations are the multiple endpoints of calculated 20, 50 and 80 % TOSC and the DT50 (first derivative of TOSC of 50 %) since it was shown that there is no linear relationship between the different multiple endpoints(Reference Lichtenthaler and Marx187).

The chemiluminescence assay (CL) is a modification of the TRAP assay. Radical formation is followed by CL or photo-CL (PCL). CL is characterized by low emission intensity and by the fact that reactions with oxidants emit CL. The most widely used marker is luminol(Reference Lundqvist, Kricka, Stott, Thorpe and Dahlgren188, Reference Whitehead, Thorpe and Maxwell189), but also biuminescent proteins like pholasin are becoming popular(Reference Witko-Sarsat, Nguyen, Knight and Descamps-Latscha190Reference Glebska and Koppenol192). The antioxidant capacity is the time of depressed light emission, which is measured at 10 % recovery of light output.

Recently Popov et al. (Reference Popov, Völker and Lewin193) have described the PCL, a commercial test system termed PHOTOCHEM for the determination of the integral antioxidative capacity towards O2(Reference Popov, Völker and Lewin193). In a strict sense, the method measures antiradical capacity. In contrast to many other assays used to determine AOC, this procedure requires no standardisation of the pH and of the temperature. However, to date, the system is only marketed by one company (Analytic Jena, Germany) and reagents for the hydrophilic and lipophilic assays are only available from the manufacturer; furthermore, a high throughput is not possible. Ascorbic acid is normally used for the determination of hydrophilic and Trolox for the lipophilic antioxidative capacity.

Low-density lipoprotein (LDL) oxidation is based on the autoxidation of linoleic acid or LDL which is induced in vitro mainly by Cu2+ or some other azo-initiators(Reference Esterbauer, Jurgens, Quehenberger and Koller194Reference Rice-Evans, Leake, Bruckdorfer and Diplock196). LDL-oxidation is of higher physiological relevance when tested under in vivo conditions and not ex vivo. The oxidation is monitored at 234 nm for conjugated dienes or by peroxide values for lipid hydroperoxides. LDL has to be freshly isolated from blood which is a time- and material-consuming procedure which requires ultracentrifugation. During the preparation, low temperature and light protection are essential(Reference Wagner, Tomasch and Elmadfa197). Further, conjugated dienes can be formed in presence of polyunsaturated fatty acids.

Recently, fluorescence and UV based ELISA assays with plasma became available, for which no complicated and time consuming LDL isolation is needed. This methods can also be used for larger human trials. Also the procedure developed by Holvoet et al. (Reference Holvoet, Donck, Landeloos, Brouwers, Luijtens, Arnout, Lesaffre, Vanrenterghem and Collen198) who measured oxidized LDL levels by a competitive ELISA utilizing a specific murine monoclonal antibody (mAb-4E6) based on UV is employed quite often at present(Reference Holvoet, Donck, Landeloos, Brouwers, Luijtens, Arnout, Lesaffre, Vanrenterghem and Collen198). The AOC is determined in all these experiments either as AUC or as the lag time until the antioxidants are consumed. An important modification was developed by Frankel et al. (Reference Frankel, German and Davis199) who determined the secondary oxidation product hexanal from LDL. Hexanal was chosen, since it is the major oxidation product of n-6 fatty acids and is monitored with head space gas chromatography, the percentage inhibition of hexanal formation is used as a parameter for AOC. In many ex vivo studies LDL was isolated, subsequently the substances were added and tested on their ability to delay oxidation. This scenario does not reflect in vivo conditions. Furthermore, not all oxidation inducers which are used ex vivo can be used for in vivo testing(Reference Frankel and Meyer179).

The Crocin bleaching assay monitors the protection of AAPH-induced crocin bleaching, by antioxidants(Reference Bors, Michel and Saran200). Crocin is a mix of natural pigments and absorbs, similar to carotenoids, at 450 nm. Therefore, the interpretation of the results can be complicated in experiments with food samples. Initially, the test was used for the analysis of plasma samples(Reference Tubaro, Ghiselli, Rapuzzi, Maiorino and Ursini201). One of its limitations is that crocin is not commercially available, but high sample throughput with microplates is possible.

Single electron transfer methods

In these assays, the sample itself is an oxidant that abstracts an electron from the antioxidant, thereby causing colour changes which are proportional to the AOC. When the change of absorbance is plotted against the antioxidant concentration, the slope of the curve reflects the total reducing capacity. In contrast to the methods described in the last chapter, no oxygen radicals are present in the system; therefore, it can be assumed that the reducing capacity is equal to the antioxidant capacity.

The TEAC-assay is a spectrophotometric test which was developed by Miller et al. (Reference Miller, Rice-Evans, Davies, Gopinathan and Milner202). 2,2-Azinobis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) is oxidized by ROO to a green-blue radical cation. The ability of antioxidants to delay colour formation is expressed relative to Trolox. Originally, the test used metmyoglobin and H2O2, and the ABTS radical was measured at 734 nm. Meanwhile, various modifications have been developed(Reference Re, Pellegrini, Proteggente, Pannala, Yang and Rice-Evans203, Reference Van Den Berg, Haenen, Van Den Berg and Bast204). After generation of the ABTS radical, the sample to be tested is added, subsequently other chemicals like manganese dioxide, ABAP, potassium persulfate or enzymes are used to generate the ABTS radical(Reference Prior, Wu and Schaich175, Reference Miller, Rice-Evans, Davies, Gopinathan and Milner202, Reference Re, Pellegrini, Proteggente, Pannala, Yang and Rice-Evans203, Reference Miller, Sampson, Candeias, Bramley and Rice-Evans205). Temperatures higher than 37°C, which are not physiological and different absorption maxima (415, 645 734 or 815 nm) are frequently used. Depending on the protocol, the decrease or increase in ABTS radical absorbance in presence of the test sample or Trolox at a fixed time point is measured and the antioxidant capacity is calculated as Trolox equivalents. ABTS is not a physiological substance. It reacts fast with aqueous and organic solvents and substances with a low redox potential show a good response. Therefore, phenolic compounds or ascorbic acid react quite well with ABTS whereas lipophilic compounds respond more weakly. The test can be adapted to microplates and is not restricted to a narrow pH range, but high haemoglobin concentration in the plasma may interfere with the measurements.

The FRAP assay determines the reduction of 2,4,6-tripyridyl-s-triazine (TPTZ) in plasma to a coloured product(Reference Benzie and Strain206) and has also been adapted for food samples(Reference Benzie and Szeto207, Reference Ou, Huang, Hampsch-Woodill, Flanagan and Deemer208). Similar to the TEAC assay, compounds with a redox potential < 0·7 V are detected. FRAP not enable to monitor compounds that quench radicals like proteins or thiol compounds such as glutathione (similar to all the “reducing assays”) and can therefore underestimate the AOC. In order to maintain the solubility of Fe, the assay is conducted at acidic conditions (pH of 3·6). Most redox reactions take place within a few minutes, therefore both tests consider most of the substance effects (like polyphenols) but not all, since some substances have longer reaction times(Reference Prior, Wu and Schaich175). This was recently shown for polyphenols like caffeic-, tannic-, ferulic- or ascorbic acids, where the absorption steadily increased for hours(Reference Pulido, Bravo and Saura-Calixto209). Since the test reflects only the reducing potential and does not consider H transfer, it should only be considered in combination with other methods to give a more complete picture. Similar to the TEAC, it is a relatively easy test procedure, can be done manually or fully automated and requires no expensive equipment.

The copper reducing assay (CUPRAC, AOP-90) is a modification of the FRAP in which iron is replaced by Cu; Cu2+ is reduced to Cu1+(Reference Apak, Guclu, Ozyurek and Karademir210). The assay is conducted at 490 nm with bathocuprine, or at 450 nm with neocuproine. Results are expressed as uric acid equivalents. Since copper has a lower redox potential than iron, but enhances the redox cycling potential, its pro-oxidative potential can be more sensitive(Reference Prior, Wu and Schaich175). The limitations regarding the underestimation of slower reactive molecules is similar to the other assays but one of it advantages is that almost every antioxidant including thiols can be detected(Reference Prior, Wu and Schaich175).

The DPPH (2,2-diphenyl-1-picrylhydrazyl) radical used in the DPPH-assay is stable, deep purple, commercially available and has not to be generated. The test was developed by Brand-Williams et al. (Reference Brand-Williams, Cuvelier and Berset211). The loss of DPPH colour at 515 nm after reaction with test compounds is measured either by decrease of absorbance or by electron spin resonance(Reference Bondet, Brand-Williams and Berset212). The concentration of a 50 % decrease of the DPPH radical is defined as EC50, the time to reach it as TEC50. Since the DPPH assay uses a wavelength of 515 nm, it can interfere with substances with a similar spectrum like carotenoids(Reference Nomura, Kikuchi, Kubodera and Kawakami213). The test considers both, electron transfer as well as hydrogen (H) transfer reactions with the focus on the prior. Again, smaller molecules have better access to the radical site and contribute to a higher extent to the AOC(Reference Prior, Wu and Schaich175). The test procedure itself is quite simple and fast and requires only a spectrophotometric device.

For all the electron transfer tests it can be assumed that they “overestimate” smaller molecules and hydrophilic substances like ascorbic acid, uric acid or polyphenols and reflects not always the situation in the organism(Reference Huang, Ou and Prior214).

From the evaluation of the different assays it becomes clear that no single test reflects the overall antioxidative capacity of antioxidants. Both hydrophilic and lipophilic activities must be considered, as well as H transfer and single electron transfer mechanisms and additional tests which reflects the inactivation of various reactive oxygen/nitrogen species are needed to fully estimate the AOC (Table 1).

Table 1 Comparison of methods used to determine the total antioxidant capacity (TAC)

CL, chemiluminescence assay; CUPRAC, cupric reducing antioxidant capacity; DPPH, 2,2-diphenyl-2-picrylhydrazyl assay; ELISA, enzyme-linked immunosorbent assay; FRAP, ferric reducing ability of plasma; LDL, low density lipoprotein; ORAC, oxygen radical absorbance capacity test; PCL, photo-chemiluminescence assay; TEAC, trolox equivalent antioxidative capacity; TOSC, total oxidant scavenging capacity; TRAP, total radical trapping antioxidant parameter.

* +,++,+++, positive; − , −  − , −  −  − , negative; UC, ultracentrifugation.

Number of antioxidant studies conducted with the different methods identified by use of a computer aided search (Scopus database).

Oxidation of macromolecules

Biomarkers of lipid oxidation

Cell membranes are highly susceptible to LP due to their specific composition which is charactesized by high amounts of polyunsaturated fatty acids (PUFAs)(Reference Spiteller215). LP oxidation is a chain reaction (see above) and leads to structural and functional damage of membranes as well as to the formation of lipid hydroperoxides which are unstable and degrade to various secondary oxidation products.

The formation of malondialdehyde (MDA) is the most widely used parameter of PUFA peroxidation in the thiobarbituric acid-reacting substances (TBARS) assay. One of the oldest and still most widely used methods is based on the precipitation of protein nearly at boiling conditions(Reference Yagi216). The samples are heated for 1 h with TBA at low pH and the pink chromogen formed absorbs at 532 nm. The sample preparation has been criticized since it is far from physiological conditions and not the free MDA in the original sample is measured but the amount generated by decomposition of lipid peroxides during heating(Reference Gutteridge217). Furthermore, it is known that also other compounds like sugars, amino acids or bilirubin are able to react with TBA(Reference Meagher and Fitzgerald218). The sensitivity of the test can be increased by combining it with HPLC to separate such compounds before acidic heating(Reference Wong, Knight, Hopfer, Zaharia, Leach and Sunderman219).

In the last few years, several innovations have been introduced to improve the specificity of the test and to reduce known bias. In particular the temperature at the deprotenization step has been reduced to physiological conditions. In addition, several methods have been developed which do not require derivatization(Reference Karatas, Karatepe and Baysar220, Reference Wilson, Metz, Graver and Rao221) or new derivatization agents like 2,4-dinitrophenylhydrazine(Reference Sim, Salonikas, Naidoo and Wilcken222) or diaminonaphthalene(Reference Steghens, Van Kappel, Denis and Collombel223). Very recently, GC/MS based methods have been developed which possess high sensitivity showing an overestimation of MDA levels(Reference Cighetti, Allevi, Anastasia, Bortone and Paroni224, Reference Stalikas and Konidari225). Although it is questionable whether MDA measurements are a reliable method for LP, it is well documented by a large number of studies that increased levels are found in patients with ROS related diseases(Reference Del Rio, Stewart and Pellegrini226).

The first step of PUFA oxidation is the conjugation of double bonds leading to formation of conjugated dienes (CD) which absorb at around 234 nm. They can either be absorbed by lipids but also in plasma samples. The plasma preparation is more physiological and requires no heat treatment. In plasma samples, CD are usually analysed with HPLC–UV detection(Reference Ramel, Wagner and Elmadfa227). Determination of the diene levels cannot be used alone to describe oxidative stress, but when measured with HPLC–CD, the findings can support results obtained with other more reliable parameters of oxidative stress. Although MDA and CD are both primary oxidation products, they can develop differently in the same sample due to different mechanisms of formation(Reference Konig, Wagner, Elmadfa and Berg228).

Isoprostanes are stable oxidation products from arachidonic acid, initially formed from phospholipids and released into circulation before the hydrolyzed form is excreted in urine(Reference Morrow, Hill, Burk, Nammour, Badr and Roberts229). A large number of endproducts can theoretically be generated but interest has focused mainly on F-isoprostanes(Reference Milne, Musiek and Morrow230); the must promising marker for oxidative stress/injury being 8-prostaglandin F(Reference Roberts and Morrow231). At present it is regarded as one of the most reliable markers of oxidative stress, although the presence of detectable concentrations of isoprostanes in biological fluids requires continuous lipid peroxidation(Reference Young232). Several favourable characteristics make isoprostanes attractive as reliable markers for oxidative stress, i.e. they are specific oxidation products, stable, present in detectable quantities, increased strongly at in vivo oxidative stress, and their formation is modulated by antioxidants(Reference Roberts and Morrow231). Various approaches such as gas chromatography–mass spectrometry (GC–MS), GC–tandem MS, liquid chromatography–tandem MS and immunoassays are available for the detection of F-isoprostanes(Reference Schwedhelm and Boger233). The first results were produced by use of the MS technique, and various isoprostanes can be separated with this method(Reference Schwedhelm and Boger233). Recently, immunoassays have been developed which correlate apparently quite well with the results obtained with GC–MS measurements in urine but some discrepancies might occur with plasma samples when they were not tested on cross reactivity with other prostaglandin metabolites(Reference Young232). Nevertheless, their use might be appropriate in intervention studies with various blood samplings from the same subject, thereby focusing not on absolute levels but on relative changes.

The measurement of breath hydrocarbons is a non-invasive method which allows to determine LP through exhaled breath by measuring trace volatile hydrocarbons(Reference Frank, Hintze, Bimboes and Remmer234). Ethane formation results from n-3 oxidation, pentane formation is caused by n-6 oxidation. Although the data reported on their consistency to describe LP are quite convincing, the limiting factor is their detection. They are mainly employed with GC–FID, but one concern is the background level in the breath since bacteria were shown to produce significant amounts of theses hydrocarbons in vivo (Reference Romero, Bosch-Morell, Romero, Jareno, Romero, Marin and Roma235). Furthermore, the separation of different hydrocarbons is not easy due to similar boiling points(Reference Risby and Sehnert236).

Aldehydes represent stable products of PUFA oxidation. 4-Hydroxynonenal (4-HNE) and hexanal are mainly formed by n-6 fatty acid oxidation, while propanol and 4-hydroxyhexenal result from n-3 fatty acid oxidation(Reference Frankel, German and Davis199, Reference Esterbauer, Schaur and Zollner237, Reference Benedetti, Comporti and Esterbauer238). High concentrations of 4-HNE have been shown to trigger well-known toxic pathways such as induction of caspases, the laddering of genomic DNA, and release of cytochrome c from mitochondria, which may lead to cell death(Reference Cahuana, Tejedo, Jimenez, Ramirez, Sobrino and Bedoya239). The most frequently used methods for determination of the aldehydes are GC–MS, GC–head space or HPLC(Reference Holley, Walker, Cheeseman and Slater240). Also polyclonal or monoclonal antibodies directed against 4-HNE-protein conjugates are now frequently used for 4-HNE measurements(Reference Sharma, Brown, Awasthi, Yang, Sharma, Patrick, Saini, Singh, Zimniak, Singh and Awasthi241).

Biomarkers of protein oxidation

Markers of protein oxidation are less frequently used than lipid oxidation parameters (Table 2). They are mainly applied in combination with LP markers, although their formation has been associated with several diseases(Reference Agarwal242).

Table 2 Main biomarkers for lipid and protein oxidation

AOPP, advanced oxidation protein products; CD, conjugated dienes; GC/MS, gas chromatography–mass spectrometry; ELISA, enzyme-linked immunosorbent assay; 4-HNE, 4-hydroxynonenal; HPLC, high-performance liquid chromatography; MDA, malondialdehyde; TBARS, thiobarbituric acid reactive substances. Distribution of the total studies to in vitro (iv), animal (an) and human (hu) studies: iv%, percentage in vitro; an%, percentage animal studies; %hu, percentage human studies.

* The number of antioxidant studies conducted with the different methods was assessed by use of a computer aided search (Scopus database).

Formation of protein bound carbonyls is most abundant endpoint used to monitor protein oxidation(Reference Winterbourn and Buss243, Reference Levine, Wehr, Williams, Stadtman and Shacter244) by a conventional colorimetric assay using 2,4-dinitrophenylhdrazine(Reference Levine, Garland, Oliver, Amici, Climent, Lenz, Ahn, Shaltiel and Stadtman245). The test is easy to perform, but large quantities of solvents are required. Recently, an ELISA method has been developed and the results correlated well with the spectrophotometric method(Reference Alamdari, Kostidou, Paletas, Sarigianni, Konstas, Karapiperidou and Koliakos246).

Advanced oxidation protein products (AOPP) are predominantly albumin and its aggregates damaged by oxidative stress(Reference Witko-Sarsat, Friedlander, Capeillere-Blandin, Nguyen-Khoa, Nguyen, Zingraff, Jungers and Descamps-Latscha247). They contain abundantly dityrosines which cause crosslinking, disulfide bridges and carbonyl groups and are formed mainly by chlorinated oxidants such as hypochloric acid and chloramines resulting from myeloperoxidase activity(Reference Capeillere-Blandin, Gausson, Descamps-Latscha and Witko-Sarsat248). AOPP have several similar characteristics as advanced glycation endproducts-modified proteins. Induction of proinflammatory activities, adhesive molecules and cytokines is even more intensive than that caused by advanced glycation end products (AGEs). They are referred to as markers of oxidative stress as well as markers of neutrophil activation(Reference Witko-Sarsat, Friedlander, Khoa, Capeillére-Blandin, Nguyen, Canteloup, Dayer, Jungers, Drüeke and Descamps-Latscha249). Protein oxidation products mediated by chlorinated species (HOCl) generated by the enzyme myeloperoxidase were found in the extracellular matrix of human atherosclerotic plaques and increased levels of advanced oxidation protein products were postulated to be an independent risk factor for coronary artery disease(Reference Kaneda, Taguchi, Ogasawara, Aizawa and Ohno250, Reference Kalousova, Zima, Tesar, Dusilova-Sulkova and Skrha251). AOPPs are expressed as chloramine-T equivalents by measuring absorbance in acidic conditions at 340 nm in presence of potassium iodide. The test is easy to perform and can be carried out with microprobes.

Markers of oxidative DNA damage used in studies with dietary antioxidants

During the last fifty years, a broad variety of genotoxicity test procedures have been developed which are used for routine testing of chemicals, in environmental research and also in human studies concerning the impact of occupational exposure, lifestyle factors and nutrition on DNA-integrity. Due to the conservative structure of the genetic material, mutagenicity experiments can be carried out with a broad variety of indicator organisms including bacteria, yeasts, plants, invertebrates including Drosophila, laboratory rodents and also with cultured mammalian cells(Reference Kilbey, Legator, Nichols and Ramel252Reference Fahrig254). The advantages and limitations of the different methods for the detection of DNA-protective dietary factors have been described by Knasmüller and coworkers(Reference Knasmüller, Steinkellner, Majer, Nobis, Scharf and Kassie255Reference Hoelzl, Bichler, Ferk, Simic, Nersesyan, Elbling, Ehrlich, Chakraborty and Knasmuller257). One of the main problems (which is encountered also relevant for studies of the effects of dietary antioxidants) encountered in experiments with mammalian cells and lower organisms concerns the inadequate representation of the metabolism of the test compounds which may lead to results which cannot be extrapolated to humans. Nevertheless, all these methods have been used to study oxidative DNA-damage and to investigate putative protective effects of phytochemicals. At present, the most widely used endpoints are gene mutation assays with bacteria and mammalian cells (Salmonella typhimurium/microsome test, HPRT gene mutation assay), chromosome analyses in metaphase cells which can be conducted with stable cell lines (e.g. CHO, V79) and lymphocytes in vitro but can be also scored in in vivo and ex vivo experiments with blood cells. Another important endpoint are micronuclei which are formed as a consequence of chromosome breakage (clastogenicity) and aneuploidy and are less time consuming to evaluate as chromosomal aberrations(Reference Knasmüller, Majer, Buchmann, Remacle and Reusens256, Reference Fenech258). The most frequently used approaches to monitor antioxidant effects of dietary factors are described in the subsequent chapters.

The most widely used bacterial mutagenicity test procedure is the Salmonella/microsome assay, which has been developed by B. Ames in the 1970s(Reference Ames, Sutton and Harris259). The test is based on the detection of back mutations in specific genes which encode for histidine biosynthesis. One of the disadvantages of the initial set of tester strains was, that none of them was highly sensitive towards oxidative effects, therefore new polyplasmid strains (TA102 and TA104) were constructed which are in particular suitable for the detection of mutagenic effects caused by ROS(Reference Levin, Hollstein, Christman, Schwiers and Ames260). Since the target gene is located in these strains (in contrast to the classical tester strains) on plasmids, it can be easily lost and due to the high spontaneous reversion rates many groups encountered difficulties with these derivatives. Nevertheless, the Salmonella strains are at present widely used in antioxidant experiments; mutations are induced either by radiation or chemically and putative protective compounds or complex mixtures are plated on histidine free selective media plates together with the indicator bacteria. After incubation, the differences in the numbers of his+ revertants serves as an indication of protective effects. The test procedure has been standardised for routine testing of chemicals (see for example OECD guideline 471(261)) and the criteria which have been defined (sufficient number of plates, inclusion of positive and negative controls, testing of different doses) can also be applied for the detection of antioxidants. One of the problems which affects the reliability of the test results, concerns the fact that false positive effects may be obtained with compounds which cause bactericidal or bacteriostatic effects since these parameters are not monitored under standard conditions. A typical example are the protective effects seen with cinnamaldehyde(Reference Rutten and Gocke262). With complex dietary mixtures, difficulties may be encountered due to their histidine contents(Reference Wurgler, Friederich, Furer and Ganss263).

Apart from the Salmonella/microsome assay, also a number of other bacterial genotoxicity tests have been developed which are less frequently used, e.g. assays, based on the scoring of backward or forward mutations with E. coli strains, tests based on the induction of repair processes, such as umu and SOS chromotest or differential DNA-repair assays based on the comparison of the survival of strains differing in their DNA-repair capacity(Reference Knasmüller, Majer, Buchmann, Remacle and Reusens256).

Single cell gel electrophoresis (SCGE) assays are based on the determination of DNA migration in an electric field which leads to formation of COMET shaped images. In the initial version, the experiments were carried out under neutral conditions which allowed only the detection of double strand breaks(Reference Ostling and Johanson264). Subsequently, Tice et al. (Reference Tice, Agurell, Anderson, Burlinson, Hartmann, Kobayashi, Miyamae, Rojas, Ryu and Sasaki265) and Singh et al. (Reference Singh, McCoy, Tice and Schneider266) developed protocols for experiments in which the cells are lysed under alkaline conditions (pH>13), which enables the additional detection of single strand breaks and apurinic sites. One of the main advantages of this test procedure is, that it does not require cell divisions (which are a prerequisite for gene mutation and micronucleus experiments), therefore only short incubation periods are required so that not only stable cell lines can be used for in vitro studies but additionally also primary cells from different organs. Furthermore, it is also possible to carry out in vivo experiments with rodents and to study effects in a broad variety of tissues(Reference Sasaki, Sekihashi, Izumiyama, Nishidate, Saga, Ishida and Tsuda267). In most human studies, peripheral lymphocytes have been used as target cells, few experiments were conducted with exfoliated epithelial cells(Reference Dhillon, Thomas and Fenech268Reference Szeto, Benzie, Collins, Choi, Cheng, Yow and Tse271) which are problematic due to their low viability. Very recently, also results from a human intervention trail with antioxidants were reported in which bioptic material from the colon was analysed(Reference DeMarini272).

Collins et al. developed in the 1990s(Reference Collins, Duthie and Dobson273) a protocol in which isolated nuclei are treated with lesion specific enzymes (endonculease III, formadidopyrimidine glycosylase, FPG). This approach has been used intensely to study the prevention of endogenous formation of oxidised purines and pyrimidines. However, it is crucial in these experiments to determine the optimal amounts of the enzymes due to their instability. More recently, Collins and coworkers published an additional modified version of the SCGE test which enables to monitor the impact of compounds on the repair capacity of the cells(Reference Collins and Horvathova274, Reference Collins275) and it was shown in a few model studies that antioxidants may alter DNA-repair processes(Reference Chakraborty, Roy and Bhattacharya276, Reference Collins, Horska, Hotten, Riddoch and Collins277). In order to obtain information concerning alterations of the sensitivity towards exogenous ROS mediated damage, it is possible to treat the cells with H2O2 other radical generating chemicals or radiation (ROS-challenge).

In the guidelines published by Tice et al. (Reference Tice, Agurell, Anderson, Burlinson, Hartmann, Kobayashi, Miyamae, Rojas, Ryu and Sasaki265) and Hartmann et al. (Reference Hartmann, Agurell, Beevers, Brendler-Schwaab, Burlinson, Clay, Collins, Smith, Speit, Thybaud and Tice278), a number of criteria are defined which are essential to obtain reliable results. They concern for example the number of parallel cultures and cells which are required in in vitro studies, the number of animals and treatment periods and also adequate statistical methods. It was generally agreed, that different parameters such as tail lengths of the comets, as well as percentage DNA in tail and tail moment are acceptable and that apart from automated image analysis systems also manual scoring methods are acceptable(Reference Burlinson, Tice, Speit, Agurell, Brendler-Schwaab, Collins, Escobar, Honma, Kumaravel, Nakajima, Sasaki, Thybaud, Uno, Vasquez and Hartmann279). An important point which is also relevant for antioxidant studies concerns the fact that multiple doses should be tested to substantiate effects and that it is necessary to monitor acute toxic effects. It is well documented that cell death leads to degradation of DNA, therefore it is essential to determine the viability of the cells after treatment with appropriate methods in in vitro experiments and to monitor toxic effects in inner organs by histopathology(Reference Burlinson, Tice, Speit, Agurell, Brendler-Schwaab, Collins, Escobar, Honma, Kumaravel, Nakajima, Sasaki, Thybaud, Uno, Vasquez and Hartmann279) and exclude conditions under which strong acute effects are observed in animal experiments. These criteria are fulfilled in most recent studies, but not in a number of older investigations.

The SGCE technique has been used in numerous in vitro investigations to study antioxidant effects of individual compounds and complex mixtures in stable cell lines and in human lymphocytes. Typical examples are investigations of flavonoids by Anderson and coworkers with blood and sperm cells(Reference Anderson, Yu, Phillips and Schmezer280, Reference Anderson, Basaran, Dobrzynska, Basaran and Yu281), experiments concerning antioxidant effects of coffee and its constituents(Reference Bichler, Cavin, Simic, Chakraborty, Ferk, Hoelzl, Schulte-Hermann, Kundi, Haidinger, Angelis and Knasmuller282), studies on the protective effects of tea catechins(Reference Elbling, Weiss, Teufelhofer, Uhl, Knasmueller, Schulte-Hermann, Berger and Micksche283, Reference Kundu, Bhattacharya, Siddiqi and Roy284) to name only a few. Also quite a substantial number of in vivo studies with laboratory animals have been published, e.g. with carotenoids(Reference Vanitha, Murthy, Kumar, Sakthivelu, Veigas, Saibaba and Ravishankar285), quercetin(Reference Kapiszewska, Cierniak, Papiez, Pietrzycka, Stepniewski and Lomnicki286), vitamins E and C(Reference Gabbianelli, Nasuti, Falcioni and Cantalamessa287) and garlic oil(Reference Liu and Xu288).

The results of human studies are described in the reviews of Moller & Loft(Reference Moller and Loft289Reference Moller and Loft291). Most of them were conducted as intervention trails which have the advantage that inter-individual variations can be reduced. In total, 76 investigations have been published since the first trial was conducted by Pool-Zobel et al. in 1997(Reference Pool-Zobel, Bub, Muller, Wollowski and Rechkemmer292). Evidence for antioxidant effects was observed in approximately 64 % of the studies (25/39); the highest number of protective effects concerned the prevention of endogenous formation of oxidised pyrimidines, (39 % of the trials, 7/19), while in few studies evidence for reduced sensitivity towards ROS and protection against formation of oxidised purines was found. The lowest rate of protective effects was seen when the SCGE analyses were carried out under standard conditions (i.e. without ROS challenge and enzymes). Clear evidence for protective effects was observed for example in experiments with lycopene and tomatoes(Reference Riso, Visioli, Erba, Testolin and Porrini293), cruciferous and leguminous sprouts(Reference Gill, Haldar, Porter, Matthews, Sullivan, Coulter, McGlynn and Rowland294) and carotenoid supplementation(Reference Zhao, Aldini, Johnson, Rasmussen, Kraemer, Woolf, Musaeus, Krinsky, Russell and Yeum295). In our own experiments we found strong protective properties of sumach (a common spice) and its main active principle gallic acid(Reference Ferk, Knasmüller, Dusinska, Brantner, Uhl, Chakraborty, Bronzetti, Ferguson and De Flora296), wheat sprouts(Reference Ehrlich, Hoelzl, Nersesyan, Winter, Koller, Ferk, Fenech, Dusinska, Knasmüller, Ďuračková, Slámenova and Micksche101), after consumption of coffee(Reference Bichler, Cavin, Simic, Chakraborty, Ferk, Hoelzl, Schulte-Hermann, Kundi, Haidinger, Angelis and Knasmuller282) and in experiments with Brussels sprouts(Reference Hoelzl, Glatt, Meinl, Sontag, Heidinger, Kundi, Simic, Chakraborty, Bichler, Ferk, Angelis, Nersesyan and Knasmüller297). The latter observation is of particular interest; since no effects were seen in a large intervention trial in which the participants consumed 600 g of fruits and vegetables/d(Reference Moller, Vogel, Pedersen, Dragsted, Sandstrom and Loft298) our findings may be taken as an indication that coffee intake may contribute to a higher extent to ROS protection than plant derived foods.

Moller & Loft (2004) discuss in their papers also the possible shortcomings of intervention trails and emphasize the importance of controlled study designs which should include a sufficient number of participants and additionally also either placebo groups or washout periods(Reference Moller and Loft290). These latter criteria are not fulfilled in some of the older studies.

Micronucleus (MN) experiments have nowadays largely replaced conventional chromosomal aberration (CA) analyses (which have been used extensively in the past to study protective effects of antioxidants towards radiation and chemically induced DNA-damage) as they are less time consuming and laborious. Both endpoints can be used in in vitro experiments with cell lines and lymphocytes; one of the most promising newer developments is the use of human derived liver cell lines (HepG2, HepG3 etc). HepG2 cells have retained the activities of drug metabolising enzymes including those which metabolise plant specific antioxidants and thus may reflect the effects in humans more adequately than cell lines which are commonly used in genetic toxicology (for reviews see(Reference Knasmüller, Mersch-Sundermann, Kevekordes, Darroudi, Huber, Hoelzl, Bichler and Majer299Reference Mersch-Sundermann, Knasmüller, Wu, Darroudi and Kassie301)).

The most widely procedure used in animal experiments is the bone marrow MN assay. Since it is problematic to induce MN with chemical oxidants in vivo, most experiments were conducted with radiation. Standardised guidelines have been published for this test system, e.g. by OECD(302), and one of the most important parameters relevant for AO studies is the treatment period. It is also notable, that one of the main shortcomings of this test is due to the fact that reactive molecules may not reach the target cells; this explains why negative results were obtained with a number of potent genotoxic carcinogens, also antioxidant effects may not be detected due to this limitation. Typical examples for AO studies in which this assay was used are experiments with epigallocatechin gallate, vitamin C, lipoic acid, ubidecarenone or coenzyme Q(10)(Reference Hosseinimehr, Azadbakht, Mousavi, Mahmoudzadeh and Akhlaghpoor303Reference Prahalathan, Selvakumar, Varalakshmi, Kumarasamy and Saravanan308).

The development of the cytokinesis block micronucleus (CBMN) method by Fenech et al. (Reference Fenech and Morley309, Reference Fenech310) allows to monitor MN formation in peripheral blood cells in vitro and can be also used in dietary human studies with lymphocytes. It is based on the use of the mitogen phytohaemagglutinin (which stimulates nuclear division), in combination with cytochalasin B (which stops cellular division), so that MN can be scored in binucleated cells. Typical examples for the use of this approach are supplementation studies with vitamin C and intervention trials with red wine(Reference Fenech, Stockley and Aitken311Reference Greenrod, Stockley, Burcham, Abbey and Fenech313), in vitro experiments with wine specific phenolic compounds(Reference Greenrod and Fenech314) as well as tests with the antioxidant drugs amifostine and melatonin(Reference Kopjar, Miocic, Ramic, Milic and Viculin315).

More than 100 different oxidative modifications of DNA have been described in the literature(Reference Dizdaroglu316) and it is notable that oxidative adducts occur at a frequency of 1 or more orders of magnitude higher than non-oxidative adducts(Reference Ames, Shigenaga and Hagen317). The dominant oxidative modification of DNA is 8-hydroxylation of guanine, which leads to formation of 8-oxo-7,8-dihydro-2-deoxyguanosine (8-oxodG). In most studies, this oxidation product was measured in urine and in leukocytes, but it is also possible to monitor it in various inner organs, e.g. in liver, lung and kidneys(Reference Chater, Abdelmelek, Douki, Garrel, Favier, Sakly and Ben Rhouma318Reference Kim, Suzuki, Laxmi, Umemoto, Matsuda and Shibutani321). The first investigations were conducted in the late 1980s(Reference Shigenaga, Gimeno and Ames322) and a number of different methods have been developed namely gas chromatography coupled with mass spectrometry(Reference Dizdaroglu323), liquid chromatography prepurification followed by gas chromatography coupled with mass spectrometry(Reference Gackowski, Rozalski, Roszkowski, Jawien, Foksinski and Olinski324), liquid chromatography coupled with tandem mass spectrometry(Reference Cooke, Olinski and Evans325), high-performance liquid chromatography with electrochemical detection(Reference Kasai326Reference Shigenaga, Aboujaoude, Chen and Ames328), high-performance liquid chromatography with mass spectrometry(Reference Harman, Liang, Tsitouras, Gucciardo, Heward, Reaven, Ping, Ahmed and Cutler329) and enzyme-linked immunosorbent assay based methods(Reference Chiou, Chang, Chan, Wu, Tsao and Wu330).

It is well known that guanine is easily prone to oxidation if stringent precautions are not taken during preparation of samples for analysis(Reference Hofer and Moller331), therefore much of what is measured could be artefacts. In order to prepare compounds for analysis by gas chromatography coupled with mass spectrometry which enables the simultaneous analysis of a number of different adducts(Reference Wiseman, Kaur and Halliwell332), DNA is chemically hydrolysed and converted into a volatile form by derivatization with a suitable reagent. This reaction is normally carried out at high temperature. During the derivatization reaction, oxidation occurs to an extent that overwhelms the amount originally present(Reference Douki, Delatour, Bianchini and Cadet333Reference Ravanat, Turesky, Gremaud, Trudel and Stadler335). Generally speaking, gas chromatography coupled with mass spectrometry estimates of DNA oxidation have been higher than high-performance liquid chromatography with electrochemical detection estimates by about a factor of 10(Reference Douki, Delatour, Bianchini and Cadet333). The cause of this difference (an overestimate due to artificial oxidation with gas chromatography coupled with mass spectrometry vs an underestimate due to inefficient enzymatic digestion with high-performance liquid chromatography with electrochemical detection) has now been settled(Reference Ravanat, Turesky, Gremaud, Trudel and Stadler335). In addition, GC/MS analyses are difficult to perform, labour-intensive and exhibit poor sensitivity or inadequate specificity when they are used to test urinary samples(Reference Hu, Wang, Chang and Chao336). After enzymatic hydrolysis of DNA to nucleosides it is possible to analyse the probes with chromatography by high-performance liquid chromatography(Reference Shigenaga, Aboujaoude, Chen and Ames328). 8-Oxo-dG and its corresponding base 8-oxo-Gua are electrochemically active, lending themselves to sensitive electrochemical detection. The relative simplicity and sensitivity of high-performance liquid chromatography with electrochemical detection of 8-oxo-dG have made it the most popular method for monitoring of DNA-oxidation(Reference Beckman and Ames337). The high-performance liquid chromatography with electrochemical detection method itself has been criticized for its high variability(Reference Adachi, Zeisig and Moller338, Reference Finnegan, Herbert, Evans, Griffiths and Lunec339). Estimates of the ratio of 8-oxodG to dG have ranged from approximately 0·25 × 10− 5 to ⩾10− 4, possibly due to artificial oxidation(Reference Beckman and Ames337, Reference Adachi, Zeisig and Moller338). Dreher & Junod(Reference Dreher and Junod340) described in detail how the formation of artefacts that occurs with high-performance liquid chromatography with electrochemical detection can be avoided. Also this method is prone to oxidation of samples during preparation for analysis but by use of antioxidants and anaerobic conditions during isolation and hydrolysis of DNA, these effects can be minimised.

Immunoassays have, by far, demonstrated the greatest versatility in terms of the matrices to which they can be applied (urine, serum, plasma, cell culture medium, DNA hydrolysates), and simplicity of use(Reference Chiou, Chang, Chan, Wu, Tsao and Wu330), but comparisons with urinary 8-oxodG levels by chromatographic techniques revealed strong discrepancies of the results(Reference Cooke, Evans, Herbert and Lunec341Reference Yoshida, Ogawa and Kasai343). The precise reason for the higher enzyme-linked immunosorbent assay based methods values remains unknown although there is a correlation between the data obtained by this and HPLC based methods(Reference Cooke, Evans, Herbert and Lunec341Reference Yoshida, Ogawa and Kasai343). Nevertheless, this method can be applied to studies comparing relative urinary 8-oxo-dG values among several groups if the determination of the exact concentratins of 8-oxo-dG in urine is not required(Reference Shimoi, Kasai, Yokota, Toyokuni and Kinae342, Reference Yoshida, Ogawa and Kasai343).

The European Standards Committee on Oxidative DNA Damage(261, 344Reference Gedik and Collins347) studied systematically the reasons for the discrepancies of the results obtained with different methods. Comparative analyses of baseline 8-oxo-dG levels in mammalian cell DNA, by different methods in different laboratories showed that estimates of 8-oxo-dG in pig liver, using chromatographic techniques, ranged between 2.23 and 441 per 106 guanines, and in HeLa cells DNA between 1.84 and 214. In the case of chromatographic methods, it was argued that spurious oxidation during work-up has been a problem and the trustworthiest results are the lowest. Most laboratories employing HPLC–ECD were able to measure chemically induced damage with similar efficiency and dose response gradients for seven of the eight sets of results were almost identical. GC–MS and HPLC–MS/MS, employed in three laboratories, did not convincingly detect dose response effects. In another study, Gedik & Collins(Reference Gedik and Collins347) evaluated data obtained in various laboratories in studies on 8-oxodG in calf thymus DNA, pig liver, oligonucleotides, HeLa cells and lymphocytes. The authors conclude that HPLC–ECD is capable of measuring 8-oxo-dG induced experimentally in the different types of samples. On the contrary, GC–MS failed to detect a dose response of physically (photosensitizer Ro 19-8022 and visible light) induced 8-oxodG formation and was not regarded as a reliable method for measuring low levels of damage; also HPLC–MS/MS was not found capable of detecting low levels of oxidative DNA damage.

8-Oxo-dG studies were used in a number of animal studies with laboratory rodents for example with cyanidin-3-glycoside(Reference Duthie, Jenkinson, Crozier, Mullen, Pirie, Kyle, Yap, Christen and Duthie348), curcumin(Reference Tunstall, Sharma, Perkins, Sale, Singh, Farmer, Steward and Gescher349), grape juice(Reference Jung, Wallig and Singletary350), green tea extract, catechins(Reference Hong, Ryu, Kim, Lee, Lee, Kim, Yun, Ryu, Lee and Kim351, Reference Kaneko, Tahara and Takabayashi352) and coffee(Reference van Zeeland, de Groot, Hall and Donato353) to name only a few.

The formation of oxidised guanine was also monitored extensively in human studies. It was shown that certain diseases such as cancer, Parkinson's and Alzheimer's diseases, multiple sclerosis, HIV, cystic fibrosis of lung, muscular dystrophy, diabetes mellitus, rheumatoid arthritis(Reference Cooke, Olinski and Evans325) as well as occupational exposure to coal dust(Reference Schins, Schilderman and Borm354), polyaromatic hydrocarbons and aromatic amines(Reference Loft, Poulsen, Vistisen and Knudsen355, Reference Marczynski, Rihs, Rossbach, Holzer, Angerer, Scherenberg, Hoffmann, Bruning and Wilhelm356), asbestos(Reference Yoshida, Ogawa, Shioji, Yu, Shibata, Mori, Kubota, Kishida and Hisanaga357), arsenic(Reference Hu, Wang, Chang and Chao336) and smoking(Reference Hu, Wang, Chang and Chao336, Reference Loft, Astrup, Buemann and Poulsen358) lead to increased urinary excretion.

The results of dietary intervention trials are summarised in the reviews of Moller & Loft(Reference Moller and Loft289, Reference Moller and Loft290). At present results of 23 studies are available, and in 12, protective effects were found. Evidence for reduced formation was seen for example in intervention trials with individual vitamins (C and E) and with supplements containing different vitamins, red ginseng, green tea and red wine and in newer investigations with specific vegetables(Reference Thompson, Heimendinger, Diker, O'Neill, Haegele, Meinecke, Wolfe, Sedlacek, Zhu and Jiang359, Reference Thompson, Heimendinger, Gillette, Sedlacek, Haegele, O'Neill and Wolfe360). Machowetz et al. (Reference Machowetz, Poulsen, Gruendel, Weimann, Fito, Marrugat, de la Torre, Salonen, Nyyssonen, Mursu, Nascetti, Gaddi, Kiesewetter, Baumler, Selmi, Kaikkonen, Zunft, Covas and Koebnick361) published recently an interesting study in which they found a significant (13 %) reduction of the formation of oxidised bases after consumption of olive oil which could be not explained by the phenolic contents of the different oils. Examples for studies in which no effects were detected are intervention trials with β-carotene, low vitamin E levels in combination with polyunsaturated fatty acids and cranberry juice(Reference Duthie, Jenkinson, Crozier, Mullen, Pirie, Kyle, Yap, Christen and Duthie348, Reference Jenkinson, Collins, Duthie, Wahle and Duthie362).

Correlations between different markers of oxidative stress

The questions if and to which extent relationships exist between the sensitivity of commonly used biomarkers of oxidative stress and endpoints of oxidative DNA damage, is addressed in the review of Dotan et al. (Reference Dotan, Lichtenberg and Pinchuk363). The authors analysed the results of studies in which two or more methods were used under identical experimental conditions and conclude that good correlations exist between measurements of peroxidation products such as MDA, lipid hydroperoxides, F-2 isoprostanes, conjugated dienes, glutathione and protein carbonyls but not with other criteria of the oxidative status such as the concentrations of antioxidants (TAC) and DNA migration (SCGE assay).

The evaluation of recent studies with dietary antioxidants shows that in general good correlations are observed between different biomarkers in in vitro experiments while this is not the case for animal and human studies. Results obtained with curcurmin, lycopene and ellagic acid in X-radiation experiments in vitro showed that the antioxidants increased the levels of GSH and reduced lipid peroxidation (MDA formation), in parallel a decrease of MN formation was observed(Reference Srinivasan, Sudheer, Pillai, Kumar, Sudhakaran and Menon364Reference Sudheer, Muthukumaran, Devipriya and Menon366). Also in a study with colon derived cell lines (HT29, CaCo2), and phenolic apple juice extracts, improvement of the intracellular redox status (measured with dichloroflurecein assay) was paralleled by decreased DNA-damage in SCGE experiments(Reference Schaefer, Baum, Eisenbrand and Janzowski367).

In a recent Indian study(Reference Gandhi and Nair368) with mice, a clear correlation was observed between protection against radiation induced DNA-migration in lymphocytes and TBARS formation by gallic acid, while no such relations could be seen between these parameters in experiments with vitamin E deficient rats. Although the TBARS levels were significantly increased, formation of 8-oxodG and comet formation were not affected(Reference Duthie, Gardner, Morrice, Wood, Pirie, Bestwick, Milne and Duthie369).

Also in human studies controversial results were obtained in multiple endpoint studies. Consumption of anthocyanin/polyphenolic rich fruit juices reduced in an intervention trial (n = 18) DNA-migration in lymphocytes and increased the levels of reduced GSH, while other parameters (MDA in plasma, excretion of isoprostanes in urine) were not affected(Reference Weisel, Baum, Eisenbrand, Dietrich, Will, Stockis, Kulling, Rufer, Johannes and Janzowski370). Also in an earlier study (n = 27) with two juices containing either cyanidin glycoside or EGCG, a significant decrease of ENDO III lesions was observed in COMET assays, while other markers of the redox status (FRAP, TBARS, and FOX2) were not altered(Reference Bub, Watzl, Blockhaus, Briviba, Liegibel, Muller, Pool-Zobel and Rechkemmer371). In another larger study (n = 36), significant decreases of plasma antioxidant capacity and reduced ratio of GSH:GGSG were found while the concentration of reduced GSH and 8-oxo-dG in lymphocytes were not altered(Reference Janse van Rensburg, Erasmus, Loots, Oosthuizen, Jerling, Kruger, Louw, Brits and van der Westhuizen372).

Also changes of markers of oxidative DNA-damage did not correlate in all studies. In workers exposed to coke ovens and of graphite electrode producing plants significant increases of both 8-oxo-dG concentrations and FPG lesions were observed in white blood cells(Reference Marczynski, Rihs, Rossbach, Holzer, Angerer, Scherenberg, Hoffmann, Bruning and Wilhelm356). Also Gedik et al. (Reference Gedik, Boyle, Wood, Vaughan and Collins373) found a good overall correlation between 8-oxo-dG excretion in urine and FPG lesions in lymphocytes in unexposed individuals (n = 8), while no associations were detected at the individual level. On the contrary, no correlations between these two endpoints were found in a much larger study (n = 99)(Reference Hofer, Karlsson and Moller374). Only few investigations have been conducted in which MN and comet formation were studied in parallel. In in vitro experiments with radiated lymphocytes both parameters were reduced by several dietary antioxidants(Reference Srinivasan, Sudheer, Pillai, Kumar, Sudhakaran and Menon364Reference Sudheer, Muthukumaran, Devipriya and Menon366), but in a recent trial with wheat sprouts (n = 13), a significant decrease of FPG lesions was detected in lymphocytes whereas no differences in the comets were detected under standard conditions with Endo III and the frequencies of MN were also not altered in the same target cells(Reference Ehrlich, Hoelzl, Nersesyan, Winter, Koller, Ferk, Fenech, Dusinska, Knasmüller, Ďuračková, Slámenova and Micksche101). It is notable that the differences between formation of 8-oxo-dG and comet formation may be due to the fact that the latter parameter detects a broader spectrum of lesions. The discrepancies between results of MN assays and comet measurements may be due to the fact that DNA oxidation is efficiently repaired and does not necessarily lead to double strand breaks which cause formation of chromosomal aberrations and MN.

The discrepancies seen in comparative experiments raise the question which of the parameters is the most reliable. In terms of prevention of diseases the most reliable endpoints are probably those which are directly related to specific pathologic conditions. For example, oxidized LDL is known to be related to coronary heart disease(Reference Domanski375) while for chromosomal aberrations and micronuclei it is well known that they are predictive for increased cancer risks(Reference Bonassi, Znaor, Ceppi, Lando, Chang, Holland, Kirsch-Volders, Zeiger, Ban, Barale, Bigatti, Bolognesi, Cebulska-Wasilewska, Fabianova, Fucic, Hagmar, Joksic, Martelli, Migliore, Mirkova, Scarfi, Zijno, Norppa and Fenech376, Reference Norppa, Bonassi, Hansteen, Hagmar, Stromberg, Rossner, Boffetta, Lindholm, Gundy, Lazutka, Cebulska-Wasilewska, Fabianova, Sram, Knudsen, Barale and Fucic377). The association between comet formation in lymphocytes and human diseases is not fully understood at present but it is known that occupationally exposed individuals as well as individuals with diseases which are associated with elevated cancer risks show increased comet formation(Reference Hoelzl, Bichler, Ferk, Simic, Nersesyan, Elbling, Ehrlich, Chakraborty and Knasmuller257, Reference Knudsen and Hansen378, Reference Maluf and Erdtmann379).

Measurement of antioxidant enzymes and of proteins involved in cell signalling and transcription

In many AO studies, the activities of enzymes were measured, which inactivate ROS and are part of the endogenous defence system. Table 3 lists up examples of such studies, a detailed description of currently used methods for the measurement of SOD, GPx and CAT can be found in the paper of Vives-Bauza et al. (Reference Vives-Bauza, Starkov and Garcia-Arumi380).

Table 3 Overview on currently used methods for the detection of antioxidant enzymes and examples of their induction by food constituents

* SM, spectrophotometric measurement; t.o., target organ; GSH, glutathione; GSSG, oxidised form of glutathione; NADPH, nicotinamide adenine dinucleotide phosphate; n.i., not indicated; RT-PCR, real time polymerase chain reaction; WB, Western blot.

 ↑ , increase; ↓ , decrease; ↔ , unchanged; bw, body weight; h, hours; AP-1, activating protein-1; CAT, catalase; DAS, diallyl sulphide; DATS, diallyl trisulfide; EGCG, epigallocatechin-3-gallate; GPx, glutathione peroxidase; HO-1, heme oxigenase-1 protein; LPS, lipopolysaccharide; iNOS, inducible nitric oxide synthase; NAD(P)H, nicotinamide adenine dinucleotide phosphate; QR, quinone reductase; SOD, superoxide dismutase; TGF-α, transforming growth factor-α.

In many investigations it was found, that dietary factors cause an induction of SOD. For example, significant effects were seen in human trials with coffee(Reference Bichler, Cavin, Simic, Chakraborty, Ferk, Hoelzl, Schulte-Hermann, Kundi, Haidinger, Angelis and Knasmuller282), Brussels sprouts(Reference Hoelzl, Glatt, Meinl, Sontag, Heidinger, Kundi, Simic, Chakraborty, Bichler, Ferk, Angelis, Nersesyan and Knasmüller297), gallic acid(Reference Ferk, Chakraborty, Simic, Kundi, Knasmüller, Gee and Fogliano381), plant extract supplements(Reference Nelson, Bose, Grunwald, Myhill and McCord382) and phenolics rich diets(Reference Kim, Kim and Sung383). However, in some studies e.g. in an animal study by Akturk and coworkers(Reference Akturk, Demirin, Sutcu, Yilmaz, Koylu and Altuntas384), a decrease of its activity was found with antioxidants. Also in the case of heme oxygenase-1 (HO-1) induction effects were observed in most investigations (see Table 3), while glutathione peroxidase (GPx) was not altered in the majority of human and animal studies(Reference Bichler, Cavin, Simic, Chakraborty, Ferk, Hoelzl, Schulte-Hermann, Kundi, Haidinger, Angelis and Knasmuller282, Reference Ferk, Chakraborty, Simic, Kundi, Knasmüller, Gee and Fogliano381, Reference Akturk, Demirin, Sutcu, Yilmaz, Koylu and Altuntas384Reference Pedraza-Chaverri, Granados-Silvestre, Medina-Campos, Maldonado, Olivares-Corichi and Ibarra-Rubio386), one exception being experiments with Ginkgo biloba extracts administered to ethanol fed rats, in which an increase was detected(Reference Yao, Li, Song, Zhou, Sun, Zhang, Nussler and Liu387). It can be seen in Table 3 that the results of catalase (CAT) measurements with different dietary the antioxidants are inconsistent. Antioxidant diets caused an induction of CAT in a human study(Reference Nelson, Bose, Grunwald, Myhill and McCord382) while garlic feeding to rats caused a decrease of its activity(Reference Pedraza-Chaverri, Granados-Silvestre, Medina-Campos, Maldonado, Olivares-Corichi and Ibarra-Rubio386).

NADPH-quinone reductase (NADPH-QR) was used as a marker enzyme for the general induction of phase I and phase II enzymes. Prochaska & Talalay(Reference Prochaska, Santamaria and Talalay388) developed a protocol for high throughput measurements of food derived enzyme inducers with a murine hepatoma cell line (Hepa 1c1c7), subsequently, an improved protocol was developed, which enables the discrimination between monofunctional and bifunctional inducers(Reference Prochaska and Talalay389) i.e. between compounds which cause an induction of both, phase I and phase II enzymes and those which only affect the latter group. In a large number of studies, induction of NADPH-QR was observed with a variety of different dietary compounds in this experimental system; one of the most potent inducers identified was sulforaphane, which was used as a model compound in numerous subsequent experiments(Reference Talalay and Fahey390). However, comparative measurements in human derived cells (Hep G2) showed, that the measurement of NADPH-QR does not correlate quantitatively with the induction of GST, a key enzyme involved in the detoxification of many carcinogens(Reference Hintze, Wald, Zeng, Jeffery and Finley391).

Gamma glutamylcysteine synthetase (GCS) can be regarded as an “indirect” AO enzyme. It catalyses the rate limiting step of glutathione (GSH) synthesis(Reference Wild and Mulcahy392) and its activity can be measured with the protocol of Nardi and coworkers(Reference Nardi, Cipollaro and Loguercio393). This mehtod is based on the formation of c-glutamylcysteine from cysteine and glutamic acid which is quantified by derivatization with bromobimane and fluorimetric detection after separation by high performance liquid chromatography. It was shown that coffee diterpenoids and also other antioxidants such as the phenolic food additive butylated hydroxyanisole and vitamin E induce GCS(Reference Huber, Scharf, Rossmanith, Prustomersky, Grasl-Kraupp, Peter, Turesky and Schulte-Hermann394Reference Eaton and Hamel396). GSH itself is a potent antioxidant and scavenges predominantly , OH, RO and ROO∙(Reference Younes, Marquart and Schäfer397). In a number of dietary studies the ratio between oxidised and reduced GSH was monitored as a parameter of antioxidant effects. The method is based on spectrophotometric measurements with Ellmans reagent(Reference Boyne and Ellman398) and changes were observed in newer in vivo studies for example with kawheol/cafestol(Reference Huber, Scharf, Rossmanith, Prustomersky, Grasl-Kraupp, Peter, Turesky and Schulte-Hermann394), polyphenol rich cereals(Reference Alvarez, Alvarado, Mathieu, Jimenez and De la Fuente399) antioxidant (vitamins C and E, l-carnitine, and lipoic acid or additional bioflavonoids, polyphenols, and carotenoids) enriched diets(Reference Rebrin, Zicker, Wedekind, Paetau-Robinson, Packer and Sohal400) and vitamin E(Reference Lii, Ko, Chiang, Sung and Chen401). GSH is also the substrate of glutathione-S-transferases (GSTs), which are regulated by Nrf2 and catalyse the detoxification of a broad variety of xenobiotics including food specific carcinogens (for reviews see(Reference Landi402, Reference Tew and Ronai403)) and it has been postulated that GSTs are also involved in the inactivation of radicals(Reference Younes, Marquart and Schäfer397, Reference Landi402).

Increased levels of the antioxidant enzymes listed in Table 3 are also seen under oxidative stress conditions and in inflammatory and ROS-related diseases and in tumour tissues(Reference Soini, Kaarteenaho-Wiik, Paakko and Kinnula404, Reference Wenzel, Herzog, Kuntz and Daniel405). This holds also true for GCS which is increased for example by NO and in meta- and dysplastic cells(Reference Soini, Kaarteenaho-Wiik, Paakko and Kinnula404). Therefore the induction of these enzymes cannot be taken as a firm evidence for protective properties of dietary constituents and results of such measurements should be supported by additional data.

A plethora of methods has been developed to monitor the impact of antioxidants on proteins involved in cell signalling and on the activation of transcription factors. A detailed description of the currently used techniques is beyond the scope of the present article. In general, biochemical standard methods, such as Western blots, Northern blots, real time PCR and ELISA-based techniques are employed. For the measurement of kinases, e.g. mitogen activated protein kinases (MAPKs) and others, like protein kinase C (PKC) as well as phosphoinositide 3 kinase (PI3K), which are also important targets of phytochemicals(Reference Surh, Kundu, Na and Lee121, Reference Surh406) mainly Western blots with phosphorylation specific antibodies(Reference Favata, Horiuchi, Manos, Daulerio, Stradley, Feeser, Van Dyk, Pitts, Earl, Hobbs, Copeland, Magolda, Scherle and Trzaskos407, Reference Fukazawa and Uehara408) and more recently also more expensive ELISA techniques are used (for commercially available test kits and description of methods see also www.biocompare.com or www.cellsignal.com). Transcription factors like the nuclear factor kappa B (NFκB) can be determined by the luciferin/luciferase system, which is a sensitive reporter assay for gene expression. d-Luciferin is the substrate for firefly luciferase catalyzing the oxidation of luciferin to oxyluciferin in presence of ATP and magnesium, resulting in bioluminescence(Reference Bronstein, Fortin, Stanley, Stewart and Kricka409). By use of this assay, expression of NFκB can be measured in vivo simultaneously in different organs(Reference Mann410) or in stable cell cultures(Reference Hintze, Wald, Zeng, Jeffery and Finley391, Reference Lai, Jiang and Li411).

Two other enzymes, which are often monitored in antioxidant studies are cyclooxygenase 2 (COX 2) and ornithine decarboxylase (ODC). They are regarded as important makers of inflammation and tumour development.

COX 2 is an inducible enzyme which can be activated by NFκB and catalyses the cyclooxygenase pathway of prostaglandin synthesis from arachidonic acid. It is abundant in activated macrophages and other cells at sites of inflammation. COX 2 overexpression has been found in various types of human cancers(Reference Sarkar, Adsule, Li and Padhye412). Protein and m-RNA levels of COX 2 are measured with Western blots, SDS-PAGE, and Northern blots. COX 2 activity can be determined in various ways; a) oxygen consumption can be monitored using an oxygen electrode(Reference Kulmacz, Lands, Benedetto, McDonald-Gibson and Nigam413), b) by measurement of COX 2 produced prostaglandines via enzyme immunoassays (EIA)(Reference Pradelles, Grassi and Maclouf414), c) In addition, several methods for the determination of peroxidase activity of COX 2 with luminescent detection have been developed (for a detailed description of methods and commercially available test kits see e.g. www.caymanchem.com/app/template/cur-rents,010.vm/a/z; ELISA kits also can be found on www.calbiochem.com). The results obtained in experiments with dietary antioxidants on COX 2 inhibition are reviewed in the article of Surh and coworkers(Reference Surh, Chun, Cha, Han, Keum, Park and Lee415).

ODC catalyses the decarboxylation of ornithine into putrescine and is involved in cell proliferation. Its activity is rapidly modified in response to various chemical or physical stimuli(Reference Gaboriau, Havouis, Groussard, Moulinoux and Lescoat416). ODC is a marker enzyme for tumour promotion and is upregulated in transformed cells(Reference Pegg417). The reduction of its activity as well as reversal of the malignant state of cells by antioxidants has been shown in many studies(Reference Hunt and Fragonas418, Reference Friedman and Cerutti419). Various methods for ODC activity measurements were developed, including radiometric assays based on trapping the [14C]O2 generated from the decarboxylation of [1− 14C]ornithine(Reference Russell and Snyder420), the retention of 3H-putrescine from tritiated ornithine on a strong cation-exchange paper(Reference Djurhuus421) and the determination of the radiolabelled form of the enzyme-activated suicide inhibitor of ODC, difluoro-methylornithine (DFMO)(Reference Seely, Poso and Pegg422). A radio- immunoassay using purified polyclonal antibodies has also been employed in some experiments(Reference Seely and Pegg423). More recently, chemiluminescence and spectrophotometry based methods have became available(Reference Gaboriau, Havouis, Groussard, Moulinoux and Lescoat416, Reference Wang and Bachrach424).

Use of -omics based approaches for the investigation of oxidant and antioxidant effects

During the last decade, a number of new techniques has been developed and optimised which enable the simultaneous detection of a large number of alterations of biological functions. The use of these approaches in nutritional sciences has led to the formation of a new discipline termed “Nutrigenomics”(Reference Ordovas and Mooser425). The overall aim of this new field of research is to find out how dietary factors alter gene transcription, protein expression and metabolism(Reference Ordovas and Corella426) in regard to health effects. The area covers three interrelated main areas namely transcriptomics (gene expression analyses), proteomics (global protein analysis) and metabolomics (metabolite profiling). Details on the basic concepts can be found in the article of Kussmann et al. (Reference Kussmann, Affolter and Fay427).

In the following chapters we will focus on the description of results which have been obtained in studies concerning the effects of oxidative stress and with dietary antioxidants in transcriptomics, proteomics and metabolomics. The latter approach are based on the chemical characterisation of metabolites by use of nuclear magnetic resonance and/or mass spectrometry(Reference Gavaghan, Holmes, Lenz, Wilson and Nicholson428, Reference Glassbrook and Ryals429). The use of these techniques for dietary studies is described in recent reviews, e.g.(Reference Scalbert, Milenkovic, Llorach, Manach, Leroux and Ortigues-Marty430, Reference Ovesná, Slabý, Toussaint, Kodíček, Maršík, Pouchová and Vaněk431). Of particular interest for the detection of oxidant/antioxidant effects are analyses of the lipidome (i.e. of the lipids in biological liquids).

Only few dietary studies have been carried out in which the effects of plant foods, beverages and phytochemicals were investigated. Urinary patterns of three types of diets – vegetarian, low meat and high meat consumption in humans were compared, and a specific vegetarian metabolic signatures (i.e. a metabolite of microbial metabolism) were found(Reference Stella, Beckwith-Hall, Cloarec, Holmes, Lindon, Powell, van der Ouderaa, Bingham, Cross and Nicholson432). In other dietary studies with humans, e.g. with camomile tea(Reference Wang, Tang, Nicholson, Hylands, Sampson and Holmes433) and also with black and green teas(Reference Van Dorsten, Daykin, Mulder and Van Duynhoven434) changes were registered; e.g. in the latter trial a change of the excretion of dihydroxyphenylsulphates (end products of flavonoid degradation by colonic bacteria) was seen.

Other studies which are notable are the one of Solanky et al. (Reference Solanky, Bailey, Beckwith-Hall, Davis, Bingham, Holmes, Nicholson and Cassidy435) who investigated the impact of isoflavones on plasma profiles, and the investigations of Ito et al. (Reference Ito, Gonthiera, Manach, Morand, Mennen, Remesy and Scalbert436, Reference Ito, Gonthier, Manach, Morand, Mennen, Remesy and Scalbert437) who studied metabolites of dietary polyphenolics. Fardet et al. studied the excretion profiles after consumption of whole-grain and refined wheat flours(Reference Fardet, Canlet, Gottardi, Lyan, Llorach, Remesy, Mazur, Paris and Scalbert438). Overall, none of these investigations is clearly indicative for antioxidant properties of the food factors studies and the changes detected concern alterations of the composition of the gut microflora and of energy metabolism, or the identification of compound specific metabolites.

Transcriptomics

Basic principles

Subtractive and differential hybridization of messenger RNAs (mRNAs) have been the most frequently employed techniques to determine disparities in the steady state expression levels of transcripts in cells at a given time, the latter in general referred to as the transcriptome. Starting in the early eighties, cDNA (complementary DNA to mRNA) libraries have been used to identify clones representative of genes whose changes in transcription and/or mRNA stability result from variations in the proliferation state(Reference Mohn, Melby, Tewari, Laz and Taub439), the stage of development and disease(Reference DeRisi, Penland, Brown, Bittner, Meltzer, Ray, Chen, Su and Trent440, Reference Rothstein, Johnson, DeLoia, Skowronski, Solter and Knowles441), or the consequences of an agent(Reference Friedman and Weissman442). Although these techniques allow to reveal the whole transcriptome and to select for both, known and unknown differentially expressed mRNAs characteristic to open systems, these methods showed limitations due to the preferential identification of highly to moderately abundant mRNAs(Reference Liang and Pardee443). Subsequently, several more efficient procedures emerged, among them the reverse transcription-polymerase chain reaction (RT-PCR)-based representational difference analysis (RDA)(Reference Lisitsyn and Wigler444), the differential display (DD) technique and the serial analysis of gene expression (SAGE)(Reference Liang and Pardee445, Reference Velculescu, Zhang, Vogelstein and Kinzler446), which both represent open screening techniques even allowing to detect and quantify differentially expressed mRNA species of low abundance. Yet, these methods are subject to the criticism that they require considerable sequencing efforts and may yield false positive signals which demand additional experimental arrangements to get rid of them. In the mid to late nineties, DNA microarrays have been developed for the high throughput expression profiling of the transcriptome which facilitates to dissect the genetic flow of information upon various (patho)physiological stages and to identify suitable drug targets(Reference Brown and Botstein447, Reference Duggan, Bittner, Chen, Meltzer and Trent448).

DNA microarrays are currently widely used and provide the unique opportunity to detect hybridization signals to sequenced clones and collections of partially sequenced cDNAs known as expressed sequence tags (ESTs)(Reference Adams, Kelley, Gocayne, Dubnick, Polymeropoulos, Xiao, Merril, Wu, Olde and Moreno449, Reference Greely450). A variety of different cDNA microarrays with high density hybridization targets immobilized on nitrocellulose or nylon surfaces are available and frequently applied in basic research and clinical settings(Reference Mikulits, Pradet-Balade, Habermann, Beug, Garcia-Sanz and Mullner451). Advances in the microarray technology are associated with the development of different techniques to synthesize DNA probes and to deliver them by immobilization on solid surfaces(Reference Schena, Heller, Theriault, Konrad, Lachenmeier and Davis452). Beside mechanical microspotting and ink jetting of non-synthetic cDNA probes with various lengths on coated glass slides which were prepared by biochemical methods such as PCR, the photolithography technologies employs in situ synthesis of oligonucleotides.

The isolation of total RNA or mRNA by standard biochemical methods requires quality control by analyzing the respective integrity. Solid techniques are available to amplify even low amounts of the extracted RNA for subsequent labeling and hybridization on microarrays. Particular efforts have been made to separate RNA in polysome-associated species which are involved in protein synthesis and to profile translated mRNA in order to more closely depict the proteome and to identify both transciptionally and translationally controled mRNAs(Reference Pradet-Balade, Boulme, Beug, Mullner and Garcia-Sanz453Reference Petz, Kozina, Huber, Siwiec, Seipelt, Sommergruber and Mikulits455). In all experimental settings, standard operating procedures are recommended for the RNA extraction and labeling, the hybridization procedure and the subsequent monitoring of hybridization signals to obtain reproducible and comparable microarray data. Cross-analysis of hybridization signals from independent experiments performed in triplicate ensures a high reliability of results. For routine applications, e.g. GeneSpring™ provides a user-friendly solution for the computational analysis of raw data allowing hierarchical clustering of data sets. However, evaluation of whole-genome microarrays requires particular efforts with regard to the management of data. Changes in the transcriptome indicated by microarray assays depend finally on the confirmation of data by quantitative RT-PCR which is the most sensitive method for the detection of rarely expressed transcripts(Reference Bustin456). The incorporation of SYBR Green dye or the disruption of the Taqman probe exhibit fluorescence during RT-PCR, and are frequently using techniques for the quantitative analysis of mRNA expression. Together, cDNA or oligonucleotide microarrays are regarded as powerful tools for the profiling of the transcriptome, and being greatly versatile in gene throughput by customization and applicability in various experimental approaches.

Results of microarray studies concerning the transcription of genes by oxidative stress

Several reviews on the impact of oxidative stress on gene regulation have been published(Reference Allen and Tresini80, Reference Kunsch and Medford457Reference Finkel and Holbrook459), probably the most comprehensive overview is the one by Allen & Tressin(Reference Allen and Tresini80). However, most of the data contained in these surveys are derived from experiments with conventional methods (including RT-PCR). The present paper is confined to newer findings obtained with arrays which are increasingly used in the last years.

The investigations can be grouped into three categories namely in vitro experiments with stable cell lines or primary cells; animal studies and gene expression analyses with humans. Table 4 gives an overview of groups of genes which were measured in a number of studies.

Table 4 Examples for genes which are transcriptionally regulated by oxidative stress

* OS, Organ specificity.

LO, Location.

In vitro studies: An interesting and promising approach has been published by Scherf and coworkers(Reference Scherf, Ross, Waltham, Smith, Lee, Tanabe, Kohn, Reinhold, Myers, Andrews, Scudiero, Eisen, Sausville, Pommier, Botstein, Brown and Weinstein460) who assessed gene expression profiles in 60 human derived cancer cell lines for a drug discovery screen. They used relatively large arrays (8000 genes) and tested approximately 70 000 compounds; subsequently the results were clustered according to different parameters including known mechanisms of action of the different compounds. The database which emerged from this large study is available from NCI (http//dtp.nci.nih.gov) and provides a valuable source of information. It was employed for example in a study by Efferth & Oesch(Reference Efferth and Oesch461) to characterize the molecular mechanisms of the toxicity of two antimalarial drugs; the authors selected from the database 170 genes involved in oxidative defence and metabolism which they used to design a custom made array and tested the acute toxic effects of the compounds in the individual cell lines; on the basis of the patterns they found distinct differences in the mode of action of the two drugs.

All other studies with human derived cells comprised a lower number of cell lines and chemicals. Murray et al. (Reference Murray, Whitfield, Trinklein, Myers, Brown and Botstein462) compared the effects of different types of stress (heat shock, ESR stress, oxidative stress and crowding) in human fibroblasts and HeLa cells and found clear differences in the responses. On the contrary, similar transcription patterns were identified by Chuang et al. (Reference Chuang, Chen, Gadisetti, Chandramouli, Cook, Coffin, Tsai, DeGraff, Yan, Zhao, Russo, Liu and Mitchell463) who analysed the effects of three ROS generating chemicals namely H2O2, 4-HNE and tBOH in human retinal cells. Other papers with human derived cells were published by Yoneda et al. (Reference Yoneda, Chang, Chmiel, Chen and Wu464), who studied the effects of H2O2 and tobacco smoke in bronchial epithelial cells and Morgan and coworkers(Reference Morgan, Ni, Brown, Yoon, Qualls, Crosby, Reynolds, Gaskill, Anderson, Kepler, Brainard, Liv, Easton, Merrill, Creech, Sprenger, Conner, Johnson, Fox, Sartor, Richard, Kuruvilla, Casey and Benavides465) who used the human derived liver cell line HepG2 to investigate the effects of a variety of ROS generating chemicals. Except in the later study, relative large arrays (>1000 genes) were used in these investigations. In some experiments, ionising radiation was used to induce stress responses; for example Amundson et al. (Reference Amundson, Bittner, Chen, Trent, Meltzer and Fornace466) analysed the expression of genes caused by γ-radiation in twelve different human cell lines.

Another topic which was addressed in in vitro gene expression studies concerns the impact of specific cell functions (transcription factors, enzymes involved in signalling pathways) which are regulated by ROS. Investigations which fall into this category are for example analyses of the impact of PI3K in tBOH induced gene expression in IMR3 cells(Reference Li, Lee and Johnson467), experiments on the role of the BRCA1 gene in breast cancer cells(Reference Bae, Fan, Meng, Rih, Kim, Kang, Xu, Goldberg, Jaiswal and Rosen468), as well as comparative studies with cell lines differing in their p53 status, Nrf2 functions and SOD activity(Reference Amundson, Do, Vinikoor, Koch-Paiz, Bittner, Trent, Meltzer and Fornace469Reference Kirby, Menzies, Cookson, Bushby and Shaw471).

Animal experiments: Only a few studies have been carried out in which the effects of ROS were investigated in laboratory rodents have been published so far. Examples for experiments with different oxidants are described in the articles of McMillian et al. (Reference McMillian, Nie, Parker, Leone, Kemmerer, Bryant, Herlich, Yieh, Bittner, Liu, Wan, Johnson and Lord472, Reference McMillian, Nie, Parker, Leone, Bryant, Kemmerer, Herlich, Liu, Yieh, Bittner, Liu, Wan and Johnson473) who analysed gene expression patterns of oxidants in rat livers and attempted to establish compound specific expression signatures. Examples for the use of knockout animals are the investigations of Yoshihara et al. (Reference Yoshihara, Ishigaki, Yamamoto, Liang, Niwa, Takeuchi, Doyu and Sobue474) and Thimmulappa and coworkers(Reference Thimmulappa, Scollick, Traore, Yates, Trush, Liby, Sporn, Yamamoto, Kensler and Biswal475) who studied comparatively gene expression patterns in normal, SOD and Nrf2 deficient animals.

Another topic which has been addressed in a number of in vivo array studies concerns the impact of ageing on gene transcription and oxidant responses. As mentioned above, it is assumed that several age related diseases are due to increased oxidative damage and it was shown by Lee et al. (Reference Lee, Allison, Brand, Weindruch and Prolla476Reference Lee, Klopp, Weindruch and Prolla478) that transcription patterns in muscles, brain and heart of mice are age dependent. Edwards et al. (Reference Edwards, Sarkar, Klopp, Morrow, Weindruch and Prolla479) found subsequently, that paraquat induced expression patterns differ significantly depending on the age of the animals.

Human studies: Only few articles have appeared in the last years. An example for an occupational study is the one published by Wang et al. (Reference Wang, Neuburg, Li, Su, Kim, Chen and Christiani480) who investigated the responses of blood cells in metal fume exposed workers. It is assumed that ROS are at least partly involved in the toxic effects of metals and indeed the authors found that oxidative damage responsive genes were altered in their expression. Microarrays were also used in a number of studies with patients who suffered from ROS related diseases such as rheumatoid arthritis, inflammatory bowel disease, arterial fibrillation and sickle cell anaemia(Reference Kim, Lim, Lee, Shim, Ro, Park, Becker, Cho-Chung and Kim481Reference Jison, Munson, Barb, Suffredini, Talwar, Logun, Raghavachari, Beigel, Shelhamer, Danner and Gladwin483).

The results of all these investigations aimed at identifying genes which are transcriptionally regulated by ROS depend largely on the experimental model used and the most important parameters are the origin of the cells, the time schedule as well as the mechanisms by which oxidative damage is induced; also the design and size of array technique plays an important role.

The results of the in vitro experiments described above show that the patterns of gene alterations caused by oxidants depends primarily on the type of indicator cells used and to a lesser extent on the chemicals(Reference Murray, Whitfield, Trinklein, Myers, Brown and Botstein462, Reference Weigel, Handa and Hjelmeland484). On the basis of the current state of knowledge one may expect that genes which are predominately altered in their transcription are those which are regulated by ROS dependent transcription factors. Indeed, it was found in some studies that Nrf2/ARE, apoptosis and p53 regulated genes are affected(Reference Li, Lee and Johnson467, Reference Bae, Fan, Meng, Rih, Kim, Kang, Xu, Goldberg, Jaiswal and Rosen468, Reference McMillian, Nie, Parker, Leone, Kemmerer, Bryant, Herlich, Yieh, Bittner, Liu, Wan, Johnson and Lord472, Reference McMillian, Nie, Parker, Leone, Bryant, Kemmerer, Herlich, Liu, Yieh, Bittner, Liu, Wan and Johnson473). However, in some investigations only little evidence for the up regulation of such genes was detected(Reference Bae, Fan, Meng, Rih, Kim, Kang, Xu, Goldberg, Jaiswal and Rosen468, Reference Suzuki, Spitz, Gandhi, Lin and Crawford485, Reference Weindruch, Kayo, Lee, Prolla, Rosenberg and Sastre486) and often genes were identified which are involved in cell cycle regulation, cellular communication, biosynthesis and metabolism. Some of them are controlled by ROS dependent signalling pathways and transcription factors but the patterns seen in the different studies are highly divergent and it is not possible to identify specific marker genes(Reference Li, Lee and Johnson467, Reference Bae, Fan, Meng, Rih, Kim, Kang, Xu, Goldberg, Jaiswal and Rosen468, Reference Weigel, Handa and Hjelmeland484, Reference Yoneda, Peck, Chang, Chmiel, Sher, Chen, Yang, Chen and Wu487).

What are the reasons for the strong inconsistencies? As mentioned above, one of the important parameters is the origin of the cells. For example, Murray et al. (Reference Murray, Whitfield, Trinklein, Myers, Brown and Botstein462) compared the effects of ROS generating chemicals in human fibroblasts and HeLa cells under identical experimental conditions and found the former cells by far more responsive. The reasons for these discrepancies may be due to the fact that signalling pathways and transcription factors are partly tissue specific and may be impaired in cancer cells lines which are frequently used in array studies. Nrf2 and p53 are found in a broad variety of different organs(Reference Lee, Li, Johnson, Stein, Kraft, Calkins, Jakel and Johnson488), while NFκB is lacking in a number of tissues(Reference Allen and Tresini80). It is known that p53 is frequently mutated in tumours in many organs(Reference Ivyna Bong, Patricia, Pauline, Edmund and Zubaidah489Reference Stewart, Querec, Ochman, Gruver, Bao, Babb, Wong, Koutroukides, Pinnola, Klein-Szanto, Hamilton and Patriotis491), and it was shown in a comparative study with p53+ and p53 −  cell lines that loss of this function has a substantial impact on oxidative stress induced gene transcription(Reference Amundson, Do, Vinikoor, Koch-Paiz, Bittner, Trent, Meltzer and Fornace469). In addition, it is so that xenobiotic drug metabolising as well as antioxidant enzymes, which are controlled by the ARE element are highly organ specific (see Table 3). It is also well documented that stable cell lines used routinely in toxicological studies lack the activities of phase I, phase II and antioxidant enzymes which are regulated by ARE via Nrf2(Reference Knasmüller, Mersch-Sundermann, Kevekordes, Darroudi, Huber, Hoelzl, Bichler and Majer299), therefore strong efforts have been made to establish cell lines which have retained the activities of these enzymes in an inducible form. Also other factors may account for the inconsistency of the results(Reference Murray, Whitfield, Trinklein, Myers, Brown and Botstein462) found strong differences between the results of array studies conducted in vitro and in rodents, and the authors hypothesized that the fact that substantially more genes were transcriptionally regulated in vivo may be due to the inadequate representation of cell communication in cultures.

Another important parameter which has been often neglected is the time dependency of AO responses(Reference Murray, Whitfield, Trinklein, Myers, Brown and Botstein462, Reference Yoneda, Chang, Chmiel, Chen and Wu464, Reference Yoneda, Peck, Chang, Chmiel, Sher, Chen, Yang, Chen and Wu487). For example, three distinct phases could be discriminated in experiments with human bronchial epithelial cells. The first was characterised by upregulation of apoptosis related genes and MAPKs, the second by activation of proteins involved in the turnover of damaged proteins and only in the third (10 h after the challenge) activation of genes was observed which are involved in the detoxification of ROS(Reference Yoneda, Chang, Chmiel, Chen and Wu464, Reference Yoneda, Peck, Chang, Chmiel, Sher, Chen, Yang, Chen and Wu487).

Despite these inconsistencies, it is possible to define a number of genes which are typical markers of oxidative stress under experimental conditions (Table 3) and it was possible to establish distinct signatures of gene expression for different types of oxidative tress in specific experimental models such as macrophage activation, peroxisomal proliferation and ROS releasing chemicals in rat hepatocytes in vivo (Reference McMillian, Nie, Parker, Leone, Kemmerer, Bryant, Herlich, Yieh, Bittner, Liu, Wan, Johnson and Lord472, Reference McMillian, Nie, Parker, Leone, Bryant, Kemmerer, Herlich, Liu, Yieh, Bittner, Liu, Wan and Johnson473). Also in in vitro studies with a human derived liver cell line a clear difference was observed between oxidative and non-oxidative stress responses(Reference Morgan, Ni, Brown, Yoon, Qualls, Crosby, Reynolds, Gaskill, Anderson, Kepler, Brainard, Liv, Easton, Merrill, Creech, Sprenger, Conner, Johnson, Fox, Sartor, Richard, Kuruvilla, Casey and Benavides465).

It is known for other areas of genomic research that studies interrogating gene transcription in similar contexts often resulted in poor overlapping lists of regulated genes (for a general review see(Reference Jarvinen, Hautaniemi, Edgren, Auvinen, Saarela, Kallioniemi and Monni492)); are a typical example are the strongly divergent results obtained in investigations aimed at identifying retinoic acid responsive genes(Reference van der Spek, Kremer, Murry and Walker493). These inconsistencies have lead to strong efforts to elucidate the methodological reasons for these variations and it became clear that correlations are improved when low abundant genes are excluded(Reference Shippy, Sendera, Lockner, Palaniappan, Kaysser-Kranich, Watts and Alsobrook494). Different platforms were established which attempted to standardise the techniques of array based approaches. The outcome of these efforts have been published in a number of recent articles(Reference Brazma, Hingamp, Quackenbush, Sherlock, Spellman, Stoeckert, Aach, Ansorge, Ball, Causton, Gaasterland, Glenisson, Holstege, Kim, Markowitz, Matese, Parkinson, Robinson, Sarkans, Schulze-Kremer, Stewart, Taylor, Vilo and Vingron495Reference Bammler, Beyer, Bhattacharya, Boorman, Boyles, Bradford, Bumgarner, Bushel, Chaturvedi, Choi, Cunningham, Deng, Dressman, Fannin, Farin, Freedman, Fry, Harper, Humble, Hurban, Kavanagh, Kaufmann, Kerr, Jing, Lapidus, Lasarev, Li, Li, Lobenhofer, Lu, Malek, Milton, Nagalla, O'Malley, Palmer, Pattee, Paules, Perou, Phillips, Qin, Qiu, Quigley, Rodland, Rusyn, Samson, Schwartz, Shi, Shin, Sieber, Slifer, Speer, Spencer, Sproles, Swenberg, Suk, Sullivan, Tian, Tennant, Todd, Tucker, Van Houten, Weis, Xuan and Zarbl498).

Results of microarray studies with dietary antioxidants

In total, results from approximately 60 studies are available at present in which the impact of dietary antioxidants on gene expression patterns was investigated. The number of publication has almost doubled in the last two years and it is also interesting that many of the newer studies were conducted with laboratory rodents; nevertheless in vitro experiments with stable cell lines are still dominating. It is also notable, that research focused predominantly on specific types of compounds such as resveratrol, a phenolic compound from red wine, epigallocatechingalleate (EGCG), the main catechin in green tea, cucurmin contained in the spice tumeric, sulphoraphane the breakdown product of glucobrassicin (a glucosinolate found in cruciferous vegetables) and the phytoestrogen genistein which is contained in soy beans. Several reviews have been published which contain results of microarray studies with phytochemicals. The most comprehensive one by Narayanan(Reference Narayanan31) describes data obtained in twenty studies (3 in vivo, 17 in vitro) with various dietary components, the findings with EGCG are summarised in the paper of Mariappan et al. (Reference Mariappan, Winkler, Parthiban, Doss, Hescheler and Sachinidis499), results with carotenoids can be found in the overview of Elliott(Reference Elliott500).

Table 5 summarises the results of newer studies which have not been evaluated in the aforementioned articles. The first part describes results of in vitro experiments with cells lines, the second contains in vivo experiments with laboratory rodents.

Table 5 Examples for results obtained with dietary antioxidants in microarray experiments

* AS, Arrays size (genes number); SD, Sprague–Dawley

 ↓ , downregulation; ↑ , upregulation; ↔ , no changes; CDKN, cyclin-dependent kinase inhibitor; COX, cyclooxygenase; FGFR2, fibroblast growth factor receptor 2; GAPDH, glyceraldehyde-3-phosphatase dehydrogenase; LOXL, lysyl oxidase-like genes; MAPK2, mitogen-activated kinase 2; MCP-1, monocyte chemoattractant protein 1; MPK6, mitogen-activated protein kinase 6; NAD(P)H, nicotinamide adenine dinucleotide phosphate; NQO1, NAD(P)H dehydrogenase, quinone 1; TNF, tumour necrosis factor; TGFB, transforming growth factor beta; TF, transcription factor; SOD, superoxide dismutase; n.s., not statistically significant.

Apart from the in vitro and animal studies, also a few human intervention trials have been carried out with antioxidants. For example Majewitz et al. (Reference Majewicz, Rimbach, Proteggente, Lodge, Kraemer and Minihane501) conducted a study with normal and apoE smokers (which carry a specific mutation in apolipoprotein E and are at increased risk for coronary heart disease). Before and after vitamin C supplementation (60 mg/P/4 weeks) they analysed monocytes by use of a small array (225 genes). After the intervention, the expression of 22 of the genes was altered in normal smokers, 71 were altered in apoE individuals. The most interesting observation was the down regulation of TNF-β and its receptor, also genes encoding for the neutrophil growth factor receptor and the monocyte chemoattractant protein receptor were affected. These genes are involved in inflammatory responses and it is known that TNF-β is regulated by NFκB. Another human study was published by Hoffmann and coworkers(Reference Hofmann, Liegibel, Winterhalter, Bub, Rechkemmer and Pool-Zobel502) who analysed the effects of consumption of a polyphenolics mix in lymphocytes of healthy individuals by use of a small targeted array (containing 96 transcripts related to drug metabolism). Fifty six genes were found expressed in the target cells and seven of them were altered after the intervention, three of them encoded for cytochrome P450 isozymes (2F1, 3A4 and 3A5), two for them for sulfotransferases (SULT 2A1 and 1C2) the others were associated with microsomal GST, nicotinamide–N-methyltransferase and the ATP binding cassette.

All of the compounds listed in the table are potent antioxidants and it is apparent that many of the transcriptional changes which were detected concern genes which encode for AO defence, inflammatory responses, apoptosis and cell signalling and division. Nevertheless, it is not possible, to define a general signature of AO effects. The lack of a common pattern may be due to the fact that the chemical structures of the antioxidants as well as their mode of action and tissue specificity differs substantially. For example vitamins C and E differ strongly in their organ distribution and it their ability to inactive ROS species(Reference Cotgreave, Moldeus and Orrenius503). Furthermore, also the experimental design of the studies plays an important role. As described above, the expression patterns seen with resveratrol were highly sex specific, in female mice the number of genes which were transcriptionally altered in hepatic tissue was approximately 3 times higher in females as compared to males(Reference Hebbar, Shen, Hu, Kim, Chen, Korytko, Crowell, Levine and Kong504). Also organ differences have an impact on the results of array studies, for example EGCG affected in the liver of mice three times more genes as compared to the colon(Reference Funk, Frye, Oyarzo, Kuscuoglu, Wilson, McCaffrey, Stafford, Chen, Lantz, Jolad, Solyom, Kiela and Timmermann505). Another important parameter affecting the outcome is the time dependency of the expression patterns and also the dose dependency of the responses (see Table 5).

The results of microarray analyses do at present not allow to draw firm conclusions if a compound elicits ROS protective effects. For example, findings obtained with moderate doses of vitamin C in mice(Reference Selman, McLaren, Meyer, Duncan, Redman, Collins, Duthie and Speakman506) indicate that it rather induces inflammation (up regulation of COX 2) and reduces ROS protection (down regulation of SOD). Also the results obtained with diallyldisulfide in HepG2 cells were unspecific and not indicative for antioxidant properties which are well documented for this compound(Reference Belloir, Singh, Daurat, Siess and Le Bon507). However, it may be possible that general antioxidant gene expression patterns can be defined on the basis of comparative standardised experiments with a variety of compounds which are lacking at present. The currently available results show that microarray studies nevertheless provide highly useful information concerning the molecular mechanisms of prevention of ROS related diseases. Typical examples are the results of studies with prostate cancer cell lines and compounds such as resveratrol and sulforaphane which showed that these compounds interact with the transcription of genes involved in cell cycle regulation and apoptosis; these findings provide a possible explanation for protective effects of these dietary components towards prostate cancer cell line LNCaP (for review see(Reference Narayanan31)); also the assumption of cancer protective effects of EGCG and retinoids could be enhardened by results of array studies(Reference Mariappan, Winkler, Parthiban, Doss, Hescheler and Sachinidis499, Reference Elliott500).

Proteomics

Basic principles

Proteomics is a screening technology based on the separation, quantification and identification of proteins from biological samples and allows to directly identify proteins bearing oxidative modifications. Adaptive cell responses in order to cope with the cell damage imposed by oxidative stress may as well be investigated by transcriptomics. As proteins are translated by ribosomes based on the information provided by mRNA molecules, no protein can be synthesised without the appropriate RNA. On the other hand, the mere existence of mRNA does not mean that the corresponding protein gets translated. It only means that the protein might get translated upon demand. This defines one main difference between proteomics and transcriptomics. Transcriptomics shows which genes are expressed, but does not tell which proteins actually get translated. The picture gets even more complicated, when time is considered. Some proteins such as histones are almost exclusively translated during the S-phase of the cell cycle, but not in G0(Reference Schumperli508). A poor correlation of proteomics to transcriptomics data may be the consequence, because these proteins may be abundant in G0, but no corresponding mRNA may be detectable at this time. On the other hand, several regulatory proteins such as p53 may be subject to continuous protein degradation by proteasomes. Here, protein amounts may accumulate to minute and undetectable amounts only, while the mRNA may be easily detectable. This explains why proteomics may give very different results when compared to transcriptomics.

In order to assign the identity of the modified molecules, separation of proteins may be applied before the detection of oxidative modification. 2D-PAGE is still the most important technique to separate proteins according to electric charge and molecular weight(Reference Nordhoff, Egelhofer, Giavalisco, Eickhoff, Horn, Przewieslik, Theiss, Schneider, Lehrach and Gobom509). Protein spots on 2D gels can be identified by Western analysis or mass spectrometric analysis of tryptic digests and quantified by a variety of techniques including silver staining, fluorescence detection or autoradiography(Reference Gerner, Vejda, Gelbmann, Bayer, Gotzmann, Schulte-Hermann and Mikulits510). This approach is used to perform comparative analysis of protein fractions isolated from e.g. treated and untreated cells. Spot patterns can be compared in order to detect alterations in relative amounts or, quite relevant to oxidative stress, protein modifications accompanied by changes of the molecular charge of the protein. Application of immunological detection of carbonyl groups or nitro-tyrosine by Western analysis may subsequently identify the modified proteins. Thus, application of 2D-PAGE may enable the detection of upregulated and/or modified proteins consequent to oxidative stress. The main draw-back of 2D-PAGE is its limited resolution and sometimes poor reproducibility concealing many important effects(Reference Rabilloud511). The number of proteins accessible via 2D-PAGE is usually in the order of several hundreds and hardly exceeds thousand different proteins(Reference Nordhoff, Egelhofer, Giavalisco, Eickhoff, Horn, Przewieslik, Theiss, Schneider, Lehrach and Gobom509).

Another essential technique for proteome analysis is mass spectrometry. Upon tryptic digestion, peptides can be obtained from isolated proteins which can be forwarded to mass analysis. Determination of the molecular weight of the peptides by e.g. MALDI-TOF provides a fingerprint of peptide masses for each protein(Reference Gras, Muller, Gasteiger, Gay, Binz, Bienvenut, Hoogland, Sanchez, Bairoch, Hochstrasser and Appel512). As tryptic peptides may be predicted from databases for all known proteins, a best-fit search can be performed resulting in candidates which show theoretical peptide masses comparable to those identified in the experiment. This technique is called fingerprint analysis and can be applied only to well-purified proteins as potentially obtained from 2D-PAGE. Minimal contamination of peptide preparations may render fingerprint data not accessible for meaningful interpretation and are a main drawback of this method.

Much more robust and powerful is the identification of peptide sequences obtained from peptide fragmentation analysis. Peptides can be fragmented specifically in a mass spectrometer resulting in the breakage of a single peptide bond per molecule(Reference Fountain, Lee and Lubman513). Ordering of the resulting peptide fragments may generate mass differences according to single amino acids in the peptide. As a result, not only total masses of peptides are determined as in case of fingerprint analysis, but complete amino acid sequences are obtained, which allow identification of proteins with much higher confidence. Furthermore, amino acid modifications may be determined by fragmentation analysis. When applied to isolated spots from 2D-gels, proteins can be identified with very high confidence. Furthermore, specific modifications such as oxidation of methionine, cysteine, tryptophan or others can be determined(Reference Spickett, Pitt, Morrice and Kolch514). On the other hand, the determination of amino acid sequences renders separation of proteins before tryptic digest unnecessary. Protein mixtures may be digested and then peptide mixtures separated by e.g. nano-flow chromatography(Reference Wolters, Washburn and Yates515). Peptide sequences may be determined successively and peptides derived from one protein sorted after the experiment. This powerful technique is called shotgun analysis and enables the identification of more proteins as 2D-PAGE/MS(Reference Rabilloud511). As long as peptide modifications are detected, protein modification can also be assigned. The major drawback of this approach is the limited sequence coverage rendering a lot of information missing and the inapplicability of immunological detection methods(Reference Bodnar, Blackburn, Krise and Moseley516).

As a consequence, the experimental design studies has to be carefully prepared for proteome. First of all, it should be clear what to look at. In case of assessment of the extent of damage in response to a defined challenge, a bulk analysis summarizing the extent of e.g. carbonyl formation with chemical or immunological methods may be appropriate. When the identity of target proteins appears desirable, 2D-PAGE can be used to assign the identity to the modified proteins. Here, the poor access of e.g. membrane proteins by most proteome analysis techniques poses(Reference Watarai, Inagaki, Kubota, Fuju, Nagafune, Yamaguchi and Kadoya517) a severe limitation. Mass analysis of the tryptic digest of an isolated 2D-spot may easily identify the corresponding protein because a few peptide fragmentation data may be sufficient for unequivocal protein identification. When the target amino acids of a modified protein are desired, full sequence coverage of the identified peptides is desired, but hard to realise. Peptides have to be ionised before they can be analysed by mass spectrometry. Ionisation is an unpredictable event and may be strongly dependent on chemical properties of the peptide. Consequently, only a subset of the peptides generated by proteolytic digests can usually be detected by mass spectrometry(Reference Zhu, Kim, Yoo, Miller and Lubman518).

Results of proteomic studies concerning oxidative stress

In order to identify proteins related to oxidative stress, several strategies were employed. A direct consequence of ROS-mediated damage may be amino acid modifications such as oxidation or nitration. Such modifications are accessible via peptide fragmentation analysis by mass spectrometry. While this method is very exact providing direct proof of oxidation together with the identification of the target protein and target amino acid, it has several draw-backs. First, it is almost impossible to survey the total amino acid sequence of proteins. Consequently, a negative result is hardly a proof for the absence of modifications. Second, proteins are very sensitive to artifacts introduced by sample handling procedures. For the proteome analysis of cells, they have to be lysed to extract the proteins for further analyses. Upon cell lysis, organelles get ruptured, calcium is set free, enzymes may get activated and many biochemical reactions including oxidations may get out of control. Also separation of proteins by 2D-PAGE exposes proteins to oxygen. This can be directly observed by the detection of oxidized methionine in samples from the same cell lysis, which were processed by 2D-PAGE and shotgun analysis in parallel. The proteins separated by 2D-PAGE generally show more methionine oxidation compared to the same proteins analysed by shotgun proteomics(Reference Manzanares, Rodriguez-Capote, Liu, Haines, Ramos, Zhao, Doherty-Kirby, Lajoie and Possmayer519). Thus, it is essential to most accurately control all experimental conditions and run comparative experiments strictly in parallel. Actually, several amino acid modifications consequent to oxidative stress are detectable by mass spectrometry, including oxidation of cysteine(Reference Paron, D'Elia, D'Ambrosio, Scaloni, D'Aurizio, Prescott, Damante and Tell520, Reference Rabilloud, Heller, Gasnier, Luche, Rey, Aebersold, Benahmed, Louisot and Lunardi521), methionine and tryptophan(Reference Manzanares, Rodriguez-Capote, Liu, Haines, Ramos, Zhao, Doherty-Kirby, Lajoie and Possmayer519), and nitration of tyrosine(Reference Koeck, Fu, Hazen, Crabb, Stuehr and Aulak522). Beside direct amino acid modification, the formation of disulfide bridges by oxidation may form protein adducts(Reference Brennan, Wait, Begum, Bell, Dunn and Eaton523) or protein glutathione adducts(Reference Fratelli, Demol, Puype, Casagrande, Eberini, Salmona, Bonetto, Mengozzi, Duffieux, Miclet, Bachi, Vandekerckhove, Gianazza and Ghezzi524). Amino-acid adducts can also be detected with antibodies specific for e.g. nitrotyrosine and carbonyl adducts(Reference Pocernich, Poon, Boyd-Kimball, Lynn, Nath, Klein and Butterfield525, Reference Stagsted, Bendixen and Andersen526). As mentioned above, the assignment of the target protein identity might be a daunting task and the assignment of the modified amino acid within the sequence is impossible. On the other hand, modified peptides which may be missed by MS-based analysis might be easily detected with antibodies.

In contrast to the identification of proteins directly targeted by oxidative stress, an alternative approach is the investigation of the response of cells via up-regulation and down-regulation of regulatory proteins(Reference Mathers, Fraser, McMahon, Saunders, Hayes and McLellan82). Here, rather indirect effects are observed, which may be consequent to transcriptional alterations of genes helping the cell to manage the stress situation and, on the other hand, proteasomal degradation of damaged proteins. As a consequence, the readout of such an experiment may become very complex, because we can see a lot of protein alterations, but we do not know where they are coming from. To reduce the complexity of the biological system, prokaryotes and yeast were used to investigate the basic responses to oxidative stress(Reference Godon, Lagniel, Lee, Buhler, Kieffer, Perrot, Boucherie, Toledano and Labarre527, Reference Mostertz, Scharf, Hecker and Homuth528). These works indicate that the expected up-regulation of antioxidant proteins and chaperones may be accompanied by substantial alterations in the metabolic state of the cells. Again, several approaches are feasible to investigate cellular responses. First, cells can be treated with sub-lethal concentrations of agents mediating oxidative stress such as H2O2. After a defined period of time, treated and untreated samples are forwarded to proteome analysis and the results compared in a quantitative fashion. Several studies with human monocytes and epithelial cells demonstrated that again, besides the obvious target proteins such as redox regulators and chaperones, a broad variety of other protein families seems to be involved in regulatory processes consequent to oxidative stress including cytoskeletal proteins and glycolytic proteins(Reference Chan, Gharbi, Gaffney, Cramer, Waterfield and Timms529, Reference Seong, Kim, Choi, Oh, Kim, Lee and Um530).

Second, cells which are very sensitive to oxidative stress may be compared to others which were made resistant e.g. by chronic exposure with increasing H2O2 concentrations(Reference Keightley, Shang and Kinter531). Here, differentially expressed proteins may include those which actually mediate resistance. Obviously, proteins differentially expressed due to different differentiation or activation states of the investigated cells may repress the identification of the true players related to oxidative stress control. Rather multiple comparisons of various cell systems or additional functional assays may allow to identify the relevant proteins. For example, it was demonstrated that enzyme aldose reductase identified by comparative analysis indeed mediates protection in Chinese hamster fibroblasts(Reference Keightley, Shang and Kinter531).

Finally, the involvement of oxidative stress in human diseases has been investigated by analysis of clinical material and it was shown that proteomics may help to understand patho-physiological processes relevant to the disease state. The involvement and relevance of protein oxidation in brain tissue in Alzheimer's disease has been clearly demonstrated(Reference Ding, Dimayuga and Keller532, Reference Sultana, Perluigi and Butterfield533). Specific oxidative modifications may also be involved in obesity(Reference Grimsrud, Picklo, Griffin and Bernlohr534), asthma(Reference Ghosh, Janocha, Aronica, Swaidani, Comhair, Xu, Zheng, Kaveti, Kinter, Hazen and Erzurum535), and various forms of inflammatory diseases(Reference Dalle-Donne, Scaloni, Giustarini, Cavarra, Tell, Lungarella, Colombo, Rossi and Milzani536).

Results of proteomic studies with dietary antioxidants

Although numerous position papers emphasized the importance of proteomics techniques in the investigations of health effects of dietary factors(Reference Go, Butrum and Wong537Reference Opii, Joshi, Head, Milgram, Muggenburg, Klein, Pierce, Cotman and Butterfield546), the number of studies which contain data on the effects of food related compounds on protein patterns is strikingly low. One of the reasons may be that proteomics requires, unlike other techniques described in this article, a sound financial background to establish a proteomics facility. Most of the existing labs are specialised in the analysis of specific cells and tissues. Table 6 summarizes examples for results obtained in recent studies.

Table 6 Examples for results obtained with dietary antioxidants in recent proteomics studies

SwissProt Accession numbers: VDAC-1, P21796; VDAC-2, P45880; HSPBP1, Q9NZL4; GrpE, Q9HAV7; DEAD, Q92499; TGFβ, PO1137; Rho GDP, P52565; forkhead TF, Q08050.

* AO, antioxidant; bw, body weight; CE, capillary electrophoresis; 2 DE, two-dimensional electrophoresis; 2DE-SDS-PAGE, two-dimensional electrophoresis-sodium dodecylsulfate polyacrylamide gel electrophoresis; ESI, electro spray ionisation; h, hour; LC–MS/MS, liquid chromatography ion trap tandem mass spectrometry; MALDI-TOF MS, matrix assisted laser desorption ionization time-of-flight mass spectrometry; n.i., not indicated; ox-LDL, oxidised low density lipoproteins; s.c., subcutaneous.

SD, Sprague–Dawley, pts, patients, t.o., target organ.

 ↓ , downregulation; ↑ , upregulation; ↔ , no changes; TF, transcription factor; SOD, superoxide dismutase; GAPDH, glyceraldehyde-3-phosphatase dehydrogenase; n.s., not significant.

It is quite clear that in most in vitro studies more drastic effects were observed than in human trials. This discrepancy may be not only due to the differences in the effects of the compounds tested, but also to the use of high doses in the experiments with isolated cells. Unphysiological test conditions my lead to secondary effects which may be due to cell death and not caused by the test compound itself. Furthermore, in some of the studies high abundant proteins (α-2 HS glycoprotein) with high inter-individual variations were postulated to be biomarkers for dietary effects. Many of the compounds/diets altered the expression of proteins, which play a role in apoptosis, cytoskeleton structure and energy metabolism, while no substantial alterations of enzymes were seen which are involved in ROS defence, except SOD, which was found upregulated in some of the studies.

Metabolomics

Impact of the experimental design on the outcome of antioxidant studies

In the last chapters we described extensively the technical difficulties of AO studies and the limitations of the reliability of different methods. Also the experimental design has a strong impact on the outcome and on the predictive value of investigations concerning antioxidant properties of dietary compounds which are conducted to find out if beneficial effects can be expected in humans.

General problems of the experimental designs of AO studies

Fig. 4 summarises the advantages and disadvantages of different experimental models. At present around 90 % of AO experiments are conducted in vitro with stable cell lines or with subcelluar fractions.

Fig. 4 Advantages and disadvantages of different experimental used to investigate ROS – protective effects of phytochemicals.

The classical strategy used to identify dietary antioxidants is the screening of a large number of potential candidates in high throughput systems and in vitro by use of stable cell lines and subsequently investigate the most promising compounds in more reliable models (e.g. in animals studies and in human intervention trials). This approach is justified on the basis of the assumption that the key mode of action of dietary antioxidants is the direct (non-enzymatic) inactivation of ROS which can be easily detected in this simple systems. However, it became clear over the last years that the mechanisms by which dietary compounds protect against ROS are by far more complex and that indirect modes of action such as induction of enzymes and other defence mechanisms play an important role. For example, the coffee diterpeonids cawheol and kafestol do not protect human lymphocytes under in vitro conditions against ROS mediated DNA damage(Reference Bichler, Cavin, Simic, Chakraborty, Ferk, Hoelzl, Schulte-Hermann, Kundi, Haidinger, Angelis and Knasmuller282) while Huber et al. (Reference Huber, Scharf, Rossmanith, Prustomersky, Grasl-Kraupp, Peter, Turesky and Schulte-Hermann394) found in experiments with rats that they are potent inducers of the enzyme gamma-glutamylcysteine synthetase (GCS) which catalyses the rate limiting step of the synthesis of glutathione which is a potent antioxidant(Reference Seifried, Anderson, Fisher and Milner547). It is conceivable that the increased GSH levels seen in the plasma of coffee drinkers(Reference Grubben, Nagengast, Katan and Peters548) and the protection against oxidative DNA damage observed in a human intervention trial and in animal experiments(Reference Bichler, Cavin, Simic, Chakraborty, Ferk, Hoelzl, Schulte-Hermann, Kundi, Haidinger, Angelis and Knasmuller282, Reference Hoelzl, Bichler, Ferk, Ehrlich, Cavin, Simic, Edelbauer, Grasl-Kraupp, Knasmüller, Ďuračková and Knasmüller549) with coffee are due to this mechanism.

AO enzymes and also transcription factors which regulate cellular defence mechanisms may be not represented adequately in stable cancer cell lines which are used in AO studies. For example, it is known that that p53 controls the transcription of a variety of genes which encode for responses towards antioxidants(Reference Li and Prives550). Since >50 % of human tumours are p53 deficient due to mutations(Reference Levesque and Eastman551), cell lines derived from them will not adequately reflect AO responses. Also the origin of the cells is important, while p53 and NrF2 are found in many organs, NFκB is not or only weakly expressed in certain tissues(Reference Allen and Tresini80); also the activities of AO enzymes show strong organ specificities (see Table 4). As mentioned above, the differences in the transcription pattern of genes in studies with oxidants and antioxidants support the assumption that the cellular responses depend largely on the type of indicator cells used.

Another important issue concerns the representation of phase I and phase II enzymes which metabolise dietary antioxidants and therefore may alter their protective properties; furthermore also reactions catalysed by the intestinal microflora are not reflected under in vitro conditions. Polyphenolics are extensively metabolised in the gut and in the body and non-conjugated metabolites most often account for a minor fraction of the circulating metabolites(Reference Manach, Williamson, Morand, Scalbert and Remesy552). In the case of isoquercitrin it was shown that completely different conjugates are formed under in vitro conditions (i.e. after exposure to colon cell lines) as in the serum of rats(Reference Depeint, Gee, Williamson and Johnson553, Reference Depeint554). Hydroxycinnamic acids which are potent oxidants in coffee are glucuronated in the body(Reference Nardini, Cirillo, Natella and Scaccini555, Reference Clifford556), but stable cell lines and peripheral lymphocytes which are used in in vitro experiments lack glucuronosyltransferase (UGT) activity which catalyses this reaction(Reference Mitoma and Fukuda557, Reference Hu and Wells558). In most cases, it is not known if and to which extent the AO properties of conjugates differ from that of the parent compounds, but for glucuronides of isoflavones and epicatechin it has been shown that they provide no protection against oxidative stress(Reference Zhang, Song, Cunnick, Murphy and Hendrich559, Reference Spencer, Schroeter, Crossthwaithe, Kuhnle, Williams and Rice-Evans560) while the parent compounds are potent antioxidants. The inadequate representation of drug metabolising enzymes is a general problem which is also encountered in acute toxicity and genotoxicity experiments with stable cells lined and various attempts were made to solve it. For example, the cultivation conditions of primary hepatocytes which possess a broad spectrum of xenobiotic drug metabolising enzymes have been improved(Reference Eckl and Raffelsberger561, Reference Fahrig, Rupp, Steinkamp-Zucht and Bader562), other solutions may be the establishment of hepatoma cell lines which have retained the activities of several enzymes in an inducible form, (for review see(Reference Knasmüller, Mersch-Sundermann, Kevekordes, Darroudi, Huber, Hoelzl, Bichler and Majer299)) and the construction of genetically engineered lines which express human phase I and phase II enzymes(Reference Dogra, Doehmer, Glatt, Molders, Siegert, Friedberg, Seidel and Oesch563, Reference Muckel, Frandsen and Glatt564).

The cultivation conditions of the cells may have a strong impact on the outcome of antioxidant studies. It is known that the composition of the medium affects the sensitivity of cells towards ROS. For example when lymphocytes are treated with ROS-generating chemicals in serum, they are up to five times less sensitive as compared to treatment in regular medium (L. Elbling, personal communication). When coffee or coffee specific compounds (chlorogenic and caffeic acid) were tested at high concentrations in genotoxicity tests with bacteria or human lymphocytes in vitro, strong effects were observed which were attributed to formation of H2O2, while with lower doses clear antioxidant effects were detectable and it was postulated by Aeschbacher et al. (Reference Aeschbacher, Wolleb, Loliger, Spadone and Liardon565) that adverse effects do not take place under realistic in vivo conditions.

On the basis of the results of in vitro experiments it was possible to identify a number of highly potent antioxidants such as chlorophylls(Reference Kumar, Shankar and Sainis566), curcumin(Reference Su, Chen, Lin, Wu and Chung567), EGCG(Reference Ogasawara, Matsunaga and Suzuki568) and anthocyanins(Reference Heinonen569) which are currently sold as food supplements and/or used for the production of functional foods(Reference Shahidi, Remacle and Reusens570). However, the results of experiments in which protection of oxidative DNA damage was monitored with foods containing these compounds are not promising, i.e. negative results were for example obtained with green vegetables, EGCG supplemented products(Reference Moller and Loft289, Reference Moller, Vogel, Pedersen, Dragsted, Sandstrom and Loft298, Reference Watzl and Leitzmann571) and anthocyan rich blueberries(Reference Duthie, Jenkinson, Crozier, Mullen, Pirie, Kyle, Yap, Christen and Duthie348). The reasons for these disappointing results are probably due to the fact that the active compounds which have a large molecular configuration are only poorly absorbed, therefore protective effects can be expected in the digestive tract but not in inner organs. In recent investigations, we found in a human intervention trial (SCGE experiments) with lymphocytes that gallic acid (GA), a small phenolic molecule, contained in specific plant foods reduces the formation of oxidised bases in peripheral lymphocytes with 30–40 fold higher efficiency as vitamins E and C while under in vitro conditions similar protective effects were observed(Reference Ferk, Knasmüller, Dusinska, Brantner, Uhl, Chakraborty, Bronzetti, Ferguson and De Flora296, Reference Ferk, Chakraborty, Simic, Kundi, Knasmüller and Schulte-Herman572). One of the reasons for the strong antioxidant properties of GA may be its high absorption rate(Reference Shahrzad, Aoyagi, Winter, Koyama and Bitsch573).

Overall, the predictive value of results obtained in in vivo experiments with rodents are higher as those of in vitro findings but one of the problems encountered in the interpretation of animal derived results is due to the fact that putative antioxidants were often administered at high doses which are irrelevant for humans.

The major problem of human studies concerns the fact that most experiments are carried out with peripheral blood cells and plasma and it remains unclear if and to which extent protection against ROS mediated damage can be expected in inner organs. In the case of GA we observed significant reduction of radical induced damage in a variety of inner organs in animal experiments(Reference Ferk, Chakraborty, Simic, Kundi, Knasmüller, Gee and Fogliano381, Reference Ferk, Chakraborty, Simic, Kundi, Knasmüller and Schulte-Herman572), also for a number of other compounds such as lycopene and vitamins and E it is known that the reduction of damage seen in blood cells in humans is paralleled by protective effects in a variety of tissues(Reference Ellinger, Ellinger and Stehle574, Reference Domenici, Vannucchi, Jordao, Meirelles and Vannucchi575).

An important question which has been neglected in human intervention trials concerns the impact of the overall antioxidant status of the participants on the outcome of protection studies. It can be expected that individuals which consume AO rich diets respond less sensitive. In the case of folic acid it has been shown that supplementation is only effective in regard to improvement of the DNA stability in individuals with low intake levels(Reference Beetstra, Thomas, Salisbury, Turner and Fenech576). On the basis of these findings Fenech(Reference Fenech577) developed the “genome health clinic and genome health nutrigenomics“ concept which postulates individualised supplementation strategies on the basis of biochemical and DNA-stability measurements and can be also applied to antioxidants(Reference Kazimirova, Barancokova, Krajcovicova-Kudlackova, Volkovova, Staruchova, Valachovicova, Paukova, Blazicek, Wsolova and Dusinska578, Reference Fenech579).

Justification of heath claims for antioxidant properties of dietary factors?

Growing consumer concerns about the health attributes of food products and supplements and the increased production of health foods have led to extensive discussions of the scientific evidence required for health claims(Reference Katan and de Roos580). Already in 2001, the global sales of functional foods were estimated USD 47,6 billion worldwide(Reference Sloan581). As a consequence, authorities such as the US Food and Drug Administration and the European Commission have issued new rules directed at inaccuracies, confusion and false information related to functional and dose related risk reduction claims(Reference Derby and Levy582Reference Nickelson584). The EU regulation on nutrition and health claims made on foods (No. 1924/2006) entered into force on July 1st, 2007 and it is clearly stated in article 6 that claims have to be “based on and substantiated by generally accepted scientific evidence”.

The evaluation of the methods which are currently used to investigate the antioxidant properties of foods and food constituents show that not all approaches provide reliable results. Data from in vitro experiments cannot be extrapolated to the human situation in general, while the value of results obtained in animal and human studies depends largely on the experimental design and on the techniques employed. As mentioned above, frequently used methods such as TBARS measurements and the quantification of protein bound carbonyls and breath hydrocarbons do not provide firm evidence for AO effects, also some TAC measurements, which are conducted under unphyiological conditions (e.g. TOSC, CUPRAC), may not be reliable(Reference Aruoma171, Reference Hoelzl, Bichler, Ferk, Simic, Nersesyan, Elbling, Ehrlich, Chakraborty and Knasmuller257). Despite the uncertainities concerning the reliability of TAC measurements, results of such experiments can support the overall evidence for ROS protective effects but claims should be not solely based on such studies. Probably more relevant information can be expected from HPLC based isoprostane measurements as a marker for lipid peroxidation and from results of experiments concerning prevention of oxidative DNA-damage (HPLC based determination of oxidised bases, SCGE experiments with restriction enzymes). In this context it is notable that there is strong evidence that a number of ROS related diseases (including cancer) are causally directly related to damage of the genetic material (see introduction section).

As mentioned above, measurements of the induction of antioxidant enzymes do not provide reliable information for protective effects as they can also be induced by ROS themselves and their up regulation may reflect prooxidant effects.

Although the results obtained with -omics techniques no not allow to establish antioxidant specific patterns of gene transcription and protein alterations they can provide evidence for protective effects of antioxidants in specific experimental settings.

For example, it was found in a few studies that the transcription of genes caused by oxidative stress or by ROS related diseases can be normalised by dietary antioxidants which strongly supports the assumption of protective properties.

A promising strategy for the justification of health claims may be the combination of different methods to a test battery, but the selection of the individual approaches and techniques to be included will be a matter of intense debate.

Acknowledgements

The publication of this paper was made possible by the financial support of the European Co-operation in the field of Scientific and Technical (COST) Research Action 926 “Impact of new technologies on the health benefits and safety of bioactive plant compounds” (2004–2008). The authors had no conflicts of interest to disclose.

References

1Blomhoff, R (2005) Dietary antioxidants and cardiovascular disease. Curr Opin Lipidol 16, 4754.CrossRefGoogle ScholarPubMed
2Elahi, MM & Matata, BM (2006) Free radicals in blood: evolving concepts in the mechanism of ischemic heart disease. Arch Biochem Biophys 450, 7888.CrossRefGoogle ScholarPubMed
3Lefer, DJ & Granger, DN (2000) Oxidative stress and cardiac disease. Am J Med 109, 315323.CrossRefGoogle ScholarPubMed
4Agarwal, A, Sharma, RK, Nallella, KP, Thomas, AJ Jr, Alvarez, JG & Sikka, SC (2006) Reactive oxygen species as an independent marker of male factor infertility. Fertil Steril 86, 878885.Google Scholar
5Pasqualotto, FF, Sharma, RK, Kobayashi, H, Nelson, DR, Thomas, AJ Jr & Agarwal, A (2001) Oxidative stress in normospermic men undergoing infertility evaluation. J Androl 22, 316322.CrossRefGoogle ScholarPubMed
6Von Kobbe, C, May, A, Grandori, C & Bohr, VA (2004) Werner syndrome cells escape hydrogen peroxide-induced cell proliferation arrest. FASEB J 18, 19701972.Google Scholar
7Yan, T, Li, S, Jiang, X & Oberley, LW (1999) Altered levels of primary antioxidant enzymes in progeria skin fibroblasts. Biochem Biophys Res Commun 257, 163167.CrossRefGoogle ScholarPubMed
8Kohen, R & Nyska, A (2002) Oxidation of biological systems: oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol 30, 620650.Google Scholar
9Gracy, RW, Talent, JM, Kong, Y & Conrad, CC (1999) Reactive oxygen species: the unavoidable environmental insult? Mutat Res 428, 1722.CrossRefGoogle ScholarPubMed
10Calabrese, V, Guagliano, E, Sapienza, M, Mancuso, C, Butterfield, DA & Stella, AM (2006) Redox regulation of cellular stress response in neurodegenerative disorders. Ital J Biochem 55, 263282.Google ScholarPubMed
11Zimmermann, JS & Kimmig, B (1998) Pharmacological management of acute radiation morbidity. Strahlenther Onkol 174, Suppl. 3, 6265.Google ScholarPubMed
12Sarma, L & Kesavan, PC (1993) Protective effects of vitamins C and E against gamma-ray-induced chromosomal damage in mouse. Int J Radiat Biol 63, 759764.CrossRefGoogle Scholar
13Gebhart, E (1974) Antimutagens. Data and problems. Humangenetik 24, 132.Google Scholar
14Li, L, Tsao, R, Yang, R, Liu, C, Zhu, H & Young, JC (2006) Polyphenolic profiles and antioxidant activities of heartnut (Juglans ailanthifolia Var. cordiformis) and Persian walnut (Juglans regia L.). J Agric Food Chem 54, 80338040.CrossRefGoogle ScholarPubMed
15Anagnostopoulou, MA, Kefalas, P, Kokkalou, E, Assimopoulou, AN & Papageorgiou, VP (2005) Analysis of antioxidant compounds in sweet orange peel by HPLC–diode array detection–electrospray ionization mass spectrometry. Biomed Chromatogr 19, 138148.Google Scholar
16Minoggio, M, Bramati, L, Simonetti, P, Gardana, C, Iemoli, L, Santangelo, E, Mauri, PL, Spigno, P, Soressi, GP & Pietta, PG (2003) Polyphenol pattern and antioxidant activity of different tomato lines and cultivars. Ann Nutr Metab 47, 6469.Google Scholar
17Erdman, JW Jr, Balentine, D, Arab, L, Beecher, G, Dwyer, JT, Folts, J, Harnly, J, Hollman, P, Keen, CL, Mazza, G, Messina, M, Scalbert, A, Vita, J & Williamson, G & Burrowes, J (2007) Flavonoids and heart health: proceedings of the ILSI North America Flavonoids Workshop, May 31–June 1, 2005, Washington, DC. J Nutr 137, 718S737S.CrossRefGoogle ScholarPubMed
18Kim, JM, Chang, HJ, Kim, WK, Chang, N & Chun, HS (2006) Structure–activity relationship of neuroprotective and reactive oxygen species scavenging activities for allium organosulfur compounds. J Agric Food Chem 54, 65476553.CrossRefGoogle ScholarPubMed
19Iwai, K, Kishimoto, N, Kakino, Y, Mochida, K & Fujita, T (2004) In vitro antioxidative effects and tyrosinase inhibitory activities of seven hydroxycinnamoyl derivatives in green coffee beans. J Agric Food Chem 52, 48934898.CrossRefGoogle ScholarPubMed
20Fujioka, K & Shibamoto, T (2006) Quantitation of volatiles and nonvolatile acids in an extract from coffee beverages: correlation with antioxidant activity. J Agric Food Chem 54, 60546058.CrossRefGoogle Scholar
21Lopez-Velez, M, Martinez-Martinez, F & Del Valle-Ribes, C (2003) The study of phenolic compounds as natural antioxidants in wine. Crit Rev Food Sci Nutr 43, 233244.Google Scholar
22Hidalgo, FJ, Leon, MM & Zamora, R (2006) Antioxidative activity of amino phospholipids and phospholipid/amino acid mixtures in edible oils as determined by the Rancimat method. J Agric Food Chem 54, 54615467.CrossRefGoogle ScholarPubMed
23Luximon-Ramma, A, Neergheen, VS, Bahorun, T, Crozier, A, Zbarsky, V, Datla, KP, Dexter, DT & Aruoma, OI (2006) Assessment of the polyphenolic composition of the organic extracts of Mauritian black teas: a potential contributor to their antioxidant functions. Biofactors 27, 7991.Google Scholar
24Antonious, GF, Kochhar, TS, Jarret, RL & Snyder, JC (2006) Antioxidants in hot pepper: variation among accessions. J Environ Sci Health B 41, 12371243.CrossRefGoogle ScholarPubMed
25Masuda, Y, Kikuzaki, H, Hisamoto, M & Nakatani, N (2004) Antioxidant properties of gingerol related compounds from ginger. Biofactors 21, 293296.CrossRefGoogle ScholarPubMed
26Somparn, P, Phisalaphong, C, Nakornchai, S, Unchern, S & Morales, NP (2007) Comparative antioxidant activities of curcumin and its demethoxy and hydrogenated derivatives. Biol Pharm Bull 30, 7478.CrossRefGoogle ScholarPubMed
27Hsu, CY, Yang, CM, Chen, CM, Chao, PY & Hu, SP (2005) Effects of chlorophyll-related compounds on hydrogen peroxide induced DNA damage within human lymphocytes. J Agric Food Chem 53, 27462750.CrossRefGoogle ScholarPubMed
28Xiong, S, Melton, LD, Easteal, AJ & Siew, D (2006) Stability and antioxidant activity of black currant anthocyanins in solution and encapsulated in glucan gel. J Agric Food Chem 54, 62016208.Google Scholar
29Semba, RD, Lauretani, F & Ferrucci, L (2007) Carotenoids as protection against sarcopenia in older adults. Arch Biochem Biophys 458, 141145.Google Scholar
30Aruoma, OI, Sun, B, Fujii, H, Neergheen, VS, Bahorun, T, Kang, KS & Sung, MK (2006) Low molecular proanthocyanidin dietary biofactor Oligonol: its modulation of oxidative stress, bioefficacy, neuroprotection, food application and chemoprevention potentials. Biofactors 27, 245265.CrossRefGoogle ScholarPubMed
31Narayanan, BA (2006) Chemopreventive agents alters global gene expression pattern: predicting their mode of action and targets. Curr Cancer Drug Targets 6, 711727.CrossRefGoogle ScholarPubMed
32Abdelwahed, A, Bouhlel, I, Skandrani, I, Valenti, K, Kadri, M, Guiraud, P, Steiman, R, Mariotte, AM, Ghedira, K, Laporte, F, Dijoux-Franca, MG & Chekir-Ghedira, L (2007) Study of antimutagenic and antioxidant activities of gallic acid and 1,2,3,4,6-pentagalloylglucose from Pistacia lentiscus. Confirmation by microarray expression profiling. Chem Biol Interact 165, 113.CrossRefGoogle ScholarPubMed
33Scandalios, JG (2002) Oxidative stress responses – what have genome-scale studies taught us? Genome Biol 3, 1019.CrossRefGoogle ScholarPubMed
34Choe, E & Min, DB (2006) Chemistry and reactions of reactive oxygen species in foods. Crit Rev Food Sci Nutr 46, 122.CrossRefGoogle ScholarPubMed
35Foksinski, M, Gackowski, D, Rozalski, R, Siomek, A, Guz, J, Szpila, A, Dziaman, T & Olinski, R (2007) Effects of basal level of antioxidants on oxidative DNA damage in humans. Eur J Nutr 46, 174180.CrossRefGoogle ScholarPubMed
36Karihtala, P & Soini, Y (2007) Reactive oxygen species and antioxidant mechanisms in human tissues and their relation to malignancies. Apmis 115, 81103.Google Scholar
37Fridovich, I (1986) Biological effects of the superoxide radical. Arch Biochem Biophys 247, 111.CrossRefGoogle ScholarPubMed
38Halliwell, B & Gutteridge, JM (1999) Free Radicals in Biology and Medicine, 3rd ed.Avon, England: Oxford University Press.Google Scholar
39Beckman, JS & Koppenol, WH (1996) Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and the ugly. Am J Physiol 271, C1424C1437.Google Scholar
40Czapski, G & Goldstein, S (1995) The role of the reactions of NO with superoxide and oxygen in biological systems: a kinetic approach. Free Radic Biol Med 19, 785794.Google Scholar
41Shadyro, OI, Yurkova, IL & Kisel, MA (2002) Radiation-induced peroxidation and fragmentation of lipids in a model membrane. Int J Radiat Biol 78, 211217.CrossRefGoogle Scholar
42Pentland, AP (1994) Active oxygen mechanisms of UV inflammation. Adv Exp Med Biol 366, 8797.Google Scholar
43Koren, HS (1995) Associations between criteria air pollutants and asthma. Environ Health Perspect 103, Suppl. 6, 235242.Google Scholar
44Victorin, K (1994) Review of the genotoxicity of nitrogen oxides. Mutat Res 317, 4355.CrossRefGoogle ScholarPubMed
45Yu, TW & Anderson, D (1997) Reactive oxygen species-induced DNA damage and its modification: a chemical investigation. Mutat Res 379, 201210.Google Scholar
46Wiseman, A, Ridgway, T, Goldfarb, PS & Woods, L (2002) Are food and environmental toxicants ‘overdetected’ by bioassays? Trends Biotechnol 20, 1315.CrossRefGoogle Scholar
47Parke, DV & Sapota, A (1996) Chemical toxicity and reactive oxygen species. Int J Occup Med Environ Health 9, 331340.Google Scholar
48Ames, BN (1986) Food constituents as a source of mutagens, carcinogens, and anticarcinogens. Prog Clin Biol Res 206, 332.Google Scholar
49Kanner, J & Lapidot, T (2001) The stomach as a bioreactor: dietary lipid peroxidation in the gastric fluid and the effects of plant-derived antioxidants. Free Radic Biol Med 31, 13881395.CrossRefGoogle ScholarPubMed
50Richter, C, Gogvadze, V, Laffranchi, R, Schlapbach, R, Schweizer, M, Suter, M, Walter, P & Yaffee, M (1995) Oxidants in mitochondria: from physiology to diseases. Biochim Biophys Acta 1271, 6774.CrossRefGoogle ScholarPubMed
51Fleury, C, Mignotte, B & Vayssiere, JL (2002) Mitochondrial reactive oxygen species in cell death signaling. Biochimie 84, 131141.Google Scholar
52Dhalla, NS, Temsah, RM & Netticadan, T (2000) Role of oxidative stress in cardiovascular diseases. J Hypertens 18, 655673.Google Scholar
53Forman, HJ & Torres, M (2001) Signaling by the respiratory burst in macrophages. IUBMB Life 51, 365371.Google Scholar
54Hawkins, CL, Brown, BE & Davies, MJ (2001) Hypochlorite- and hypobromite-mediated radical formation and its role in cell lysis. Arch Biochem Biophys 395, 137145.CrossRefGoogle ScholarPubMed
55Rodrigues, MR, Rodriguez, D, Henrique Catalani, L, Russo, M & Campa, A (2003) Interferon-gamma independent oxidation of melatonin by macrophages. J Pineal Res 34, 6974.Google Scholar
56Davies, KJ (1987) Protein damage and degradation by oxygen radicals. I. General aspects. J Biol Chem 262, 98959901.CrossRefGoogle ScholarPubMed
57Grune, T, Reinheckel, T & Davies, KJ (1997) Degradation of oxidized proteins in mammalian cells. FASEB J 11, 526534.CrossRefGoogle ScholarPubMed
58Levine, RL & Stadtman, ER (2001) Oxidative modification of proteins during aging. Exp Gerontol 36, 14951502.Google Scholar
59Stadtman, ER (2004) Role of oxidant species in aging. Curr Med Chem 11, 11051112.Google Scholar
60Kushnareva, Y, Murphy, AN & Andreyev, A (2002) Complex I-mediated reactive oxygen species generation: modulation by cytochrome c and NAD(P)+ oxidation–reduction state. Biochem J 368, 545553.CrossRefGoogle ScholarPubMed
61Squadrito, GL & Pryor, WA (1998) Oxidative chemistry of nitric oxide: the roles of superoxide, peroxynitrite, and carbon dioxide. Free Radic Biol Med 25, 392403.Google Scholar
62Younes, M & Siegers, CP (1984) Interrelation between lipid peroxidation and other hepatotoxic events. Biochem Pharmacol 33, 20012003.Google Scholar
63Horton, AA & Fairhurst, S (1987) Lipid peroxidation and mechanisms of toxicity. Crit Rev Toxicol 18, 2779.Google Scholar
64Zock, PL & Katan, MB (1998) Diet, LDL oxidation, and coronary artery disease. Am J Clin Nutr 68, 759760.Google Scholar
65Forrester, JS & Shah, PK (2006) Emerging strategies for increasing high-density lipoprotein. Am J Cardiol 98, 15421549.Google Scholar
66Moller, P & Wallin, H (1998) Adduct formation, mutagenesis and nucleotide excision repair of DNA damage produced by reactive oxygen species and lipid peroxidation product. Mutat Res 410, 271290.CrossRefGoogle ScholarPubMed
67Barzilai, A & Yamamoto, K (2004) DNA damage responses to oxidative stress. DNA Repair (Amst) 3, 11091115.CrossRefGoogle ScholarPubMed
68Niles, JC, Wishnok, JS & Tannenbaum, SR (2006) Peroxynitrite-induced oxidation and nitration products of guanine and 8-oxoguanine: structures and mechanisms of product formation. Nitric Oxide 14, 109121.CrossRefGoogle ScholarPubMed
69Tsutsui, H, Ide, T & Kinugawa, S (2006) Mitochondrial oxidative stress, DNA damage, and heart failure. Antioxid Redox Signal 8, 17371744.CrossRefGoogle ScholarPubMed
70Vaca, CE, Wilhelm, J & Harms-Ringdahl, M (1988) Interaction of lipid peroxidation products with DNA. A review. Mutat Res 195, 137149.Google Scholar
71Ehrenberg, L, Osterman-Golkar, S, Segerbck, D, Svensson, K & Calleman, CJ (1977) Evaluation of genetic risks of alkylating agents. III. Alkylation of haemoglobin after metabolic conversion of ethene to ethene oxide in vivo. Mutat Res 45, 175184.CrossRefGoogle ScholarPubMed
72Segerback, D (1983) Alkylation of DNA and hemoglobin in the mouse following exposure to ethene and ethene oxide. Chem Biol Interact 45, 139151.Google Scholar
73Doublie, S, Bandaru, V, Bond, JP & Wallace, SS (2004) The crystal structure of human endonuclease VIII-like 1 (NEIL1) reveals a zincless finger motif required for glycosylase activity. Proc Natl Acad Sci U S A 101, 1028410289.Google Scholar
74Sanderson, RJ, Bennett, SE, Sung, JS & Mosbaugh, DW (2001) Uracil-initiated base excision DNA repair synthesis fidelity in human colon adenocarcinoma LoVo and Escherichia coli cell extracts. Prog Nucleic Acid Res Mol Biol 68, 165188.Google Scholar
75Krokan, HE, Drablos, F & Slupphaug, G (2002) Uracil in DNA – occurrence, consequences and repair. Oncogene 21, 89358948.Google Scholar
76Cooke, MS, Evans, MD, Dizdaroglu, M & Lunec, J (2003) Oxidative DNA damage: mechanisms, mutation, and disease. FASEB J 17, 11951214.Google Scholar
77Kim, KS, Chang, YJ, Chung, YJ, Park, CU & Seo, HY (2007) Enhanced expression of high-affinity iron transporters via H-ferritin production in yeast. J Biochem Mol Biol 40, 8287.Google ScholarPubMed
78Valko, M, Rhodes, CJ, Moncol, J, Izakovic, M & Mazur, M (2006) Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact 160, 140.CrossRefGoogle ScholarPubMed
79Dalton, TP, Shertzer, HG & Puga, A (1999) Regulation of gene expression by reactive oxygen. Annu Rev Pharmacol Toxicol 39, 67101.Google Scholar
80Allen, RG & Tresini, M (2000) Oxidative stress and gene regulation. Free Radic Biol Med 28, 463499.Google Scholar
81Martindale, JL & Holbrook, NJ (2002) Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol 192, 115.Google Scholar
82Mathers, J, Fraser, JA, McMahon, M, Saunders, RD, Hayes, JD & McLellan, LI (2004) Antioxidant and cytoprotective responses to redox stress. Biochem Soc Symp, 157176.Google ScholarPubMed
83Owuor, ED & Kong, AN (2002) Antioxidants and oxidants regulated signal transduction pathways. Biochem Pharmacol 64, 765770.Google Scholar
84Valko, M, Leibfritz, D, Moncol, J, Cronin, MT, Mazur, M & Telser, J (2007) Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 39, 4484.CrossRefGoogle ScholarPubMed
85Ma, Q, Battelli, L & Hubbs, AF (2006) Multiorgan autoimmune inflammation, enhanced lymphoproliferation, and impaired homeostasis of reactive oxygen species in mice lacking the antioxidant-activated transcription factor Nrf2. Am J Pathol 168, 19601974.Google Scholar
86Poli, G, Leonarduzzi, G, Biasi, F & Chiarpotto, E (2004) Oxidative stress and cell signalling. Curr Med Chem 11, 11631182.Google Scholar
87Abe, J & Berk, BC (1999) Fyn and JAK2 mediate Ras activation by reactive oxygen species. J Biol Chem 274, 2100321010.CrossRefGoogle ScholarPubMed
88Esposito, F, Chirico, G, Montesano Gesualdi, N, Posadas, I, Ammendola, R, Russo, T, Cirino, G & Cimino, F (2003) Protein kinase B activation by reactive oxygen species is independent of tyrosine kinase receptor phosphorylation and requires SRC activity. J Biol Chem 278, 2082820834.CrossRefGoogle ScholarPubMed
89Gopalakrishna, R & Jaken, S (2000) Protein kinase C signaling and oxidative stress. Free Radic Biol Med 28, 13491361.CrossRefGoogle ScholarPubMed
90Kyriakis, JM & Avruch, J (2001) Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev 81, 807869.Google Scholar
91Lopez-Ilasaca, M, Crespo, P, Pellici, PG, Gutkind, JS & Wetzker, R (1997) Linkage of G protein-coupled receptors to the MAPK signaling pathway through PI 3-kinase gamma. Science 275, 394397.Google Scholar
92Iles, KE & Forman, HJ (2002) Macrophage signaling and respiratory burst. Immunol Res 26, 95105.Google Scholar
93Torres, M & Forman, HJ (1999) Activation of several MAP kinases upon stimulation of rat alveolar macrophages: role of the NADPH oxidase. Arch Biochem Biophys 366, 231239.CrossRefGoogle ScholarPubMed
94Rao, A, Luo, C & Hogan, PG (1997) Transcription factors of the NFAT family: regulation and function. Annu Rev Immunol 15, 707747.Google Scholar
95Pinkus, R, Weiner, LM & Daniel, V (1996) Role of oxidants and antioxidants in the induction of AP-1, NF-kappaB, and glutathione S-transferase gene expression. J Biol Chem 271, 1342213429.Google Scholar
96Amiri, KI & Richmond, A (2005) Role of nuclear factor-kappa B in melanoma. Cancer Metastasis Rev 24, 301313.CrossRefGoogle ScholarPubMed
97Hofseth, LJ, Hussain, SP & Harris, CC (2004) P53: 25 years after its discovery. Trends Pharmacol Sci 25, 177181.Google Scholar
98Jin, HO, An, S, Lee, HC, Woo, SH, Seo, SK, Choe, TB, Yoo, DH, Lee, SB, Um, HD, Lee, SJ, Park, MJ, Kim, JI, Hong, SI, Rhee, CH & Park, IC (2007) Hypoxic condition- and high cell density-induced expression of Redd1 is regulated by activation of hypoxia-inducible factor-1alpha and Sp1 through the phosphatidylinositol 3-kinase/Akt signalling pathway. Cell Signal 19, 13931403.Google Scholar
99Li, MH, Jang, JH & Surh, YJ (2005) Nitric oxide induces apoptosis via AP-1-driven upregulation of COX-2 in rat pheochromocytoma cells. Free Radic Biol Med 39, 890899.Google Scholar
100Sablina, AA, Budanov, AV, Ilyinskaya, GV, Agapova, LS, Kravchenko, JE & Chumakov, PM (2005) The antioxidant function of the p53 tumor suppressor. Nat Med 11, 13061313.Google Scholar
101Ehrlich, V, Hoelzl, C, Nersesyan, A, Winter, H, Koller, V, Ferk, F, Fenech, M, Dusinska, M & Knasmüller, S (2007) Wheat sprout consumption and DNA stability: results from an controlled intervention trial. In Synthetic and Natural Compounds in Cancer Therapy and Prevention, p. 15 [Ďuračková, Z, Slámenova, D and Micksche, M et al. , editors]. Bratislava, Slovakia.Google Scholar
102Jauliac, S, Lopez-Rodriguez, C, Shaw, LM, Brown, LF, Rao, A & Toker, A (2002) The role of NFAT transcription factors in integrin-mediated carcinoma invasion. Nat Cell Biol 4, 540544.Google Scholar
103Semenza, GL (2000) HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Appl Physiol 88, 14741480.Google Scholar
104Huang, HC, Nguyen, T & Pickett, CB (2000) Regulation of the antioxidant response element by protein kinase C-mediated phosphorylation of NF-E2-related factor 2. Proc Natl Acad Sci USA 97, 1247512480.CrossRefGoogle ScholarPubMed
105Nguyen, T, Yang, CS & Pickett, CB (2004) The pathways and molecular mechanisms regulating Nrf2 activation in response to chemical stress. Free Radic Biol Med 37, 433441.Google Scholar
106Favreau, LV & Pickett, CB (1995) The rat quinone reductase antioxidant response element. Identification of the nucleotide sequence required for basal and inducible activity and detection of antioxidant response element-binding proteins in hepatoma and non-hepatoma cell lines. J Biol Chem 270, 2446824474.CrossRefGoogle ScholarPubMed
107Gupta, M, Dobashi, K, Greene, EL, Orak, JK & Singh, I (1997) Studies on hepatic injury and antioxidant enzyme activities in rat subcellular organelles following in vivo ischemia and reperfusion. Mol Cell Biochem 176, 337347.CrossRefGoogle ScholarPubMed
108Klaunig, JE & Kamendulis, LM (2004) The role of oxidative stress in carcinogenesis. Annu Rev Pharmacol Toxicol 44, 239267.Google Scholar
109Michiels, C, Raes, M, Toussaint, O & Remacle, J (1994) Importance of Se-glutathione peroxidase, catalase, and Cu/Zn-SOD for cell survival against oxidative stress. Free Radic Biol Med 17, 235248.CrossRefGoogle ScholarPubMed
110Fridovich, I (1986) Superoxide dismutases. Adv Enzymol Relat Areas Mol Biol 58, 6197.Google Scholar
111Kwak, MK, Wakabayashi, N, Itoh, K, Motohashi, H, Yamamoto, M & Kensler, TW (2003) Modulation of gene expression by cancer chemopreventive dithiolethiones through the Keap1-Nrf2 pathway. Identification of novel gene clusters for cell survival. J Biol Chem 278, 81358145.Google Scholar
112Thimmulappa, RK, Mai, KH, Srisuma, S, Kensler, TW, Yamamoto, M & Biswal, S (2002) Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray. Cancer Res 62, 51965203.Google ScholarPubMed
113McMahon, M, Itoh, K, Yamamoto, M & Hayes, JD (2003) Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression. J Biol Chem 278, 2159221600.Google Scholar
114Itoh, K, Wakabayashi, N, Katoh, Y, Ishii, T, Igarashi, K, Engel, JD & Yamamoto, M (1999) Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev 13, 7686.Google Scholar
115Kwak, MK & Kensler, TW (2006) Induction of 26S proteasome subunit PSMB5 by the bifunctional inducer 3-methylcholanthrene through the Nrf2-ARE, but not the AhR/Arnt-XRE, pathway. Biochem Biophys Res Commun 345, 13501357.CrossRefGoogle Scholar
116He, X, Lin, GX, Chen, MG, Zhang, JX & Ma, Q (2007) Protection against chromium (VI)-induced oxidative stress and apoptosis by Nrf2. Recruiting Nrf2 into the nucleus and disrupting the nuclear Nrf2/Keap1 association. Toxicol Sci 98, 298309.CrossRefGoogle ScholarPubMed
117He, X, Chen, MG, Lin, GX & Ma, Q (2006) Arsenic induces NAD(P)H-quinone oxidoreductase I by disrupting the Nrf2 × Keap1 × Cul3 complex and recruiting Nrf2 × Maf to the antioxidant response element enhancer. J Biol Chem 281, 2362023631.Google Scholar
118Mann, GE, Niehueser-Saran, J, Watson, A, Gao, L, Ishii, T, de Winter, P & Siow, RC (2007) Nrf2/ARE regulated antioxidant gene expression in endothelial and smooth muscle cells in oxidative stress: implications for atherosclerosis and preeclampsia. Sheng Li Xue Bao 59, 117127.Google Scholar
119Kundu, JK & Surh, YJ (2005) Breaking the relay in deregulated cellular signal transduction as a rationale for chemoprevention with anti-inflammatory phytochemicals. Mutat Res 591, 123146.CrossRefGoogle ScholarPubMed
120Chen, C & Kong, AN (2004) Dietary chemopreventive compounds and ARE/EpRE signaling. Free Radic Biol Med 36, 15051516.Google Scholar
121Surh, YJ, Kundu, JK, Na, HK & Lee, JS (2005) Redox-sensitive transcription factors as prime targets for chemoprevention with anti-inflammatory and antioxidative phytochemicals. J Nutr 135, 2993S3001S.Google Scholar
122Stange, EF (2006) Review Article: the effect of aminosalicylates and immunomodulation on cancer risk in inflammatory bowel disease. Aliment Pharmacol Ther 24, Suppl. 3, 6467.CrossRefGoogle ScholarPubMed
123Chia, VM, Newcomb, PA, Bigler, J, Morimoto, LM, Thibodeau, SN & Potter, JD (2006) Risk of microsatellite-unstable colorectal cancer is associated jointly with smoking and nonsteroidal anti-inflammatory drug use. Cancer Res 66, 68776883.Google Scholar
124Kong, AN, Owuor, E, Yu, R, Hebbar, V, Chen, C, Hu, R & Mandlekar, S (2001) Induction of xenobiotic enzymes by the MAP kinase pathway and the antioxidant or electrophile response element (ARE/EpRE). Drug Metab Rev 33, 255271.Google Scholar
125Nishinaka, T, Ichijo, Y, Ito, M, Kimura, M, Katsuyama, M, Iwata, K, Miura, T, Terada, T & Yabe-Nishimura, C (2007) Curcumin activates human glutathione S-transferase P1 expression through antioxidant response element. Toxicol Lett 170, 238247.Google Scholar
126Tanigawa, S, Fujii, M & Hou, DX (2007) Action of Nrf2 and Keap1 in ARE-mediated NQO1 expression by quercetin. Free Radic Biol Med 42, 16901703.CrossRefGoogle ScholarPubMed
127Gonzalez-Gallego, J, Sanchez-Campos, S & Tunon, MJ (2007) Anti-inflammatory properties of dietary flavonoids. Nutr Hosp 22, 287293.Google Scholar
128Hong, F, Freeman, ML & Liebler, DC (2005) Identification of sensor cysteines in human Keap1 modified by the cancer chemopreventive agent sulforaphane. Chem Res Toxicol 18, 19171926.Google Scholar
129Hong, F, Sekhar, KR, Freeman, ML & Liebler, DC (2005) Specific patterns of electrophile adduction trigger Keap1 ubiquitination and Nrf2 activation. J Biol Chem 280, 3176831775.Google Scholar
130Wakabayashi, N, Dinkova-Kostova, AT, Holtzclaw, WD, Kang, MI, Kobayashi, A, Yamamoto, M, Kensler, TW & Talalay, P (2004) Protection against electrophile and oxidant stress by induction of the phase 2 response: fate of cysteines of the Keap1 sensor modified by inducers. Proc Natl Acad Sci U S A 101, 20402045.Google Scholar
131Eggler, AL, Liu, G, Pezzuto, JM, van Breemen, RB & Mesecar, AD (2005) Modifying specific cysteines of the electrophile-sensing human Keap1 protein is insufficient to disrupt binding to the Nrf2 domain Neh2. Proc Natl Acad Sci U S A 102, 1007010075.Google Scholar
132Le, SB, Hailer, MK, Buhrow, S, Wang, Q, Flatten, K, Pediaditakis, P, Bible, KC, Lewis, LD, Sausville, EA, Pang, YP, Ames, MM, Lemasters, JJ, Holmuhamedov, EL & Kaufmann, SH (2007) Inhibition of mitochondrial respiration as a source of adaphostin-induced reactive oxygen species and cytotoxicity. J Biol Chem aufmann 282, 88608872.Google Scholar
133Wiseman, A (2006) Oxygen-induced reperfusion-injury is caused by ROS: amelioration is possible by recombinant-DNA antioxidant enzymes and mimics in selected tissues. Med Hypotheses 66, 329331.Google Scholar
134Criddle, DN, Gillies, S, Baumgartner-Wilson, HK, Jaffar, M, Chinje, EC, Passmore, S, Chvanov, M, Barrow, S, Gerasimenko, OV, Tepikin, AV, Sutton, R & Petersen, OH (2006) Menadione-induced reactive oxygen species generation via redox cycling promotes apoptosis of murine pancreatic acinar cells. J Biol Chem 281, 4048540492.CrossRefGoogle ScholarPubMed
135McCarthy, S, Somayajulu, M, Sikorska, M, Borowy-Borowski, H & Pandey, S (2004) Paraquat induces oxidative stress and neuronal cell death; neuroprotection by water-soluble coenzyme Q10. Toxicol Appl Pharmacol 201, 2131.Google Scholar
136Srinivas, P, Gopinath, G, Banerji, A, Dinakar, A & Srinivas, G (2004) Plumbagin induces reactive oxygen species, which mediate apoptosis in human cervical cancer cells. Mol Carcinog 40, 201211.Google Scholar
137Phillips, BJ, Anderson, D & Gangolli, SD (1986) Influence of phagocyte-derived active oxygen species in tissue responses to tumour promoters and irritants. Food Chem Toxicol 24, 681683.Google Scholar
138Minamiyama, Y, Takemura, S, Hai, S, Suehiro, S & Okada, S (2006) Vitamin E deficiency accelerates nitrate tolerance via a decrease in cardiac P450 expression and increased oxidative stress. Free Radic Biol Med 40, 808816.CrossRefGoogle Scholar
139Isomura, H, Fujie, K, Shibata, K, Inoue, N, Iizuka, T, Takebe, G, Takahashi, K, Nishihira, J, Izumi, H & Sakamoto, W (2004) Bone metabolism and oxidative stress in postmenopausal rats with iron overload. Toxicology 197, 93100.Google Scholar
140Gregorevic, P, Lynch, GS & Williams, DA (2001) Hyperbaric oxygen modulates antioxidant enzyme activity in rat skeletal muscles. Eur J Appl Physiol 86, 2427.CrossRefGoogle ScholarPubMed
141He, SX, Luo, JY, Wang, YP, Wang, YL, Fu, H, Xu, JL, Zhao, G & Liu, EQ (2006) Effects of extract from Ginkgo biloba on carbon tetrachloride-induced liver injury in rats. World J Gastroenterol 12, 39243928.Google Scholar
142Bruck, R, Ashkenazi, M, Weiss, S, Goldiner, I, Shapiro, H, Aeed, H, Genina, O, Helpern, Z & Pines, M (2007) Prevention of liver cirrhosis in rats by curcumin. Liver Int 27, 373383.Google Scholar
143Wirtz, S, Neufert, C, Weigmann, B & Neurath, MF (2007) Chemically induced mouse models of intestinal inflammation. Nat Protoc 2, 541546.Google Scholar
144Fujimoto, K, Arakawa, S, Shibaya, Y, Miida, H, Ando, Y, Yasumo, H, Hara, A, Uchiyama, M, Iwabuchi, H, Takasaki, W, Manabe, S & Yamoto, T (2006) Characterization of phenotypes in Gstm1-null mice by cytosolic and in vivo metabolic studies using 1,2-dichloro-4-nitrobenzene. Drug Metab Dispos 34, 14951501.CrossRefGoogle ScholarPubMed
145Erker, L, Schubert, R, Elchuri, S, Huang, TT, Tarin, D, Mueller, K, Zielen, S, Epstein, CJ & Wynshaw-Boris, A (2006) Effect of the reduction of superoxide dismutase 1 and 2 or treatment with alpha-tocopherol on tumorigenesis in Atm-deficient mice. Free Radic Biol Med 41, 590600.Google Scholar
146Ashe, KH (2006) Molecular basis of memory loss in the Tg2576 mouse model of Alzheimer's disease. J Alzheimers Dis 9, 123126.Google Scholar
147Hamaue, N, Ogata, A, Terado, M, Ohno, K, Kikuchi, S, Sasaki, H, Tashiro, K, Hirafuji, M & Minami, M (2006) Brain catecholamine alterations and pathological features with aging in Parkinson disease model rat induced by Japanese encephalitis virus. Neurochem Res 31, 14511455.CrossRefGoogle ScholarPubMed
148Zha, Y, Le, VT, Higami, Y, Shimokawa, I, Taguchi, T & Razzaque, MS (2006) Life-long suppression of growth hormone-insulin-like growth factor I activity in genetically altered rats could prevent age-related renal damage. Endocrinology 147, 56905698.CrossRefGoogle ScholarPubMed
149Larsen, E, Kwon, K, Coin, F, Egly, JM & Klungland, A (2004) Transcription activities at 8-oxoG lesions in DNA. DNA Repair (Amst) 3, 14571468.Google Scholar
150Xie, Y, Yang, H, Cunanan, C, Okamoto, K, Shibata, D, Pan, J, Barnes, DE, Lindahl, T, McIlhatton, M, Fishel, R & Miller, JH (2004) Deficiencies in mouse Myh and Ogg1 result in tumor predisposition and G to T mutations in codon 12 of the K-ras oncogene in lung tumors. Cancer Res 64, 30963102.Google Scholar
151Glasziou, PP & Sanders, SL (2002) Investigating causes of heterogeneity in systematic reviews. Stat Med 21, 15031511.Google Scholar
152Hartmann, A, Niess, AM, Grunert-Fuchs, M, Poch, B & Speit, G (1995) Vitamin E prevents exercise-induced DNA damage. Mutat Res 346, 195202.Google Scholar
153Sacheck, JM, Milbury, PE, Cannon, JG, Roubenoff, R & Blumberg, JB (2003) Effect of vitamin E and eccentric exercise on selected biomarkers of oxidative stress in young and elderly men. Free Radic Biol Med 34, 15751588.Google Scholar
154Dennog, C, Radermacher, P, Barnett, YA & Speit, G (1999) Antioxidant status in humans after exposure to hyperbaric oxygen. Mutat Res 428, 8389.Google Scholar
155Astley, S, Langrish-Smith, A, Southon, S & Sampson, M (1999) Vitamin E supplementation and oxidative damage to DNA and plasma LDL in type 1 diabetes. Diabetes Care 22, 16261631.Google Scholar
156Lean, ME, Noroozi, M, Kelly, I, Burns, J, Talwar, D, Sattar, N & Crozier, A (1999) Dietary flavonols protect diabetic human lymphocytes against oxidative damage to DNA. Diabetes 48, 176181.Google Scholar
157Sampson, MJ, Astley, S, Richardson, T, Willis, G, Davies, IR, Hughes, DA & Southon, S (2001) Increased DNA oxidative susceptibility without increased plasma LDL oxidizability in Type II diabetes: effects of alpha-tocopherol supplementation. Clin Sci (Lond) 101, 235241.Google Scholar
158Jaruga, P, Jaruga, B, Gackowski, D, Olczak, A, Halota, W, Pawlowska, M & Olinski, R (2002) Supplementation with antioxidant vitamins prevents oxidative modification of DNA in lymphocytes of HIV-infected patients. Free Radic Biol Med 32, 414420.Google Scholar
159Soccio, M, Toniato, E, Evangelista, V, Carluccio, M & De Caterina, R (2005) Oxidative stress and cardiovascular risk: the role of vascular NAD(P)H oxidase and its genetic variants. Eur J Clin Invest 35, 305314.Google Scholar
160Hercberg, S, Czernichow, S & Galan, P (2006) Antioxidant vitamins and minerals in prevention of cancers: lessons from the SU.VI.MAX study. Br J Nutr 96, Suppl. 1, S28S30.Google Scholar
161Flora, SJ (2007) Role of free radicals and antioxidants in health and disease. Cell Mol Biol (Noisy-le-grand) 53, 12.Google Scholar
162Kan, E, Undeger, U, Bali, M & Basaran, N (2002) Assessment of DNA strand breakage by the alkaline COMET assay in dialysis patients and the role of vitamin E supplementation. Mutat Res 520, 151159.Google Scholar
163Evans, MD, Cooke, MS, Akil, M, Samanta, A & Lunec, J (2000) Aberrant processing of oxidative DNA damage in systemic lupus erythematosus. Biochem Biophys Res Commun 273, 894898.Google Scholar
164Berliner, LJ, Khramtsov, V, Fujii, H & Clanton, TL (2001) Unique in vivo applications of spin traps. Free Radic Biol Med 30, 489499.Google Scholar
165Khan, N, Wilmot, CM, Rosen, GM, Demidenko, E, Sun, J, Joseph, J, O'Hara, J, Kalyanaraman, B & Swartz, HM (2003) Spin traps: in vitro toxicity and stability of radical adducts. Free Radic Biol Med 34, 14731481.Google Scholar
166Bottle, SE, Hanson, GR & Micallef, AS (2003) Application of the new EPR spin trap 1,1,3-trimethylisoindole N-oxide (TMINO) in trapping HO. and related biologically important radicals. Org Biomol Chem 1, 25852589.Google Scholar
167Karoui, H, Clément, J-L, Rockenbauer, A, Siri, D & Tordo, P (2004) Synthesis and structure of 5,5-diethoxycarbonyl-1-pyrroline N-oxide (DECPO). Application to superoxide radical trapping. Tetrahedron Lett 45, 149152.Google Scholar
168Stolze, K, Udilova, N, Rosenau, T, Hofinger, A & Nohl, H (2003) Spin trapping of superoxide, alkyl- and lipid-derived radicals with derivatives of the spin trap EPPN. Biochem Pharmacol 66, 17171726.Google Scholar
169Zhao, H, Joseph, J, Zhang, H, Karoui, H & Kalyanaraman, B (2001) Synthesis and biochemical applications of a solid cyclic nitrone spin trap: a relatively superior trap for detecting superoxide anions and glutathiyl radicals. Free Radic Biol Med 31, 599606.Google Scholar
170Halliwell, B & Whiteman, M (2004) Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br J Pharmacol 142, 231255.Google Scholar
171Aruoma, OI (2003) Methodological considerations for characterizing potential antioxidant actions of bioactive components in plant foods. Mutat Res 523–524, 920.Google Scholar
172Ghiselli, A, Serafini, M, Maiani, G, Azzini, E & Ferro-Luzzi, A (1995) A fluorescence-based method for measuring total plasma antioxidant capability. Free Radic Biol Med 18, 2936.Google Scholar
173Glazer, AN (1990) Phycoerythrin fluorescence-based assay for reactive oxygen species. Methods Enzymol 186, 161168.Google Scholar
174Cao, G, Alessio, HM & Cutler, RG (1993) Oxygen-radical absorbance capacity assay for antioxidants. Free Radic Biol Med 14, 303311.Google Scholar
175Prior, RL, Wu, X & Schaich, K (2005) Standardized methods for the determination of antioxidant capacity and phenolics in foods and dietary supplements. J Agric Food Chem 53, 42904302.Google Scholar
176Huang, D, Ou, B, Hampsch-Woodill, M, Flanagan, JA & Deemer, EK (2002) Development and validation of oxygen radical absorbance capacity assay for lipophilic antioxidants using randomly methylated β-cyclodextrin as the solubility enhancer. J Agric Food Chem 50, 18151821.Google Scholar
177Huang, D, Ou, B, Hampsch-Woodill, M, Flanagan, JA & Prior, RL (2002) High-throughput assay of oxygen radical absorbance capacity (ORAC) using a multichannel liquid handling system coupled with a microplate fluorescence reader in 96-well format. J Agric Food Chem 50, 44374444.Google Scholar
178Lussignoli, S, Fraccaroli, M, Andrioli, G, Brocco, G & Bellavite, P (1999) A microplate-based colorimetric assay of the total peroxyl radical trapping capability of human plasma. Anal Biochem 269, 3844.Google Scholar
179Frankel, EN & Meyer, AS (2000) The problems of using one-dimensional methods to evaluate multifunctional food and biological antioxidants. J Sci Food Agric 80, 19251941.Google Scholar
180Wayner, DDM, Burton, GW, Ingold, KU & Locke, S (1985) Quantitative measurement of the total, peroxyl radical-trapping antioxidant capability of human blood plasma by controlled peroxidation. The important contribution made by plasma proteins. FEBS Lett 187, 3337.Google Scholar
181DeLange, RJ & Glazer, AN (1989) Phycoerythrin fluorescence-based assay for peroxy radicals: a screen for biologically relevant protective agents. Anal Biochem 177, 300306.Google Scholar
182Bartosz, G, Janaszewska, A, Ertel, D & Bartosz, M (1998) Simple determination of peroxyl radical-trapping capacity. Biochem Mol Biol Int 46, 519528.Google Scholar
183Rota, C, Chignell, CF & Mason, RP (1999) Evidence for free radical formation during the oxidation of 2′-7′-dichlorofluorescin to the fluorescent dye 2′-7′-dichlorofluorescein by horseradish peroxidase: possible implications for oxidative stress measurements. Free Radic Biol Med 27, 873881.Google Scholar
184Ghiselli, A, Serafini, M, Natella, F & Scaccini, C (2000) Total antioxidant capacity as a tool to assess redox status: critical view and experimental data. Free Radic Biol Med 29, 11061114.Google Scholar
185Winston, GW, Regoli, F, Dugas, AJ Jr, Fong, JH & Blanchard, KA (1998) A rapid gas chromatographic assay for determining oxyradical scavenging capacity of antioxidants and biological fluids. Free Radic Biol Med 24, 480493.Google Scholar
186Regoli, F & Winston, GW (1999) Quantification of total oxidant scavenging capacity of antioxidants for peroxynitrite, peroxyl radicals, and hydroxyl radicals. Toxicol Appl Pharmacol 156, 96105.Google Scholar
187Lichtenthaler, R & Marx, F (2005) Total oxidant scavenging capacities of common European fruit and vegetable juices. J Agric Food Chem 53, 103110.Google Scholar
188Lundqvist, H, Kricka, LJ, Stott, RA, Thorpe, GH & Dahlgren, C (1995) Influence of different luminols on the characteristics of the chemiluminescence reaction in human neutrophils. J Biolumin Chemilumin 10, 353359.Google Scholar
189Whitehead, TP, Thorpe, GHG & Maxwell, SRJ (1992) Enhanced chemiluminescent assay for antioxidant capacity in biological fluids. Analytica Chimica Acta 266, 265277.Google Scholar
190Witko-Sarsat, V, Nguyen, AT, Knight, J & Descamps-Latscha, B (1992) Pholasin®: a new chemiluminescent probe for the detection of chloramines derived from human phagocytes. Free Radic Biol Med 13, 8388.Google Scholar
191Swindle, EJ, Hunt, JA & Coleman, JW (2002) A comparison of reactive oxygen species generation by rat peritoneal macrophages and mast cells using the highly sensitive real-time chemiluminescent probe pholasin: inhibition of antigen-induced mast cell degranulation by macrophage-derived hydrogen peroxide. J Immunol 169, 58665873.Google Scholar
192Glebska, J & Koppenol, WH (2005) Chemiluminescence of Pholasin caused by peroxynitrite. Free Radic Biol Med 38, 10141022.Google Scholar
193Popov, I, Völker, H & Lewin, G (2001) Photochemiluminescent detection of antiradical activity. V. Application in combination with the hydrogen peroxide-initiated chemiluminescence of blood plasma proteins to evaluate antioxidant homeostasis in humans. Redox Report 6, 4348.Google Scholar
194Esterbauer, H, Jurgens, G, Quehenberger, O & Koller, E (1987) Autoxidation of human low density lipoprotein: loss of polyunsaturated fatty acids and vitamin E and generation of aldehydes. J Lipid Res 28, 495509.Google Scholar
195Quehenberger, O, Jürgens, G, Zadravec, S & Esterbauer, H (1988) Oxidation of human low density lipoprotein initiated by copper (II) chloride. Basic Life Sci 49, 387390.Google Scholar
196Rice-Evans, C, Leake, D, Bruckdorfer, R & Diplock, AT (1996) Practical approaches to low density lipoprotein oxidation: whys, wherefores and pitfalls. Free Radic Res 25, 285311.Google Scholar
197Wagner, K-H, Tomasch, R & Elmadfa, I (2001) Impact of diets containing corn oil or olive/sunflower oil mixture on the human plasma and lipoprotein lipid metabolism. Eur J Nutr 40, 161167.Google Scholar
198Holvoet, P, Donck, J, Landeloos, M, Brouwers, E, Luijtens, K, Arnout, J, Lesaffre, E, Vanrenterghem, Y & Collen, D (1996) Correlation between oxidized low density lipoproteins and von Willebrand factor renal failure. Thromb Haemost 76, 663669.Google Scholar
199Frankel, EN, German, JB & Davis, PA (1992) Headspace gas chromatography to determine human low density lipoprotein oxidation. Lipids 27, 10471051.Google Scholar
200Bors, W, Michel, C & Saran, M (1984) Inhibition of the bleaching of the carotenoid crocin. A rapid test for quantifying antioxidant activity. Biochim Biophys Acta 796, 312319.Google Scholar
201Tubaro, F, Ghiselli, A, Rapuzzi, P, Maiorino, M & Ursini, F (1998) Analysis of plasma antioxidant capacity by competition kinetics. Free Radic Biol Med 24, 12281234.Google Scholar
202Miller, NJ, Rice-Evans, C, Davies, MJ, Gopinathan, V & Milner, A (1993) A novel method for measuring antioxidant capacity and its application to monitoring the antioxidant status in premature neonates. Clin Sci 84, 407412.Google Scholar
203Re, R, Pellegrini, N, Proteggente, A, Pannala, A, Yang, M & Rice-Evans, C (1999) Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radic Biol Med 26, 12311237.Google Scholar
204Van Den Berg, R, Haenen, G, Van Den Berg, H & Bast, A (1999) Applicability of an improved Trolox equivalent antioxidant capacity (TEAC) assay for evaluation of antioxidant capacity measurements of mixtures. Food Chem 66, 511517.Google Scholar
205Miller, NJ, Sampson, J, Candeias, LP, Bramley, PM & Rice-Evans, CA (1996) Antioxidant activities of carotenes and xanthophylls. FEBS Lett 384, 240242.Google Scholar
206Benzie, IFF & Strain, JJ (1996) The ferric reducing ability of plasma (FRAP) as a measure of ‘antioxidant power’: the FRAP assay. Anal Biochem 239, 7076.Google Scholar
207Benzie, IFF & Szeto, YT (1999) Total antioxidant capacity of teas by the ferric reducing/antioxidant power assay. J Agric Food Chem 47, 633636.Google Scholar
208Ou, B, Huang, D, Hampsch-Woodill, M, Flanagan, JA & Deemer, EK (2002) Analysis of antioxidant activities of common vegetables employing oxygen radical absorbance capacity (ORAC) and ferric reducing antioxidant power (FRAP) assays: a comparative study. J Agric Food Chem 50, 31223128.Google Scholar
209Pulido, R, Bravo, L & Saura-Calixto, F (2000) Antioxidant activity of dietary polyphenols as determined by a modified ferric reducing/antioxidant power assay. J Agric Food Chem 48, 33963402.Google Scholar
210Apak, R, Guclu, K, Ozyurek, M & Karademir, SE (2004) Novel total antioxidant capacity index for dietary polyphenols and vitamins C and E, using their cupric ion reducing capability in the presence of neocuproine: CUPRAC method. J Agric Food Chem 52, 79707981.Google Scholar
211Brand-Williams, W, Cuvelier, ME & Berset, C (1995) Use of a free radical method to evaluate antioxidant activity. Lebensm Wiss Technol 28, 2530.Google Scholar
212Bondet, V, Brand-Williams, W & Berset, C (1997) Kinetics and mechanisms of antioxidant activity using the DPPH free radical method. Food Science and Technology 30, 609615.Google Scholar
213Nomura, T, Kikuchi, M, Kubodera, A & Kawakami, Y (1997) Proton-donative antioxidant activity of fucoxanthin with 1,1-diphenyl-2-picrylhydrazyl (DPPH). Biochem Mol Biol Int 42, 361370.Google Scholar
214Huang, D, Ou, B & Prior, RL (2005) The chemistry behind antioxidant capacity assays. J Agric Food Chem 53, 18411856.Google Scholar
215Spiteller, G (2006) Peroxyl radicals: inductors of neurodegenerative and other inflammatory diseases. Their origin and how they transform cholesterol, phospholipids, plasmalogens, polyunsaturated fatty acids, sugars, and proteins into deleterious products. Free Radic Biol Med 41, 362387.Google Scholar
216Yagi, K (1976) A simple fluorometric assay for lipoperoxide in blood plasma. Biochem Med 15, 212216.Google Scholar
217Gutteridge, JM (1986) Aspects to consider when detecting and measuring lipid peroxidation. Free Radic Res Commun 1, 173184.Google Scholar
218Meagher, EA & Fitzgerald, GA (2000) Indices of lipid peroxidation in vivo: strengths and limitations. Free Radic Biol Med 28, 17451750.Google Scholar
219Wong, SHY, Knight, JA, Hopfer, SM, Zaharia, O, Leach, CN Jr & Sunderman, FW Jr (1987) Lipoperoxides in plasma as measured by liquid-chromatographic separation of malondialdehyde-thiobarbituric acid adduct. Clin Chem 33, 214220.Google Scholar
220Karatas, F, Karatepe, M & Baysar, A (2002) Determination of free malondialdehyde in human serum by high-performance liquid chroma tography. Anal Biochem 311, 7679.Google Scholar
221Wilson, DW, Metz, HN, Graver, LM & Rao, PS (1997) Direct method for quantification of free malondialdehyde with high-performance capillary electrophoresis in biological samples. Clin Chem 43, 19821984.Google Scholar
222Sim, AS, Salonikas, C, Naidoo, D & Wilcken, DEL (2003) Improved method for plasma malondialdehyde measurement by high-performance liquid chromatography using methyl malondialdehyde as an internal standard. J Chromatogr B Analyt Technol Biomed Life Sci 785, 337344.Google Scholar
223Steghens, J-P, Van Kappel, AL, Denis, I & Collombel, C (2001) Diaminonaphtalene, a new highly specific regent for HPLC–UV measurement of total and free malondialdehyde in human plasma or serum. Free Radic Biol Med 31, 242249.Google Scholar
224Cighetti, G, Allevi, P, Anastasia, L, Bortone, L & Paroni, R (2002) Use of methyl malondialdehyde as an internal standard for malondialdehyde detection: validation by isotope-dilution gas chromatography–mass spectrometry. Clin Chem 48, 22662269.Google Scholar
225Stalikas, CD & Konidari, CN (2001) Analysis of malondialdehyde in biological matrices by capillary gas chromatography with electron-capture detection and mass spectrometry. Anal Biochem 290, 108115.Google Scholar
226Del Rio, D, Stewart, AJ & Pellegrini, N (2005) A review of recent studies on malondialdehyde as toxic molecule and biological marker of oxidative stress. Nutr Metab Cardiovasc Dis 15, 316328.Google Scholar
227Ramel, A, Wagner, K-H & Elmadfa, I (2004) Plasma antioxidants and lipid oxidation after submaximal resistance exercise in men. Eur J Nutr 43, 26.Google Scholar
228Konig, D, Wagner, KH, Elmadfa, I & Berg, A (2001) Exercise and oxidative stress: significance of antioxidants with reference to inflammatory, muscular, and systemic stress. Exerc Immunol Rev 7, 108133.Google Scholar
229Morrow, JD, Hill, KE, Burk, RF, Nammour, TM, Badr, KF & Roberts, LJ II (1990) A series of prostaglandin F2-like compounds are produced in vivo in humans by a non-cyclooxygenase, free radical-catalyzed mechanism. Proc Natl Acad Sci U S A 87, 93839387.Google Scholar
230Milne, GL, Musiek, ES & Morrow, JD (2005) F2-isoprostanes as markers of oxidative stress in vivo: an overview. Biomarkers 10, Suppl. 1, S10S23.Google Scholar
231Roberts, LJ II & Morrow, JD (2000) Measurement of F2-isoprostanes as an index of oxidative stress in vivo. Free Radic Biol Med 28, 505513.Google Scholar
232Young, IS (2005) Oxidative stress and vascular disease: insights from isoprostane measurement. Clin Chem 51, 1415.Google Scholar
233Schwedhelm, E & Boger, RH (2003) Application of gas chromatography–mass spectrometry for analysis of isoprostanes: their role in cardiovascular disease. Clin Chem Lab Med 41, 15521561.Google Scholar
234Frank, H, Hintze, T, Bimboes, D & Remmer, H (1980) Monitoring lipid peroxidation by breath analysis: endogenous hydrocarbons and their metabolic elimination. Toxicol Appl Pharmacol 56, 337344.Google Scholar
235Romero, FJ, Bosch-Morell, F, Romero, MJ, Jareno, EJ, Romero, B, Marin, N & Roma, J (1998) Lipid peroxidation products and antioxidants in human disease. Environ Health Perspect 106, Suppl. 5, 12291234.Google Scholar
236Risby, TH & Sehnert, SS (1999) Clinical application of breath biomarkers of oxidative stress status. Free Radic Biol Med 27, 11821192.Google Scholar
237Esterbauer, H, Schaur, RJ & Zollner, H (1991) Chemistry and Biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 11, 81128.Google Scholar
238Benedetti, A, Comporti, M & Esterbauer, H (1980) Identification of 4-hydroxynonenal as a cytotoxic product originating from the peroxidation of liver microsomal lipids. Biochim Biophys Acta 620, 281296.Google Scholar
239Cahuana, GM, Tejedo, JR, Jimenez, J, Ramirez, R, Sobrino, F & Bedoya, FJ (2003) Involvement of advanced lipooxidation end products (ALEs) and protein oxidation in the apoptotic actions of nitric oxide in insulin secreting RINm5F cells. Biochem Pharmacol 66, 19631971.Google Scholar
240Holley, AE, Walker, MK, Cheeseman, KH & Slater, TF (1993) Measurement of n-alkanals and hydroxyalkenals in biological samples. Free Radic Biol Med 15, 281289.Google Scholar
241Sharma, R, Brown, D, Awasthi, S, Yang, Y, Sharma, A, Patrick, B, Saini, MK, Singh, SP, Zimniak, P, Singh, SV & Awasthi, YC (2004) Transfection with 4-hydroxynonenal-metabolizing glutathione S-transferase isozymes leads to phenotypic transformation and immortalization of adherent cells. Eur J Biochem 271, 16901701.Google Scholar
242Agarwal, R (2004) Chronic kidney disease is associated with oxidative stress independent of hypertension. Clin Nephrol 61, 377383.Google Scholar
243Winterbourn, CC & Buss, IH (1999) Protein carbonyl measurement by enzyme-linked immunosorbent assay. Methods Enzymol 300, 106111.Google Scholar
244Levine, RL, Wehr, N, Williams, JA, Stadtman, ER & Shacter, E (2000) Determination of carbonyl groups in oxidized proteins. Methods Mol Biol 99, 1524.Google Scholar
245Levine, RL, Garland, D, Oliver, CN, Amici, A, Climent, I, Lenz, A-G, Ahn, B-W, Shaltiel, S & Stadtman, ER (1990) Determination of carbonyl content in oxidatively modified proteins. Methods Enzymol 186, 464478.Google Scholar
246Alamdari, DH, Kostidou, E, Paletas, K, Sarigianni, M, Konstas, AGP, Karapiperidou, A & Koliakos, G (2005) High sensitivity enzyme-linked immunosorbent assay (ELISA) method for measuring protein carbonyl in samples with low amounts of protein. Free Radic Biol Med 39, 13621367.Google Scholar
247Witko-Sarsat, V, Friedlander, M, Capeillere-Blandin, C, Nguyen-Khoa, T, Nguyen, AT, Zingraff, J, Jungers, P & Descamps-Latscha, B (1996) Advanced oxidation protein products as a novel marker of oxidative stress in uremia. Kidney Int 49, 13041313.Google Scholar
248Capeillere-Blandin, C, Gausson, V, Descamps-Latscha, B & Witko-Sarsat, V (2004) Biochemical and spectrophotometric significance of advanced oxidized protein products. Biochim Biophys Acta 1689, 91102.Google Scholar
249Witko-Sarsat, V, Friedlander, M, Khoa, TN, Capeillére-Blandin, C, Nguyen, AT, Canteloup, S, Dayer, J-M, Jungers, P, Drüeke, T & Descamps-Latscha, B (1998) Advanced oxidation protein products as novel mediators of inflammation and monocyte activation in chronic renal failure. J Immunol 161, 25242532.Google Scholar
250Kaneda, H, Taguchi, J, Ogasawara, K, Aizawa, T & Ohno, M (2002) Increased level of advanced oxidation protein products in patients with coronary artery disease. Atherosclerosis 162, 221225.Google Scholar
251Kalousova, M, Zima, T, Tesar, V, Dusilova-Sulkova, S & Skrha, J (2005) Advanced glycoxidation end products in chronic diseases – clinical chemistry and genetic background. Mutat Res 579, 3746.CrossRefGoogle ScholarPubMed
252Kilbey, BJ, Legator, M, Nichols, W & Ramel, C (1984) Handbook of Mutagenicity Test Procedures. Amsterdam: Elsevier Science Publisher.Google Scholar
253Venitt, SP & Parry, JM (1984) Mutagenicity Testing: A Practical Approach. Oxford: IRL-Press.Google Scholar
254Fahrig, R (1993) Mutationsforschung und genetische Toxikologie. Darmstadt: Wiss Buchges.Google Scholar
255Knasmüller, S, Steinkellner, H, Majer, BJ, Nobis, EC, Scharf, G & Kassie, F (2002) Search for dietary antimutagens and anticarcinogens: methodological aspects and extrapolation problems. Food Chem Toxicol 40, 10511062.Google Scholar
256Knasmüller, S, Majer, BJ & Buchmann, C (2004) Identifying antimutagenic constituents of food. In Functional Foods, Ageing and Degenerative Disease, pp. 581614 [Remacle, C and Reusens, B, editors]. Cambridge, England/Boca Raton, Boston, New York, Washington, DC: CRC Press.Google Scholar
257Hoelzl, C, Bichler, J, Ferk, F, Simic, T, Nersesyan, A, Elbling, L, Ehrlich, V, Chakraborty, A & Knasmuller, S (2005) Methods for the detection of antioxidants which prevent age related diseases: a critical review with particular emphasis on human intervention studies. J Physiol Pharmacol 56, Suppl. 2, 4964.Google Scholar
258Fenech, M (1990) The cytokinesis-block micronucleus assay in nucleated cells. Prog Clin Biol Res 340B, 195206.Google Scholar
259Ames, BN (1972) A bacterial system for detecting mutagens and carcinogens. In Mutagenic Effects of Environmental Contaminants, pp. 5766 [Sutton, HE and Harris, MI, editors]. New York: Academic Press.Google Scholar
260Levin, DE, Hollstein, M, Christman, MF, Schwiers, EA & Ames, BN (1982) A new Salmonella tester strain (TA102) with A X T base pairs at the site of mutation detects oxidative mutagens. Proc Natl Acad Sci U S A 79, 74457449.CrossRefGoogle Scholar
261OECD (1997) Bacterial Reverse Mutation Test. OECD Guidelines for the Testing of Chemicals. Test Guideline No. 471.Google Scholar
262Rutten, B & Gocke, E (1988) The ‘antimutagenic’ effect of cinnamaldehyde is due to a transient growth inhibition. Mutat Res 201, 97105.Google Scholar
263Wurgler, FE, Friederich, U, Furer, E & Ganss, M (1990) Salmonella/mammalian microsome assay with tetranitromethane and 3-nitro-l-tyrosine. Mutat Res 244, 714.Google Scholar
264Ostling, O & Johanson, KJ (1984) Microelectrophoretic study of radiation-induced DNA damages in individual mammalian cells. Biochem Biophys Res Commun 123, 291298.Google Scholar
265Tice, RR, Agurell, E, Anderson, D, Burlinson, B, Hartmann, A, Kobayashi, H, Miyamae, Y, Rojas, E, Ryu, JC & Sasaki, YF (2000) Single cell gel/comet assay: guidelines for in vitro and in vivo genetic toxicology testing. Environ Mol Mutagen 35, 206221.Google Scholar
266Singh, NP, McCoy, MT, Tice, RR & Schneider, EL (1988) A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res 175, 184191.Google Scholar
267Sasaki, YF, Sekihashi, K, Izumiyama, F, Nishidate, E, Saga, A, Ishida, K & Tsuda, S (2000) The comet assay with multiple mouse organs: comparison of comet assay results and carcinogenicity with 208 chemicals selected from the IARC monographs and U.S. NTP Carcinogenicity Database. Crit Rev Toxicol 30, 629799.Google Scholar
268Dhillon, VS, Thomas, P & Fenech, M (2004) Comparison of DNA damage and repair following radiation challenge in buccal cells and lymphocytes using single-cell gel electrophoresis. Int J Radiat Biol 80, 517528.Google Scholar
269Martino-Roth, MG, Viegas, J & Roth, DM (2003) Occupational genotoxicity risk evaluation through the comet assay and the micronucleus test. Genet Mol Res 2, 410417.Google Scholar
270Rojas, E, Valverde, M, Sordo, M & Ostrosky-Wegman, P (1996) DNA damage in exfoliated buccal cells of smokers assessed by the single cell gel electrophoresis assay. Mutat Res 370, 115120.Google Scholar
271Szeto, YT, Benzie, IF, Collins, AR, Choi, SW, Cheng, CY, Yow, CM & Tse, MM (2005) A buccal cell model comet assay: development and evaluation for human biomonitoring and nutritional studies. Mutat Res 578, 371381.Google Scholar
272DeMarini, DM (2006) Inhibition of fried meat-induced DNA-damage: use of cruciferous vegetables, yogurt and chlorophyllin in a dietary intervention study in humans. In 36th Annual Meeting of the European Environmental Mutagen Society. From Genes to Molecular Epidemiology. Prague, Czech Republic..Google Scholar
273Collins, AR, Duthie, SJ & Dobson, VL (1993) Direct enzymic detection of endogenous oxidative base damage in human lymphocyte DNA. Carcinogenesis 14, 17331735.Google Scholar
274Collins, AR & Horvathova, E (2001) Oxidative DNA damage, antioxidants and DNA repair: applications of the comet assay. Biochem Soc Trans 29, 337341.Google Scholar
275Collins, AR (2004) The comet assay for DNA damage and repair: principles, applications, and limitations. Mol Biotechnol 26, 249261.Google Scholar
276Chakraborty, S, Roy, M & Bhattacharya, RK (2004) Prevention and repair of DNA damage by selected phytochemicals as measured by single cell gel electrophoresis. J Environ Pathol Toxicol Oncol 23, 215226.Google Scholar
277Collins, BH, Horska, A, Hotten, PM, Riddoch, C & Collins, AR (2001) Kiwifruit protects against oxidative DNA damage in human cells and in vitro. Nutr Cancer 39, 148153.Google Scholar
278Hartmann, A, Agurell, E, Beevers, C, Brendler-Schwaab, S, Burlinson, B, Clay, P, Collins, A, Smith, A, Speit, G, Thybaud, V & Tice, RR (2003) Recommendations for conducting the in vivo alkaline comet assay. Mutagenesis 18, 4551.Google Scholar
279Burlinson, B, Tice, RR, Speit, G, Agurell, E, Brendler-Schwaab, SY, Collins, AR, Escobar, P, Honma, M, Kumaravel, TS, Nakajima, M, Sasaki, YF, Thybaud, V, Uno, Y, Vasquez, M & Hartmann, A (2007) Fourth International Workgroup on Genotoxicity testing: results of the in vivo comet assay workgroup. Mutat Res 627, 3135.Google Scholar
280Anderson, D, Yu, TW, Phillips, BJ & Schmezer, P (1994) The effect of various antioxidants and other modifying agents on oxygen-radical-generated DNA damage in human lymphocytes in the COMET assay. Mutat Res 307, 261271.Google Scholar
281Anderson, D, Basaran, N, Dobrzynska, MM, Basaran, AA & Yu, TW (1997) Modulating effects of flavonoids on food mutagens in human blood and sperm samples in the comet assay. Teratog Carcinog Mutagen 17, 4558.Google Scholar
282Bichler, J, Cavin, C, Simic, T, Chakraborty, A, Ferk, F, Hoelzl, C, Schulte-Hermann, R, Kundi, M, Haidinger, G, Angelis, K & Knasmuller, S (2007) Coffee consumption protects human lymphocytes against oxidative and 3-amino-1-methyl-5H-pyrido[4,3-b]indole acetate (Trp-P-2) induced DNA-damage: results of an experimental study with human volunteers. Food Chem Toxicol 45, 14281436.Google Scholar
283Elbling, L, Weiss, RM, Teufelhofer, O, Uhl, M, Knasmueller, S, Schulte-Hermann, R, Berger, W & Micksche, M (2005) Green tea extract and ( − )-epigallocatechin-3-gallate, the major tea catechin, exert oxidant but lack antioxidant activities. FASEB J 19, 807809.Google Scholar
284Kundu, T, Bhattacharya, RK, Siddiqi, M & Roy, M (2005) Correlation of apoptosis with comet formation induced by tea polyphenols in human leukemia cells. J Environ Pathol Toxicol Oncol 24, 115128.Google Scholar
285Vanitha, A, Murthy, KN, Kumar, V, Sakthivelu, G, Veigas, JM, Saibaba, P & Ravishankar, GA (2007) Effect of the carotenoid-producing alga, Dunaliella bardawil, on CCl4-induced toxicity in rats. Int J Toxicol 26, 159167.Google Scholar
286Kapiszewska, M, Cierniak, A, Papiez, MA, Pietrzycka, A, Stepniewski, M & Lomnicki, A (2007) Prolonged quercetin administration diminishes the etoposide-induced DNA damage in bone marrow cells of rats. Drug Chem Toxicol 30, 6781.Google Scholar
287Gabbianelli, R, Nasuti, C, Falcioni, G & Cantalamessa, F (2004) Lymphocyte DNA damage in rats exposed to pyrethroids: effect of supplementation with vitamins E and C. Toxicology 203, 1726.Google Scholar
288Liu, HG & Xu, LH (2007) Garlic oil prevents tributyltin-induced oxidative damage in vivo and in vitro. J Food Prot 70, 716721.Google Scholar
289Moller, P & Loft, S (2002) Oxidative DNA damage in human white blood cells in dietary antioxidant intervention studies. Am J Clin Nutr 76, 303310.Google Scholar
290Moller, P & Loft, S (2004) Interventions with antioxidants and nutrients in relation to oxidative DNA damage and repair. Mutat Res 551, 7989.Google Scholar
291Moller, P & Loft, S (2006) Dietary antioxidants and beneficial effect on oxidatively damaged DNA. Free Radic Biol Med 41, 388415.Google Scholar
292Pool-Zobel, BL, Bub, A, Muller, H, Wollowski, I & Rechkemmer, G (1997) Consumption of vegetables reduces genetic damage in humans: first results of a human intervention trial with carotenoid-rich foods. Carcinogenesis 18, 18471850.Google Scholar
293Riso, P, Visioli, F, Erba, D, Testolin, G & Porrini, M (2004) Lycopene and vitamin C concentrations increase in plasma and lymphocytes after tomato intake. Effects on cellular antioxidant protection. Eur J Clin Nutr 58, 13501358.Google Scholar
294Gill, CI, Haldar, S, Porter, S, Matthews, S, Sullivan, S, Coulter, J, McGlynn, H & Rowland, I (2004) The effect of cruciferous and leguminous sprouts on genotoxicity, in vitro and in vivo. Cancer Epidemiol Biomarkers Prev 13, 11991205.Google Scholar
295Zhao, X, Aldini, G, Johnson, EJ, Rasmussen, H, Kraemer, K, Woolf, H, Musaeus, N, Krinsky, NI, Russell, RM & Yeum, KJ (2006) Modification of lymphocyte DNA damage by carotenoid supplementation in postmenopausal women. Am J Clin Nutr 83, 163169.Google Scholar
296Ferk, F, Knasmüller, S, Dusinska, M, Brantner, A, Uhl, M & Chakraborty, A (2003) DNA-protective effects of sumach (Rhus coriaria) in human and human derived cells in vitro and identifications of its active principles. In Eighth International Conference on Mechanisms of Antimutagenesis and Anticarcinogenesis, p. 47 [Bronzetti, G, Ferguson, LR and De Flora, S, editors]. Italy: Pisa.Google Scholar
297Hoelzl, C, Glatt, H-R, Meinl, W, Sontag, G, Heidinger, G, Kundi, M, Simic, T, Chakraborty, A, Bichler, J, Ferk, F, Angelis, K, Nersesyan, A & Knasmüller, S (2008) Consumption of Brussels sprouts protects peripheral human lymphocytes against 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and oxidative. Mol Nutr Food Res 53, 330341.Google Scholar
298Moller, P, Vogel, U, Pedersen, A, Dragsted, LO, Sandstrom, B & Loft, S (2003) No effect of 600 grams fruit and vegetables per day on oxidative DNA damage and repair in healthy nonsmokers. Cancer Epidemiol Biomarkers Prev 12, 10161022.Google Scholar
299Knasmüller, S, Mersch-Sundermann, V, Kevekordes, S, Darroudi, F, Huber, WW, Hoelzl, C, Bichler, J & Majer, BJ (2004) Use of human-derived liver cell lines for the detection of environmental and dietary genotoxicants; current state of knowledge. Toxicology 198, 315328.Google Scholar
300Knasmüller, S, Parzefall, W, Sanyal, R, Ecker, S, Schwab, C, Uhl, M, Mersch-Sundermann, V, Williamson, G, Hietsch, G, Langer, T, Darroudi, F & Natarajan, AT (1998) Use of metabolically competent human hepatoma cells for the detection of mutagens and antimutagens. Mutat Res 402, 185202.Google Scholar
301Mersch-Sundermann, V, Knasmüller, S, Wu, XJ, Darroudi, F & Kassie, F (2004) Use of a human-derived liver cell line for the detection of cytoprotective, antigenotoxic and cogenotoxic agents. Toxicology 198, 329340.Google Scholar
302OECD (1997) Mammalian Erythrocyte Micronucleus Test. OECD Guidelines for the Testing of Chemicals. Test Guideline No. 474.Google Scholar
303Hosseinimehr, SJ, Azadbakht, M, Mousavi, SM, Mahmoudzadeh, A & Akhlaghpoor, S (2007) Radioprotective effects of hawthorn fruit extract against gamma irradiation in mouse bone marrow cells. J Radiat Res (Tokyo) 48, 6368.Google Scholar
304Isbrucker, RA, Bausch, J, Edwards, JA & Wolz, E (2006) Safety studies on epigallocatechin gallate (EGCG) preparations. Part 1: genotoxicity. Food Chem Toxicol 44, 626635.Google Scholar
305Misra, S & Choudhury, RC (2006) Vitamin C modulation of cisplatin-induced cytogenotoxicity in bone marrow, spermatogonia and its transmission in the male germline of Swiss mice. J Chemother 18, 182187.Google Scholar
306Kitano, M, Hosoe, K, Fukutomi, N, Hidaka, T, Ohta, R, Yamakage, K & Hara, T (2006) Evaluation of the mutagenic potential of ubidecarenone using three short-term assays. Food Chem Toxicol 44, 364370.Google Scholar
307Subapriya, R, Kumaraguruparan, R, Abraham, SK & Nagini, S (2005) Protective effects of ethanolic neem leaf extract on DMBA-induced genotoxicity and oxidative stress in mice. J Herb Pharmacother 5, 3950.Google Scholar
308Prahalathan, C, Selvakumar, E, Varalakshmi, P, Kumarasamy, P & Saravanan, R (2006) Salubrious effects of lipoic acid against adriamycin-induced clastogenesis and apoptosis in Wistar rat bone marrow cells. Toxicology 222, 225232.Google Scholar
309Fenech, M & Morley, AA (1985) Measurement of micronuclei in lymphocytes. Mutat Res 147, 2936.Google Scholar
310Fenech, M (2000) The in vitro micronucleus technique. Mutat Res 455, 8195.Google Scholar
311Fenech, M, Stockley, C & Aitken, C (1997) Moderate wine consumption protects against hydrogen peroxide-induced DNA damage. Mutagenesis 12, 289296.Google Scholar
312Crott, JW & Fenech, M (1999) Effect of vitamin C supplementation on chromosome damage, apoptosis and necrosis ex vivo. Carcinogenesis 20, 10351041.Google Scholar
313Greenrod, W, Stockley, CS, Burcham, P, Abbey, M & Fenech, M (2005) Moderate acute intake of de-alcoholized red wine, but not alcohol, is protective against radiation-induced DNA damage ex vivo – results of a comparative in vivo intervention study in younger men. Mutat Res 591, 290301.Google Scholar
314Greenrod, W & Fenech, M (2003) The principal phenolic and alcoholic components of wine protect human lymphocytes against hydrogen peroxide- and ionizing radiation-induced DNA damage in vitro. Mutagenesis 18, 119126.Google Scholar
315Kopjar, N, Miocic, S, Ramic, S, Milic, M & Viculin, T (2006) Assessment of the radioprotective effects of amifostine and melatonin on human lymphocytes irradiated with gamma-rays in vitro. Arh Hig Rada Toksikol 57, 155163.Google Scholar
316Dizdaroglu, M (1994) Chemical determination of oxidative DNA damage by gas chromatography–mass spectrometry. Methods Enzymol 234, 316.Google Scholar
317Ames, BN, Shigenaga, MK & Hagen, TM (1993) Oxidants, antioxidants, and the degenerative diseases of aging. Proc Natl Acad Sci U S A 90, 79157922.Google Scholar
318Chater, S, Abdelmelek, H, Douki, T, Garrel, C, Favier, A, Sakly, M & Ben Rhouma, K (2006) Exposure to static magnetic field of pregnant rats induces hepatic GSH elevation but not oxidative DNA damage in liver and kidney. Arch Med Res 37, 941946.Google Scholar
319Hays, AM, Srinivasan, D, Witten, ML, Carter, DE & Lantz, RC (2006) Arsenic and cigarette smoke synergistically increase DNA oxidation in the lung. Toxicol Pathol 34, 396404.Google Scholar
320Hofer, T, Seo, AY, Prudencio, M & Leeuwenburgh, C (2006) A method to determine RNA and DNA oxidation simultaneously by HPLC–ECD: greater RNA than DNA oxidation in rat liver after doxorubicin administration. Biol Chem 387, 103111.Google Scholar
321Kim, SY, Suzuki, N, Laxmi, YR, Umemoto, A, Matsuda, T & Shibutani, S (2006) Antiestrogens and the formation of DNA damage in rats: a comparison. Chem Res Toxicol 19, 852858.Google Scholar
322Shigenaga, MK, Gimeno, CJ & Ames, BN (1989) Urinary 8-hydroxy-2′-deoxyguanosine as a biological marker of in vivo oxidative DNA damage. Proc Natl Acad Sci USA 86, 96979701.Google Scholar
323Dizdaroglu, M (1990) Gas chromatography–mass spectrometry of free radical-induced products of pyrimidines and purines in DNA. Methods Enzymol 193, 842857.Google Scholar
324Gackowski, D, Rozalski, R, Roszkowski, K, Jawien, A, Foksinski, M & Olinski, R (2001) 8-Oxo-7,8-dihydroguanine and 8-oxo-7,8-dihydro-2′-deoxyguanosine levels in human urine do not depend on diet. Free Radic Res 35, 825832.Google Scholar
325Cooke, MS, Olinski, R & Evans, MD (2006) Does measurement of oxidative damage to DNA have clinical significance? Clin Chim Acta 365, 3049.Google Scholar
326Kasai, H (2003) A new automated method to analyze urinary 8-hydroxydeoxyguanosine by a high-performance liquid chromatography–electrochemical detector system. J Radiat Res (Tokyo) 44, 185189.Google Scholar
327Loft, S, Deng, XS, Tuo, J, Wellejus, A, Sorensen, M & Poulsen, HE (1998) Experimental study of oxidative DNA damage. Free Radic Res 29, 525539.Google Scholar
328Shigenaga, MK, Aboujaoude, EN, Chen, Q & Ames, BN (1994) Assays of oxidative DNA damage biomarkers 8-oxo-2′-deoxyguanosine and 8-oxoguanine in nuclear DNA and biological fluids by high-performance liquid chromatography with electrochemical detection. Methods Enzymol 234, 1633.Google Scholar
329Harman, SM, Liang, L, Tsitouras, PD, Gucciardo, F, Heward, CB, Reaven, PD, Ping, W, Ahmed, A & Cutler, RG (2003) Urinary excretion of three nucleic acid oxidation adducts and isoprostane F(2)alpha measured by liquid chromatography–mass spectrometry in smokers, ex-smokers, and nonsmokers. Free Radic Biol Med 35, 13011309.Google Scholar
330Chiou, CC, Chang, PY, Chan, EC, Wu, TL, Tsao, KC & Wu, JT (2003) Urinary 8-hydroxydeoxyguanosine and its analogs as DNA marker of oxidative stress: development of an ELISA and measurement in both bladder and prostate cancers. Clin Chim Acta 334, 8794.Google Scholar
331Hofer, T & Moller, L (1998) Reduction of oxidation during the preparation of DNA and analysis of 8-hydroxy-2′-deoxyguanosine. Chem Res Toxicol 11, 882887.Google Scholar
332Wiseman, H, Kaur, H & Halliwell, B (1995) DNA damage and cancer: measurement and mechanism. Cancer Lett 93, 113120.Google Scholar
333Douki, T, Delatour, T, Bianchini, F & Cadet, J (1996) Observation and prevention of an artefactual formation of oxidized DNA bases and nucleosides in the GC–EIMS method. Carcinogenesis 17, 347353.Google Scholar
334Poulsen, HE, Weimann, A & Loft, S (1999) Methods to detect DNA damage by free radicals: relation to exercise. Proc Nutr Soc 58, 10071014.Google Scholar
335Ravanat, JL, Turesky, RJ, Gremaud, E, Trudel, LJ & Stadler, RH (1995) Determination of 8-oxoguanine in DNA by gas chromatography–mass spectrometry and HPLC–electrochemical detection: overestimation of the background level of the oxidized base by the gas chromatography–mass spectrometry assay. Chem Res Toxicol 8, 10391045.Google Scholar
336Hu, CW, Wang, CJ, Chang, LW & Chao, MR (2006) Clinical-scale high-throughput analysis of urinary 8-oxo-7,8-dihydro-2′-deoxyguanosine by isotope-dilution liquid chromatography–tandem mass spectrometry with on-line solid-phase extraction. Clin Chem 52, 13811388.Google Scholar
337Beckman, KB & Ames, BN (1997) Oxidative decay of DNA. J Biol Chem 272, 1963319636.Google Scholar
338Adachi, S, Zeisig, M & Moller, L (1995) Improvements in the analytical method for 8-hydroxydeoxyguanosine in nuclear DNA. Carcinogenesis 16, 253258.Google Scholar
339Finnegan, MT, Herbert, KE, Evans, MD, Griffiths, HR & Lunec, J (1996) Evidence for sensitisation of DNA to oxidative damage during isolation. Free Radic Biol Med 20, 9398.Google Scholar
340Dreher, D & Junod, AF (1996) Role of oxygen free radicals in cancer development. Eur J Cancer 32A, 3038.Google Scholar
341Cooke, MS, Evans, MD, Herbert, KE & Lunec, J (2000) Urinary 8-oxo-2′-deoxyguanosine – source, significance and supplements. Free Radic Res 32, 381397.Google Scholar
342Shimoi, K, Kasai, H, Yokota, N, Toyokuni, S & Kinae, N (2002) Comparison between high-performance liquid chromatography and enzyme-linked immunosorbent assay for the determination of 8-hydroxy-2′-deoxyguanosine in human urine. Cancer Epidemiol Biomarkers Prev 11, 767770.Google Scholar
343Yoshida, R, Ogawa, Y & Kasai, H (2002) Urinary 8-oxo-7, 8-dihydro-2′-deoxyguanosine values measured by an ELISA correlated well with measurements by high-performance liquid chromatography with electrochemical detection. Cancer Epidemiol Biomarkers Prev 11, 10761081.Google Scholar
344OECD (2003) Measurement of DNA oxidation in human cells by chromatographic and enzymic methods. Free Radic Biol Med 34, 10891099.Google Scholar
345OECD (2002) Comparative analysis of baseline 8-oxo-7,8-dihydroguanine in mammalian cell DNA, by different methods in different laboratories: an approach to consensus. Carcinogenesis 23, 21292133.Google Scholar
346Collins, AR, Cadet, J, Moller, L, Poulsen, HE & Vina, J (2004) Are we sure we know how to measure 8-oxo-7,8-dihydroguanine in DNA from human cells? Arch Biochem Biophys 423, 5765.Google Scholar
347Gedik, CM & Collins, A (2005) Establishing the background level of base oxidation in human lymphocyte DNA: results of an interlaboratory validation study. FASEB J 19, 8284.Google Scholar
348Duthie, SJ, Jenkinson, AM, Crozier, A, Mullen, W, Pirie, L, Kyle, J, Yap, LS, Christen, P & Duthie, GG (2006) The effects of cranberry juice consumption on antioxidant status and biomarkers relating to heart disease and cancer in healthy human volunteers. Eur J Nutr 45, 113122.Google Scholar
349Tunstall, RG, Sharma, RA, Perkins, S, Sale, S, Singh, R, Farmer, PB, Steward, WP & Gescher, AJ (2006) Cyclooxygenase-2 expression and oxidative DNA adducts in murine intestinal adenomas: modification by dietary curcumin and implications for clinical trials. Eur J Cancer 42, 415421.Google Scholar
350Jung, KJ, Wallig, MA & Singletary, KW (2006) Purple grape juice inhibits 7,12-dimethylbenz[a]anthracene (DMBA)-induced rat mammary tumorigenesis and in vivo DMBA–DNA adduct formation. Cancer Lett 233, 279288.Google Scholar
351Hong, JT, Ryu, SR, Kim, HJ, Lee, JK, Lee, SH, Kim, DB, Yun, YP, Ryu, JH, Lee, BM & Kim, PY (2000) Neuroprotective effect of green tea extract in experimental ischemia-reperfusion brain injury. Brain Res Bull 53, 743749.Google Scholar
352Kaneko, T, Tahara, S & Takabayashi, F (2003) Protective effect of fluvastatin, an HMG-CoA reductase inhibitor, on the formation of 8-oxo-2′-deoxyguanosine in the nuclear DNA of hamster pancreas after a single administration of N-nitrosobis(2-oxopropyl)amine. Biol Pharm Bull 26, 12451248.Google Scholar
353van Zeeland, AA, de Groot, AJ, Hall, J & Donato, F (1999) 8-Hydroxydeoxyguanosine in DNA from leukocytes of healthy adults: relationship with cigarette smoking, environmental tobacco smoke, alcohol and coffee consumption. Mutat Res 439, 249257.Google Scholar
354Schins, RP, Schilderman, PA & Borm, PJ (1995) Oxidative DNA damage in peripheral blood lymphocytes of coal workers. Int Arch Occup Environ Health 67, 153157.Google Scholar
355Loft, S, Poulsen, HE, Vistisen, K & Knudsen, LE (1999) Increased urinary excretion of 8-oxo-2′-deoxyguanosine, a biomarker of oxidative DNA damage, in urban bus drivers. Mutat Res 441, 1119.Google Scholar
356Marczynski, B, Rihs, HP, Rossbach, B, Holzer, J, Angerer, J, Scherenberg, M, Hoffmann, G, Bruning, T & Wilhelm, M (2002) Analysis of 8-oxo-7,8-dihydro-2′-deoxyguanosine and DNA strand breaks in white blood cells of occupationally exposed workers: comparison with ambient monitoring, urinary metabolites and enzyme polymorphisms. Carcinogenesis 23, 273281.Google Scholar
357Yoshida, R, Ogawa, Y, Shioji, I, Yu, X, Shibata, E, Mori, I, Kubota, H, Kishida, A & Hisanaga, N (2001) Urinary 8-oxo-7, 8-dihydro-2′-deoxyguanosine and biopyrrins levels among construction workers with asbestos exposure history. Ind Health 39, 186188.Google Scholar
358Loft, S, Astrup, A, Buemann, B & Poulsen, HE (1994) Oxidative DNA damage correlates with oxygen consumption in humans. FASEB J 8, 534537.Google Scholar
359Thompson, HJ, Heimendinger, J, Diker, A, O'Neill, C, Haegele, A, Meinecke, B, Wolfe, P, Sedlacek, S, Zhu, Z & Jiang, W (2006) Dietary botanical diversity affects the reduction of oxidative biomarkers in women due to high vegetable and fruit intake. J Nutr 136, 22072212.Google Scholar
360Thompson, HJ, Heimendinger, J, Gillette, C, Sedlacek, SM, Haegele, A, O'Neill, C & Wolfe, P (2005) In vivo investigation of changes in biomarkers of oxidative stress induced by plant food rich diets. J Agric Food Chem 53, 61266132.Google Scholar
361Machowetz, A, Poulsen, HE, Gruendel, S, Weimann, A, Fito, M, Marrugat, J, de la Torre, R, Salonen, JT, Nyyssonen, K, Mursu, J, Nascetti, S, Gaddi, A, Kiesewetter, H, Baumler, H, Selmi, H, Kaikkonen, J, Zunft, HJ, Covas, MI & Koebnick, C (2007) Effect of olive oils on biomarkers of oxidative DNA stress in Northern and Southern Europeans. FASEB J 21, 4552.Google Scholar
362Jenkinson, AM, Collins, AR, Duthie, SJ, Wahle, KW & Duthie, GG (1999) The effect of increased intakes of polyunsaturated fatty acids and vitamin E on DNA damage in human lymphocytes. FASEB J 13, 21382142.Google Scholar
363Dotan, Y, Lichtenberg, D & Pinchuk, I (2004) Lipid peroxidation cannot be used as a universal criterion of oxidative stress. Prog Lipid Res 43, 200227.Google Scholar
364Srinivasan, M, Sudheer, AR, Pillai, KR, Kumar, PR, Sudhakaran, PR & Menon, VP (2007) Lycopene as a natural protector against gamma-radiation induced DNA damage, lipid peroxidation and antioxidant status in primary culture of isolated rat hepatocytes in vitro. Biochim Biophys Acta 1770, 659665.Google Scholar
365Srinivasan, M, Rajendra Prasad, N & Menon, VP (2006) Protective effect of curcumin on gamma-radiation induced DNA damage and lipid peroxidation in cultured human lymphocytes. Mutat Res 611, 96103.Google Scholar
366Sudheer, AR, Muthukumaran, S, Devipriya, N & Menon, VP (2007) Ellagic acid, a natural polyphenol protects rat peripheral blood lymphocytes against nicotine-induced cellular and DNA damage in vitro: with the comparison of N-acetylcysteine. Toxicology 230, 1121.Google Scholar
367Schaefer, S, Baum, M, Eisenbrand, G & Janzowski, C (2006) Modulation of oxidative cell damage by reconstituted mixtures of phenolic apple juice extracts in human colon cell lines. Mol Nutr Food Res 50, 413417.Google Scholar
368Gandhi, NM & Nair, CK (2005) Protection of DNA and membrane from gamma radiation induced damage by gallic acid. Mol Cell Biochem 278, 111117.Google Scholar
369Duthie, SJ, Gardner, PT, Morrice, PC, Wood, SG, Pirie, L, Bestwick, CC, Milne, L & Duthie, GG (2005) DNA stability and lipid peroxidation in vitamin E-deficient rats in vivo and colon cells in vitro – modulation by the dietary anthocyanin, cyanidin-3-glycoside. Eur J Nutr 44, 195203.Google Scholar
370Weisel, T, Baum, M, Eisenbrand, G, Dietrich, H, Will, F, Stockis, JP, Kulling, S, Rufer, C, Johannes, C & Janzowski, C (2006) An anthocyanin/polyphenolic-rich fruit juice reduces oxidative DNA damage and increases glutathione level in healthy probands. Biotechnol J 1, 388397.Google Scholar
371Bub, A, Watzl, B, Blockhaus, M, Briviba, K, Liegibel, U, Muller, H, Pool-Zobel, BL & Rechkemmer, G (2003) Fruit juice consumption modulates antioxidative status, immune status and DNA damage. J Nutr Biochem 14, 9098.Google Scholar
372Janse van Rensburg, C, Erasmus, E, Loots, DT, Oosthuizen, W, Jerling, JC, Kruger, HS, Louw, R, Brits, M & van der Westhuizen, FH (2005) Rosa roxburghii supplementation in a controlled feeding study increases plasma antioxidant capacity and glutathione redox state. Eur J Nutr 44, 452457.Google Scholar
373Gedik, CM, Boyle, SP, Wood, SG, Vaughan, NJ & Collins, AR (2002) Oxidative stress in humans: validation of biomarkers of DNA damage. Carcinogenesis 23, 14411446.Google Scholar
374Hofer, T, Karlsson, HL & Moller, L (2006) DNA oxidative damage and strand breaks in young healthy individuals: a gender difference and the role of life style factors. Free Radic Res 40, 707714.Google Scholar
375Domanski, MJ (2007) Primary prevention of coronary artery disease. N Engl J Med 357, 15431545.Google Scholar
376Bonassi, S, Znaor, A, Ceppi, M, Lando, C, Chang, WP, Holland, N, Kirsch-Volders, M, Zeiger, E, Ban, S, Barale, R, Bigatti, MP, Bolognesi, C, Cebulska-Wasilewska, A, Fabianova, E, Fucic, A, Hagmar, L, Joksic, G, Martelli, A, Migliore, L, Mirkova, E, Scarfi, MR, Zijno, A, Norppa, H & Fenech, M (2007) An increased micronucleus frequency in peripheral blood lymphocytes predicts the risk of cancer in humans. Carcinogenesis 28, 625631.Google Scholar
377Norppa, H, Bonassi, S, Hansteen, IL, Hagmar, L, Stromberg, U, Rossner, P, Boffetta, P, Lindholm, C, Gundy, S, Lazutka, J, Cebulska-Wasilewska, A, Fabianova, E, Sram, RJ, Knudsen, LE, Barale, R & Fucic, A (2006) Chromosomal aberrations and SCEs as biomarkers of cancer risk. Mutat Res 600, 3745.Google Scholar
378Knudsen, LE & Hansen, AM (2007) Biomarkers of intermediate endpoints in environmental and occupational health. Int J Hyg Environ Health 210, 461470.Google Scholar
379Maluf, SW & Erdtmann, B (2001) Genomic instability in Down syndrome and Fanconi anemia assessed by micronucleus analysis and single-cell gel electrophoresis. Cancer Genet Cytogenet 124, 7175.Google Scholar
380Vives-Bauza, C, Starkov, A & Garcia-Arumi, E (2007) Measurements of the antioxidant enzyme activities of superoxide dismutase, catalase, and glutathione peroxidase. Methods Cell Biol 80, 379393.Google Scholar
381Ferk, F, Chakraborty, A, Simic, T, Kundi, M & Knasmüller, S (2006) Detection of a “super antioxidant” in sumac (Rhus coriaria), a common spice and identification of gallic acid as its active principle. In COST 925/927 “Molecular and Physiological Effects of Bioactive Food Components”, p. 114 [Gee, J and Fogliano, V, editors]. Vienna, Austria.Google Scholar
382Nelson, SK, Bose, SK, Grunwald, GK, Myhill, P & McCord, JM (2006) The induction of human superoxide dismutase and catalase in vivo: a fundamentally new approach to antioxidant therapy. Free Radic Biol Med 40, 341347.Google Scholar
383Kim, HY, Kim, OH & Sung, MK (2003) Effects of phenol-depleted and phenol-rich diets on blood markers of oxidative stress, and urinary excretion of quercetin and kaempferol in healthy volunteers. J Am Coll Nutr 22, 217223.Google Scholar
384Akturk, O, Demirin, H, Sutcu, R, Yilmaz, N, Koylu, H & Altuntas, I (2006) The effects of diazinon on lipid peroxidation and antioxidant enzymes in rat heart and ameliorating role of vitamin E and vitamin C. Cell Biol Toxicol 22, 455461.Google Scholar
385Hoelzl, C, Lorenz, O, Haudek, V, Gundacker, N, Knasmuller, S & Gerner, C (2008) Proteome alterations induced in human white blood cells by consumption of Brussels sprouts: results of a pilot intervention study. Proteomics Clin Appl 2, 108117.Google Scholar
386Pedraza-Chaverri, J, Granados-Silvestre, MD, Medina-Campos, ON, Maldonado, PD, Olivares-Corichi, IM & Ibarra-Rubio, ME (2001) Post-transcriptional control of catalase expression in garlic-treated rats. Mol Cell Biochem 216, 919.Google Scholar
387Yao, P, Li, K, Song, F, Zhou, S, Sun, X, Zhang, X, Nussler, AK & Liu, L (2007) Heme oxygenase-1 upregulated by Ginkgo biloba extract: potential protection against ethanol-induced oxidative liver damage. Food Chem Toxicol 45, 13331342.Google Scholar
388Prochaska, HJ, Santamaria, AB & Talalay, P (1992) Rapid detection of inducers of enzymes that protect against carcinogens. Proc Natl Acad Sci USA 89, 23942398.Google Scholar
389Prochaska, HJ & Talalay, P (1988) Regulatory mechanisms of monofunctional and bifunctional anticarcinogenic enzyme inducers in murine liver. Cancer Res 48, 47764782.Google Scholar
390Talalay, P & Fahey, JW (2001) Phytochemicals from cruciferous plants protect against cancer by modulating carcinogen metabolism. J Nutr 131, 3027S3033S.Google Scholar
391Hintze, KJ, Wald, KA, Zeng, H, Jeffery, EH & Finley, JW (2003) Thioredoxin reductase in human hepatoma cells is transcriptionally regulated by sulforaphane and other electrophiles via an antioxidant response element. J Nutr 133, 27212727.Google Scholar
392Wild, AC & Mulcahy, RT (2000) Regulation of gamma-glutamylcysteine synthetase subunit gene expression: insights into transcriptional control of antioxidant defenses. Free Radic Res 32, 281301.Google Scholar
393Nardi, G, Cipollaro, M & Loguercio, C (1990) Assay of gamma-glutamylcysteine synthetase and glutathione synthetase in erythrocytes by high-performance liquid chromatography with fluorimetric detection. J Chromatogr 530, 122128.Google Scholar
394Huber, WW, Scharf, G, Rossmanith, W, Prustomersky, S, Grasl-Kraupp, B, Peter, B, Turesky, RJ & Schulte-Hermann, R (2002) The coffee components kahweol and cafestol induce gamma-glutamylcysteine synthetase, the rate limiting enzyme of chemoprotective glutathione synthesis, in several organs of the rat. Arch Toxicol 75, 685694.Google Scholar
395Ip, C (1984) Comparative effects of antioxidants on enzymes involved in glutathione metabolism. Life Sci 34, 25012506.Google Scholar
396Eaton, DL & Hamel, DM (1994) Increase in gamma-glutamylcysteine synthetase activity as a mechanism for butylated hydroxyanisole-mediated elevation of hepatic glutathione. Toxicol Appl Pharmacol 126, 145149.Google Scholar
397Younes, M (2004) Freie Radikale und reaktive Sauerstoffspezies. In Lehrbuch der Toxikologie, pp. 117130 [Marquart, H and Schäfer, S, editors]. Stuttgart: Wissenschaftliche Verlagsgesellschaft mbH Stuttgart.Google Scholar
398Boyne, AF & Ellman, GL (1972) A methodology for analysis of tissue sulfhydryl components. Anal Biochem 46, 639653.Google Scholar
399Alvarez, P, Alvarado, C, Mathieu, F, Jimenez, L & De la Fuente, M (2006) Diet supplementation for 5 weeks with polyphenol-rich cereals improves several functions and the redox state of mouse leucocytes. Eur J Nutr 45, 428438.Google Scholar
400Rebrin, I, Zicker, S, Wedekind, KJ, Paetau-Robinson, I, Packer, L & Sohal, RS (2005) Effect of antioxidant-enriched diets on glutathione redox status in tissue homogenates and mitochondria of the senescence-accelerated mouse. Free Radic Biol Med 39, 549557.Google Scholar
401Lii, CK, Ko, YJ, Chiang, MT, Sung, WC & Chen, HW (1998) Effect of dietary vitamin E on antioxidant status and antioxidant enzyme activities in Sprague–Dawley rats. Nutr Cancer 32, 95100.Google Scholar
402Landi, S (2000) Mammalian class theta GST and differential susceptibility to carcinogens: a review. Mutat Res 463, 247283.Google Scholar
403Tew, KD & Ronai, Z (1999) GST function in drug and stress response. Drug Resist Updat 2, 143147.Google Scholar
404Soini, Y, Kaarteenaho-Wiik, R, Paakko, P & Kinnula, V (2003) Expression of antioxidant enzymes in bronchial metaplastic and dysplastic epithelium. Lung Cancer 39, 1522.Google Scholar
405Wenzel, U, Herzog, A, Kuntz, S & Daniel, H (2004) Protein expression profiling identifies molecular targets of quercetin as a major dietary flavonoid in human colon cancer cells. Proteomics 4, 21602174.Google Scholar
406Surh, YJ (2003) Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer 3, 768780.Google Scholar
407Favata, MF, Horiuchi, KY, Manos, EJ, Daulerio, AJ, Stradley, DA, Feeser, WS, Van Dyk, DE, Pitts, WJ, Earl, RA, Hobbs, F, Copeland, RA, Magolda, RL, Scherle, PA & Trzaskos, JM (1998) Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem 273, 1862318632.Google Scholar
408Fukazawa, H & Uehara, Y (2000) U0126 reverses Ki-ras-mediated transformation by blocking both mitogen-activated protein kinase and p70 S6 kinase pathways. Cancer Res 60, 21042107.Google Scholar
409Bronstein, I, Fortin, J, Stanley, PE, Stewart, GS & Kricka, LJ (1994) Chemiluminescent and bioluminescent reporter gene assays. Anal Biochem 219, 169181.Google Scholar
410Mann, DA (2002) The NFkappaB luciferase mouse: a new tool for real time measurement of NFkappaB activation in the whole animal. Gut 51, 769770.Google Scholar
411Lai, C, Jiang, X & Li, X (2006) Development of luciferase reporter-based cell assays. Assay Drug Dev Technol 4, 307315.Google Scholar
412Sarkar, FH, Adsule, S, Li, Y & Padhye, S (2007) Back to the future: COX-2 inhibitors for chemoprevention and cancer therapy. Mini Rev Med Chem 7, 599608.Google Scholar
413Kulmacz, RJ & Lands, WEM (1987). Cyclo-oxygenase: measurement, purification and properties. In Prostaglandins and Related Substances: A Practical Approach, pp. 209227 [Benedetto, C, McDonald-Gibson, RG and Nigam, S et al. , editors]. Washington, DC: IRL Press.Google Scholar
414Pradelles, P, Grassi, J & Maclouf, J (1990) Enzyme immunoassays of eicosanoids using acetylcholinesterase. Methods Enzymol 187, 2434.Google Scholar
415Surh, YJ, Chun, KS, Cha, HH, Han, SS, Keum, YS, Park, KK & Lee, SS (2001) Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat Res 480–481, 243268.Google Scholar
416Gaboriau, F, Havouis, R, Groussard, K, Moulinoux, JP & Lescoat, G (2005) Measurement of ornithine decarboxylase activity in cell extracts using mass spectrometry detection of dansylated putrescine. Anal Biochem 341, 385387.Google Scholar
417Pegg, AE (2006) Regulation of ornithine decarboxylase. J Biol Chem 281, 1452914532.Google Scholar
418Hunt, NH & Fragonas, JC (1992) Effects of anti-oxidants on ornithine decarboxylase in mitogenically-activated T lymphocytes. Biochim Biophys Acta 1133, 261267.Google Scholar
419Friedman, J & Cerutti, P (1983) The induction of ornithine decarboxylase by phorbol 12-myristate 13-acetate or by serum is inhibited by antioxidants. Carcinogenesis 4, 14251427.Google Scholar
420Russell, D & Snyder, SH (1968) Amine synthesis in rapidly growing tissues: ornithine decarboxylase activity in regenerating rat liver, chick embryo, and various tumors. Proc Natl Acad Sci U S A 60, 14201427.Google Scholar
421Djurhuus, R (1981) Ornithine decarboxylase (EC 4.1.1.1.17) assay based upon the retention of putrescine by a strong cation-exchange paper. Anal Biochem 113, 352355.Google Scholar
422Seely, JE, Poso, H & Pegg, AE (1983) Labeling and quantitation of ornithine decarboxylase protein by reaction with alpha-[5-14C]difluoromethylornithine. Methods Enzymol 94, 206209.Google Scholar
423Seely, JE & Pegg, AE (1983) Changes in mouse kidney ornithine decarboxylase activity are brought about by changes in the amount of enzyme protein as measured by radioimmunoassay. J Biol Chem 258, 24962500.Google Scholar
424Wang, Y & Bachrach, U (2000) A luminescence-based test for determining ornithine decarboxylase activity. Anal Biochem 287, 299302.Google Scholar
425Ordovas, JM & Mooser, V (2004) Nutrigenomics and nutrigenetics. Curr Opin Lipidol 15, 101108.Google Scholar
426Ordovas, JM & Corella, D (2004) Nutritional genomics. Annu Rev Genomics Hum Genet 5, 71118.Google Scholar
427Kussmann, M, Affolter, M & Fay, LB (2005) Proteomics in nutrition and health. Comb Chem High Throughput Screen 8, 679696.Google Scholar
428Gavaghan, CL, Holmes, E, Lenz, E, Wilson, ID & Nicholson, JK (2000) An NMR-based metabonomic approach to investigate the biochemical consequences of genetic strain differences: application to the C57BL10J and Alpk:ApfCD mouse. FEBS Lett 484, 169174.Google Scholar
429Glassbrook, N & Ryals, J (2001) A systematic approach to biochemical profiling. Curr Opin Plant Biol 4, 186190.Google Scholar
430Scalbert, A, Milenkovic, D, Llorach, R, Manach, C & Leroux, C (2007) Nutrigenomics: techniques and applications. In Energy and protein metabolism and nutrition. EAAP publication No. 124, pp. 259276 [Ortigues-Marty, I, editor]. Wageningen: Wageningen Academic Publishers.Google Scholar
431Ovesná, J, Slabý, O, Toussaint, O, Kodíček, M, Maršík, P, Pouchová, V & Vaněk, T (2008) High throughput ‘omics’ approaches to assess the effects of phytochemicals in human health studies. Br J Nutr. 99, E-Suppl.1, ES127ES134..Google Scholar
432Stella, C, Beckwith-Hall, B, Cloarec, O, Holmes, E, Lindon, JC, Powell, J, van der Ouderaa, F, Bingham, S, Cross, AJ & Nicholson, JK (2006) Susceptibility of human metabolic phenotypes to dietary modulation. J Proteome Res 5, 27802788.Google Scholar
433Wang, Y, Tang, H, Nicholson, JK, Hylands, PJ, Sampson, J & Holmes, E (2005) A metabonomic strategy for the detection of the metabolic effects of chamomile (Matricaria recutita L.) ingestion. J Agric Food Chem 53, 191196.Google Scholar
434Van Dorsten, FA, Daykin, CA, Mulder, TP & Van Duynhoven, JP (2006) Metabonomics approach to determine metabolic differences between green tea and black tea consumption. J Agric Food Chem 54, 69296938.Google Scholar
435Solanky, KS, Bailey, NJ, Beckwith-Hall, BM, Davis, A, Bingham, S, Holmes, E, Nicholson, JK & Cassidy, A (2003) Application of biofluid 1H nuclear magnetic resonance-based metabonomic techniques for the analysis of the biochemical effects of dietary isoflavones on human plasma profile. Anal Biochem 323, 197204.Google Scholar
436Ito, H, Gonthiera, MP, Manach, C, Morand, C, Mennen, L, Remesy, C & Scalbert, A (2004) High-throughput profiling of dietary polyphenols and their metabolites by HPLC–ESI–MS–MS in human urine. Biofactors 22, 241243.Google Scholar
437Ito, H, Gonthier, MP, Manach, C, Morand, C, Mennen, L, Remesy, C & Scalbert, A (2005) Polyphenol levels in human urine after intake of six different polyphenol-rich beverages. Br J Nutr 94, 500509.Google Scholar
438Fardet, A, Canlet, C, Gottardi, G, Lyan, B, Llorach, R, Remesy, C, Mazur, A, Paris, A & Scalbert, A (2007) Whole-grain and refined wheat flours show distinct metabolic profiles in rats as assessed by a 1H NMR-based metabonomic approach. J Nutr 137, 923929.Google Scholar
439Mohn, KL, Melby, AE, Tewari, DS, Laz, TM & Taub, R (1991) The gene encoding rat insulinlike growth factor-binding protein 1 is rapidly and highly induced in regenerating liver. Mol Cell Biol 11, 13931401.Google Scholar
440DeRisi, J, Penland, L, Brown, PO, Bittner, ML, Meltzer, PS, Ray, M, Chen, Y, Su, YA & Trent, JM (1996) Use of a cDNA microarray to analyse gene expression patterns in human cancer. Nat Genet 14, 457460.Google Scholar
441Rothstein, JL, Johnson, D, DeLoia, JA, Skowronski, J, Solter, D & Knowles, B (1992) Gene expression during preimplantation mouse development. Genes Dev 6, 11901201.Google Scholar
442Friedman, J & Weissman, I (1991) Two cytoplasmic candidates for immunophilin action are revealed by affinity for a new cyclophilin: one in the presence and one in the absence of CsA. Cell 66, 799806.Google Scholar
443Liang, P & Pardee, AB (2003) Analysing differential gene expression in cancer. Nat Rev Cancer 3, 869876.Google Scholar
444Lisitsyn, N & Wigler, M (1993) Cloning the differences between two complex genomes. Science 259, 946951.Google Scholar
445Liang, P & Pardee, AB (1992) Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science 257, 967971.Google Scholar
446Velculescu, VE, Zhang, L, Vogelstein, B & Kinzler, KW (1995) Serial analysis of gene expression. Science 270, 484487.Google Scholar
447Brown, PO & Botstein, D (1999) Exploring the new world of the genome with DNA microarrays. Nat Genet 21, 3337.Google Scholar
448Duggan, DJ, Bittner, M, Chen, Y, Meltzer, P & Trent, JM (1999) Expression profiling using cDNA microarrays. Nat Genet 21, 1014.Google Scholar
449Adams, MD, Kelley, JM, Gocayne, JD, Dubnick, M, Polymeropoulos, MH, Xiao, H, Merril, CR, Wu, A, Olde, B, Moreno, RF, et al. (1991) Complementary DNA sequencing: expressed sequence tags and human genome project. Science 252, 16511656.Google Scholar
450Greely, HT (2001) Human genome diversity: what about the other human genome project? Nat Rev Genet 2, 222227.Google Scholar
451Mikulits, W, Pradet-Balade, B, Habermann, B, Beug, H, Garcia-Sanz, JA & Mullner, EW (2000) Isolation of translationally controlled mRNAs by differential screening. FASEB J 14, 16411652.Google Scholar
452Schena, M, Heller, RA, Theriault, TP, Konrad, K, Lachenmeier, E & Davis, RW (1998) Microarrays: biotechnology's discovery platform for functional genomics. Trends Biotechnol 16, 301306.Google Scholar
453Pradet-Balade, B, Boulme, F, Beug, H, Mullner, EW & Garcia-Sanz, JA (2001) Translation control: bridging the gap between genomics and proteomics? Trends Biochem Sci 26, 225229.Google Scholar
454Waerner, T, Alacakaptan, M, Tamir, I, Oberauer, R, Gal, A, Brabletz, T, Schreiber, M, Jechlinger, M & Beug, H (2006) ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell 10, 227239.Google Scholar
455Petz, M, Kozina, D, Huber, H, Siwiec, T, Seipelt, J, Sommergruber, W & Mikulits, W (2007) The leader region of laminin B1 mRNA confers cap-independent translation. Nucleic Acids Res 35, 24732482.Google Scholar
456Bustin, SA (2000) Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays. J Mol Endocrinol 25, 169193.Google Scholar
457Kunsch, C & Medford, RM (1999) Oxidative stress as a regulator of gene expression in the vasculature. Circ Res 85, 753766.Google Scholar
458MacDonald, J, Galley, HF & Webster, NR (2003) Oxidative stress and gene expression in sepsis. Br J Anaesth 90, 221232.Google Scholar
459Finkel, T & Holbrook, NJ (2000) Oxidants, oxidative stress and the biology of ageing. Nature 408, 239247.Google Scholar
460Scherf, U, Ross, DT, Waltham, M, Smith, LH, Lee, JK, Tanabe, L, Kohn, KW, Reinhold, WC, Myers, TG, Andrews, DT, Scudiero, DA, Eisen, MB, Sausville, EA, Pommier, Y, Botstein, D, Brown, PO & Weinstein, JN (2000) A gene expression database for the molecular pharmacology of cancer. Nat Genet 24, 236244.Google Scholar
461Efferth, T & Oesch, F (2004) Oxidative stress response of tumor cells: microarray-based comparison between artemisinins and anthracyclines. Biochem Pharmacol 68, 310.Google Scholar
462Murray, JI, Whitfield, ML, Trinklein, ND, Myers, RM, Brown, PO & Botstein, D (2004) Diverse and specific gene expression responses to stresses in cultured human cells. Mol Biol Cell 15, 23612374.Google Scholar
463Chuang, YY, Chen, Y, Gadisetti, V, Chandramouli, VR, Cook, JA, Coffin, D, Tsai, MH, DeGraff, W, Yan, H, Zhao, S, Russo, A, Liu, ET & Mitchell, JB (2002) Gene expression after treatment with hydrogen peroxide, menadione, or t-butyl hydroperoxide in breast cancer cells. Cancer Res 62, 62466254.Google Scholar
464Yoneda, K, Chang, MM, Chmiel, K, Chen, Y & Wu, R (2003) Application of high-density DNA microarray to study smoke- and hydrogen peroxide-induced injury and repair in human bronchial epithelial cells. J Am Soc Nephrol 14, S284S289.Google Scholar
465Morgan, KT, Ni, H, Brown, HR, Yoon, L, Qualls, CW Jr, Crosby, LM, Reynolds, R, Gaskill, B, Anderson, SP, Kepler, TB, Brainard, T, Liv, N, Easton, M, Merrill, C, Creech, D, Sprenger, D, Conner, G, Johnson, PR, Fox, T, Sartor, M, Richard, E, Kuruvilla, S, Casey, W & Benavides, G (2002) Application of cDNA microarray technology to in vitro toxicology and the selection of genes for a real-time RT-PCR-based screen for oxidative stress in Hep-G2 cells. Toxicol Pathol 30, 435451.Google Scholar
466Amundson, SA, Bittner, M, Chen, Y, Trent, J, Meltzer, P & Fornace, AJ Jr (1999) Fluorescent cDNA microarray hybridization reveals complexity and heterogeneity of cellular genotoxic stress responses. Oncogene 18, 36663672.Google Scholar
467Li, J, Lee, JM & Johnson, JA (2002) Microarray analysis reveals an antioxidant responsive element-driven gene set involved in conferring protection from an oxidative stress-induced apoptosis in IMR-32 cells. J Biol Chem 277, 388394.Google Scholar
468Bae, I, Fan, S, Meng, Q, Rih, JK, Kim, HJ, Kang, HJ, Xu, J, Goldberg, ID, Jaiswal, AK & Rosen, EM (2004) BRCA1 induces antioxidant gene expression and resistance to oxidative stress. Cancer Res 64, 78937909.Google Scholar
469Amundson, SA, Do, KT, Vinikoor, L, Koch-Paiz, CA, Bittner, ML, Trent, JM, Meltzer, P & Fornace, AJ Jr (2005) Stress-specific signatures: expression profiling of p53 wild-type and -null human cells. Oncogene 24, 45724579.Google Scholar
470Li, Z, Khaletskiy, A, Wang, J, Wong, JY, Oberley, LW & Li, JJ (2001) Genes regulated in human breast cancer cells overexpressing manganese-containing superoxide dismutase. Free Radic Biol Med 30, 260267.Google Scholar
471Kirby, J, Menzies, FM, Cookson, MR, Bushby, K & Shaw, PJ (2002) Differential gene expression in a cell culture model of SOD1-related familial motor neurone disease. Hum Mol Genet 11, 20612075.Google Scholar
472McMillian, M, Nie, A, Parker, JB, Leone, A, Kemmerer, M, Bryant, S, Herlich, J, Yieh, L, Bittner, A, Liu, X, Wan, J, Johnson, MD & Lord, P (2005) Drug-induced oxidative stress in rat liver from a toxicogenomics perspective. Toxicol Appl Pharmacol 207, 171178.Google Scholar
473McMillian, M, Nie, AY, Parker, JB, Leone, A, Bryant, S, Kemmerer, M, Herlich, J, Liu, Y, Yieh, L, Bittner, A, Liu, X, Wan, J & Johnson, MD (2004) A gene expression signature for oxidant stress/reactive metabolites in rat liver. Biochem Pharmacol 68, 22492261.Google Scholar
474Yoshihara, T, Ishigaki, S, Yamamoto, M, Liang, Y, Niwa, J, Takeuchi, H, Doyu, M & Sobue, G (2002) Differential expression of inflammation- and apoptosis-related genes in spinal cords of a mutant SOD1 transgenic mouse model of familial amyotrophic lateral sclerosis. J Neurochem 80, 158167.Google Scholar
475Thimmulappa, RK, Scollick, C, Traore, K, Yates, M, Trush, MA, Liby, KT, Sporn, MB, Yamamoto, M, Kensler, TW & Biswal, S (2006) Nrf2-dependent protection from LPS induced inflammatory response and mortality by CDDO-imidazolide. Biochem Biophys Res Commun 351, 883889.Google Scholar
476Lee, CK, Allison, DB, Brand, J, Weindruch, R & Prolla, TA (2002) Transcriptional profiles associated with aging and middle age-onset caloric restriction in mouse hearts. Proc Natl Acad Sci U S A 99, 1498814993.Google Scholar
477Lee, CK, Weindruch, R & Prolla, TA (2000) Gene-expression profile of the ageing brain in mice. Nat Genet 25, 294297.Google Scholar
478Lee, CK, Klopp, RG, Weindruch, R & Prolla, TA (1999) Gene expression profile of aging and its retardation by caloric restriction. Science 285, 13901393.Google Scholar
479Edwards, MG, Sarkar, D, Klopp, R, Morrow, JD, Weindruch, R & Prolla, TA (2003) Age-related impairment of the transcriptional responses to oxidative stress in the mouse heart. Physiol Genomics 13, 119127.Google Scholar
480Wang, Z, Neuburg, D, Li, C, Su, L, Kim, JY, Chen, JC & Christiani, DC (2005) Global gene expression profiling in whole-blood samples from individuals exposed to metal fumes. Environ Health Perspect 113, 233241.Google Scholar
481Kim, YH, Lim, DS, Lee, JH, Shim, WJ, Ro, YM, Park, GH, Becker, KG, Cho-Chung, YS & Kim, MK (2003) Gene expression profiling of oxidative stress on atrial fibrillation in humans. Exp Mol Med 35, 336349.Google Scholar
482Heller, RA, Schena, M, Chai, A, Shalon, D, Bedilion, T, Gilmore, J, Woolley, DE & Davis, RW (1997) Discovery and analysis of inflammatory disease-related genes using cDNA microarrays. Proc Natl Acad Sci U S A 94, 21502155.Google Scholar
483Jison, ML, Munson, PJ, Barb, JJ, Suffredini, AF, Talwar, S, Logun, C, Raghavachari, N, Beigel, JH, Shelhamer, JH, Danner, RL & Gladwin, MT (2004) Blood mononuclear cell gene expression profiles characterize the oxidant, hemolytic, and inflammatory stress of sickle cell disease. Blood 104, 270280.Google Scholar
484Weigel, AL, Handa, JT & Hjelmeland, LM (2002) Microarray analysis of H2O2-, HNE-, or tBH-treated ARPE-19 cells. Free Radic Biol Med 33, 14191432.Google Scholar
485Suzuki, T, Spitz, DR, Gandhi, P, Lin, HY & Crawford, DR (2002) Mammalian resistance to oxidative stress: a comparative analysis. Gene Expr 10, 179191.Google Scholar
486Weindruch, R, Kayo, T, Lee, CK & Prolla, TA (2002) Effects of caloric restriction on gene expression. In Nutrition and Ageing: Nestle Nutr Workshop Ser Clin Perform Programme, pp. 1732 [Rosenberg, I and Sastre, A, editors]. Basel/Vevey: Nestec LTD/S. Karger AG.Google Scholar
487Yoneda, K, Peck, K, Chang, MM, Chmiel, K, Sher, YP, Chen, J, Yang, PC, Chen, Y & Wu, R (2001) Development of high-density DNA microarray membrane for profiling smoke- and hydrogen peroxide-induced genes in a human bronchial epithelial cell line. Am J Respir Crit Care Med 164, S85S89.Google Scholar
488Lee, JM, Li, J, Johnson, DA, Stein, TD, Kraft, AD, Calkins, MJ, Jakel, RJ & Johnson, JA (2005) Nrf2, a multi-organ protector? FASEB J 19, 10611066.Google Scholar
489Ivyna Bong, PN, Patricia, L, Pauline, B, Edmund, SU & Zubaidah, Z (2006) Quantitative analysis of the expression of p53 gene in colorectal carcinoma by using real-time PCR. Trop Biomed 23, 5359.Google Scholar
490De Flora, S, Izzotti, A, D'Agostini, F, Bennicelli, C, You, M, Lubet, RA & Balansky, RM (2005) Induction and modulation of lung tumors: genomic and transcriptional alterations in cigarette smoke-exposed mice. Exp Lung Res 31, 1935.Google Scholar
491Stewart, SL, Querec, TD, Ochman, AR, Gruver, BN, Bao, R, Babb, JS, Wong, TS, Koutroukides, T, Pinnola, AD, Klein-Szanto, A, Hamilton, TC & Patriotis, C (2004) Characterization of a carcinogenesis rat model of ovarian preneoplasia and neoplasia. Cancer Res 64, 81778183.Google Scholar
492Jarvinen, AK, Hautaniemi, S, Edgren, H, Auvinen, P, Saarela, J, Kallioniemi, OP & Monni, O (2004) Are data from different gene expression microarray platforms comparable? Genomics 83, 11641168.Google Scholar
493van der Spek, PJ, Kremer, A, Murry, L & Walker, MG (2003) Are gene expression microarray analyses reliable? A review of studies of retinoic acid responsive genes. Genomics Proteomics Bioinformatics 1, 914.Google Scholar
494Shippy, R, Sendera, TJ, Lockner, R, Palaniappan, C, Kaysser-Kranich, T, Watts, G & Alsobrook, J (2004) Performance evaluation of commercial short-oligonucleotide microarrays and the impact of noise in making cross-platform correlations. BMC Genomics 5, 61.Google Scholar
495Brazma, A, Hingamp, P, Quackenbush, J, Sherlock, G, Spellman, P, Stoeckert, C, Aach, J, Ansorge, W, Ball, CA, Causton, HC, Gaasterland, T, Glenisson, P, Holstege, FC, Kim, IF, Markowitz, V, Matese, JC, Parkinson, H, Robinson, A, Sarkans, U, Schulze-Kremer, S, Stewart, J, Taylor, R, Vilo, J & Vingron, M (2001) Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet 29, 365371.Google Scholar
496Irizarry, RA, Warren, D, Spencer, F, Kim, IF, Biswal, S, Frank, BC, Gabrielson, E, Garcia, JG, Geoghegan, J, Germino, G, Griffin, C, Hilmer, SC, Hoffman, E, Jedlicka, AE, Kawasaki, E, Martinez-Murillo, F, Morsberger, L, Lee, H, Petersen, D, Quackenbush, J, Scott, A, Wilson, M, Yang, Y, Ye, SQ & Yu, W (2005) Multiple-laboratory comparison of microarray platforms. Nat Methods 2, 345350.Google Scholar
497Larkin, JE, Frank, BC, Gavras, H, Sultana, R & Quackenbush, J (2005) Independence and reproducibility across microarray platforms. Nat Methods 2, 337344.Google Scholar
498Bammler, T, Beyer, RP, Bhattacharya, S, Boorman, GA, Boyles, A, Bradford, BU, Bumgarner, RE, Bushel, PR, Chaturvedi, K, Choi, D, Cunningham, ML, Deng, S, Dressman, HK, Fannin, RD, Farin, FM, Freedman, JH, Fry, RC, Harper, A, Humble, MC, Hurban, P, Kavanagh, TJ, Kaufmann, WK, Kerr, KF, Jing, L, Lapidus, JA, Lasarev, MR, Li, J, Li, YJ, Lobenhofer, EK, Lu, X, Malek, RL, Milton, S, Nagalla, SR, O'Malley, JP, Palmer, VS, Pattee, P, Paules, RS, Perou, CM, Phillips, K, Qin, LX, Qiu, Y, Quigley, SD, Rodland, M, Rusyn, I, Samson, LD, Schwartz, DA, Shi, Y, Shin, JL, Sieber, SO, Slifer, S, Speer, MC, Spencer, PS, Sproles, DI, Swenberg, JA, Suk, WA, Sullivan, RC, Tian, R, Tennant, RW, Todd, SA, Tucker, CJ, Van Houten, B, Weis, BK, Xuan, S & Zarbl, H (2005) Standardizing global gene expression analysis between laboratories and across platforms. Nat Methods 2, 351356.Google Scholar
499Mariappan, D, Winkler, J, Parthiban, V, Doss, MX, Hescheler, J & Sachinidis, A (2006) Dietary small molecules and large-scale gene expression studies: an experimental approach for understanding their beneficial effects on the development of malignant and non-malignant proliferative diseases. Curr Med Chem 13, 14811489.Google Scholar
500Elliott, R (2005) Mechanisms of genomic and non-genomic actions of carotenoids. Biochim Biophys Acta 1740, 147154.Google Scholar
501Majewicz, J, Rimbach, G, Proteggente, AR, Lodge, JK, Kraemer, K & Minihane, AM (2005) Dietary vitamin C down-regulates inflammatory gene expression in apoE4 smokers. Biochem Biophys Res Commun 338, 951955.Google Scholar
502Hofmann, T, Liegibel, U, Winterhalter, P, Bub, A, Rechkemmer, G & Pool-Zobel, BL (2006) Intervention with polyphenol-rich fruit juices results in an elevation of glutathione S-transferase P1 (hGSTP1) protein expression in human leucocytes of healthy volunteers. Mol Nutr Food Res 50, 11911200.Google Scholar
503Cotgreave, IA, Moldeus, P & Orrenius, S (1988) Host biochemical defense mechanisms against prooxidants. Annu Rev Pharmacol Toxicol 28, 189212.Google Scholar
504Hebbar, V, Shen, G, Hu, R, Kim, BR, Chen, C, Korytko, PJ, Crowell, JA, Levine, BS & Kong, AN (2005) Toxicogenomics of resveratrol in rat liver. Life Sci 76, 22992314.Google Scholar
505Funk, JL, Frye, JB, Oyarzo, JN, Kuscuoglu, N, Wilson, J, McCaffrey, G, Stafford, G, Chen, G, Lantz, RC, Jolad, SD, Solyom, AM, Kiela, PR & Timmermann, BN (2006) Efficacy and mechanism of action of turmeric supplements in the treatment of experimental arthritis. Arthritis Rheum 54, 34523464.Google Scholar
506Selman, C, McLaren, JS, Meyer, C, Duncan, JS, Redman, P, Collins, AR, Duthie, GG & Speakman, JR (2006) Life-long vitamin C supplementation in combination with cold exposure does not affect oxidative damage or lifespan in mice, but decreases expression of antioxidant protection genes. Mech Ageing Dev 127, 897904.Google Scholar
507Belloir, C, Singh, V, Daurat, C, Siess, MH & Le Bon, AM (2006) Protective effects of garlic sulfur compounds against DNA damage induced by direct- and indirect-acting genotoxic agents in HepG2 cells. Food Chem Toxicol 44, 827834.Google Scholar
508Schumperli, D (1988) Multilevel regulation of replication-dependent histone genes. Trends Genet 4, 187191.Google Scholar
509Nordhoff, E, Egelhofer, V, Giavalisco, P, Eickhoff, H, Horn, M, Przewieslik, T, Theiss, D, Schneider, U, Lehrach, H & Gobom, J (2001) Large-gel two-dimensional electrophoresis-matrix assisted laser desorption/ionization-time of flight-mass spectrometry: an analytical challenge for studying complex protein mixtures. Electrophoresis 22, 28442855.Google Scholar
510Gerner, C, Vejda, S, Gelbmann, D, Bayer, E, Gotzmann, J & Schulte-Hermann, R & Mikulits, W (2002) Concomitant determination of absolute values of cellular protein amounts, synthesis rates, and turnover rates by quantitative proteome profiling. Mol Cell Proteomics 1, 528537.Google Scholar
511Rabilloud, T (2002) Two-dimensional gel electrophoresis in proteomics: old, old fashioned, but it still climbs up the mountains. Proteomics 2, 310.Google Scholar
512Gras, R, Muller, M, Gasteiger, E, Gay, S, Binz, PA, Bienvenut, W, Hoogland, C, Sanchez, JC, Bairoch, A, Hochstrasser, DF & Appel, RD (1999) Improving protein identification from peptide mass fingerprinting through a parameterized multi-level scoring algorithm and an optimized peak detection. Electrophoresis 20, 35353550.Google Scholar
513Fountain, ST, Lee, H & Lubman, DM (1994) Ion fragmentation activated by matrix-assisted laser desorption/ionization in an ion-trap/reflectron time-of-flight device. Rapid Commun Mass Spectrom 8, 407416.Google Scholar
514Spickett, CM, Pitt, AR, Morrice, N & Kolch, W (2006) Proteomic analysis of phosphorylation, oxidation and nitrosylation in signal transduction. Biochim Biophys Acta 1764, 18231841.Google Scholar
515Wolters, DA, Washburn, MP & Yates, JR 3rd (2001) An automated multidimensional protein identification technology for shotgun proteomics. Anal Chem 73, 56835690.Google Scholar
516Bodnar, WM, Blackburn, RK, Krise, JM & Moseley, MA (2003) Exploiting the complementary nature of LC/MALDI/MS/MS and LC/ESI/MS/MS for increased proteome coverage. J Am Soc Mass Spectrom 14, 971979.Google Scholar
517Watarai, H, Inagaki, Y, Kubota, N, Fuju, K, Nagafune, J, Yamaguchi, Y & Kadoya, T (2000) Proteomic approach to the identification of cell membrane proteins. Electrophoresis 21, 460464.Google Scholar
518Zhu, K, Kim, J, Yoo, C, Miller, FR & Lubman, DM (2003) High sequence coverage of proteins isolated from liquid separations of breast cancer cells using capillary electrophoresis-time-of-flight MS and MALDI-TOF MS mapping. Anal Chem 75, 62096217.Google Scholar
519Manzanares, D, Rodriguez-Capote, K, Liu, S, Haines, T, Ramos, Y, Zhao, L, Doherty-Kirby, A, Lajoie, G & Possmayer, F (2007) Modification of tryptophan and methionine residues is implicated in the oxidative inactivation of surfactant protein B. Biochemistry 46, 56045615.Google Scholar
520Paron, I, D'Elia, A, D'Ambrosio, C, Scaloni, A, D'Aurizio, F, Prescott, A, Damante, G & Tell, G (2004) A proteomic approach to identify early molecular targets of oxidative stress in human epithelial lens cells. Biochem J 378, 929937.Google Scholar
521Rabilloud, T, Heller, M, Gasnier, F, Luche, S, Rey, C, Aebersold, R, Benahmed, M, Louisot, P & Lunardi, J (2002) Proteomics analysis of cellular response to oxidative stress. Evidence for in vivo overoxidation of peroxiredoxins at their active site. J Biol Chem 277, 1939619401.Google Scholar
522Koeck, T, Fu, X, Hazen, SL, Crabb, JW, Stuehr, DJ & Aulak, KS (2004) Rapid and selective oxygen-regulated protein tyrosine denitration and nitration in mitochondria. J Biol Chem 279, 2725727262.Google Scholar
523Brennan, JP, Wait, R, Begum, S, Bell, JR, Dunn, MJ & Eaton, P (2004) Detection and mapping of widespread intermolecular protein disulfide formation during cardiac oxidative stress using proteomics with diagonal electrophoresis. J Biol Chem 279, 4135241360.Google Scholar
524Fratelli, M, Demol, H, Puype, M, Casagrande, S, Eberini, I, Salmona, M, Bonetto, V, Mengozzi, M, Duffieux, F, Miclet, E, Bachi, A, Vandekerckhove, J, Gianazza, E & Ghezzi, P (2002) Identification by redox proteomics of glutathionylated proteins in oxidatively stressed human T lymphocytes. Proc Natl Acad Sci U S A 99, 35053510.Google Scholar
525Pocernich, CB, Poon, HF, Boyd-Kimball, D, Lynn, BC, Nath, A, Klein, JB & Butterfield, DA (2005) Proteomic analysis of oxidatively modified proteins induced by the mitochondrial toxin 3-nitropropionic acid in human astrocytes expressing the HIV protein tat. Brain Res Mol Brain Res 133, 299306.Google Scholar
526Stagsted, J, Bendixen, E & Andersen, HJ (2004) Identification of specific oxidatively modified proteins in chicken muscles using a combined immunologic and proteomic approach. J Agric Food Chem 52, 39673974.Google Scholar
527Godon, C, Lagniel, G, Lee, J, Buhler, JM, Kieffer, S, Perrot, M, Boucherie, H, Toledano, MB & Labarre, J (1998) The H2O2 stimulon in Saccharomyces cerevisiae. J Biol Chem 273, 2248022489.Google Scholar
528Mostertz, J, Scharf, C, Hecker, M & Homuth, G (2004) Transcriptome and proteome analysis of Bacillus subtilis gene expression in response to superoxide and peroxide stress. Microbiology 150, 497512.Google Scholar
529Chan, HL, Gharbi, S, Gaffney, PR, Cramer, R, Waterfield, MD & Timms, JF (2005) Proteomic analysis of redox- and ErbB2-dependent changes in mammary luminal epithelial cells using cysteine- and lysine-labelling two-dimensional difference gel electrophoresis. Proteomics 5, 29082926.Google Scholar
530Seong, JK, Kim, DK, Choi, KH, Oh, SH, Kim, KS, Lee, SS & Um, HD (2002) Proteomic analysis of the cellular proteins induced by adaptive concentrations of hydrogen peroxide in human U937 cells. Exp Mol Med 34, 374378.Google Scholar
531Keightley, JA, Shang, L & Kinter, M (2004) Proteomic analysis of oxidative stress-resistant cells: a specific role for aldose reductase overexpression in cytoprotection. Mol Cell Proteomics 3, 167175.Google Scholar
532Ding, Q, Dimayuga, E & Keller, JN (2007) Oxidative damage, protein synthesis, and protein degradation in Alzheimer's disease. Curr Alzheimer Res 4, 7379.Google Scholar
533Sultana, R, Perluigi, M & Butterfield, DA (2006) Protein oxidation and lipid peroxidation in brain of subjects with Alzheimer's disease: insights into mechanism of neurodegeneration from redox proteomics. Antioxid Redox Signal 8, 20212037.Google Scholar
534Grimsrud, PA, Picklo, MJ Sr, Griffin, TJ & Bernlohr, DA (2007) Carbonylation of adipose proteins in obesity and insulin resistance: identification of adipocyte fatty acid-binding protein as a cellular target of 4-hydroxynonenal. Mol Cell Proteomics 6, 624637.Google Scholar
535Ghosh, S, Janocha, AJ, Aronica, MA, Swaidani, S, Comhair, SA, Xu, W, Zheng, L, Kaveti, S, Kinter, M, Hazen, SL & Erzurum, SC (2006) Nitrotyrosine proteome survey in asthma identifies oxidative mechanism of catalase inactivation. J Immunol 176, 55875597.Google Scholar
536Dalle-Donne, I, Scaloni, A, Giustarini, D, Cavarra, E, Tell, G, Lungarella, G, Colombo, R, Rossi, R & Milzani, A (2005) Proteins as biomarkers of oxidative/nitrosative stress in diseases: the contribution of redox proteomics. Mass Spectrom Rev 24, 5599.Google Scholar
537Go, VL, Butrum, RR & Wong, DA (2003) Diet, nutrition, and cancer prevention: the postgenomic era. J Nutr 133, 3830S3836S.Google Scholar
538Davis, CD & Milner, J (2004) Frontiers in nutrigenomics, proteomics, metabolomics and cancer prevention. Mutat Res 551, 5164.Google Scholar
539Guengerich, FP (2001) Functional genomics and proteomics applied to the study of nutritional metabolism. Nutr Rev 59, 259263.Google Scholar
540Wang, J, Li, D, Dangott, LJ & Wu, G (2006) Proteomics and its role in nutrition research. J Nutr 136, 17591762.Google Scholar
541Trujillo, E, Davis, C & Milner, J (2006) Nutrigenomics, proteomics, metabolomics, and the practice of dietetics. J Am Diet Assoc 106, 403413.Google Scholar
542Keusch, GT (2006) What do -omics mean for the science and policy of the nutritional sciences? Am J Clin Nutr 83, 520S522S.Google Scholar
543Kim, H (2005) New nutrition, proteomics, and how both can enhance studies in cancer prevention and therapy. J Nutr 135, 27152718.Google Scholar
544Fuchs, D, Winkelmann, I, Johnson, IT, Mariman, E, Wenzel, U & Daniel, H (2005) Proteomics in nutrition research: principles, technologies and applications. Br J Nutr 94, 302314.Google Scholar
545Kussmann, M & Affolter, M (2006) Proteomic methods in nutrition. Curr Opin Clin Nutr Metab Care 9, 575583.Google Scholar
546Opii, WO, Joshi, G, Head, E, Milgram, NW, Muggenburg, BA, Klein, JB, Pierce, WM, Cotman, CW & Butterfield, DA (2008) Proteomic identification of brain proteins in the canine model of human aging following a long-term treatment with antioxidants and a program of behavioral enrichment: relevance to Alzheimer's disease. Neurobiol Aging 29, 5170.Google Scholar
547Seifried, HE, Anderson, DE, Fisher, EI & Milner, JA (2007) A review of the interaction among dietary antioxidants and reactive oxygen species. J Nutr Biochem 18, 567579.Google Scholar
548Grubben, MJ, Nagengast, FM, Katan, MB & Peters, WH (2001) The glutathione biotransformation system and colorectal cancer risk in humans. Scand J Gastroenterol Suppl, 6876.Google Scholar
549Hoelzl, C, Bichler, J, Ferk, F, Ehrlich, V, Cavin, C, Simic, T, Edelbauer, L, Grasl-Kraupp, B & Knasmüller, S (2007) Antimutagenic and anticarcinogenic properties of coffee. In The Activity of Natural Compounds in Diseses and Therapy, pp. 127138 [Ďuračková, Z and Knasmüller, S, editors]. Bratislava: Slovak Academic Press.Google Scholar
550Li, Y & Prives, C (2007) Are interactions with p63 and p73 involved in mutant p53 gain of oncogenic function? Oncogene 26, 22202225.Google Scholar
551Levesque, AA & Eastman, A (2007) p53-based cancer therapies: is defective p53 the Achilles heel of the tumor? Carcinogenesis 28, 1320.Google Scholar
552Manach, C, Williamson, G, Morand, C, Scalbert, A & Remesy, C (2005) Bioavailability and bioefficacy of polyphenols in humans I. Review of 97 bioavailability studies. Am J Clin Nutr 81, 230S242S.Google Scholar
553Depeint, F, Gee, JM, Williamson, G & Johnson, IT (2002) Evidence for consistent patterns between flavonoid structures and cellular activities. Proc Nutr Soc 61, 97103.Google Scholar
554Depeint, F (2003) Dietary phytochemicals and their effect on colon cell proliferation. PhD thesis, Institute of Food Research.Google Scholar
555Nardini, M, Cirillo, E, Natella, F & Scaccini, C (2002) Absorption of phenolic acids in humans after coffee consumption. J Agric Food Chem 50, 57355741.Google Scholar
556Clifford, MN (2004) Diet-derived phenols in plasma and tissues and their implications for health. Planta Med 70, 11031114.Google Scholar
557Mitoma, J & Fukuda, M (2006) Expression of specific carbohydrates by transfection with carbohydrate modifying enzymes. Methods Enzymol 416, 293304.Google Scholar
558Hu, Z & Wells, PG (2004) Human interindividual variation in lymphocyte UDP-glucuronosyltransferases as a determinant of in vitro benzo[a]pyrene covalent binding and cytotoxicity. Toxicol Sci 78, 3240.Google Scholar
559Zhang, Y, Song, TT, Cunnick, JE, Murphy, PA & Hendrich, S (1999) Daidzein and genistein glucuronides in vitro are weakly estrogenic and activate human natural killer cells at nutritionally relevant concentrations. J Nutr 129, 399405.Google Scholar
560Spencer, JP, Schroeter, H, Crossthwaithe, AJ, Kuhnle, G, Williams, RJ & Rice-Evans, C (2001) Contrasting influences of glucuronidation and O-methylation of epicatechin on hydrogen peroxide-induced cell death in neurons and fibroblasts. Free Radic Biol Med 31, 11391146.Google Scholar
561Eckl, PM & Raffelsberger, I (1997) The primary rat hepatocyte micronucleus assay: general features. Mutat Res 392, 117124.Google Scholar
562Fahrig, R, Rupp, M, Steinkamp-Zucht, A & Bader, A (1998) Use of primary rat and human hepatocyte sandwich cultures for activation of indirect carcinogens: monitoring of DNA strand breaks and gene mutations in co-cultured cells. Toxicol In Vitro 12, 431444.Google Scholar
563Dogra, S, Doehmer, J, Glatt, H, Molders, H, Siegert, P, Friedberg, T, Seidel, A & Oesch, F (1990) Stable expression of rat cytochrome P-450IA1 cDNA in V79 Chinese hamster cells and their use in mutagenicity testing. Mol Pharmacol 37, 608613.Google Scholar
564Muckel, E, Frandsen, H & Glatt, HR (2002) Heterologous expression of human N-acetyltransferases 1 and 2 and sulfotransferase 1A1 in Salmonella typhimurium for mutagenicity testing of heterocyclic amines. Food Chem Toxicol 40, 10631068.Google Scholar
565Aeschbacher, HU, Wolleb, U, Loliger, J, Spadone, JC & Liardon, R (1989) Contribution of coffee aroma constituents to the mutagenicity of coffee. Food Chem Toxicol 27, 227232.Google Scholar
566Kumar, SS, Shankar, B & Sainis, KB (2004) Effect of chlorophyllin against oxidative stress in splenic lymphocytes in vitro and in vivo. Biochim Biophys Acta 1672, 100111.Google Scholar
567Su, CC, Chen, GW, Lin, JG, Wu, LT & Chung, JG (2006) Curcumin inhibits cell migration of human colon cancer colo 205 cells through the inhibition of nuclear factor kappa B/p65 and down-regulates cyclooxygenase-2 and matrix metalloproteinase-2 expressions. Anticancer Res 26, 12811288.Google Scholar
568Ogasawara, M, Matsunaga, T & Suzuki, H (2007) Differential effects of antioxidants on the in vitro invasion, growth and lung metastasis of murine colon cancer cells. Biol Pharm Bull 30, 200204.Google Scholar
569Heinonen, M (2007) Antioxidant activity and antimicrobial effect of berry phenolics – a Finnish perspective. Mol Nutr Food Res 51, 684691.Google Scholar
570Shahidi, F (2004) Food phenolics and cancer chmoprevention. In Funktional Foods, Aging and Degenerative Disesase, pp. 669680 [Remacle, C and Reusens, B, editors]. Cambridge, England: Woodhead Publishing Limited.Google Scholar
571Watzl, B & Leitzmann, C (2005) Bioaktive Substanzen in Lebensmitteln. Stuttgart: Hippokrates Verlag.Google Scholar
572Ferk, F, Chakraborty, A, Simic, T, Kundi, M & Knasmüller, S (2006) Antioxidant and free radical scavenging activities of sumac (Rhus coriaria) and identification of gallic acid as its active principle. In 12th Scientific Symposium of the Austrian Pharmacological Society (APHAR). Joint Meeting with the Austrian Society for Toxicology (ASTOX), p. 11 [Schulte-Herman, R, editor]. Vienna, Austria.Google Scholar
573Shahrzad, S, Aoyagi, K, Winter, A, Koyama, A & Bitsch, I (2001) Pharmacokinetics of gallic acid and its relative bioavailability from tea in healthy humans. J Nutr 131, 12071210.Google Scholar
574Ellinger, S, Ellinger, J & Stehle, P (2006) Tomatoes, tomato products and lycopene in the prevention and treatment of prostate cancer: do we have the evidence from intervention studies? Curr Opin Clin Nutr Metab Care 9, 722727.Google Scholar
575Domenici, FA, Vannucchi, MT, Jordao, AA Jr, Meirelles, MS & Vannucchi, H (2005) DNA oxidative damage in patients with dialysis treatment. Ren Fail 27, 689694.Google Scholar
576Beetstra, S, Thomas, P, Salisbury, C, Turner, J & Fenech, M (2005) Folic acid deficiency increases chromosomal instability, chromosome 21 aneuploidy and sensitivity to radiation-induced micronuclei. Mutat Res 578, 317326.Google Scholar
577Fenech, M (2005) The Genome Health Clinic and Genome Health Nutrigenomics concepts: diagnosis and nutritional treatment of genome and epigenome damage on an individual basis. Mutagenesis 20, 255269.Google Scholar
578Kazimirova, A, Barancokova, M, Krajcovicova-Kudlackova, M, Volkovova, K, Staruchova, M, Valachovicova, M, Paukova, V, Blazicek, P, Wsolova, L & Dusinska, M (2006) The relationship between micronuclei in human lymphocytes and selected micronutrients in vegetarians and non-vegetarians. Mutat Res 611, 6470.Google Scholar
579Fenech, M (2001) Recommended dietary allowances (RDAs) for genomic stability. Mutat Res 480-481, 5154.Google Scholar
580Katan, MB & de Roos, NM (2003) Public health. Toward evidence-based health claims for foods. Science 299, 206207.Google Scholar
581Sloan, AE (2002) The natural and organic foods marketplace. Food Technol 56, 32.Google Scholar
582Derby, BM & Levy, AS (2005) Working Paper: Effects of Strength of Science Disclaimers on the Communication Impact of Health Claims. DoSS US Food and Drug Administration, editor.Google Scholar
583Regulation (2006) (EC) No. 1924/2006 of the European Parliament and the Council of 20 December 2006 on Nutrition and Health Claims made on Foods. Official J Eur Union 404, L12/13L12/17.Google Scholar
584Nickelson, L (2005) Legal context of consumer research on health claims and other food. Public Meeting: Assessing Consumer Perceptions of Health Claims, Colleage Park, Marylandhttp://www.cfsan.fda.gov~dms/qhctran.html#legal.Google Scholar
585Zhan, CD, Sindhu, RK, Pang, J, Ehdaie, A & Vaziri, ND (2004) Superoxide dismutase, catalase and glutathione peroxidase in the spontaneously hypertensive rat kidney: effect of antioxidant-rich diet. J Hypertens 22, 20252033.Google Scholar
586McCord, JM & Fridovich, I (1969) Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J Biol Chem 244, 60496055.Google Scholar
587Woolliams, JA, Wiener, G, Anderson, PH & McMurray, CH (1983) Variation in the activities of glutathione peroxidase and superoxide dismutase and in the concentration of copper in the blood in various breed crosses of sheep. Res Vet Sci 34, 253256.Google Scholar
588Prohaska, JR (1983) Changes in tissue growth, concentrations of copper, iron, cytochrome oxidase and superoxide dismutase subsequent to dietary or genetic copper deficiency in mice. J Nutr 113, 20482058.Google Scholar
589Marklund, S & Marklund, G (1974) Involvement of the superoxide anion radical in the autoxidation of pyrogallol and a convenient assay for superoxide dismutase. Eur J Biochem 47, 469474.Google Scholar
590DiSilvestro, RA, Goodman, J, Dy, E & Lavalle, G (2005) Soy isoflavone supplementation elevates erythrocyte superoxide dismutase, but not plasma ceruloplasmin in postmenopausal breast cancer survivors. Breast Cancer Res Treat 89, 251255.Google Scholar
591Claiborne, A (1985) Catalase activity. In Handbook of Methods for Oxygen Free Radical Research, pp. 283284 [Greenwald, RA, editor]. Boca Raton: CRC Press.Google Scholar
592Beers, RF Jr & Sizer, IW (1952) A spectrophotometric method for measuring the breakdown of hydrogen peroxide by catalase. J Biol Chem 195, 133140.Google Scholar
593Aebi, H (1984) Catalase in vitro. Methods Enzymol 105, 121126.Google Scholar
594Price, VE, Sterling, WR, Tarantola, VA, Hartley, RW Jr & Rechcigl, M Jr (1962) The kinetics of catalase synthesis and destruction in vivo. J Biol Chem 237, 34683475.Google Scholar
595Paglia, DE & Valentine, WN (1967) Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase. J Lab Clin Med 70, 158169.Google Scholar
596Lawrence, RA & Burk, RF (1976) Glutathione peroxidase activity in selenium-deficient rat liver. Biochem Biophys Res Commun 71, 952958.Google Scholar
597Sazuka, Y, Tanizawa, H & Takino, Y (1989) Effect of adriamycin on the activities of superoxide dismutase, glutathione peroxidase and catalase in tissues of mice. Jpn J Cancer Res 80, 8994.Google Scholar
598Chow, S, Patel, H & Hedley, DW (2001) Measurement of MAP kinase activation by flow cytometry using phospho-specific antibodies to MEK and ERK: potential for pharmacodynamic monitoring of signal transduction inhibitors. Cytometry 46, 7278.Google Scholar
599Yao, P, Nussler, A, Liu, L, Hao, L, Song, F, Schirmeier, A & Nussler, N (2007) Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. J Hepatol 47, 253261.Google Scholar
600Woo, J, Iyer, S, Cornejo, MC, Mori, N, Gao, L, Sipos, I, Maines, M & Buelow, R (1998) Stress protein-induced immunosuppression: inhibition of cellular immune effector functions following overexpression of haem oxygenase (HSP 32). Transpl Immunol 6, 8493.Google Scholar
601Motterlini, R, Foresti, R, Bassi, R & Green, CJ (2000) Curcumin, an antioxidant and anti-inflammatory agent, induces heme oxygenase-1 and protects endothelial cells against oxidative stress. Free Radic Biol Med 28, 13031312.Google Scholar
602Benson, AM, Hunkeler, MJ & Talalay, P (1980) Increase of NAD(P)H:quinone reductase by dietary antioxidants: possible role in protection against carcinogenesis and toxicity. Proc Natl Acad Sci USA 77, 52165220.Google Scholar
603Nho, CW & Jeffery, E (2001) The synergistic upregulation of phase II detoxification enzymes by glucosinolate breakdown products in cruciferous vegetables. Toxicol Appl Pharmacol 174, 146152.Google Scholar
604De Long, MJ, Prochaska, HJ & Talalay, P (1986) Induction of NAD(P)H:quinone reductase in murine hepatoma cells by phenolic antioxidants, azo dyes, and other chemoprotectors: a model system for the study of anticarcinogens. Proc Natl Acad Sci U S A 83, 787791.Google Scholar
605Talalay, P, Dinkova-Kostova, AT & Holtzclaw, WD (2003) Importance of phase 2 gene regulation in protection against electrophile and reactive oxygen toxicity and carcinogenesis. Adv Enzyme Regul 43, 121134.Google Scholar
606Fahey, JW, Zhang, Y & Talalay, P (1997) Broccoli sprouts: an exceptionally rich source of inducers of enzymes that protect against chemical carcinogens. Proc Natl Acad Sci U S A 94, 1036710372.Google Scholar
607Rafi, MM & Shafaie, Y (2007) Dietary lutein modulates inducible nitric oxide synthase (iNOS) gene and protein expression in mouse macrophage cells (RAW 264.7). Mol Nutr Food Res 51, 333340.Google Scholar
608Calvisi, DF, Ladu, S, Hironaka, K, Factor, VM & Thorgeirsson, SS (2004) Vitamin E down-modulates iNOS and NADPH oxidase in c-Myc/TGF-alpha transgenic mouse model of liver cancer. J Hepatol 41, 815822.Google Scholar
609Hevel, JM & Marletta, MA (1994) Nitric-oxide synthase assays. Methods Enzymol 233, 250258.Google Scholar
610Chan, MM, Huang, HI, Fenton, MR & Fong, D (1998) In vivo inhibition of nitric oxide synthase gene expression by curcumin, a cancer preventive natural product with anti-inflammatory properties. Biochem Pharmacol 55, 19551962.Google Scholar
611Keum, YS, Han, YH, Liew, C, Kim, JH, Xu, C, Yuan, X, Shakarjian, MP, Chong, S & Kong, AN (2006) Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. Pharm Res 23, 25862594.Google Scholar
612Hou, DX, Fukuda, M, Fujii, M & Fuke, Y (2000) Transcriptional regulation of nicotinamide adenine dinucleotide phosphate: quinone oxidoreductase in murine hepatoma cells by 6-(methylsufinyl)hexyl isothiocyanate, an active principle of wasabi (Eutrema wasabi Maxim). Cancer Lett 161, 195200.Google Scholar
613Hollander, PM & Ernster, L (1975) Studies on the reaction mechanism of DT diaphorase. Action of dead-end inhibitors and effects of phospholipids. Arch Biochem Biophys 169, 560567.Google Scholar
614Singh, SV, Pan, SS, Srivastava, SK, Xia, H, Hu, X, Zaren, HA & Orchard, JL (1998) Differential induction of NAD(P)H:quinone oxidoreductase by anti-carcinogenic organosulfides from garlic. Biochem Biophys Res Commun 244, 917920.Google Scholar
615Nair, HK, Rao, KV, Aalinkeel, R, Mahajan, S, Chawda, R & Schwartz, SA (2004) Inhibition of prostate cancer cell colony formation by the flavonoid quercetin correlates with modulation of specific regulatory genes. Clin Diagn Lab Immunol 11, 6369.Google Scholar
616Murtaza, I, Marra, G, Schlapbach, R, Patrignani, A, Kunzli, M, Wagner, U, Sabates, J & Dutt, A (2006) A preliminary investigation demonstrating the effect of quercetin on the expression of genes related to cell-cycle arrest, apoptosis and xenobiotic metabolism in human CO115 colon-adenocarcinoma cells using DNA microarray. Biotechnol Appl Biochem 45, 2936.Google Scholar
617Suzuki, K, Koike, H, Matsui, H, Ono, Y, Hasumi, M, Nakazato, H, Okugi, H, Sekine, Y, Oki, K, Ito, K, Yamamoto, T, Fukabori, Y, Kurokawa, K & Yamanaka, H (2002) Genistein, a soy isoflavone, induces glutathione peroxidase in the human prostate cancer cell lines LNCaP and PC-3. Int J Cancer 99, 846852.Google Scholar
618Weinreb, O, Mandel, S & Youdim, MB (2003) cDNA gene expression profile homology of antioxidants and their antiapoptotic and proapoptotic activities in human neuroblastoma cells. FASEB J 17, 935937.Google Scholar
619Weinreb, O, Mandel, S & Youdim, MB (2003) Gene and protein expression profiles of anti- and pro-apoptotic actions of dopamine, R-apomorphine, green tea polyphenol ( − )-epigallocatechine-3-gallate, and melatonin, Ann N Y Acad Sci 993, 351361; discussion 387–393.Google Scholar
620Guo, S, Yang, S, Taylor, C & Sonenshein, GE (2005) Green tea polyphenol epigallocatechin-3 gallate (EGCG) affects gene expression of breast cancer cells transformed by the carcinogen 7, 12-dimethylbenz[a]anthracene. J Nutr 135, 2978S2986S.Google Scholar
621Wolfram, S, Raederstorff, D, Preller, M, Wang, Y, Teixeira, SR, Riegger, C & Weber, P (2006) Epigallocatechin gallate supplementation alleviates diabetes in rodents. J Nutr 136, 25122518.Google Scholar
622Knowles, LM & Milner, JA (2003) Diallyl disulfide induces ERK phosphorylation and alters gene expression profiles in human colon tumor cells. J Nutr 133, 29012906.Google Scholar
623Zhou, Z, Tan, HL, Xu, BX, Ma, ZC, Gao, Y & Wang, SQ (2005) Microarray analysis of altered gene expression in diallyl trisulfide-treated HepG2 cells. Pharmacol Rep 57, 818823.Google Scholar
624King, AA, Shaughnessy, DT, Mure, K, Leszczynska, J, Ward, WO, Umbach, DM, Xu, Z, Ducharme, D, Taylor, JA, DeMarini, DM & Klein, CB (2007) Antimutagenicity of cinnamaldehyde and vanillin in human cells: global gene expression and possible role of DNA damage and repair. Mutat Res 616, 6069.Google Scholar
625Yang, SH, Kim, JS, Oh, TJ, Kim, MS, Lee, SW, Woo, SK, Cho, HS, Choi, YH, Kim, YH, Rha, SY, Chung, HC & An, SW (2003) Genome-scale analysis of resveratrol – induced gene expression profile in human ovarian cancer cells using a cDNA microarray. Int J Oncol 22, 741750.Google Scholar
626Chalabi, N, Delort, L, Le Corre, L, Satih, S, Bignon, YJ & Bernard-Gallon, D (2006) Gene signature of breast cancer cell lines treated with lycopene. Pharmacogenomics 7, 663672.Google Scholar
627Lunec, J, Halligan, E, Mistry, N & Karakoula, K (2004) Effect of vitamin E on gene expression changes in diet-related carcinogenesis. Ann N Y Acad Sci 1031, 169183.Google Scholar
628Veeriah, S, Kautenburger, T, Habermann, N, Sauer, J, Dietrich, H, Will, F & Pool-Zobel, BL (2006) Apple flavonoids inhibit growth of HT29 human colon cancer cells and modulate expression of genes involved in the biotransformation of xenobiotics. Mol Carcinog 45, 164174.Google Scholar
629Golkar, L, Ding, XZ, Ujiki, MB, Salabat, MR, Kelly, DL, Scholtens, D, Fought, AJ, Bentrem, DJ, Talamonti, MS, Bell, RH & Adrian, TE (2007) Resveratrol inhibits pancreatic cancer cell proliferation through transcriptional induction of macrophage inhibitory cytokine-1. J Surg Res 138, 163169.Google Scholar
630Takahashi, Y, Lavigne, JA, Hursting, SD, Chandramouli, GV, Perkins, SN, Barrett, JC & Wang, TT (2004) Using DNA microarray analyses to elucidate the effects of genistein in androgen-responsive prostate cancer cells: identification of novel targets. Mol Carcinog 41, 108119.Google Scholar
631Powolny, A, Takahashi, K, Hopkins, RG & Loo, G (2003) Induction of GADD gene expression by phenethylisothiocyanate in human colon adenocarcinoma cells. J Cell Biochem 90, 11281139.Google Scholar
632van Erk, MJ, Roepman, P, van der Lende, TR, Stierum, RH, Aarts, JM, van Bladeren, PJ & van Ommen, B (2005) Integrated assessment by multiple gene expression analysis of quercetin bioactivity on anticancer-related mechanisms in colon cancer cells in vitro. Eur J Nutr 44, 143156.Google Scholar
633Barella, L, Muller, PY, Schlachter, M, Hunziker, W, Stocklin, E, Spitzer, V, Meier, N, de Pascual-Teresa, S, Minihane, AM & Rimbach, G (2004) Identification of hepatic molecular mechanisms of action of alpha-tocopherol using global gene expression profile analysis in rats. Biochim Biophys Acta 1689, 6674.Google Scholar
634Rota, C, Barella, L, Minihane, AM, Stocklin, E & Rimbach, G (2004) Dietary alpha-tocopherol affects differential gene expression in rat testes. IUBMB Life 56, 277280.Google Scholar
635Shen, G, Xu, C, Hu, R, Jain, MR, Nair, S, Lin, W, Yang, CS, Chan, JY & Kong, AN (2005) Comparison of ( − )-epigallocatechin-3-gallate elicited liver and small intestine gene expression profiles between C57BL/6J mice and C57BL/6J/Nrf2 ( − / − ) mice. Pharm Res 22, 18051820.Google Scholar
636Dolara, P, Luceri, C, Filippo, CD, Femia, AP, Giovannelli, L, Caderni, G, Cecchini, C, Silvi, S, Orpianesi, C & Cresci, A (2005) Red wine polyphenols influence carcinogenesis, intestinal microflora, oxidative damage and gene expression profiles of colonic mucosa in F344 rats. Mutat Res 591, 237246.Google Scholar
637Kim, S, Sohn, I, Lee, YS & Lee, YS (2005) Hepatic gene expression profiles are altered by genistein supplementation in mice with diet-induced obesity. J Nutr 135, 3341.Google Scholar
638Hishikawa, K, Nakaki, T & Fujita, T (2005) Oral flavonoid supplementation attenuates atherosclerosis development in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 25, 442446.Google Scholar
639Li, N, Guo, R, Li, W, Shao, J, Li, S, Zhao, K, Chen, X, Xu, N, Liu, S & Lu, Y (2006) A proteomic investigation into a human gastric cancer cell line BGC823 treated with diallyl trisulfide. Carcinogenesis 27, 12221231.Google Scholar
640Lee, SC, Chan, J, Clement, MV & Pervaiz, S (2006) Functional proteomics of resveratrol-induced colon cancer cell apoptosis: caspase-6-mediated cleavage of lamin A is a major signaling loop. Proteomics 6, 23862394.Google Scholar
641Narayanan, NK, Narayanan, BA & Nixon, DW (2004) Resveratrol-induced cell growth inhibition and apoptosis is associated with modulation of phosphoglycerate mutase B in human prostate cancer cells: two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis and mass spectrometry evaluation. Cancer Detect Prev 28, 443452.Google Scholar
642Ramljak, D, Romanczyk, LJ, Metheny-Barlow, LJ, Thompson, N, Knezevic, V, Galperin, M, Ramesh, A & Dickson, RB (2005) Pentameric procyanidin from Theobroma cacao selectively inhibits growth of human breast cancer cells. Mol Cancer Ther 4, 537546.Google Scholar
643Park, S, Lee, J & Yeom, CH (2006) A proteomic approach to the identification of early molecular targets changed by l-ascorbic acid in NB4 human leukemia cells. J Cell Biochem 99, 16281641.Google Scholar
644Wang, Y, He, QY, Chen, H & Chiu, JF (2007) Synergistic effects of retinoic acid and tamoxifen on human breast cancer cells: proteomic characterization. Exp Cell Res 313, 357368.Google Scholar
645Herzog, A, Kindermann, B, Doring, F, Daniel, H & Wenzel, U (2004) Pleiotropic molecular effects of the pro-apoptotic dietary constituent flavone in human colon cancer cells identified by protein and mRNA expression profiling. Proteomics 4, 24552464.Google Scholar
646Fuchs, D, de Pascual-Teresa, S, Rimbach, G, Virgili, F, Ambra, R, Turner, R, Daniel, H & Wenzel, U (2005) Proteome analysis for identification of target proteins of genistein in primary human endothelial cells stressed with oxidized LDL or homocysteine. Eur J Nutr 44, 95104.Google Scholar
647Deshane, J, Chaves, L, Sarikonda, KV, Isbell, S, Wilson, L, Kirk, M, Grubbs, C, Barnes, S, Meleth, S & Kim, H (2004) Proteomics analysis of rat brain protein modulations by grape seed extract. J Agric Food Chem 52, 78727883.Google Scholar
648Breikers, G, van Breda, SG, Bouwman, FG, van Herwijnen, MH, Renes, J, Mariman, EC, Kleinjans, JC & van Delft, JH (2006) Potential protein markers for nutritional health effects on colorectal cancer in the mouse as revealed by proteomics analysis. Proteomics 6, 28442852.Google Scholar
649Rowell, C, Carpenter, DM & Lamartiniere, CA (2005) Chemoprevention of breast cancer, proteomic discovery of genistein action in the rat mammary gland. J Nutr 135, 2953S2959S.Google Scholar
650Mitchell, BL, Yasui, Y, Lampe, JW, Gafken, PR & Lampe, PD (2005) Evaluation of matrix-assisted laser desorption/ionization-time of flight mass spectrometry proteomic profiling: identification of alpha 2-HS glycoprotein B-chain as a biomarker of diet. Proteomics 5, 22382246.Google Scholar
651Weissinger, EM, Nguyen-Khoa, T, Fumeron, C, Saltiel, C, Walden, M, Kaiser, T, Mischak, H, Drueke, TB, Lacour, B & Massy, ZA (2006) Effects of oral vitamin C supplementation in hemodialysis patients: a proteomic assessment. Proteomics 6, 9931000.Google Scholar
652Aldred, S, Sozzi, T, Mudway, I, Grant, MM, Neubert, H, Kelly, FJ & Griffiths, HR (2006) Alpha tocopherol supplementation elevates plasma apolipoprotein A1 isoforms in normal healthy subjects. Proteomics 6, 16951703.Google Scholar
Figure 0

Fig. 1 Different form of reactive oxygen species and their interaction with organic molecules.

Figure 1

Fig. 2 Impact of ROS on cell signalling and activation of transcription factors.

Figure 2

Fig. 3 Overview of different methods used for the detection of dietary antioxidants.

Figure 3

Table 1 Comparison of methods used to determine the total antioxidant capacity (TAC)

Figure 4

Table 2 Main biomarkers for lipid and protein oxidation

Figure 5

Table 3 Overview on currently used methods for the detection of antioxidant enzymes and examples of their induction by food constituents

Figure 6

Table 4 Examples for genes which are transcriptionally regulated by oxidative stress

Figure 7

Table 5 Examples for results obtained with dietary antioxidants in microarray experiments

Figure 8

Table 6 Examples for results obtained with dietary antioxidants in recent proteomics studies

Figure 9

Fig. 4 Advantages and disadvantages of different experimental used to investigate ROS – protective effects of phytochemicals.