Skip to main content

Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases

Abstract

Background

The production of peroxide and superoxide is an inevitable consequence of aerobic metabolism, and while these particular 'reactive oxygen species' (ROSs) can exhibit a number of biological effects, they are not of themselves excessively reactive and thus they are not especially damaging at physiological concentrations. However, their reactions with poorly liganded iron species can lead to the catalytic production of the very reactive and dangerous hydroxyl radical, which is exceptionally damaging, and a major cause of chronic inflammation.

Review

We review the considerable and wide-ranging evidence for the involvement of this combination of (su)peroxide and poorly liganded iron in a large number of physiological and indeed pathological processes and inflammatory disorders, especially those involving the progressive degradation of cellular and organismal performance. These diseases share a great many similarities and thus might be considered to have a common cause (i.e. iron-catalysed free radical and especially hydroxyl radical generation).

The studies reviewed include those focused on a series of cardiovascular, metabolic and neurological diseases, where iron can be found at the sites of plaques and lesions, as well as studies showing the significance of iron to aging and longevity. The effective chelation of iron by natural or synthetic ligands is thus of major physiological (and potentially therapeutic) importance. As systems properties, we need to recognise that physiological observables have multiple molecular causes, and studying them in isolation leads to inconsistent patterns of apparent causality when it is the simultaneous combination of multiple factors that is responsible.

This explains, for instance, the decidedly mixed effects of antioxidants that have been observed, since in some circumstances (especially the presence of poorly liganded iron) molecules that are nominally antioxidants can actually act as pro-oxidants. The reduction of redox stress thus requires suitable levels of both antioxidants and effective iron chelators. Some polyphenolic antioxidants may serve both roles.

Understanding the exact speciation and liganding of iron in all its states is thus crucial to separating its various pro- and anti-inflammatory activities. Redox stress, innate immunity and pro- (and some anti-)inflammatory cytokines are linked in particular via signalling pathways involving NF-kappaB and p38, with the oxidative roles of iron here seemingly involved upstream of the IkappaB kinase (IKK) reaction. In a number of cases it is possible to identify mechanisms by which ROSs and poorly liganded iron act synergistically and autocatalytically, leading to 'runaway' reactions that are hard to control unless one tackles multiple sites of action simultaneously. Some molecules such as statins and erythropoietin, not traditionally associated with anti-inflammatory activity, do indeed have 'pleiotropic' anti-inflammatory effects that may be of benefit here.

Conclusion

Overall we argue, by synthesising a widely dispersed literature, that the role of poorly liganded iron has been rather underappreciated in the past, and that in combination with peroxide and superoxide its activity underpins the behaviour of a great many physiological processes that degrade over time. Understanding these requires an integrative, systems-level approach that may lead to novel therapeutic targets.

Peer Review reports

Background and preamble

The 'balkanisation' of the literature is in part due to the amount of it (some 25,000 journals with presently 2.5 million peer-reviewed papers per year, i.e. ~5 per minute [1]), with a number http://www.nlm.nih.gov/bsd/medline_cit_counts_yr_pub.html increasing by something approaching 2 per minute at PubMed/Medline alone. In addition, the disconnect between the papers in the literature (usually as pdf files) and the metadata describing them (author, journal, year, pages, etc) is acute and badly needs filling [2]. Without solving this problem, and without automation of the processes of reading, interpreting and exploiting this literature and its metadata in a digital format, we cannot make use of the existing tools for text mining and natural language processing (e.g. [35]), for joining disparate concepts [6], for literature-based discovery (e.g. [711], and for studies of bibliometrics [12, 13], literature dynamics [14], knowledge domains [15], detecting republication [16] and so on. Until we recognise these possibilities we are unlikely to seek to realise them.

The present article (and see [17] for a preprint) serves to show some of the benefits than can accrue from a more overarching view of the otherwise highly disparate literature in a particular domain (see also [18]), but was done 'the hard way', i.e. with a few bibliographic and bibliometric tools but without the kind of automation implied above. For the record, the main tools used (see a review in [2]) were Web of Knowledge and Scopus for literature and citation searching, supplemented by Google Scholar. Some use was also made of ARROWSMITH [6, 19, 20] and GOPubMed [21], as well as various workflows in the Taverna environment [2226], including the BioAID_DiseaseDiscovery workflow http://www.myexperiment.org/workflows/72 written by Marco Roos. Citations and attendant metadata were stored in Endnote (latterly version X).

Introduction

Even under 'normal' conditions, as well as during ischaemia when tissue oxygenation levels are low, the redox poise of the mitochondrial respiratory chain is such that the normally complete four-electron reduction of dioxygen to water is also accompanied by the production, at considerable rates (ca 1–4% of O2 reduced), of partially reduced forms of dioxygen such as hydrogen peroxide and superoxide (e.g. [2745]). These 1- and 2-electron reductions of O2 are necessarily exacerbated when the redox poise of the b-type cytochromes is low, for instance when substrate supplies are in excess or when the terminal electron acceptor O2 is abnormally low due to hypoxia or ischaemia. Various other oxygenases, oxidases and peroxidases can also lead directly to the production of such 'reduced' forms of dioxygen in vivo (e.g. [4648]), with H2O2 from xanthine oxidase being especially implicated in ischaemia/reperfusion injury (e.g. [47, 4954]). These molecules (peroxide and superoxide) can cause or contribute to various kinds of oxidative stress. However, this is mainly not in fact because they can react directly with tissue components themselves, since they are comparatively non-toxic, cells have well-known means of dealing with them [55], and they are even used in cellular signalling (e.g. [5660]). Much more importantly, it is because they can react with other particular species to create far more reactive and damaging products such as hydroxyl radicals, with all these agents nevertheless being known collectively (and indiscriminately) as reactive oxygen species (ROSs). Possibly the commonest means by which such much more damaging species, in particular the hydroxyl radical, are created is by reaction with unliganded or incompletely liganded iron ions [6163]. The themes of this review are thus (i) that it is this combination of poorly liganded iron species, coupled to the natural production of ROSs, that is especially damaging, (ii) that the role of iron has received far less attention than has the general concept of ROSs, albeit the large literature that we review, and (iii) that this basic combination underpins a great many (and often similar) physiological changes leading to a variety of disease manifestations, and in particular those where the development of the disease is manifestly progressive and degenerative.

An overview of the structure of the review is given in Fig 1, in the form of a 'mind map' [64]. The main literature review for this meta-analysis was completed on June 30th, 2008, with some updates being added following the refereeing process.

Figure 1
figure 1

An overview of this article, set out in the form of a 'mind map' [64].

Some relevant chemistry of iron and reduced forms of oxygen

While superoxide and peroxide are the proximate forms of incomplete O2 reduction in biology, a reaction catalysed by the enzyme superoxide dismutase [65] serves to equilibrate superoxide and peroxide:

(1)

Arguably the most important reaction of hydrogen peroxide with (free or poorly liganded) Fe(II) is the Fenton reaction [66], leading to the very reactive and damaging hydroxyl radical (OH)

(2)

Superoxide can also react with ferric iron in the Haber-Weiss reaction [67] to produce Fe(II) again, thereby effecting redox cycling:

(3)

Ascorbate can replace O2 •- within the cell for reducing the Fe(III) to Fe(II) [68]. Further reactions, that are not the real focus here, follow from the ability of hydroxyl radicals and indeed Fe(n) directly to interact with many biological macro- and small molecules, especially including DNA, proteins and unsaturated lipids. Thus [6973], Fe(II) and certain Fe(II) chelates react with lipid hydroperoxides (ROOH), as they do with hydrogen peroxide, splitting the O--O bond. This gives RO, an alkoxyl radical, which can also abstract H from polyunsaturated fatty acids and from hydroperoxides. The resulting peroxyl radicals ROO can continue propagation of lipid peroxidation. Oxidative stress also leads to considerable DNA damage [7476] and to the polymerisation and denaturation of proteins [7779] and proteolipids that can together form insoluble structures typically known as lipofucsin (see e.g. [80, 81]) or indeed plaques. Such plaques can also entrap the catalysts of their formation, and thereby point them up. Some of the evidence for these is described below. Many small molecule metabolic markers for this kind of oxidative stress induced by the hydroxyl radical and other 'reactive oxygen species' (ROSs) are known [43, 8289], and include 8-oxo-guanine [9094], 8-hydroxy guanine [95], 8-hydroxy-2'-deoxy-guanosine [96, 97], 8-oxo-GTP [98], 4-hydroxy-2-hexenal [99], 4-hydroxy-nonenal [100], 4-hydroperoxy-2-nonenal, various isoprostanes [101107], 7-keto-cholesterol [108], many other cholesterol derivatives [109], malondialdehyde [110], neopterin [111], nitrotyrosine [112115] and thymidine glycol [116, 117]. Note that the trivial names in common use for this kind of metabolite are not helpful and may even be ambiguous or misleading, and it is desirable (e.g. [118]) to refer to such molecules using terminology that relates them either to molecules identified in persistent curated datbases [119] such as ChEBI [120] or KEGG [121], or better to describe them via database-independent encodings such as SMILES [122] or InChI [123128] strings. (There are other oxidative markers that may be less direct, such as the ratio of 6-keto-prostaglandin F1α to thromboxane B2 [129], but these are not our focus here.)

Overall, it is in fact well established that the interactions between 'iron' sensu lato and partly reduced forms of oxygen can lead to the production of the very damaging hydroxyl radical (e.g. [43, 130139]), and that this radical in particular probably underpins or mediates many higher-level manifestations of tissue damage, disease, organ failure and ultimately death[36, 137, 140143]. While the role of ROSs generally in these processes has been widely discussed, the general recognition of the importance of inadequately liganded iron in each of them has perhaps been less than fully appreciated. One of our tasks here will therefore be to stress this role of 'iron', and to assess the various means of chelating 'iron' such that it does not in fact do this. (Throughout we use 'iron' to refer to forms of Fe(n, n > 0) with unspecified ligands, though we absolutely stress that it is the exact speciation and liganding that determines the reactivity of 'iron' in catalysing reactions such as that of hydroxyl radical formation, and indeed its bioavailability generally – inadequate liganding of iron in the required forms can be a cause of anaemia even if the total amount of 'iron' is plentiful.)

For completeness we note the reactions catalysed by superoxide dismutase

(4)

and by catalase

(5)

These together, were their activity in the relevant locations sufficiently great, might serve to remove (su)peroxide from cells completely.

In addition to reactive oxygen species there are ions such as the perferryl ion (Fe-O) [144] and reactive nitrogen species [60, 145147]. These latter are mainly formed from the natural radical NO, an important inflammatory mediator [148], with peroxynitrite production (from the reaction of NO and superoxide) [46, 149154] leading to nitrotyrosine [112], or nitro-fatty acid [155, 156] or protein cystein nitrosylation [157, 158] being a common means of their detection downstream. Other toxic products of the reactions of NO include NO2, N2O3, and S-nitrosothiols [159], and the sequelae of some of these may also involve iron [160].

Overall, we recognise that these kinds of inflammatory, oxidative stress-related reactions are accumulative and somewhat irreversible [161], that they are consequently age-related, and (see [162165] and later), and that most diseases and causes of mortality that are prevalent in the developed world are in this sense largely manifestations of this kind of aging.

Ligands and siderophores

As well as the reactions described above, ferrous ions will react with oxygen under aerobic conditions to produce ferric ions, and in natural environments there is little to stop this. Consequently, and because these reflect fundamental physicochemical properties of such ions, the problems of both solubility and toxicity were faced by bacteria (and indeed fungi [166169]) long ago in evolution, and were solved by their creation and excretion of (mainly ferric-)iron chelators known as siderophores [170189] (and for haemophores see [190]). These typically have extremely tight binding constants (Kf > 1030 [191]) and can solubilise and sequester iron such that it can be internalised via suitable transporter molecules within the bacterial plasma membrane [192]. Bacterial and fungal siderophores usually form hexadentate octahedral complexes with ferric iron and typically employ hydroxamates, α-hydroxycarboxylates and catechols as extremely effective Fe3+ ligands [182]. Since bacterial growth requires iron, it is unsurprising that siderophores are effectively virulence factors (e.g. [174, 193196]). While upwards of 500 microbial siderophores have been identified [182], with new ones still appearing (some via genomic analyses, e.g. [197]), and with the most common one in medical use, desferrioxamine or DFO, being such a bacterial product (see below), it is an astonishing fact that no human siderophore has been chemically identified, even though such activities were detected nearly 30 years ago [198, 199] (see also [200205]). As noted by Kaplan [206], "a discovery that mammals produce siderophores would lead to an epochal change in the paradigm of mammalian iron homeostasis." To this end, some recent events have begun to change matters, and our overall knowledge of the regulation of iron metabolism, considerably.

Mammalian iron metabolism

The total body iron in an adult male is 3000 to 4000 mg and the daily iron requirement for erythropoiesis, the major 'sink', is about 20 mg [207]. However, the loss of iron in a typical adult male is very small [208, 209] and can be met by absorbing just 1 – 2 mg of iron per day [210, 211]. The careful conservation and recycling of iron – mainly from degrading erythrocytes – is in fact essential because typical human diets contain just enough iron to replace the small losses, although when dietary iron is more abundant, absorption must be (and is) attenuated since higher levels than necessary lead to iron overload and many distressing sequelae contingent on the radical production described above.

A variety of aspects of mammalian iron metabolism have been reviewed in detail elsewhere (e.g. [134, 139, 195, 212241]), including a series on 'iron imports' [242248], and for our present purposes (Fig 2) mainly involves the intestinal (mainly duodenal) uptake of Fe(II) (produced from Fe(III) using a luminal ferrireductase) via a divalent metal ion transporter DMT1/DCT1/NRAMP [249, 250] and its subsequent binding as Fe(III) to transferrin (Tf). The intestinal uptake of haem (heme) occurs via the heme carrier protein-1 (HCP1) [251] and it is thereby internalized, while the iron in heme is liberated by heme oxygenase-1 (HO1) [252254]. Haem is synthesised in many tissues, especially liver and erythroid cells [255]. Vesicular routes of intestinal transfer may also occur [256, 257]. Low MW cytoplasmic chelators such as citrate can bind iron fairly weakly and thereby contribute to a labile iron pool (LIP) in the cytoplasm and especially the lysosomes and mitochondria (see [258262]), while ferritin [263] too can bind cytoplasmic iron (via a chaperone [264]) and is seen as a good overall marker of iron status [265267]. Iron(II) is subsequently exported through the basolateral membrane of the enterocyte by ferroportin-1 (FPN1) [268270]. Ferroportin may also contribute to uptake in enterocytes [271]. Fe(III) may then be produced by hephaestin (Hp) [272] before it is bound by transferrin (Tf), which is the main but not sole means of binding Fe(III) when it is transported through the circulation, with major iron storage taking place in the liver. Similar processes occur in the peripheral tissues, with significant transfer of iron from transferrin occurring via the transferrin receptor [273].

Figure 2
figure 2

Schematic overview of the main elements considered to participate in mammalian iron metabolism.

'Free' haem appears in the circulation (it may have a signalling role [274]) and elsewhere largely because of erythrocyte degradation, and it can also greatly amplify the cellular damage caused by ROSs [275], and its degradation pathway via haem oxygenase [276, 277] to biliverdin and then using biliverdin reductase to form bilirubin generates 'free' (and potentially redox-active) iron. It would appear, not least because biliverdin has powerful antioxidant properties, that haem oxygenase is more protective than damaging [253, 278282], even though one of the products of its reaction is Fe that must eventually be liganded (or e.g. incorporated into ferritin). (Another product is the gas CO, that has been proposed as a measure of oxidative stress in the lung [283].)

All of the above obviously ignores both some important aspects of the speciation and liganding of iron, as well as the tissue distribution of the specific proteins involved – for which latter global information will shortly emerge [284] (http://www.proteinatlas.org/ and see later). It also ignores any discussion on the genetic regulation of iron metabolism (e.g. [285288]), which is not our main focus.

However, one molecule in particular, hepcidin, has recently emerged as a 'master regulator' of regulation at the physiological level, and we describe some of these new developments.

Hepcidin

In the liver and elsewhere, many aspects of iron metabolism are regulated by a recently discovered 25-amino acid polypeptide called hepcidin [207, 241, 245, 271, 289327] that acts in part as a negative regulator of iron efflux [328] by causing the internalisation of ferroportin [329333]. Hepcidin is produced, partly under the regulation of a receptor called hemojuvelin (e.g. [334]), via an 84-aa precursor called pre-pro-hepcidin and a 60 mer called pro-hepcidin [304, 335, 336] although the active agent is considered to be the 25 mer referred to above, and with the inactive precursors appearing not to be useful markers [337, 338].

Strikingly, anaemia and anoxia both suppress hepcidin production [245, 339, 340] (Fig 3), such that just while superoxide production is being enhanced by the anoxia there is more iron being absorbed from the intestine and effluxed into the circulation. In view of the inter-reactivity of superoxide and iron this could be anticipated to enhance free radical formation, leading to a positive feedback loop in which the problems are amplified: ischaemia/anoxia changes Fe(n) distribution leading to differential reactivity with the products of anoxia and thus further free radical production. However, hepcidin is overexpressed in inflammatory disease and is an early inflammatory marker [245, 341345]. Its expression is positively controlled inter alia by SMAD4, and loss of hepatic SMAD4 is thus associated with dramatically decreased expression of hepcidin in liver and increased duodenal expression of a variety of genes involved in intestinal iron absorption, including Dcytb, DMT1 and ferroportin, leading to iron overload [346]. STAT3 is another positive effector of hepcidin expression [347, 348], and ROSs inhibit this effect [349], thereby creating a link between ROSs and Fe metabolism. To understand the exact roles of hepcidin in iron metabolism, it is going to be especially important to understand where it is expressed; fortunately, such studies are beginning to emerge [350].

Figure 3
figure 3

Some effects of hepcidin, summarizing the fact that hypoxic condition can suppress it and thus lead to hyperferraemia. Since hypoxic conditions can also lead to ROS production the hypoxia-mediated regulation of hepcidin can have especially damaging effects.

Overall there is a complex interplay between positive and negative regulation and the organismal distribution of iron caused by changes in hepcidin concentration [351], with in many cases the hypoxic response (decreased hepcidin) seeming to dominate that due to inflammation (increased hepcidin) even when iron levels are high [352, 353]. Specifically, lowered hepcidin causes hyperferraemia. Hepcidin is also activated by p53 [354], and may play a role in the degradation of atherosclerotic plaques [355]. Another recently discovered protein that is crucially involved in human iron metabolism is NGAL or siderocalin, and while there is some evidence for their co-regulation [356], they have normally been studied separately.

NGAL (also known as lipocalin-2 or siderocalin)

Lipocalins [357] are a diverse group of ligand-binding proteins that share a conserved structure even in the absence of significant sequence conservation. This core structure includes an eight-stranded anti- parallel β barrel that defines a calyx, or cup-shaped structure, enclosing the ligand binding site.

NGAL – neutrophil gelatinase-associated lipocalin – is a 21 kDal glycoprotein first isolated by Kjeldsen and colleagues in 1993 [358]. Synonyms include lipocalin 2, siderocalin, Lcn2, α2-microglobulin-related protein or neu-related lipocalin (in rats) [359, 360] and (in mice) 24p3 or uterocalin [361]. Although lipocalins are well known to be involved in the sequestration and transport of a variety of ligands, the natural ligand of NGAL (as is the case with many lipocalins) was not initially known even in terms of its chemical class. This changed with the seminal paper of Goetz and colleagues [362] (and see [206]) who purified recombinant NGAL from a particular strain of E. coli and found that its structure contained a negatively charged ferric siderophore with a subnanomolar dissociation constant that it had extracted from its bacterial host, and that the apo form of this molecule could also act as a potent bacteriostatic agent by sequestering iron (see also [363367]). A companion paper [368] showed that the iron-delivering activity was expressed in mammalian cells. The structure of NGAL is now known [369] and one of its interaction partners is a matrix metalloproteinase [370] to which it can presumably donate a metal ion and in the complex decrease its degradation [371].

The finding that NGAL was one of the most highly expressed proteins following ischaemia-reperfusion injury in kidney cells [372374], and prognostic of kidney damage long before the more traditional marker creatinine was raised significantly, has led to considerable interest in this protein, especially as a marker of renal injury [375389], and perhaps as a therapeutic [375]. Devireddy and colleagues [390] identified a receptor that internalizes 24p3, and internalization of iron bound to 24p3 prevents apoptosis. In contrast, internalization of the apo form of 24p3 that does not contain iron led to cellular iron efflux and apoptosis via the proapoptotic protein Bim [391]. In humans the megalin receptor can bind siderocalin (and its siderophore payload) and mediate its intracellular uptake [392]. Oxidative stress can also induce its expression [393], and it is protective against it [394].

Exogenously administered NGAL also markedly upregulates heme oxygenase-1, a proven multifunctional protective agent in experimental Acute Kidney Injury (AKI) that is thought to work by limiting iron uptake, promoting intracellular iron release, enhancing production of antioxidants such as biliverdin and carbon monoxide, and inducing the cell cycle regulatory protein p21 [279, 395, 396]. Because of this multifaceted protective action, NGAL has emerged as a prime therapeutic target in ischaemic AKI [379].

Structural and direct binding studies have suggested that siderocalin tends (although not exclusively) to bind catecholate-type ligands, rather than hydroxamate- or carboxylate-based siderophores, at least when tested with microbially derived siderophores [362, 363, 365] (but cf. [369] for claims, disputed [360] and not now accepted, as to the binding of bacterially derived formyl peptides!). The role of NGAL, as a siderophore-binding agent, is thus consistent with the widespread recognition that iron-induced radical generation is intimately involved in a variety of renal and other diseases [397, 398]. However, while it is certainly the case that siderocalin can reduce the virulence of bacteria when it binds the relevant bacterial siderophores [362367] and that bacteria can 'evade' this by synthesising siderophores that siderocalin cannot bind (e.g. [186, 187, 399401]), it is questionable whether the only role of siderocalin lies in fact in its antibacterial activity. Rather we would suggest that its main role is in sequestrating iron via a human siderophore to stop inappropriately liganded iron from producing damaging oxygen radicals. Consistent with this iron-liganding role for human biology is the fact that the tissue most highly expressing NGAL under normal conditions is bone marrow [360, 402], the site of erythropoiesis. The liganding can be extensive; as Goetz and colleagues [362] note, "During inflammation, concentrations of NGAL can increase to levels, with concentrations approaching 20–30 nM in the serum [403], presumably adequate to bind all available iron as ferric siderophore complexes".

Significant changes in NGAL expression have also been observed, for instance, during kinase-mediated signalling [404, 405], in cardiovascular disease [406409] and in cancer [410412].

These findings on the kidney and the role of NGAL, together with the important knowledge that its chief ligand is probably an unknown human siderophore (Figs 2, 4), thus lead us to consider the role of this system (and unliganded iron generally) in a whole series of other diseases that all share many characteristics of oxidative stress and inflammation (see also [413]). A similar thesis, albeit with comparatively little stress on iron, is the leitmotif of Finch's recent detailed monograph [163]. The theme of these sections is thus to stress the fact that while the role of ROSs in general in such syndromes has been pointed up previously, that of iron as a major culprit has not so generally yet been stressed, notwithstanding that there is in fact a great deal of pertinent literature that we here highlight as the focus of this review.

Figure 4
figure 4

Overview of the roles of ischaemia, ROSs, poorly liganded iron and the iron metabolism regulators HGAL and hepicidin in effecting inflammation as a physiological level.

Some disease manifestations in which iron may be implicated

Preeclampsia (PE)

Another important disease that shares many of the same properties (or at least sequelae) of renal impairment, and may have the same fundamental aetiology, is pre-eclampsia. This is the most significant cause of morbidity and mortality in pregnant women [414]. The chief clinical manifestations at time of diagnosis are a raised blood pressure (hypertension) [415] and proteinuria, together with raised creatinine, consistent with the reversible existence (since it is relieved upon delivery of the baby) of renal impairment. However, prognostic markers that might manifest early in pregnancy are lacking, and would be highly desirable. There is widespread agreement [416] that a poor connection of the placenta to the uterus leads to ischaemia and thus oxidative stress, with a substantial involvement of apoptosis during the placental remodelling [417423]. Since preeclampsia-like syndromes can be induced in pregnant animals by surgical restriction of the uteroplacental blood supply [424], it is presumed that blood-borne agents arising from the ischaemic placenta are the cause of the generalized endothelial cell damage and inflammatory responses that give rise to the symptoms of hypertension, proteinuria, and sudden oedema characteristic of preeclampsia [70]. Indeed, many studies implicate oxidative stress as a substantial contributor to this [425489], while some have noted the importance of iron status [70, 133, 450, 490511], and so far as is known the transporters of iron in the placenta are similar to those in other cells [512]. Oxidative stress of this type is of course inflammatory in nature and inflammation is observed in PE [472, 476, 484, 486, 513519]. We suggest strongly that it is the combination of inadequately liganded Fe(II or III) and superoxide/peroxide leading to OH formation that is the chief mechanistic cause of the downstream events that manifest in PE, and that appropriate removal by liganding/chelation or otherwise of these ions would prove of therapeutic benefit. (Iron status has also been implicated in other pregnancy and neonatal disorders [520524].) There is evidence too for the involvement of the radical NO [456, 525].

We note that it is quite common nevertheless for iron to be prescribed during pregnancy, especially during its latter stages [526, 527], and that this does of course lead to oxidative stress [528, 529].

Oxidative stress is caused both by the initial rate of production of superoxide and the rate of their conversion into OH radicals. The former can be induced by hypoxic conditions such as occur at high altitude, and one prediction, that is borne out [487, 530], is that PE should therefore be more prevalent at high altitude. Erythropoietin may be a marker for oxidative stress in pre-eclampsia [531].

Regarding the second stage, predictions include that PE should be more common in those suffering from diseases of iron metabolism. Although such mothers are of course less well a priori, this prediction is borne out for α-thalassemia [532, 533] although not, interestingly, for haemochromatosis [534]. We note in this context that thalassaemia not only predisposes towards PE but is known in general to cause hepcidin to decrease and NGAL to increase [352, 353, 356, 535], with consequent and inevitable iron dysregulation.

Another prediction is then that hepcidin should be changed in pre-eclampsia. Although no serum measurements have been reported to date, it is of extreme interest that – while they took it to be an antimicrobial peptide rather than an iron regulator – a recent study by Knox and colleagues of placental gene expression in a mouse model of PE showed that hepcidin expression increased by a greater factor than that of any other gene save one [536], consistent with the view that major changes in the regulation of iron liganding and metabolism underpin PE.

Finally, we note that NGAL is significantly implicated in pregnancy, and was even named uterocalin in mice to reflect its high expression in the uterus [361, 537539]. A very recent study [540] suggests that it may be a useful second trimester biomarker for pre-eclampsia.

Diabetes

Type 2 diabetes and insulin resistance are known complications of pregnancy (e.g. [541545]), and also predispose towards PE. In a similar vein, various types of pregnancy-related intrauterine growth restriction predispose towards diabetes in later life [546, 547], pointing up the progressive nature of these syndromes. Metabolic biomarkers for the one can thus be predictive of the other [548], consistent with a common cause. Certainly ROSs are known to play a substantive role in both insulin resistance [549556] and in a variety of diabetic sequelae [95, 557559], and mitochondrial dysfunction may be an early step in this [560]. Some anti-diabetic drugs, such as the 'glitazones' that are considered to act on Peroxisome Proliferator Activated Receptor (PPAR)γ, may also act by decreasing ROS production (e.g. [561565]), and are even active aganst cerebral ischaemia and stroke [566569]. As with most if not all of the other diseases we review here, studies of pro-inflammatory markers (such as TNF-α, IL-1 and C-reactive protein [570]) during the development of diabetes show its aetiology to be inflammatory in nature [553, 571590]. Iron 'excess' is also a known feature of gestational diabetes [591593], and is a clear risk factor for the disease even in 'normal' populations [594603], and indeed diabetes is a classical consequence of iron overloading as seen in hereditary haemochromatosis [604]. Serum ferritin and body iron stores are strongly associated with diabetes [603, 605609], including prospectively [610], while changes in visfatin are also intimately involved in changes of iron metabolism (with pro-hepcidin being elevated) [611]. Most importantly, lipocalin 2 (siderocalin/NGAL) is strongly associated with the development of diabetes [612, 613]. Lowering iron improves insulin sensitivity [598, 614], and metallothionein is also protective [615619]. There seems little doubt that iron status is a major determinant of the development of type 2 diabetes [620].

Non-transferrin-bound iron is also considerably elevated in type 2 diabetes [621], and this too is exacerbated by vitamin C. Iron metabolism is substantially deranged in type 2 diabetes and the metabolism of glucose (a reducing sugar) interacts significantly with iron metabolism [598]. Iron is also strongly implicated in non-alcoholic steatohepatitis, considered an early marker of insulin resistance [622624]. Well-known diabetic complications include retinopathies, and it is noteworthy that elevated levels of ferritin can lead to cataract formation [625, 626].

The metabolic syndrome

Although some of its origins may be pre-natal [547], many of the features of these diseases are also seen in the (so-called) Metabolic Syndrome [627631]. Thus, serum ferritin is also related to insulin resistance [606, 632, 633] and iron levels are raised [624, 634, 635]. Of course diabetes and the Metabolic Syndrome are also closely coupled, so it is reasonable that features observed in the one may be observed during the development of the other. The metabolic syndrome is also an independent indicator for chronic kidney disease [636] and may be related to liver steatosis [637]. Metabolic disorders of this type too are closely intertwined with inflammation [575, 581, 587, 638], that is of course stimulated by ROSs whose generation is increased by high-fat diets [639]. Thus, our role here is to point up the existence of a considerable body of more-than-circumstantial evidence that here too the progressive and damaging nature of these diseases may be caused, in part, by inappropriately chelated iron.

Obesity

"As previously pointed out by Booth et al. [640], 100% of the increase in the prevalence of Type 2 diabetes and obesity in the United States during the latter half of the 20th century must be attributed to a changing environment interacting with genes, because 0% of the human genome has changed during this time period." [629]

It is well known that there has been a staggering increase in the prevalence of obesity, diabetes, and especially type 2 diabetes, in the last 50 years or so, and that this increase is expected to continue (e.g. [641643] and http://www.who.int/diabetes/). Equally, it is now well known that obesity, metabolic syndrome, diabetes and cardiovascular diseases are all more or less related to each other [643], and the question arises here as to whether dysfunctional iron metabolism might be a feature of each of them. In the case of obesity per se, however, we see no major evidence as yet for a causative role of deranged iron metabolism or chelation in causing obesity. Indeed, while they are related [644], what little evidence there is [645, 646] suggests that the converse may be true, i.e. that changes in iron metabolism might be consequent upon obesity (possibly via peroxide generation [639]). Importantly, considerable evidence suggests that obesity and inflammation are significantly related [163, 486, 575, 581, 642, 647664], not least because adipocytes produce and release various adipokines including pro-inflammatory cytokines such as IL-6 and TNF-α [575, 649, 650, 665671]. It is likely that it is the combination of overfeeding-induced obesity and inflammation (partly induced by the obesity itself [672]) that leads to diabetes [673]. Certainly there is evidence for increased ROS production in obese mice, possibly mediated in part via the fatty acid-induced activation of NAPH oxidase [674], while obesity is linked [675, 676] to urinary levels of 8-epi-PGF, a well established marker of oxidative stress (qv). Fig 5 summarises the above in a manner that stresses the roles of iron, overfeeding and inflammation in the genesis of these processes, and notes that interference in several of these steps is likely to be required to limit their progression to best advantage.

Figure 5
figure 5

Role of inflammation caused by hydroxyl radical formation in the interactive development of obesity, the metabolic syndrome and diabetes. Intervention at multiple steps is likely to be most beneficial in alleviating this kind of progression.

Hypertension

As well its significance in pre-eclampsia (see above), hypertension is a well known risk factor for many cardiovascular and related disease (e.g. [677]), and there is considerable evidence that its underlying cause is inflammatory in nature [678686], is related to the metabolic syndrome and obesity (e.g. [648, 687691]), and may be mediated mainly via ROSs [692]. There is evidence that some of its sequelae may be mediated via iron [693, 694].

Cardiovascular diseases

It is well known that elevated iron stores can predispose to coronary artery disease and thence myocardial infarction. The 'iron hypothesis' of the benefits of some iron depletion due to menstruation was devised to account for the lowering of heart-disease risk in young women (that disappears in those post-menopause) and was proposed by Jerome Sullivan in 1981 [695698] (and see also [699, 700]). (In this sense, the lack of menstruation during pregnancy would predispose to a comparative abundance of iron, as is indeed found – see above.) It is of particular interest that the well-known adverse vascular effects of homocysteine (in inhibiting flow-mediated dilatation) are in fact iron-dependent [701703], and that reducing homocysteine (e.g. by folate supplementation) in the absence of lowering iron has shown no clinical benefit to date [704], thereby suggestion iron mediation. By contrast, iron stores represent an established risk factor for cardiovascular disease [705].

Of course many factors such as lipid levels, stress, smoking and so are well-known risk factors for cardiovascular, coronary artery disease and related diseases. Indeed kidney disease is well established as a risk factor for cardiovascular disease [706708] (and indeed stroke [709]), all consistent with their having in part a common cause – we believe inflammation). Our purpose here, within the spirit of this review, is to indicate the evidence for the involvement of inappropriately chelated iron in cardiovascular diseases too. There is no doubt that the iron-mediated causal chain of ischaemia → (su)peroxide → OH radical formation occurs during the development of heart disease, especially during reperfusion injury [710713], and suitable iron chelators inhibit this [714, 715] (see also [716, 717], and for thalassaemia [718]). Iron is also involved in the protection that can be produced by ischaemic preconditioning [719, 720]. Erythropoietin, a hormone with multiple effects that may involve iron metabolism, is also protective [721, 722].

Heart failure

The sequelae of heart failure are complex, and involve a chronic and continuing worsening of a variety of physiological properties. ROSs are certainly involved here, since allopurinol (a potent inhibitor of xanthine oxidase) improves prognosis considerably [723], and uric acid is a well known biomarker for heart failure (see e.g. [724, 725]). Biopyrrins, degradation products of bilirubin and thus markers of oxidative stress are also considerably increased [726]. Anaemia is a common occurrence (and risk factor) in heart failure [727729], again implying a role for dysregulated iron metabolism and a need to understand the exact speciation of iron in chronic anaemias linked to inflammatory diseases [730].

It is next on the formation of atherosclerotic plaques that our attention is here focussed.

Atherosclerosis

Atherosclerosis is a progressive inflammatory disease[731762] characterized by the accumulation of both oxidised lipids and various fibrous elements in arteries, often as plaques [763, 764]. Iron and oxidised lipids are both found in atherosclerotic lesions [141, 765777], and iron depletion by dietary or other means delays this [778782]. There is a correlation between iron status and atherosclerosis [766, 776, 783794], evidently caused in part by the known ability of poorly liganded iron to effect lipid [765, 784] and protein peroxidation, and by the effects of primed neutrophils [795] and transferrin [758]. In this context, exogenous ferric iron is deleterious to endothelial function [796], while iron chelation improves it [797800]. However, phlebotomy provided no clinical benefit here [801]. Note that iron levels in plaques correlate with the amount of oxidised proteins therein [771], and that in one study [767], the EPR-detectable iron (essentially Fe(III)) in atherosclerotic tissue was seventeen times greater than that in the equivalent healthy tissue; this is not a small effect. (Iron, as part of the general ROS cascade, has also been implicated in gallstone formation, where melatonin has proved protective [802, 803].)

Statins, typically developed on the basis of their ability to inhibit the enzyme HMG-CoA redutase and thus decrease serum cholesterol, are well established to have benefits in terms of decreasing the adverse events of various types of cardiovascular disease [804], albeit that in many populations (e.g. [805807]) cholesterol alone is a poor predictor of cardiovascular disease, especially in the normal range. However, a known target of statins different from HMGCoA reductase is the β2-integrin leukocyte function antigen-1 (LFA-1) [808, 809] and in this context, it is important to note that the clinical benefits of the statins are certainly not due solely to their cholesterol-lowering ability via the inhibition of HMG-CoA reductase (see e.g. [163, 281, 650, 807, 808, 810840]), and different statins can cause a variety of distinct expression profiles [841] that are inconsistent with a unitary mode of action. The apparent paradox [842] that lipid-lowering statins do indeed exhibit epidemiological disease-lowering benefit, while having little effect on plaques, is arguably well explained, especially within the context of the present review, via their additional anti-inflammatory effects [817, 819, 843863, 163, 807, 823825, 830, 836, 837, 864892], acting upstream of the nuclear transcription factor NF-κB (and see later). It is also extremely relevant, for instance, that some statins have metal chelating properties [893].

It has been pointed out that many measures of iron stress are inappropriate, since it is only the redox-active form of iron that is likely involved in oxidative stress. Serum ferritin is considered by some to be the most reliable marker of iron status in general [894], although it is not well correlated with iron distribution in the heart [895], for instance. What is clear, however, from the above is that deranged iron metabolism is intimately and causally involved in the formation of atherosclerotic lesions, and that appropriate iron chelation can help both to prevent and to reverse this.

Iron status is also closely involved in other chronic vascular diseases, and in the behaviour of wounds [896899].

Stroke

Stroke is caused by ischaemia, leading to inflammation [900] and to the formation of ROS and other damaging free radicals [901] in the brain (which is high in metal ions [902]), and is exacerbated by existing inflammation – see e.g. [903, 904]. Thus, another prediction is that iron excess should also aggravate the sequelae of stroke, and that appropriate chelation or free radical trapping agents should mitigate these effects. These predictions are indeed borne out [138, 905914]. It is also of considerable interest that plasma NGAL levels are increased in stroke [406]; it is noteworthy that this can be seen in plasma despite the localised origin of the disease.

A variety of other studies have shown the beneficial treatments in stroke models of anti-inflammatory and antioxidant treatment, i.e. treatments that lower the amount of ROSs (e.g. [915922]), as well as of preconditioning [923]. Given its role in iron metabolism, it is of considerable interest that erythropoietin also seems to be very effective in protecting against brain ischaemia/reperfusion injury and stroke [924939], by a mechanism independent of erythropoiesis [940942], and one that appears to involve anti-inflammatory activity [943].

Alzheimer's, Parkinson's and other major neurodegenerative diseases

Oxidative stress and inflammation are early events of neurodegenerative diseases [920, 944957] such as Alzheimer's disease (e.g. [958973]), where plaque formation precedes neurodegeneration [974]. Iron (and in some cases copper) is also strongly implicated in a variety of neurodegenerative diseases [944, 981, 945, 958, 982985, 962, 986989, 950, 9901021, 917, 10221034, 278, 10351055, 43, 10561059, 285, 1060, 972, 10611065, 141, 975, 10661087].

Indeed Thompson and colleagues comment [136] that "The underlying pathogenic event in oxidative stress is cellular iron mismanagement" and stress that "Multiple lines of evidence implicate redox-active transition metals, such as iron and copper, as mediators of oxidative stress and ROS production in neurodegenerative diseases". There is also ample evidence for its presence in the plaques characteristic of Alzheimer's disease [1004, 1013, 1027, 1088], just as in those of atherosclerosis (see above). Note too that iron can catalyse the oxidation of dopamine to a quinine form that can bind covalently to and then aggregate proteins 1089. Kostoff 1090 has used a very interesting literature-based discovery approach to highlight the role of oxidative stress in the development of Parkinson's disease.

Other papers highlight the role of iron in multiple sclerosis [899, 991, 10911098] and in prion diseases [967, 1099, 1100]. However, a particularly clear example of iron-mediated neurodegeneration is given by the sequelae consequent upon lesions in a protein known as frataxin involved in the disease Friedreich's ataxia (FA).

Friedreich's ataxia

A number of repiratory chain components contain non-heme iron, and the question arises as to how they acquire it 1101. Frataxin is a mitochondrial iron chaperone protein [11021109], involved in the safe insertion of Fe(II) during the production of Fe-S centres in the mitochondrial respiratory chain 1110. As are some other aspects of iron metabolism 1111, it is highly conserved in eukaryotes from yeast to humans [1112, 1113], a fact that made the unravelling of its function considerably easier [11141123]. Friedreich's ataxia (FA) is a neurodegenerative disorder that arises from a genetic deficit of frataxin activity, whether by a missense mutation or, much more commonly, via the addition of GAA trinucleotide repeats [1107, 11241127]. As well as the neurodegeneration and measurable iron deposition, clinical symptoms include cardiac hypertrophy 1128 and (pre-)diabetes 1129, consistent with the general thesis described here that all are in part manifestations of iron dysregulation, and in which suitable chelation may be beneficial [1130, 1131] (but cf. 1132).

ROSs are undoubtedly involved in FA [1133, 1134], specifically via Fenton chemistry [1135, 1136], since the attenuation of H2O2 production (but not of superoxide) 1110 ameliorates the disease 1137. The deficit in frataxin causes both an increase in ROS (H2O2) production via the mitochondrial electron transport deficiency 1138 as well as a dysregulation in iron metabolism, potentially a very damaging synergistic combination (see later). Elements of its (in)activity that are seen as paradoxical 1139 are in fact easily explained when one recognises that it is the combination of free iron with H2O2 that is especially damaging. The neonatally lethal GRACILE syndrome is also caused by a failure of iron chaperoning into mitochondrial complex III due to mutations in the BCS1L gene [1140, 1141].

Amyotrophic lateral sclerosis (ALS) or Lou Gehrig's disease

ALS is another progressive inflammatory 1142 disease in which motor neuron death causes irreversible wasting of skeletal muscles. It has largely defied efforts to uncover the genetic basis of any predisposition 1143, save for a very clear association with defects in a Cu/Zn superoxide dismutase [33, 11441146] that can obviously lead to an increase in the steady-state levels of superoxide (and hence hydroxyl radical formation). There is also significant evidence for the involvement of iron [1147, 1148]. Drug therapies have to date shown rather limited benefits, and more in mouse models of Cu/Zn SOD deficiencies than in humans, though iron chelation therapy does not seem to have figured heavily, and it is recognised that combination therapies might offer better prognoses 1149.

Aging

Aging or senescence is defined as a decline in performance and fitness with advancing age 1150. Iron stores tend to increase with age [11511155], partly due to dietary reasons 1156 (and see [1157, 1158]), as does anaemia [1159, 1160]. So too does the expression of NGAL/Lcn2/siderocalin, a process that can be reversed by melatonin 1161. Mainstream theories of aging [163, 165, 11621180] recognise the relationship between progressive inflammation, cellular damage and repair and the higher-level manifestations of the aging process, and ('the free radical theory of aging' [1163, 1181]) ROSs are of course strongly implicated as partial contributors to the aging process (e.g. [31, 43, 980, 1163, 11821206]). Needless to say, not least because of the low steady-state net rate of generation of the various ROSs 1207, few studies have managed to be very specific mechanistically 1208, but it should be clear that all ROSs are not created equal and we need here to concentrate mainly on the 'nasty' parts of ROS metabolism, and in particular on the hydroxyl radical as generated via poorly liganded iron and on peroxynitrite, and to have the greatest effects we need to inhibit both their generation and their reactivity (see Systems Biology section, below). The iron content of cells also increases as cells age normally 1209. As many diseases increase with age, probably via mechanisms highlighted herein, treating aging can thereby treat disease [162], and it is important to recognise that most 'diseases' are in fact consequences of aging (despite the considerably greater historical focus on the former).

Frailty

One issue of aging is not that just it happens but that it can manifest in a series of essentially undesirable physiological changes, referred to as frailty 1210, in which ROSs have also been strongly implicated. Indeed, there are many parts of physiology and metabolism that lose functionality during aging (e.g. the cardiovascular system 1211 and of course cognitive function 1212), and iron metabolism is known to change considerably as humans age [980, 1213], with anaemia a typical accompaniment of aging 1214. The question then arises as to how much of this deranged iron metabolism is causal in accelerated aging, and this is not easy to state at this time. However, lowering iron does increase the lifespan of Drosophila 1215 and yeast 1105. At all events, the purpose of this rather brief section is to point out to researchers in aging, frailty and gerontology generally the relevance of inadequately controlled iron metabolism as a major part of ROS-induced injuries that may accelerate the aging process.

Longevity

Although aging and longevity are not of course the same thing, and longevity is not a 'disease', studies of aging are often performed with the intention of improving our understanding of longevity 1216, and certainly longevity is linked to age-related disease 1217. However, the longevity of many organisms can be varied by the manipulation of any number of diseases or processes. This said, caloric or dietary restriction is a well-known contributor to longevity (indeed the only reliable one in pretty well all species [12181223] – although possibly not H. sapiens 1180), and appears to act at the root of the processes involved 1224. Caloric restriction appears to be associated with a considerably lowered rate of production of ROSs and accrual of ROS-induced damage [1207, 12251228], and this is to be expected on general grounds in that a restriction of substrate supply will make the redox poise of mitochondria 1229 more oxiding and thereby minimise the amount of 1- and 2-electron reductions of O2 to form peroxide and superoxide. It is therefore also of great interest that caloric restriction also benefits iron status 1230 and that it is this improved iron status that in part promotes longevity 1231. Caloric restriction has also been shown to effect a differential stress response between normal and tumour cells 1232, although this study did not look at relative iron status.

Antioxidants can also influence lifespan. Thus (rather high doses of) melatonin extended the lifespan (and stress resistance) of Drosophila [1191, 12331235] while that of Caenorhadbitis elegans could be extended by mimics 1236 of SOD and catalase 1187, and by a variety of antioxidant and other pharmacological agents 1237.

As an example, let us consider C. elegans. Mutants with a decreased activity of the insulin-like growth factor signalling pathway (e.g. daf2 mutants that have a greater amount of the DAF16 FOXO-like transcription factor) can live for nearly twice as long as wild types [1238, 1239] and produce more catalase, superoxide dismutase (sod-3) 1240 and glutathione-S-transferase.

Overall, it is the potent combination of oxidative stress, already leading to damaging peroxides and radicals, and its catalysis and further reactions caused by inappropriately chelated iron, that causes a 'double whammy'. Indeed, iron, copper and H2O2 have been referred to as the 'toxic triad' 1032. While there is comparatively little that we can do about the production of superoxide and peroxide, we can (by pharmacological or dietary means) try and improve the speciation of iron ions.

Rheumatoid arthritis

One disease whose aetiology is well known to be bound up with ROSs is rheumatoid arthritis (RA) [36, 12411246]. What is known of the role of iron metabolism? Generally an overall low iron status – anaemia – is a characteristic of rheumatoid arthritis [12471251], whereas by contrast iron is elevated in the synovial fluid of arthritic joints [12521255]. This suggests a significant derangement of iron metabolism in RA as well, and a mechanism [12561259] in which elevated superoxide liberates free iron from ferritin in synovial fluid (and elsewhere 1260), thereby catalysing further the damaging production of hydroxyl radicals. This autocatalytic process (Fig 6) is, even in principle, especially destructive (and may account for species differences in sensitivity to iron loading 1261). Note that erythrocytes when oxidized can also release free iron [524]. Natural antioxidants such as vitamin E are also lowered 1262. There is some evidence that appropriate iron chelators can ameliorate the symptoms of RA 1263, though membrane-impermeant chelators such as desferroxamine cannot 1264.

Figure 6
figure 6

Catalysis and autocatalysis in the Haber-Weiss and Fenton reactions leading to the production of the hydroxyl radical, including the liberation by superoxide of free iron from ferritin.

One interesting feature of RA is that in 75% of women it is strongly ameliorated during pregnancy [12651267]; although the multifactorial nature of this observation has made a mechanistic interpretation difficult, from everything that we have seen so far it would be surprising if changes in iron metabolism were not strongly involved.

An interesting related feature is the 'restless legs syndrome' [12681270], that is often associated with iron deficiency and pregnancy. Serum transferrin receptor seems to be a rather sensitive measure of this iron deficiency [273, 1271, 1272]. There are relationships with other syndromes that we discuss here, such as cardiovascular disease 1273, but in the present review it is of especial interest that a dysregulation of iron metabolism appears to play a significant role 1274

Lupus (Systemic Lupus Erythematosus)

Lupus, or Systemic Lupus Erythematosus (SLE) [1275, 1276] describes a syndrome, somewhat related to arthritis and rheumatism, of broadly auto-immunological or inflammatory origin, and with a large variety of manifestations (e.g. [12771285]), but characterised in particular by fatigue 1286, often as a result of anaemia. This of course points to a certain level of iron dysregulation (of any number of causes), and there is certain some evidence for this [1274, 1287]. Thus, while anaemia is a feature of the disease, serum ferritin may be raised in SLE 1288, and some of the usual lipid markers of oxidative stress (that can be a result of hydroxyl radical production catalysed by poorly liganded iron) are also present 1289.

There is also a very interesting linkage between SLE and vitamin D metabolism 1290, something that has also come up in relation to the statins and atherosclerosis ([832, 1291], but cf. 1292), and indeed it there is evidence that statins may be of benefit in the treatment and even reversal of lupus [1293, 1294]. Indeed, from a much more general point of view, there is precedent for these kinds of linkages being used to uncover unknown mediators in disease states that may be worth pursuing (e.g. [6, 19, 20, 1295, 1296]).

Asthma

Asthma is a well-known inflammatory disease, and has been linked with ROS generation 1297 catalysed by iron [276, 12981300].

Inflammatory bowel diseases (IBD)

By definition, IBD such as Crohn's disease and ulcerative colitis are inflammatory diseases, and while the inflammation and ROS production are well established here, their origins are somewhat uncertain [13011304]. They are frequently accompanied by anaemia, implying a derangement in iron absorption and/or metabolism [1305, 1306], and very probably absorption [1307, 1308]. The anaemia may be monitored by ferritin and transferrin receptor levels, and its correction is possible by iron supplementation plus erythropoietin [13091315]. One may suppose that some of the issues here relate to iron speciation, that is usually not measured in these studies.

Age-related macular degeneration

Age-related macular degeneration (AMD) 1316 is now the leading cause of blindness and visual disability in the elderly in developed countries [13171320]. Many components of atherosclerotic plaques have also been demonstrated in drusen 1321, a characteristic of AMD and, as here, it is reasonable to propose a common mechanism of pathogenesis between AMD and atherosclerosis. Retinal iron levels increase with age 1322, iron is significantly implicated in AMD [13231329], and iron chelation may help to reverse the process 1330. Dietary antioxidants are also protective 1331. The source of the iron appears to be excess angiogenesis and leakage from blood vessels catalysed by VEGF, and a PEGylated aptamer [13321334] against VEGF (pegaptanib) or a monoclonal antibody (ranibizumab) have shown significant promise in the treatment of macular degeneration [13351340]. Plausibly a combination therapy with one of these plus a suitable iron chelator might be even more effective.

Psoriasis

Psoriasis is an anflammatory disease in which the production of free radicals and ROSs are strongly implicated [13411343]. Here too there is clear evidence for the involvement of a deranged iron metabolism [13431345]. Early attempts at therapy with a series of unusual iron chelators (that unfortunately had side effects) 1346 do not seem to have been followed up.

Gout

Gout is another important inflammatory disease, characterised by the accumulation of uric acid 1347. There is considerable evidence that this too is a disease of iron overload, and that uric acid accumulation – as both an antioxidant and an iron chelator 1348 – is in response to the iron overload [1349, 1350] and with highly beneficial remission of gouty symptoms occurring on depletion of iron by phlebotomy 1351.

Alcoholic and other forms of liver disease

It is known that with chronic excess, either iron or alcohol alone may individually injure the liver and other organs, and that in combination, each exaggerates the adverse effects of the other. Specifically, in alcoholic liver disease, both iron and alcohol contribute to the production of hepatic fibrosis [13521358]. Iron overload is well known to lead to hepatotoxicity [13591362] and liver cancer [1363, 1364], and lowering or chelating it is protective [1365, 1366]. Hepcidin may be involved here 1367.

Chronic obstructive pulmonary disorder (COPD) and related lung diseases

Chronic obstructive pulmonary disease (COPD) is a progressive and chronic disease which is characterised by an apparently inexorable decline in respiratory function, exercise capacity, and health status. It is also characterised by periods in which the symptoms are considerably exacerbated [13681371]. Such an acute exacerbation of COPD (AECOPD) is defined 1372 as "a sustained worsening of the patient's condition from the stable state and beyond normal day to day variations, that is acute in onset and necessitates a change in regular medication in a patient with underlying COPD". Smoking is a major source of free radicals (and indeed metals 1373), and is a major cause of COPD. Consequently, there is considerable evidence for the evidence for the involvement of inflammation and ROSs in both the 'stable' and 'exacerbated' stages [13741381].

Needless to say, there is also considerable evidence for the significance of exposure to iron 1382 (as well as exposure to other toxic metals 1383) in the development of COPD and other lung diseases 1384. Haem oxygenase also appears to be a significant culprit [280], and lung (lavage) iron is increased 1385 while transferrin levels can be considerably lower 1386.

Other lung diseases in which ROS and iron have been implicated include Adult Respiratory Distress Syndrome [13871390] and cystic fibrosis 1391.

Smoking

Tobacco smoke contains many unpleasant and carcinogenic compounds, and that tobacco smoking is a leading cause of carcinoma of the lung and indeed of other organs has become well known since the pioneering epidemiological studies of Doll, Peto and colleagues (e.g. [13921395]). Our purpose here is to point out that many of the particles associated with smoking (and also ingested from other sources) are heavily laden with micro-particulate iron, which, as a major catalyst of hydroxyl radical production, undoubtedly is a substantial contributor as well (see e.g. [1373, 1384, 13961400]).

Cancer and oncology

In addition to the issues of smoking, the development of cancer can certainly contain an inflammatory component (e.g. [14011425]), and indeed the long-term use of prophylactic anti-inflammatory aspirin lowers colon cancer incidence by 40% (age-adjusted relative risk = 0.6) 1426 (though note that this may have other side-effects 1427). (The well-known association between infectious agents such as H. pylori [14281430] and e.g. bowel cancer is probably initiated by chronic inflammation.) Given that cells require iron, restricting its supply can also limit the growth of cells, including tumour cells [14311442]. Conversely the iron carrier NGAL is overexpressed in tumours [410, 411, 1443], a process mediated via NF-κB 1444 (and see later). Further, the roles of iron, not least in the mutagenic effects of metal-catalysed Fenton chemistry, are also of significance in promoting oncogenesis [14451461]. Iron chelators [1438, 1462] are thus a doubly attractive component of anti-cancer therapeutics. The mutagenic, carcinogenic and disease-causing actions of asbestos and related fibres may also be due in significant measure to the ability of the Fe(n) that they contain to catalyse hydroxyl radical production [14631475], while there seem to be complex relations between the likelihood of apoptosis and the differential concentrations of superoxide and H2O2 [1476, 1477]. Overall, it is becoming increasingly widely recognised that anti-inflammatory agents have a role to play in the treatment of cancers; we would suggest that iron chelation may be a useful component of such treatments.

Malaria

Just as do tumour cells, the malarial parasite Plasmodium falciparum requires considerable iron for growth, and there is evidence that lowering the amount of available iron provides a promising route to antimalarials (e.g. [14781494], but cf. 1495). Note in this context that iron-catalysed radical formation is also significantly involved in the antimalarial (i.e. cytotoxic) mode of action of artemisinin [1494, 14961504], and this reaction is in fact inhibited by iron chelators 1505 such that a combined artemisinin-chelator therapy would be contraindicated.

Antimicrobials

Lower down the evolutionary scale, and as presaged earlier in the section on bacterial siderophores, microbes require iron for growth, its presence may be limiting even at the scale of global CO2 fixation [15061509], its excess can in some circumstances 1510 correlate with infectivity or virulence (see above and e.g. [180, 193, 1493, 15111531]), and its chelation in a form not available to bacteria offers a route to at least a bacteriostatic kind of antibiotic or to novel therapies based on the lowering of iron available to microbes by using hepcidin 1532 or NGAL 1533. Iron chelators are also inhibitory to trypanosomes [1534, 1535], and changes in iron metabolism are also associated with viral infections 1536.

Sepsis leading to organ failure and death: severe inflammatory response syndrome

It is well known that one consequence of bacterial infection (sepsis) can be septic shock, that this can be mimicked by the Gram-negative bacterial outer membrane component LPS (lipopolysaccharide), and that in the worst cases this leads via multiple organ failure to death. However, whatever LPS does it is quite independent of the present of viable (i.e. growing or culturable – see [1537, 1538]) bacteria as the same phenomena leading to multiple organ failure (MOF) are seen without infection [1539, 1540]. Consequently, the recognition of a series of symptoms contingent on this initial inflammatory response has led to the development of the idea of a Systemic Inflammatory Response Syndrome (SIRS) [15411551] that leads to the MOF, both via apoptosis 1552 and necrosis [1553, 1554]. There is by now little doubt that these phenomena too are associated with the hyperproduction of ROSs [1546, 1551, 15551570]. Circulating free iron is raised in sepsis and related conditions [1400, 15711573]. Direct assays of oxidant induced cell death indicate that most 'free' iron is concentrated in lysosomes [15741577], that its decompartmentation is substantially involved 1570, and that its chelation can thus prevent cell death [15781581].

Many circulating inflammatory factors have been identified as important in the development of septic shock, including cytokines such as Tissue Necrosis Factor (TNF) 1582, and cellular responses via the Toll-Like Receptor are clearly involved in this process [1583, 1584]. However, we would argue that since antibodies against TNF do not inhibit the sequelae of septic shock such as multiple organ failure, the truly damaging agents are caused elsewhere and are likely to involve the iron-mediated production of damaging hydroxyl radicals (see also 1563).

In this regard, it is especially interesting that the antioxidant melatonin is particularly effective in preventing septic shock [15851587], and a variety of suitable antioxidants have shown potential here [1573, 15881591], notably in combination with iron chelators [1592, 1593] (and see also [1594, 1595]). As with quite a number of the indications given above, a further link with Fe metabolism is seen in the protective effects of erythropoietin [1596, 1597].

Pro- and anti-oxidants and their contributions to cellular physiology

A very great many cellular metabolites are redox active within the range of redox potentials realistically accessible to biology (including some molecules such as proline 1598 that are not commonly considered to be redox-active), and it is not our purpose here to list them extensively. Not only their redox potential and status but even their absolute amounts can have profound effects on metabolism 1599). Our chief point here is that it is the intersection of iron metabolism and oxygen reduction that needs to be the focus, with the 'iron'-catalysed production of hydroxyl radical being the nexus, with the standard redox potential of a redox couple per se being less significant in absolute terms, and the redox potential that a particular redox couple 'feels' being dependent in a complex manner on a variety of thermodynamic and kinetic factors 1229. Thus, although ascorbate is 'reducing' and an 'antioxidant', its reaction with O2, especially when catalysed by Fe(II), produces superoxide and thence OH radicals that may be pro-oxidant. It is this kind of stepwise multi-electron-transfer phenomenon that explains the otherwise possibly puzzling observation of the oxidant-induced reduction of respiratory chain components (see e.g. [1600, 1601]). Consequently, it is extremely unwise to make pronouncements on the role of 'ROSs' without being quite explicit about which ones are meant.

Thus anything – even an antioxidant – that e.g. by reaction with O2 produces superoxide, peroxide and hydroxyl radicals will turn out to be a pro-oxidant if the flux to superoxide and in particular to hydroxyl radicals is stimulated. Thermodynamically, the 1-electron reduction by ascorbate of dioxygen is disfavoured, with the 2-electron reduction to peroxide being the thermodynamically preferred route. However, such reactions are heavily restricted kinetically in the absence of any catalysts [130]. It is an unfortunate fact that the oxygen-mediated "autoxidation" of ascorbate does in fact occur at considerable rates when it is accelerated by the presence of iron or other transition metal ions [130, 16021606]. In a similar vein, 'free' or inadequately liganded Fe(II) catalyses the production of hydroxyl radicals from oxygen plus a variety of natural biomolecules, including adrenaline (epinephrine) 1607, haemin 1608, and even peptides such as the amyloid-β involved in the development of Alzheimer's disease [1009, 1027]. Dietary antioxidants (see below) can therefore act in complex and synergistic ways depending on iron status 1609. In this regard, the idea of using elemental iron plus ascorbate in food supplements 1610 does not seem a good one.

It should be noted that there are also occasions, e.g. in the decomposition of refractory polymers such as lignin, where such radical production is involved beneficially 1611.

Finally, a variety of molecules can trap hydroxyl radicals, including hippurate 1612, melatonin [1233, 1585, 1586, 16131631] and salicylate 1632.

Antioxidants as therapeutic agents? Should we be including iron chelators in such clinical trials?

Given the wide recognition of the importance of ROSs in a variety of diseases as described above, many investigators have considered the use of known antioxidants such as vitamins C (ascorbate) and E (α-tocopherol) in preventative therapy. Although there have been some successes (e.g. 1633), the results have generally been decidedly mixed, with little clinical benefit (or even actual disbenefit) following from their administration [692, 1588, 16341639], e.g. for ALS 1640, atherosclerosis [106, 764], cardiovascular disease [16411645], neuroprotection 1646, macular degeneration 1647, pre-eclampsia [16481652], critical care medicine 1653, aging [1165, 1168, 1654, 1655], lung disease 1656, elective surgery involving ischaemia-reperfusion 1657, all-cause mortality [1637, 1658], etc. One interpretation for these disappointing results, that is consistent with the general theme of this review, involves the recognition that a variety of antioxidants can in fact act as pro-oxidants and thus actually promote the production of OH radicals in the presence of inappropriately or inadequately liganded Fe(II) [130, 528, 1602, 1604, 1659, 1660]. (One might also comment that the intracellular location of the antioxidants may be an issue, and that the view that targeting them to mitochondria may well have considerable merit [16611663].) Thus any predictions about the utility or otherwise of antioxidants need to take into account the amount of 'free' iron present. In particular, we would suggest that future trials of this type might beneficially include appropriate iron chelators, whether alone or with anti oxidants.

Liganding and reactivity of Fe(n)

Given the damage that iron-mediated OH radical can create, the question arises as to whether appropriate chelators can inhibit this by inhibiting the production of OH, and while the answer is 'yes' the interpretation of the relevant experiments is not always as clear cut as one would wish [43]. This is because the OH radical is so reactive that its production is normally assessed by addition of the putative chelator and observation of its effect on the rate of reaction of a target molecule such as salicylate with the OH generated. The ability of a chelator to inhibit such a reaction can then occur not only via a reduction in the rate of OH production but by trapping the OH itself, as well as by other mechanisms 1664. This said, there is little doubt that iron chelators can be highly protective, and it is many ways very surprising that their use is not more widespread.

We begin by noting that the reactivity of iron does vary greatly depending upon its liganding environment [71]. Cheng et al. state [72] "Oxygen ligands prefer Fe(III); thus, the reduction potential of the iron is decreased. Conversely, nitrogen and sulfur ligands stabilize Fe(II); thus, the reduction potential of the iron is increased. Therefore, chelators with oxygen ligands, such as citrate, promote the oxidation of Fe(II) to Fe(III), while chelators that contain nitrogen ligands, such as phenanthroline, inhibit the oxidation of Fe(II). Many chelators, such as EDTA and Desferal (DFO), will bind both Fe(II) and Fe- (III); however, the stability constants are much greater for the Fe(III)-chelator complexes. Therefore, these chelators will bind Fe(II) and subsequently promote the oxidation of the Fe(II) to Fe(III) with the concomitant reduction of molecular oxygen to partially reduced oxygen species. Since the maximal coordination number of iron is six, the hexadentate chelators can provide more consistently inert complexes due to their ability to completely saturate the coordination sphere of the iron atom and, consequently, deactivate the "free iron" completely. For example, DFO is a very effective antioxidant in clinical application because of its potential to markedly decrease the redox activity of iron [137]." However, it is not easy to make hexadentate ligands orally active 1665.

Iron typically can coordinate 6 ligands in an octahedral arrangement. Preferential chelation of the Fe(II) or the Fe(III) form necessarily changes its redox potential as a result of Le Chatelier's principle, and from Marcus theory [16661669] the rate of outer-sphere electron transfer reactions is typically related to differences in the free energy change, i.e. the differences in redox potentials of the interacting partners. In addition, it widely recognised that [137] "The tight binding of low molecular {weight} chelators via coordinating ligands such as O, N, S to iron blocks the iron's ability to catalyze redox reactions. Since the maximal coordination number of iron is six, it is often argued that the hexadentate chelators can provide more consistently inert complexes due to their ability to completely saturate the coordination sphere of the Fe atom. Consequently, a chelator molecule that binds to all six sites of the Fe ion completely deactivates the "free iron". Such chelators are termed "hexidentate" {sic}, of which desferrioxamine is an example. There are many Fe chelators that inhibit the reactions of Fe, oxygen, and their metabolites. For example, desferrioxamine ... (DFO) markedly decreases the redox activity of Fe(III) and is a very effective antioxidant through its ability to bind Fe."

By contrast, bidentate or tridentate chelators that bind to only 2 or 3 of the available iron chelation sites, especially when they bind to both Fe(II) and Fe(III), can in fact catalyse redox cycling and thereby promote free radical generation [1437, 1665, 1670, 1671]. Thus, the most potent iron chelators will normally be hexadentate (but may consequently strip iron from iron-containing enzyme and thereby have deleterious side effects). Bi- or tri-dentate ligands should therefore be at saturating concentrations for maximum effect.

Generally, the harder ligands that favour Fe(III) involve O whereas softer ligands that bind Fe(II) involve N and S. The type of ligand also influences the absorption spectrum of the ferric form of the chelator, such that conclusions can be drawn about the types of group involved in the complex. These charge transfer bands that appear on ligand binding are at around 340 nm for carboxylates, around 425 nm for trishydroxamates, 470 nm for bis-hydroxamates, 515 nm for monohydroxamates, around 480 nm for tris-catecholates, 560 nm for bis-catecholates and 680 nm for mono-catecholates [173]. In addition, for tris-bidentate complexes the complex can, on an octahedral arrangement, have two different configurations, a left-handed propeller, termed the Λ-configuration, and a righthanded propeller, the Δ-configuration [173].

Iron chelators – those approved and used clinically

A number of reviews (e.g. [1437, 1438, 16721678]) cover aspects of iron chelators that have had or may have utility clinically.

Whitnall and Richardson 1062 list a number of useful features of an experimentally (and clinically) useful iron chelator. Thus, "A compound suitable for the treatment of neurodegenerative disease should possess a number of qualities, namely (1) strong affinity for FeIII, (2) low molecular weight, (3) lipophilicity high enough to accommodate permeation of cell membranes and the BBB, (4) oral activity, and (5) minimal toxicity 1062. Also, partly because there are few trivalent ions other than Fe(III) that the body actually needs, the major synthetic focus has been on the design of FeIII-selective chelators which feature "hard" oxygen donor atoms. Additionally, under aerobic conditions, high-affinity FeIII chelators will tend to chelate FeII to facilitate autoxidation, such that high-affinity FeIII-selective compounds will beneficially bind both FeIII and FeII under most physiological conditions" 1062. (Note that liophilicity per se may not be relevant, as drugs require carriers to cross membranes [18], and promiscuity and off-target effects increase with lipophilicity 1679.)

Desferrioxamines are nonpeptide hydroxamate siderophores composed of alternating dicarboxylic acid and diamine units. linked by amide bonds. They are produced by many Streptomyces species 1680. Desferrioxamine B is a linear (acyclic) substance produced (industrially) by the actinobacterium Streptomyces pilosus 1681, and is widely used as an iron chelator for the prevention and treatment of the effects of iron overload. It is commercially available as desferal (desferrioxamine methane sulphonate), also known as deferoxamine in the USA. It has been very effective in the treatment of a number of diseases, leading to the view that such molecules should have considerable therapeutic potential. A significant disadvantage of DFO is that it does not seem to cross the intestine intact (despite the rather catholic substrate specificity of intestinal peptide transporters [16821685]) and must therefore be given intravenously or subcutaneously. By contrast, another chelator known as Deferriprone or L1 does appear to cross cell membranes, but it is only bidentate.

Those with approval for clinical use are few in number and we deal with them first. Table 1 compares them with the 'ideal' properties of a clinically useful iron chelator, while Fig 7 gives the structure of the three most common, viz. desferal (deferoxamine), ferriprox (L1 or deferiprone) and exjade (ICL670 or deferasirox). (Dexrazoxane, a hexadentate chelator, is also marketed 1686.)

Figure 7
figure 7

Some iron chelators that are in clinical use (left hand side) or that have been proposed.

Table 1 Comparison of the main available iron chelators to an ideal chelation drug (modified from 2469)

Desferal (deferoxamine) is the most used chelator for historical reasons. It is hexadentate but is not orally bioavailable. Ferriprox (deferiprone) is a bidentate ligand (1,2-dimethyl, 3-hydroxypyridin-4-one). It is orally bioavailable although comparatively high doses are required, and it postdates desferal. "Deferiprone has high affinity for iron and interacts with almost all the iron pools at the molecular, cellular, tissue and organ levels. Doses of 50–120 mg/kg/day appear to be effective in bringing patients to negative iron balance" 1687. It can have somewhat better properties than desferal 1688. Finally, Exjade (ICL670) (deferasirox) [16891702] is the most recent chelator approved for clinical use, and is tridentate. It is orally active, and there is a large bibliography at http://www.exjade.com/utils/bibliography.jsp?disclaimer=Y. The recommended initial daily dose of Exjade is 20 mg/kg body weight.

It is clear from Table 1 that in the time evolution from deferoxamine through deferiprone to deferasirox there has been a noticeable improvement in the general properties of iron chelators, although there are few published data on the quantitative structure-activity relationships of candidate molecules that might allow one to design future ones rationally. What is certainly clear is that there is a trade-off in properties, and that appropriate chelators will keep iron levels intermediate, i.e. not too low and not too high (a 'Goldilocks' strategy, if you will), and that hexadentate molecules may correspondingly be too tightly binding and strip iron from important molecules that need it. What is particularly important, as well as a good plasma half-life, is the ability to cross cell membranes, as this is necessary both for oral administration and for ensuring that the chelator in question actually accesses the intracellular 'free' iron pools of interest. Which carriers are used for this in humans in vivo is presently uncertain [18, 1703].

Drugs that have been approved for clinical use for other purposes, but that also happen to be iron chelators

The high investment of time, money and intellectual activity necessary to get a drug approved clinically has led to a number of strategies to exploit those that already have been approved and are thus considered 'safe'. One such strategy is the combination therapy of approved drugs that can yet serve for novel indications (e.g. [17041708]). Another strategy is to look for antioxidant or iron-binding chemical motifs in drugs that have already been approved for other purposes 1709 (or to measure such properties directly).

Clioquinol (CQ) [1062, 1674, 1710, 1711] (Fig 7) is one existing (anti-parasitic) drug that has been proposed for use as an iron chelator, as it contains the known iron-chelating 8-hydroxyquinoline moiety. It has indeed enjoyed some success in this role. However, clioquinol toxicity has been reported if it is used over an extended period 1712 and this may be due to the formation of a Zn-clioquinol chelate 1713.

A particular attraction of such existing drugs is that they are likely to have favourable pharmacokinetics and pharmacodynamics, and in particular are likely to be cell-permeable. Note that despite a widespread assumption that lipophilicity or log P is sufficient to account for drug distribution this is not in fact the case, as there are hundreds of natural transporters that drugs can use (e.g. [18, 1714]). For instance, the iron-chelating 8-hydroxy quinoline motif contained in molecules such as clioquinol is also present in the tryptophan catabolite xanthurenic acid (Fig 7), and it is likely that transmembrane transport of the synthetic drug molecule occurs via natural carriers whose 'normal' role is to transport endogenous but structurally related molecules [18, 1703].

Iron chelators that have been studied but not yet approved

Given the importance of the field, many academic investigators have sought to develop their own iron chelators that might exhibit the desirable properties listed above. One class of molecule includes isonicotinylhydrazones. Thus, pyridoxal isonicotinyl hydrazone (PIH) [1672, 17151720] is a promising molecule (also proposed in anti-cancer therapy), although it is hydrolysed both in vivo and in vitro 1721. Other analogues include salicylaldehyde (SIH) 1722 and 2-hydroxy-1-napthylaldehyde (NIH) isonicotinyl hydrazones. PIH was disclosed before being patented, and is thus seen as having no pharmaceutical (company) interest. Various other derivatives are therefore being considered [1035, 1462], including pyrazinylketone isonicotinoyl hydrazones 1723.

A variety of 8-hydroxyquinolines (8HQs) 1724 have been considered, although as with other bidentate and tridentate ligands that cannot necessarily effect complete liganding of iron there is always a danger that inadequate concentrations might be pro-oxidant (e.g. [1725, 1726]). One molecule, VK-28, combines various pertinent moieties and has shown some promise in the treatment of neurological disorders [17271730]. This strategy of combining drug elements that can hit multiple targets ('polypharmacology' [807, 17311734]) has much to commend it, including on theoretical grounds, and we discuss these in the section on systems biology below. Another 8HQ that has elicited interest is O-trensox [1431, 17351742].

Other ligands or motifs that might be considered include di-2-pyridylketone-4,4,-dimethyl-3-thiosemicarbazone (Dp44mT), that has been shown to be effective against tumours 1743, 2,2'-dipyridyl, 1,10-phenanthroline [1744, 1745], 2-benzoylpyridine thiosemicarbazones 1746 and thiohydrazones 1747. HBED (Fig 7) (N,N'-bis-(2-hydroxybenzyl)ethylenediamine-N,N'-diacetic acid) forms a 1:1 complex with Fe(III) but is probably only tetradentate 1748. It seems not to be very orally active 1749 but may be more effective than is DFO [17501752]. Poly-hydroxylated 1,4-naphthoquinones occur as sea urchin pigments and have shown protective effects 1753.

Continuing the theme of polypharmacology, R-(α)-lipoic acid [17541757] is also an antioxidant, that may in addition act by stimulating other anti-oxidant pathways 1758. Finally, one interesting area is that of prochelators (e.g. 1759) in which the oxidant itself triggers the formation of a chelator able to inhibit the Fenton reaction.

Utility of iron chelators in disease amelioration

Therapeutic uses of iron chelators have been widely and usefully reviewed (e.g. [1437, 1489, 1676, 17601767]). Many problems remain, such as bioavailability, mis-dosing 1768 (too little iron as well as too much of it can be bad), toxicity, selectivity and so on, and their design is consequently highly non-trivial [1665, 1670]. Nevertheless, iron chelators have demonstrated therapeutic benefits in Alzheimer's [1674, 1710, 17691771], Parkinson's [1037, 1729, 1772], cold-induced brain injury [1773, 1774], coronary disease [714, 797], renal diseases 1775, various kinds of infection 1763 and of course in iron overload diseases [1762, 1767].

As mentioned above, one interesting strategy is to devise chelators that are only activated by oxidative stress [1760, 17761779]. Another is to seek to combine different kinds of functionality in the same molecule. To this end, Youdim and colleagues in particular have developed a series of multifunctional 8-hydroxyquinoline 1740 derivatives that are effective bidentate iron chelators and that seem to show considerable promise in the treatment of a variety of neurodegenerative diseases [1037, 17271729, 17801784] (see also US Patent 20060234927). In this case the antioxidative mechanism is clearly via chelation since such (8-hydroxyquinoline) molecules are poor scavengers of radicals directly 1785, a fact that also makes them useful scientific tools. As bidentate ligands they cross both cell membranes and the BBB fairly easily (though lipophilicity per se seems not to be important for the biological activity of 8-hydroxyquinoline chelators [1737, 1739]). Importantly, the comparatively weak bidentate binders seem not to have major long-term effects if used carefully [1483, 1762, 1786, 1787]

Interaction of xenobiotics with iron metabolism

As Cherny and colleagues point out 1710, there are many US Pharmacopaeia-registered drugs that, while not being termed chelators, do in fact have both chelating properties and favourable toxicity profiles. Thus we need to recognise potentially both positive and negative interactions between drugs in general and iron metabolism. Any drug that can bind iron can also catalyse the formation of free radicals. Thus, gentamicin can form a gentamicin-iron complex that can lead to toxic symptoms such as hearing loss; this is reversed by iron chelators [1788, 1789]. Existing drugs other than iron chelators may also have effects on iron metabolism 1790, and iron can catalyse their oxidation 1791. It is not, of course, news that drugs have multiple effects. In this context, we reiterate that some statins, for instance, have chelating properties [893].

Other toxicants might also mediate their damaging effects through iron-catalysed radical formation [1792, 1793]. This in addition to the well-known iron-catalysed, radical-mediated mechanism of toxicity of the viologens such as diquat and paraquat [17941799] (whose herbicidal activity is in fact inhibited by iron chelators 1800) and of adriamycin [1801, 1802]. As mentioned above, the carcinogenic action of asbestos may also be due to the ability of the Fe(n) that it contains to catalyse hydroxyl radical production [1469, 1475], while carcinogenic mycotoxins such as aflatoxin may interact synergistically with iron 1803.

Dietary sources of iron chelators

There is also a considerable and positive role for nutrients in terms of their chelation of iron. Indeed, polyphenolic compounds, many of which have known health benefits [18041813], are widely used as food antioxidants [1814, 1815]. There is of course considerable epidemiological evidence for the benefits of consuming fruit and vegetables that are likely to contain such antioxidants (e.g. [18161819]), and – although possibly a minimum – this has been popularised as the 'five a day' message (e.g. http://www.fruitsandveggiesmatter.gov/ and http://www.5aday.nhs.uk/). Even though elements of the 'Mediterranean' diet that are considered to be beneficial are usually assumed to be so on the basis of their antioxidant capabilities (but cf. 1820), many of the polyphenolic compounds (e.g. flavones, isoflavones, stilbenes, flavanones, catechins (flavan-3-ols), chalcones, tannins and anthocyanidins) [18211828] so implicated may also act to chelate iron as well [1073, 18291843]. This is reasonable given that many of these polyphenols and flavonoid compounds [1821, 18441853] have groups such as the catechol moiety that are part of the known iron-binding elements of microbial siderophores. Examples include flavones such as quercetin [914, 1813, 1829, 18541864], rutin [1829, 1857, 1858, 1865, 1866], baicalin [1860, 1867], curcumin [1813, 18681872], kolaviron 1873, flavonol 1874, floranol 1875, xanthones such as mangiferin [18761879], morin 1876, catechins [1073, 1807, 1838, 1854, 1880, 1881] and theaflavins 1882, as well as procyanidins [1835, 1883] and melatonin [1628, 18841887]. However, the celebrated (trans-)-resveratrol molecule [18881902] may act mainly via other pathways.

A considerable number of studies with non-purified dietary constituents containing the above polyphenolic components have also shown promise in inhibiting diseases in which oxidative stress is implicated [1825, 19031906]. For instance in stroke and related neuronal aging and stress conditions, preventative activity can be found in blueberries [19071913] (and see 1914), Ginkgo biloba extract (EGb 761) [1910, 1915, 1916], grapes 1917, green tea [1807, 19181921], Mangifera indica extract 1879, strawberries 1907, spinach 1907 and Crataegus [922], while combinations of some these components ('protandim') have been claimed to reduce ROS levels by stimulating the production of catalase and SOD 1922. As with pharmaceutical drugs [18, 19231925], there are significant problems with bioavailability [1926, 1927], although the necessary measurements are starting to come forward [1804, 1809, 19261932]. There is now increasing evidence for the mechanisms with which these dietary components and related natural products and derivatives (often with anti-inflammatory, anti-mutagenic or anti-carcinogenic properties) interact with well recognised cellular signalling pathways (e.g. [1402, 19351944, 1895, 19451952, 1896, 1410, 19531960, 1413, 19611981, 1913, 1982, 1900, 1933, 19831990]).

Role of iron-generated ROSs in cellular signalling and oxidative stress

Thus, although this is not the focus of the present more physiologically based review, we recognise that many of relationships between ROSs and oxidative stress and overt progressive diseases may be mediated via the inflammatory signalling pathways involved in 'innate immunity' [900, 19911993]. NF-κB is an important transcription factor, and the NF-κB system is intimately involved in this signalling [588, 672, 719, 1408, 1409, 1454, 19942019].

In the NF-κB system (e.g. [20202024]) (Fig 8), NF-κB is normally held inactive in the cytoplasm by binding to an inhibitor IκB (often IκBα). Pro-inflammatory cytokines such as TNF-α, LPS [20252030] and IL-1 2031 act by binding to receptors at the cell surface and initiating signalling pathways that lead to the activation of a particular kinase, IκB kinase or IKK. This kinase phosphorylates the IκB causing it to be released (and ubiquitinated and degraded by the proteasome), allowing the NF-κB to be translocated to the nucleus where it can activate as many as 300 genes. Simple models of the NF-κB system show the main control elements [2032, 2033] and their synergistic interaction 2034. The NF-κB system is implicated in apoptosis [2035, 2036], aging 1199, and in diseases such as cancer [1405, 1444, 1454, 1808, 20372040], arthritis [20402043] and a variety of other diseases 2044. Antioxidants such as vitamin E [552, 2017] and melatonin [20452049] are at least partially protective. Oxidative stress seems to act upstream of IKK 2014, on IκBα directly 2050 and in the p38 MAP kinase pathway [1993, 2014, 2051], and there is also evidence that at least some of the statins act on the PI3K-akt and NF-κB pathways too [819, 883, 20522060]. A considerable number of inhibitors of the NF-κB system exist 2055, many exhibiting cross-reactivity 1734.

Figure 8
figure 8

An overview of cellular signaling using the NF-κB and p38 systems. Note that some of the extracellular effectors that mediate NF-κB activation are themselves produced and secreted as a result of the activation, potentially creating an autocatalytic system.

The induction of NF-κB by ROSs appears to involve a coupling via the glutathione system [2007, 2035, 2036, 20612078] (and see also [2079, 2080]).

A variety of studies have shown that iron is involved in these signalling processes [1839, 1996, 2015, 20812085], probably again acting upstream of the IKK [557, 2083, 2086, 2087].

Interestingly, there is interplay between the NF-κB pathway and the regulation of NGAL [404, 405, 2088, 2089], ferritin 2030 and hepcidin 2090, presumably acting as a negative feedback as the cell tries to control and ligand its free Fe(n) in the face of oxidative stress caused by the release of free iron 2091.

The systems biology of ROS generation and activity

It is not news that most major changes in physiological states have multigenic or multifactorial origins (e.g. [2092, 2093]). This means, as an inevitable consequence, that we need to recognize that their observation requires a systems approach, and that most diseases are therefore in fact to be seen as systems or network diseases [631, 20942102]. Changes in individual reaction steps (or even single manipulations) can change the levels of scores or hundreds of transcripts 2103, proteins 2104 or metabolites [725]. In this regard, small molecule (metabolomics) measurements have especial advantages for capturing network organisation, including on theoretical grounds [21052112].

If we consider just one variable of present relevance, the quantity of hydroxyl radical, the amount that is able to react with proteins, lipids and DNA is clearly determined by a huge number of reactions, whether directly or otherwise – not only the concentrations of reagents and enzyme that directly catalyse its formation and destruction but by everything else that affects their concentration and activity, and so on. This is of course well established in biology in the formalism of metabolic control analysis (MCA) (see e.g. [21132120]), and was recognized over 30 years ago in Morris' prescient review on anaerobes 2121. Modern systems theories of aging (e.g. [1175, 1180, 2120, 2122] and above) (Fig 9) also recognize physiological progression as being determined in terms of a balance between 'good' and 'bad' factors. MCA and related formalisms can be seen as theories of sensitivity analysis, which in many cases can be normalized such that an overall output function can be described quantitatively in terms of the relative contributions of each of its component steps (e.g. [21232127]). In MCA the normalized local sensitivities are known as control coefficients, and the sum of the concentration-control coefficients = 0, in other words in the steady state the rate of production and consumption of a particular entity is in balance and all reactions can contribute to it to some degree. The concentration-control coefficient describes this degree quantitatively. It is now possible to produce appropriate quantitative representations of metabolic networks using quite sparse kinds of information (in fact just the stoichiometry and network structure 2128), and thereby provide initial estimates for more sophisticated fitting algorithms (e.g. [21292132]. Indeed, the analysis of the properties and behaviour of networks is at the core of modern systems biology (e.g. [2095, 21332140]).

Figure 9
figure 9

General view of the role of iron, antioxidants and ROSs in aging and degenerative processes. Some of the decay may be ameliorated by lifestyle and dietary means. Based in part on 1175.

A corollary of such considerations is that to decrease the amount of damage caused by OH (or any other) radicals we need both to decrease their production and increase their removal to harmless substances 2141, and that on general grounds [17061708, 2142] such a strategy (for instance of combining a cell-permeable iron chelator with a cell-permeable antioxidant) might be expected to give a synergistic response. Even determining the means of cell permeability and tissue distribution turns out to be a systems biology problem in which we need to know the nature and activity of all the carriers that are involved [18, 1703, 1714, 2143]. At all events, it is undoubtedly the case that the steady-state rate of production of a molecule such as the hydroxyl radical is controlled or affected by a considerable number of steps. These minimally include the multiple reactions of the mitochondrial respiratory chain and the various oxidases producing superoxide and peroxide, the activities of catalase and SOD enzymes that together can remove them, protective reactions such as heat-shock proteins, and most pertinently to the present review a large number of reactions involved in the metabolism and safe liganding of iron that help determine the rate at which OH is produced.

It is also pertinent to enquire as to why we are now seeing so many of these progressive diseases, and as to what may be their causes. Undoubtedly the simple fact of improved longevity is one [165] as damage accumulates. However, we note that anything that decreases the amount of unliganded iron, such as decreasing the total dietary iron intake e.g. from red meat, must be helpful [1156, 2144].

Anti-inflammatory cytokines; the example of erythropoietin

We have above adduced considerable evidence that decreasing the amount of hydroxyl radical by any means is valuable, whether by removing initially generated ROSs such as superoxide and peroxide or by chelating poorly liganded iron in a way that stops these ROSs forming the hydroxyl radical. While pro-inflammatory cytokines can themselves increase ROS production and modulate the activities of signaling pathways such as NF-κB and p38, there are also anti-inflammatory cytokines. A particularly interesting example is that of erythropoietin (also discussed above as being protective in a number of iron-mediated diseases).

Erythropoietin was originally recognized via its role in erythropoiesis [21452147] (hence its name, of course), but it has become evident that it has many other roles, and in particular it is observed phenomenologically that erythropoietin (and non-erthyropoetic derivatives) is protective in a number of inflammatory conditions that accompany many diseases such as those listed above [940, 942, 21482155]. These included cardiovascular disease [721, 722, 21562175], stroke and other related neurological diseases [924926, 928, 929, 933, 937, 938, 2155, 21762204], diabetic neuropathy 2205, kidney injury [2173, 22062212], intestinal injury 2213 and shock (both septic and non-septic) [1596, 1597, 2214].

The question then arises as to how it is doing this mechanistically, and the proximate answer is that it (and other anti-inflammatory agents, e.g. [1808, 2215, 2216]) seem to act via many of the same signalling pathways as do inflammatory agents [943, 2150, 22172226]. There is evidence that it can help maintain superoxide dismutase activity [2214, 2227], invoke haem oxygenase 2228, and in particular – from the perspective of this review – that it may remove poorly liganded iron 2229 and interact with hydroxyl radical directly [22302233].

It is notable that appropriate levels of erythropoietin appear not only to be efficacious but to be safe, even in pregnancy [22342241]. Erythropoietin may itself be a marker of hypoxia and oxidative stress in pregnancy [531, 22422245], consistent with a view that the body is attempting to deal with these problems by creating anti-inflammatory cytokines.

Hypoxia-inducible factor (HIF)

Although I am mainly not concentrating on genetic regulatory aspects in this review, the HIF [2246, 2247] does deserve some mention, since many of the syndromes described above are accompanied by hypoxia, and this causes levels of the HIF to increase. HIF is a transcription factor that can activate a considerable number of genes, including VEGF [1951, 22462250]. In contrast to the constitutive expression of HIF-1α, HIF-1β protein levels are regulated in response to the cellular oxygen concentration 2251. The active HIF is the HIF-1αβ heterodimer 2252. HIF couples anoxia to innate immunity via the NF-κB system 2253. In particular, HIF effects (via hepcidin) the mobilisation or iron and can cause the expression of inflammatory cytokines such as IL-1, IL-6 and TNF-α [22542256] under conditions (hypoxia) where superoxide and peroxide production are likely to be increased, and consequently increases sepsis (in that HIF-knockout mice are resistant to LPS-induced sepsis [2254, 2255]). By contrast, induction of HIF (and the genes that it activates) can effect neuroprotection [2252, 2257]. HIF also appears to have a significant role in placental development, and defective HIF expression may be involved in pre-eclampsia and intra-uterine growth retardation [435, 2246, 2258]. Qutub and colleagues provide useful models [2259, 2260] of HIF activation under a variety of conditions of iron, O2, 2-oxoglutarate and other factors.

Autocatalysis, positive feedback and Systems Biology

What has emerged in recent years is a recognition that the structure (i.e. topology) of the modules of metabolic and signalling networks, somewhat independent of the individual activities of their components, can have a profound controlling influence on their behaviour (e.g. [2107, 2134, 2135, 2261, 2262]). Classically, negative feedback structures are considered to confer stability, while positive feedbacks tend to have the opposite effect. However, negative feedbacks with delay loops can cause oscillations [2022, 2109] while some kinds of positive feedback loops can confer stability [2262, 2263]. However, there is no doubt that structures in which a damaging agent causes the production of a second damaging agent that itself catalyses the production of the first or a separate damaging agent can exhibit a runaway kind of damage. This is exactly what can happen with iron and superoxide since Fe(n) can be liberated from ferritin by superoxide radicals and then catalyse the production of further hydroxyl radical by increasing the amount of free iron (Fig 6). A similar effect can occur with Fe-S proteins in SOD-deficiency 1148, with the degradation of mitochondria by radical damage leading to further production of radicals [28, 30, 2264], and the effects of oxidative stress on iron storage 2265. This again illustrates the importance of acting at multiple points in a network to control these kinds of damage. Exactly the same is true of the IL-1 and TNF-α systems in which IL-1 or TNF-α (oxidative stress) acting on one cell can effect the secretion of further IL-1/TNF-α that can act on adjacent cells (Fig 8), of the hypoxia-dependent increase in both ROSs and serum iron mediated by hepcidin (Fig 3), the autocatalytic synergy between overfeeding, inflammation and (pre-)diabetes, and of the peroxide/iron pair that are liberated when frataxin is deficient (see above). It is these kinds of synergistic effects and autocatalytic cycles that are the hallmark of the major and progressive effects on human physiology that are seen in these kinds of system. Indeed, one might comment that such multi-site and autocatalytic effects are required to overwhelm normal defences precisely because human metabolic and signalling systems are 'robust' systems that have evolved topologies that are resistant or robust to parameter changes (see e.g. [22662291]).

Predictive biology

It is often considered (e.g. [2292, 2293]) that a desirable feature of a scientific idea is its ability to make useful predictions, and while this is not in fact a particularly well founded philosophical principle, it probably is of value to set out a couple of 'predictions' that follow from the present analysis. One prominent feature of the above is the primacy of the iron-catalysed production of the damaging hydroxyl radical, and thus a test of the involvement of these kinds of reactions in the various physiological and pathological states to which we allude is the prediction that they should be accompanied by markers of oxidative stress characteristic of reactions of endogenous metabolites with the hydroxyl radical. While it is not that easy to disentangle the complex reactions of ROSs with biomolecules [43, 1055], at least the following appear to be a result of reactions involving OH [2294, 2295]: 8-oxo-2'-deoxyguanosine (oxo8dG) [98, 2296, 2297], 8-oxo-7,8- dihydro-2'-deoxyguanosine [90, 92] and thymine glycol [92, 2294].

Another set of predictions from the systems biology perspective [17041708, 1731, 2138, 2291, 22982310] is that combinations of chemical agents (or manipulations such as those of transcription factors that affect multiple steps in a pathway 1906 or modulation of multiple gene products 2311, or both [2312, 2313]) will be far more efficacious, for instance in modulating iron-catalysed oxidative stress and its sequelae, than will be the use of 'magic bullet' single agents. Such combinations of 'causes' do not have to be guessed a priori but can be obtained via inferencing techniques (e.g. [23142320]) – for a recent example see 2321. The nonlinear behaviour of biochemical networks also serves to explain the bell-shaped dose-response curves underpinning the hormesis [23222326] often observed.

Iron-mediated oxidative stress is arguably the cause of much of the inflammation typically observed in biological systems, often further mediated via pro-inflammatory cytokines. Another major prediction that comes from the above then is that molecules that are anti-inflammatory, whether widely recognised as such or not, should have beneficial effects in syndromes for which they have not necessarily been tested. An obvious set of candidates in this regard are to be found among the statins, since it is now clear that they have important anti-inflammatory properties (see above). Thus, there are already indications that as well as their established benefits in cardiovascular disease (e.g. [804, 2327]) they may exert benefit in a huge variety of syndromes [838], including sepsis [839, 2060, 23282340], heart failure 2341, pain perception 2342, lupus and related diseases [1293, 2343], diabetes [877, 2344], rheumatoid arthritis [866, 869, 890, 23452350], kidney disease [23512353], inflammatory skin disease 2354, emphysema 2355, ischaemia-reperfusion injury 2356, stroke [864, 872, 23572364], traumatic brain injury [23652367], neurodegenerative diseases [860862, 920, 1294, 2059, 23682384], neurotoxicity 2385 and cancer [23862400].

Concluding remarks and quo vadis

"Actually, the orgy of fact extraction in which everybody is currently engaged has, like most consumer economies, accumulated a vast debt. This is a debt of theory and some of us are soon going to have an exciting time paying it back – with interest, I hope." 2401.

"But one thing is certain: to understand the whole you must look at the whole" 2402

"If you are not thinking about your experiments on a whole-genome level you are going to be a dinosaur". J. Stamatoyannopoulos, quoted in 2403.

While it is less common for scientists to publish 'negative' results ('there was no effect of some agent on some process'), and there has been a tendency to seek to falsify specific hypotheses rather than to paint a big picture 2404, there is no doubt that the sheer weight of positive evidence can be persuasive in leading one to a view. As Bertrand Russell put it 2405, "When one admits that nothing is certain one must, I think, also admit that some things are much more nearly certain than others." However, as mentioned above, the huge volume of scientific activity has in many ways led to a 'balkanisation' of the literature 2406 in which scientists deal with the problem of the deluge of published papers by necessarily ignoring most of them. This is no longer realistic (nor necessary) in an age of post-genomics, the internet, Web 2.0 and systems biology, and when we are starting to move to integrative (if distributed) models of organisms (including humans) at a whole organ, genome or whole organism scale [118, 2135, 24072416]. The 'digital human' is thus an important research goal [2410, 24152417]. Expression profiling atlases are becoming increasingly widespread (e.g. [24182423]), and one can anticipate using these straightforwardly to extend these 'generalised' (sub)cellular network models in a tissue-specific manner 2424. With the ability to exchange models of biochemical networks in a principled way [24252428], when they are marked up appropriately (e.g. [3, 4]), we can expect to begin to reason about them automatically [118, 2429], such that we may soon look forward to an era in which we can recognise the commonalities across a variety of different subfields – a specific message of the present overview. Thus, while iron and metabolism should be considered in the context of other processes that may be contributing to the disorders discussed, and it is evident that they are intimately involved in many disease processes, therapies derived for one of the inflammatory diseases listed above may well have benefit in some of the others where their underlying 'causes' are the same. The 'mass collaboration' agenda (e.g. [24302435]), in which dispersed agents contribute their different skills to the solution of a complex problem, may well help this happen effectively. Developments in distributed workflow technology, such as the Taverna [2226, 2436, 2437]http://taverna.sourceforge.net/ and the myExperiment 2438 http://www.myexperiment.org/ environments, represent an intellectually important approach. Important too to this endeavour will be Open Access initiatives [1, 2439, 2440] and institutional and other repositories [2441, 2442] of full-text papers. This will help to build an accurate picture of the biochemical networks operating in both normal and diseased states (and see 2443), preferably marked up semantically as in [118], and hence, by modelling them 2263, where best to consider interventions. In order to develop and exploit this distributed approach, it will also be necessary for those generating them to make their data and metadata available (preferably in a semantically marked up way), probably as Web Services (e.g. [24, 24442452]), and to give greater scientific weight to those involved in bio-curation [119] as they will be an increasingly important part of the scientific landscape. Modern sequencing instruments, for instance (e.g. [24532457]) are generating quality data at truly enormous rates 2458, and innovative but computationally demanding algorithms are required to deal with them (e.g. 2459). In particular, however, we need tools for manipulating and visualising biochemical models [2110, 2410, 2429, 2460]. As well as storing these models (e.g. as SBML or CellML 2461) in a file format, it is also convenient to store them in a database format, such as the B-net database developed by Mendes and colleagues [118, 2462]. Federated annotation protocols such as the Distributed Annotaton Scheme (see e.g. 2463) allow data from heterogeneous sources to be combined, while other integrative/distributed architectures such as ONDEX 2464 and Utopia [24652468] are similarly showing considerable promise for integrative systems biology.

Given the widely dispersed communities that have been referenced herein, and the future requirement for integrating knowledge generated throughput the world, a programme for understanding the combinatorial roles of poorly liganded iron and reactive oxygen species in the aetiology of many diseases, as set out in the above, appears to be prototypical for the kinds of new approaches to doing science that we may anticipate in the eras of Web 2.0 and the Semantic Web.

Abbreviations

AECOPD:

Acute Exacerbation of Chronic Obstructive Pulmonary Disorder

AKI:

Acute Kidney Injury

ALS:

Amyotrophic Lateral Sclerosis

AMD:

Age-related Macular Degeneration

BBB:

Blood-brain barrier

ChEBI:

Chemical Entities of Biological Interest

CQ:

Clioquinol

COPD:

Chronic Obstructive Pulmonary Disorder

DFO:

Desferrioxamine

Dp44mT:

di-2-pyridylketone-4,4,-dimethyl-3-thiosemicarbazone

EDTA:

Ethylenediamine-tetraacetic Acid

FA:

Friedrich's ataxia

FPN1:

Ferroportin-1

HBED:

N,N'-bis-(2-hydroxybenzyl)ethylenediamine-N,N'-diacetic acid

HCP1:

Heme Carrier Protein-1

HIF:

Hypoxia-Inducible Factor

HO1:

Heme Oxygenase-1

HMG-CoA:

Hydroxymethyl glutaryl Coenzyme A

Hp:

Hephaestin

8HQs:

A variety of 8-hydroxyquinolines

IBD:

Inflammatory Bowel Disease

InChI:

International Chemical Identifier

IKK:

IκB kinase

KEGG:

Kyoto Encyclopedia of Genes and Genomes

L1:

Deferriprone

LFA-1:

Leukocyte Function Antigen-1

LIP:

Labile Iron Pool

LIPID:

Long-Term Intervention with Pravastatin in Ischaemic Disease

LPS:

Lipopolysaccharide

MOF:

Multiple organ failure

NF-κB:

Nuclear Factor κB

NGAL:

Neutrophil Gelatinase-Associated Lipocalin (Also known as lipocalin-2 or siderocalin)

NIH:

2-hydroxy-1-napthylaldehyde isonicotinyl hydrazone

PE:

Pre-eclampsia

PIH:

Pyridoxal isonicotinyl hydrazone

PPAR:

Peroxisome Proliferator Activated Receptor

RA:

Rheumatoid Arthritis

ROS:

Reactive Oxygen Species

SBML:

Systems Biology Markup Language

SIH:

Salicylaldehyde isonicotinyl hydrazone

SIRS:

Systemic Inflammatory Response Syndrome

SLE:

Systemic Lupus Erythematosus

SMILES:

Simplified Molecular Input Line Entry Specification

SOD:

Superoxide Dismutase

Tf:

Transferrin

TNF:

Tissue Necrosis Factor

VEGF:

Vascular Endothelial Growth Factor.

References

  1. Harnad S, Brody T, Vallieres F, Carr L, Hitchcock S, Gingras Y, Oppenheim C, Hajjem C, Hilf ER: The access/impact problem and the green and gold roads to open access: An update. Serials Review. 2008, 34: 36-40.

    Google Scholar 

  2. Hull D, Pettifer SR, Kell DB: Defrosting the digital library: bibliographic tools for the next generation web. PLoS Comput Biol. 2008, 4: e1000204-doi:10.1371/journal.pcbi.1000204

    PubMed  PubMed Central  Google Scholar 

  3. Ananiadou S, McNaught J: Text mining in biology and biomedicine. 2006, Artech House, London

    Google Scholar 

  4. Ananiadou S, Kell DB, Tsujii Ji: Text Mining and its potential applications in Systems Biology. Trends Biotechnol. 2006, 24: 571-579.

    CAS  PubMed  Google Scholar 

  5. Jensen LJ, Saric J, Bork P: Literature mining for the biologist: from information retrieval to biological discovery. Nat Rev Genet. 2006, 7: 119-29.

    CAS  PubMed  Google Scholar 

  6. Smalheiser NR: The Arrowsmith project: 2005 status report. LNCS. 2005, 3735: 26-43.

    Google Scholar 

  7. Hristovski D, Peterlin B, Mitchell JA, Humphrey SM: Using literature-based discovery to identify disease candidate genes. Int J Med Inform. 2005, 74: 289-98.

    PubMed  Google Scholar 

  8. Kostoff RN: Systematic acceleration of radical discovery and innovation in science and technology. Technol Forecasting Soc Change. 2006, 73: 923-936.

    Google Scholar 

  9. Yetisgen-Yildiz M, Pratt W: Using statistical and knowledge-based approaches for literature-based discovery. J Biomed Informatics. 2006, 39: 600-611.

    Google Scholar 

  10. Kostoff RN: Validating discovery in literature-based discovery. J Biomed Inform. 2007, 40: 448-50. author reply 450–2.

    PubMed  Google Scholar 

  11. Kostoff RN, Briggs MB, Solka JL, Rushenberg RL: Literature-related discovery (LRD): Methodology. Technol Forecast Soc Change. 2008

    Google Scholar 

  12. Cronin B: Bibliometrics and beyond: Some thoughts on web-based citation analysis. Journal of Information Science. 2001, 27: 1-7.

    Google Scholar 

  13. Kasztler A, Leitner KH: Bibliometric analysis and visualisation of intellectual capital. J Universal Comp Sci. 2002, 8: 516-525.

    Google Scholar 

  14. Tabah AN: Literature dynamics: Studies on growth, diffusion, and epidemics. Annu Rev Inf Sci Technol. 1999, 34: 249-286.

    Google Scholar 

  15. Börner K, Chen CM, Boyack KW: Visualizing knowledge domains. Annual Review of Information Science and Technology. 2003, 37: 179-255.

    Google Scholar 

  16. Errami M, Hicks JM, Fisher W, Trusty D, Wren JD, Long TC, Garner HR: Déjà vu: a study of duplicate citations in Medline. Bioinformatics. 2008, 24: 243-9.

    CAS  PubMed  Google Scholar 

  17. Kell DB: Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. 2008, [http://arxiv.org/ftp/arxiv/papers/0808/0808.1371.pdf]

    Google Scholar 

  18. Dobson PD, Kell DB: Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule?. Nat Rev Drug Discov. 2008, 7: 205-220.

    CAS  PubMed  Google Scholar 

  19. Smalheiser NR, Torvik VI, Bischoff-Grethe A, Burhans LB, Gabriel M, Homayouni R, Kashef A, Martone ME, Perkins GA, Price DL, Talk AC, West R: Collaborative development of the Arrowsmith two node search interface designed for laboratory investigators. J Biomed Discov Collab. 2006, 1: 8.

    PubMed  PubMed Central  Google Scholar 

  20. Smalheiser NR, Swanson DR: Using ARROWSMITH: a computer-assisted approach to formulating and assessing scientific hypotheses. Comput Methods Programs Biomed. 1998, 57: 149-53.

    CAS  PubMed  Google Scholar 

  21. Doms A, Schroeder M: GoPubMed: exploring PubMed with the Gene Ontology. Nucleic Acids Res. 2005, 33: W783-6.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Oinn T, Addis M, Ferris J, Marvin D, Senger M, Greenwood M, Carver T, Glover K, Pocock MR, Wipat A, Li P: Taverna: a tool for the composition and enactment of bioinformatics workflows. Bioinformatics. 2004, 20: 3045-54.

    CAS  PubMed  Google Scholar 

  23. Oinn T, Li P, Kell DB, Goble C, Goderis A, Greenwood M, Hull D, Stevens R, Turi D, Zhao J: Taverna/myGrid: aligning a workflow system with the life sciences community. Workflows for e-Science: scientific workflows for Grids. Edited by: Taylor IJ, Deelman E, Gannon DB, Shields M. 2007, Springer, Guildford, 300-319.

    Google Scholar 

  24. Hull D, Wolstencroft K, Stevens R, Goble C, Pocock MR, Li P, Oinn T: Taverna: a tool for building and running workflows of services. Nucleic Acids Res. 2006, 34: W729-32.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Oinn T, Greenwood M, Addis M, Alpdemir MN, Ferris J, Glover K, Goble C, Goderis A, Hull D, Marvin D, Li P, Lord P, Pocock MR, Senger M, Stevens R, Wipat A, Wroe C: Taverna: lessons in creating a workflow environment for the life sciences. Concurrency and Computation: Practice & Experience. 2006, 18: 1067-1100.

    Google Scholar 

  26. Goble C, Wolstencroft K, Goderis A, Hull D, Zhao J, Alper P, Lord P, Wroe C, Belhajjame K, Turi D, Stevens R, Oinn T, De Roure D: Knowledge discovery for biology with Taverna: producing and consuming semantics in the Web of Science. Semantic Web: revolutionising knowledge discovery in the life sciences. Edited by: Baker CJO, Cheung K-H. 2007, Springer, New York

    Google Scholar 

  27. Chance B, Sies H, Boveris A: Hydroperoxide metabolism in mammalian organs. Physiol Rev. 1979, 59: 527-605.

    CAS  PubMed  Google Scholar 

  28. Lenaz G: Role of mitochondria in oxidative stress and ageing. Biochim Biophys Acta. 1998, 1366: 53-67.

    CAS  PubMed  Google Scholar 

  29. Barja G: Mitochondrial oxygen radical generation and leak: sites of production in states 4 and 3, organ specificity, and relation to aging and longevity. J Bioenerg Biomembr. 1999, 31: 347-66.

    CAS  PubMed  Google Scholar 

  30. Kowaltowski AJ, Vercesi AE: Mitochondrial damage induced by conditions of oxidative stress. Free Radic Biol Med. 1999, 26: 463-71.

    CAS  PubMed  Google Scholar 

  31. Cadenas E, Davies KJ: Mitochondrial free radical generation, oxidative stress, and aging. Free Radic Biol Med. 2000, 29: 222-30.

    CAS  PubMed  Google Scholar 

  32. Lenaz G, D'Aurelio M, Merlo Pich M, Genova ML, Ventura B, Bovina C, Formiggini G, Parenti Castelli G: Mitochondrial bioenergetics in aging. Biochim Biophys Acta. 2000, 1459: 397-404.

    CAS  PubMed  Google Scholar 

  33. Raha S, Robinson BH: Mitochondria, oxygen free radicals, disease and ageing. Trends Biochem Sci. 2000, 25: 502-8.

    CAS  PubMed  Google Scholar 

  34. Lenaz G: The mitochondrial production of reactive oxygen species: mechanisms and implications in human pathology. IUBMB Life. 2001, 52: 159-64.

    CAS  PubMed  Google Scholar 

  35. Raha S, Robinson BH: Mitochondria, oxygen free radicals, and apoptosis. Am J Med Genet. 2001, 106: 62-70.

    CAS  PubMed  Google Scholar 

  36. Dröge W: Free radicals in the physiological control of cell function. Physiol Rev. 2002, 82: 47-95.

    PubMed  Google Scholar 

  37. Taylor ER, Hurrell F, Shannon RJ, Lin TK, Hirst J, Murphy MP: Reversible glutathionylation of complex I increases mitochondrial superoxide formation. J Biol Chem. 2003, 278: 19603-10.

    CAS  PubMed  Google Scholar 

  38. Turrens JF: Mitochondrial formation of reactive oxygen species. J Physiol. 2003, 552: 335-44.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Adam-Vizi V: Production of reactive oxygen species in brain mitochondria: Contribution by electron transport chain and non-electron transport chain sources. Antioxidants & Redox Signaling. 2005, 7: 1140-1149.

    CAS  Google Scholar 

  40. Andreyev AY, Kushnareva YE, Starkov AA: Mitochondrial metabolism of reactive oxygen species. Biochemistry (Mosc). 2005, 70: 200-14.

    CAS  Google Scholar 

  41. Balaban RS, Nemoto S, Finkel T: Mitochondria, oxidants, and aging. Cell. 2005, 120: 483-495.

    CAS  PubMed  Google Scholar 

  42. Adam-Vizi V, Chinopoulos C: Bioenergetics and the formation of mitochondrial reactive oxygen species. Trends Pharmacol Sci. 2006, 27: 639-45.

    CAS  PubMed  Google Scholar 

  43. Halliwell B, Gutteridge JMC: Free Radicals in Biology and Medicine. 2006, Oxford University Press, Oxford, 4

    Google Scholar 

  44. Orrenius S, Gogvadze A, Zhivotovsky B: Mitochondrial oxidative stress: Implications for cell death. Annual Review of Pharmacology and Toxicology. 2007, 47: 143-183.

    CAS  PubMed  Google Scholar 

  45. Fato R, Bergamini C, Leoni S, Strocchi P, Lenaz G: Generation of reactive oxygen species by mitochondrial complex I: implications in neurodegeneration. Neurochem Res. 2008, 33: 2487-501.

    CAS  PubMed  Google Scholar 

  46. Babior BM: Phagocytes and oxidative stress. Am J Med. 2000, 109: 33-44.

    CAS  PubMed  Google Scholar 

  47. Cave AC, Brewer AC, Narayanapanicker A, Ray R, Grieve DJ, Walker S, Shah AM: NADPH oxidases in cardiovascular health and disease. Antioxidants & Redox Signaling. 2006, 8: 691-728.

    CAS  Google Scholar 

  48. Bedard K, Krause KH: The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007, 87: 245-313.

    CAS  PubMed  Google Scholar 

  49. Brown JM, Terada LS, Grosso MA, Whitmann GJ, Velasco SE, Patt A, Harken AH, Repine JE: Xanthine oxidase produces hydrogen peroxide which contributes to reperfusion injury of ischemic, isolated, perfused rat hearts. J Clin Invest. 1988, 81: 1297-1301.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Granger DN: Role of xanthine oxidase and granulocytes in ischemia reperfusion injury. Amer J Physiol. 1988, 255: H1269-H1275.

    CAS  PubMed  Google Scholar 

  51. Linas SL, Whittenburg D, Repine JE: Role of xanthine oxidase in ischemia reperfusion injury. Amer J Physiol. 1990, 258: F711-F716.

    CAS  PubMed  Google Scholar 

  52. Thompson-Gorman SL, Zweier JL: Evaluation of the role of xanthine oxidase in myocardial reperfusion injury. J Biol Chem. 1990, 265: 6656-6663.

    CAS  PubMed  Google Scholar 

  53. Adkins WK, Taylor AE: Role of xanthine oxidase and neutrophils in ischemia-reperfusion injury in rabbit lung. J Appl Physiol. 1990, 69: 2012-2018.

    CAS  PubMed  Google Scholar 

  54. Müller MJ, Vollmar B, Friedl HP, Menger MD: Xanthine oxidase and superoxide radicals in portal triad crossclamping-induced microvascular reperfusion injury of the liver. Free Radic Biol Med. 1996, 21: 189-97.

    PubMed  Google Scholar 

  55. Imlay JA: Cellular defenses against superoxide and hydrogen peroxide. Annu Rev Biochem. 2008, 77: 755-76.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Sundaresan M, Yu ZX, Ferrans VJ, Irani K, Finkel T: Requirement for generation of H2O2 for platelet-derived growth factor signal transduction. Science. 1995, 270: 296-9.

    CAS  PubMed  Google Scholar 

  57. Finkel T: Oxidant signals and oxidative stress. Curr Opin Cell Biol. 2003, 15: 247-54.

    CAS  PubMed  Google Scholar 

  58. Rhee SG: Cell signaling. H2O2, a necessary evil for cell signaling. Science. 2006, 312: 1882-3.

    PubMed  Google Scholar 

  59. Miller EW, Tulyathan O, Isacoff EY, Chang CJ: Molecular imaging of hydrogen peroxide produced for cell signaling. Nat Chem Biol. 2007, 3: 263-7.

    CAS  PubMed  Google Scholar 

  60. Forman HJ, Fukuto JM, Miller T, Zhang H, Rinna A, Levy S: The chemistry of cell signaling by reactive oxygen and nitrogen species and 4-hydroxynonenal. Arch Biochem Biophys. 2008, 477: 183-95.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Halliwell B, Gutteridge JM: Oxygen toxicity, oxygen radicals, transition metals and disease. Biochem J. 1984, 219: 1-14.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Halliwell B, Gutteridge JMC: Role of free radicals and catalytic metal ions in human disease – an overview. Meth Enzymol. 1990, 186: 1-85.

    CAS  PubMed  Google Scholar 

  63. Galaris D, Pantopoulos K: Oxidative stress and iron homeostasis: mechanistic and health aspects. Crit Rev Clin Lab Sci. 2008, 45: 1-23.

    CAS  PubMed  Google Scholar 

  64. Buzan T: How to mind map. 2002, Thorsons, London

    Google Scholar 

  65. Fridovich I: Superoxide radical and superoxide dismutases. Annu Rev Biochem. 1995, 64: 97-112.

    CAS  PubMed  Google Scholar 

  66. Wardman P, Candeias LP: Fenton chemistry: An introduction. Rad Res. 1996, 145: 523-531.

    CAS  Google Scholar 

  67. Kehrer JP: The Haber-Weiss reaction and mechanisms of toxicity. Toxicology. 2000, 149: 43-50.

    CAS  PubMed  Google Scholar 

  68. Hershko C, Weatherall DJ: Iron-chelating therapy. Crit Rev Clin Lab Sci. 1988, 26: 303-45.

    CAS  PubMed  Google Scholar 

  69. Minotti G: Sources and role of iron in lipid peroxidation. Chem Res Toxicol. 1993, 6: 134-146.

    CAS  PubMed  Google Scholar 

  70. Rayman MP, Barlis J, Evans RW, Redman CW, King LJ: Abnormal iron parameters in the pregnancy syndrome preeclampsia. Am J Obstet Gynecol. 2002, 187: 412-8.

    CAS  PubMed  Google Scholar 

  71. Welch KD, Davis TZ, Van Eden ME, Aust SD: Deleterious iron-mediated oxidation of biomolecules. Free Radic Biol Med. 2002, 32: 577-83.

    CAS  PubMed  Google Scholar 

  72. Cheng Z, Li Y: What is responsible for the initiating chemistry of iron-mediated lipid peroxidation: an update. Chem Rev. 2007, 107: 748-66.

    CAS  PubMed  Google Scholar 

  73. Tsukahara H: Biomarkers for oxidative stress: Clinical application in pediatric medicine. Curr Med Chem. 2007, 14: 339-351.

    CAS  PubMed  Google Scholar 

  74. Evans MD, Dizdaroglu M, Cooke MS: Oxidative DNA damage and disease: induction, repair and significance. Mut Res. 2004, 567: 1-61.

    CAS  Google Scholar 

  75. Valko M, Izakovic M, Mazur M, Rhodes CJ, Telser J: Role of oxygen radicals in DNA damage and cancer incidence. Mol Cell Biochem. 2004, 266: 37-56.

    CAS  PubMed  Google Scholar 

  76. David SS, O'Shea VL, Kundu S: Base-excision repair of oxidative DNA damage. Nature. 2007, 447: 941-50.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Stadtman ER, Oliver CN: Metal-catalyzed oxidation of proteins – physiological consequences. J Biol Chem. 1991, 266: 2005-2008.

    CAS  PubMed  Google Scholar 

  78. Davies MJ, Fu S, Wang H, Dean RT: Stable markers of oxidant damage to proteins and their application in the study of human disease. Free Radic Biol Med. 1999, 27: 1151-63.

    CAS  PubMed  Google Scholar 

  79. Davies MJ: The oxidative environment and protein damage. Biochim Biophys Acta. 2005, 1703: 93-109.

    CAS  PubMed  Google Scholar 

  80. Terman A, Brunk UT: Lipofuscin. Int J Biochem Cell Biol. 2004, 36: 1400-1404.

    CAS  PubMed  Google Scholar 

  81. Jung T, Bader N, Grune T: Lipofuscin: formation, distribution, and metabolic consequences. Ann N Y Acad Sci. 2007, 1119: 97-111.

    CAS  PubMed  Google Scholar 

  82. Halliwell B: Free radicals and antioxidants: a personal view. Nutr Rev. 1994, 52: 253-65.

    CAS  PubMed  Google Scholar 

  83. De Zwart LL, Meerman JHN, Commandeur JNM, Vermeulen NPE: Biomarkers of free radical damage applications in experimental animals and in humans. Free Rad Biol Med. 1999, 26: 202-226.

    CAS  PubMed  Google Scholar 

  84. Kohen R, Nyska A: Oxidation of biological systems: oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol. 2002, 30: 620-50.

    CAS  PubMed  Google Scholar 

  85. Tarpey MM, Wink DA, Grisham MB: Methods for detection of reactive metabolites of oxygen and nitrogen: in vitro and in vivo considerations. Am J Physiol Regul Integr Comp Physiol. 2004, 286: R431-44.

    CAS  PubMed  Google Scholar 

  86. Halliwell B, Whiteman M: 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. 2004, 142: 231-55.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Cherubini A, Ruggiero C, Polidori MC, Mecocci P: Potential markers of oxidative stress in stroke. Free Radical Biology and Medicine. 2005, 39: 841-852.

    CAS  PubMed  Google Scholar 

  88. Shulaev V, Oliver DJ: Metabolic and proteomic markers for oxidative stress. New tools for reactive oxygen species research. Plant Physiol. 2006, 141: 367-72.

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Serkova NJ, Reisdorph NA, van Patot MCT: Metabolic markers of hypoxia: Systems biology application in biomedicine. Toxicology Mechanisms and Methods. 2008, 18: 81-95.

    CAS  PubMed  Google Scholar 

  90. Aust AE, Eveleigh JF: Mechanisms of DNA oxidation. Proc Soc Exp Biol Med. 1999, 222: 246-52.

    CAS  PubMed  Google Scholar 

  91. Cooke MS, Lunec J, Evans MD: Progress in the analysis of urinary oxidative DNA damage. Free Radic Biol Med. 2002, 33: 1601-1614.

    CAS  PubMed  Google Scholar 

  92. Dizdaroglu M, Jaruga P, Birincioglu M, Rodriguez H: Free radical-induced damage to DNA: Mechanisms and measurement. Free Radical Biology and Medicine. 2002, 32: 1102-1115.

    CAS  PubMed  Google Scholar 

  93. Neeley WL, Essigmann JM: Mechanisms of formation, genotoxicity, and mutation of guanine oxidation products. Chem Res Toxicol. 2006, 19: 491-505.

    CAS  PubMed  Google Scholar 

  94. Hwang ES, Bowen PE: DNA damage, a biomarker of carcinogenesis: Its measurement and modulation by diet and environment. Crit Rev Food Sci Nutrition. 2007, 47: 27-50.

    CAS  Google Scholar 

  95. Shin CS, Moon BS, Park KS, Kim SY, Park SJ, Chung MH, Lee HK: Serum 8-hydroxy-guanine levels are increased in diabetic patients. Diabetes Care. 2001, 24: 733-7.

    CAS  PubMed  Google Scholar 

  96. Loft S, Fischer-Nielsen A, Jeding IB, Vistisen K, Poulsen HE: 8-Hydroxydeoxyguanosine as a urinary biomarker of oxidative DNA damage. J Toxicol Environ Health. 1993, 40: 391-404.

    CAS  PubMed  Google Scholar 

  97. Migliore L, Fontana I, Colognato R, Coppede F, Siciliano G, Murri L: Searching for the role and the most suitable biomarkers of oxidative stress in Alzheimer's disease and in other neurodegenerative diseases. Neurobiol Aging. 2005, 26: 587-95.

    CAS  PubMed  Google Scholar 

  98. Bolin C, Cardozo-Pelaez F: Assessing biomarkers of oxidative stress: Analysis of guanosine and oxidized guanosine nucleotide triphosphates by high performance liquid chromatography with electrochemical detection. J Chromatogr B Analyt Technol Biomed Life Sci. 2007

    Google Scholar 

  99. Yamada S, Funada T, Shibata N, Kobayashi M, Kawai Y, Tatsuda E, Furuhata A, Uchida K: Protein-bound 4-hydroxy-2-hexenal as a marker of oxidized n-3 polyunsaturated fatty acids. J Lipid Res. 2004, 45: 626-634.

    CAS  PubMed  Google Scholar 

  100. Petersen DR, Doorn JA: Reactions of 4-hydroxynonenal with proteins and cellular targets. Free Radic Biol Med. 2004, 37: 937-45.

    CAS  PubMed  Google Scholar 

  101. Montuschi P, Ciabattoni G, Paredi P, Pantelidis P, du Bois RM, Kharitonov SA, Barnes PJ: 8-Isoprostane as a biomarker of oxidative stress in interstitial lung diseases. Amer J Resp Crit Care Med. 1998, 158: 1524-1527.

    CAS  Google Scholar 

  102. Montuschi P, Collins JV, Ciabattoni G, Lazzeri N, Corradi M, Kharitonov SA, Barnes PJ: Exhaled 8-isoprostane as an in vivo biomarker of lung oxidative stress in patients with COPD and healthy smokers. Amer J Resp Crit Care Med. 2000, 162: 1175-1177.

    CAS  Google Scholar 

  103. Schwedhelm E, Boger RH: Application of gas chromatography-mass spectrometry for analysis of isoprostanes: their role in cardiovascular disease. Clin Chem Lab Med. 2003, 41: 1552-61.

    CAS  PubMed  Google Scholar 

  104. Davì G, Falco A, Patrono C: Determinants of F2-isoprostane biosynthesis and inhibition in man. Chem Phys Lipids. 2004, 128: 149-63.

    PubMed  Google Scholar 

  105. Montuschi P, Barnes PJ, Roberts LJ: Isoprostanes: markers and mediators of oxidative stress. FASEB J. 2004, 18: 1791-800.

    CAS  PubMed  Google Scholar 

  106. Morrow JD: Quantification of isoprostanes as indices of oxidant stress and the risk of atherosclerosis in humans. Arterioscler Thromb Vasc Biol. 2005, 25: 279-86.

    CAS  PubMed  Google Scholar 

  107. Montuschi P, Barnes P, Roberts LJ: Insights into oxidative stress: the isoprostanes. Curr Med Chem. 2007, 14: 703-17.

    CAS  PubMed  Google Scholar 

  108. Mori TA, Croft KD, Puddey IB, Beilin LJ: Analysis of native and oxidized low-density lipoprotein oxysterols using gas chromatography mass spectrometry with selective ion monitoring. Redox Report. 1996, 2: 25-34.

    CAS  Google Scholar 

  109. Murphy RC, Johnson KM: Cholesterol, reactive oxygen species, and the formation of biologically active mediators. J Biol Chem. 2008, 283: 15521-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Janero DR: Malondialdehyde and thiobarbituric acid reactivity as diagnostic indexes of lipid peroxidation and peroxidative tissue injury. Free Radic Biol Med. 1990, 9: 515-540.

    CAS  PubMed  Google Scholar 

  111. Gieseg SP, Crone EM, Flavall EA, Amit Z: Potential to inhibit growth of atherosclerotic plaque development through modulation of macrophage neopterin/7,8-dihydroneopterin synthesis. Br J Pharmacol. 2008, 153: 627-35.

    CAS  PubMed  Google Scholar 

  112. Herce-Pagliai C, Kotecha S, Shuker DEG: Analytical methods for 3-nitrotyrosine as a marker of exposure to reactive nitrogen species: A review. Nitric Oxide-Biology and Chemistry. 1998, 2: 324-336.

    CAS  Google Scholar 

  113. Bian K, Gao Z, Weisbrodt N, Murad F: The nature of heme/iron-induced protein tyrosine nitration. Proc Natl Acad Sci USA. 2003, 100: 5712-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Duncan MW: A review of approaches to the analysis of 3-nitrotyrosine. Amino Acids. 2003, 25: 351-361.

    CAS  PubMed  Google Scholar 

  115. Ryberg H, Caidahl K: Chromatographic and mass spectrometric methods for quantitative determination of 3-nitrotyrosine in biological samples and their application to human samples. J Chromatogr B. 2007, 851: 160-171.

    CAS  Google Scholar 

  116. Cao EH, Wang JJ: Oxidative Damage to DNA – Levels of Thymine Glycol and Thymidine Glycol in Neoplastic Human Urines. Carcinogenesis. 1993, 14: 1359-1362.

    CAS  PubMed  Google Scholar 

  117. Makropoulos W, Kocher K, Heintz B, Schwarz ER, Mertens PR, Stefanidis I: Urinary thymidine glycol as a biomarker for oxidative stress after kidney transplantation. Renal Failure. 2000, 22: 499-510.

    CAS  PubMed  Google Scholar 

  118. Herrgård MJ, Swainston N, Dobson P, Dunn WB, Arga KY, Arvas M, Blüthgen N, Borger S, Costenoble R, Heinemann M, Hucka M, Le Novère N, Li P, Liebermeister W, Mo ML, Oliveira AP, Petranovic D, Pettifer S, Simeonidis E, Smallbone K, Spasić I, Weichart D, Brent R, Broomhead DS, Westerhoff HV, Kirdar B, Penttilä M, Klipp E, Palsson BØ, Sauer U, Oliver SG, Mendes P, Nielsen J, Kell DB: A consensus yeast metabolic network obtained from a community approach to systems biology. Nature Biotechnol. 2008, 26: 1155-1160.

    Google Scholar 

  119. Howe D, Costanzo M, Fey P, Gojobori T, Hannick L, Hide W, Hill DP, Kania R, Schaeffer M, St Pierre S, Twigger S, White O, Yon Rhee S: Big data: The future of biocuration. Nature. 2008, 455: 47-50.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Brooksbank C, Cameron G, Thornton J: The European Bioinformatics Institute's data resources: towards systems biology. Nucleic Acids Res. 2005, 33: D46-53.

    CAS  PubMed  Google Scholar 

  121. Kanehisa M, Goto S, Hattori M, Aoki-Kinoshita KF, Itoh M, Kawashima S, Katayama T, Araki M, Hirakawa M: From genomics to chemical genomics: new developments in KEGG. Nucleic Acids Res. 2006, 34: D354-7.

    CAS  PubMed  Google Scholar 

  122. Weininger D: SMILES, a chemical language and information system .1. Introduction to methodology and encoding rules. J Chem Inf Comput Sci. 1988, 28: 31-36.

    CAS  Google Scholar 

  123. Stein SE, Heller SR, Tchekhovski D: An open standard for chemical structure representation – the IUPAC chemical identifier. Proc 2003 Nimes Int Chem Info Conf. 2003, 131-143.

    Google Scholar 

  124. Coles SJ, Day NE, Murray-Rust P, Rzepa HS, Zhang Y: Enhancement of the chemical semantic web through the use of InChI identifiers. Organic & Biomolecular Chemistry. 2005, 3: 1832-1834.

    CAS  Google Scholar 

  125. Casher O, Rzepa HS: SemanticEye: a semantic web application to rationalize and enhance chemical electronic publishing. J Chem Inf Model. 2006, 46: 2396-411.

    CAS  PubMed  Google Scholar 

  126. Monge A, Arrault A, Marot C, Morin-Allory L: Managing, profiling and analyzing a library of 2.6 million compounds gathered from 32 chemical providers. Mol Divers. 2006, 10: 389-403.

    CAS  PubMed  Google Scholar 

  127. Knox C, Shrivastava S, Sothard P, Eisner R, Wishart DS: Biospider: a web server for automating metabolome annotations. Pac Symp Biocomputing. 2007, 12: 145-156.

    Google Scholar 

  128. Klinger R, Kolářik C, Fluck J, Hofmann-Apitius M, Friedrich CM: Detection of IUPAC and IUPAC-like chemical names. Bioinformatics. 2008, 24: i268-76.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Chavarria ME, Lara-Gonzalez L, Gonzalez-Gleason A, Garcia-Paleta Y, Vital-Reyes VS, Reyes A: Prostacyclin/thromboxane early changes in pregnancies that are complicated by preeclampsia. Am J Obstet Gynecol. 2003, 188: 986-92.

    CAS  PubMed  Google Scholar 

  130. Miller DM, Buettner GR, Aust SD: Transition metals as catalysts of "autoxidation" reactions. Free Radic Biol Med. 1990, 8: 95-108.

    CAS  PubMed  Google Scholar 

  131. Stohs SJ, Bagchi D: Oxidative mechanisms in the toxicity of metal ions. Free Rad Biol Med. 1995, 18: 321-336.

    CAS  PubMed  Google Scholar 

  132. Meneghini R: Iron homeostasis, oxidative stress, and DNA damage. Free Radical Biology and Medicine. 1997, 23: 783-792.

    CAS  PubMed  Google Scholar 

  133. Hubel CA: Dyslipidemia, iron, and oxidative stress in preeclampsia: assessment of maternal and feto-placental interactions. Semin Reprod Endocrinol. 1998, 16: 75-92.

    CAS  PubMed  Google Scholar 

  134. Aisen P, Enns C, Wessling-Resnick M: Chemistry and biology of eukaryotic iron metabolism. Int J Biochem Cell Biol. 2001, 33: 940-959.

    CAS  PubMed  Google Scholar 

  135. Reddy MB, Clark L: Iron, oxidative stress, and disease risk. Nutr Rev. 2004, 62: 120-4.

    PubMed  Google Scholar 

  136. Thompson KJ, Shoham S, Connor JR: Iron and neurodegenerative disorders. Brain Res Bull. 2001, 55: 155-64.

    CAS  PubMed  Google Scholar 

  137. Valko M, Morris H, Cronin MTD: Metals, toxicity and oxidative stress. Curr Med Chem. 2005, 12: 1161-1208.

    CAS  PubMed  Google Scholar 

  138. Carbonell T, Rama R: Iron, oxidative stress and early neurological deterioration in ischemic stroke. Current Medicinal Chemistry. 2007, 14: 857-874.

    CAS  PubMed  Google Scholar 

  139. Dunn LL, Rahmanto YS, Richardson DR: Iron uptake and metabolism in the new millennium. Trends Cell Biol. 2007, 17: 93-100.

    CAS  PubMed  Google Scholar 

  140. Weinberg ED: Exposing the hidden dangers of iron: what every medical professional should know about the impact of iron on the disease process. 2004, Cumberland House

    Google Scholar 

  141. Brewer GJ: Iron and copper toxicity in diseases of aging, particularly atherosclerosis and Alzheimer's disease. Exp Biol Med. 2007, 232: 323-335.

    CAS  Google Scholar 

  142. Valko M, Leibfritz D, Moncol J, Cronin MTD, Mazur M, Telser J: Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol. 2007, 39: 44-84.

    CAS  PubMed  Google Scholar 

  143. Winterbourn CC: Reconciling the chemistry and biology of reactive oxygen species. Nat Chem Biol. 2008, 4: 278-86.

    CAS  PubMed  Google Scholar 

  144. Qian SY, Buettner GR: Iron and dioxygen chemistry is an important route to initiation of biological free radical oxidations: an electron paramagnetic resonance spin trapping study. Free Radic Biol Med. 1999, 26: 1447-56.

    CAS  PubMed  Google Scholar 

  145. Brown GC, Borutaite V: Nitric oxide, mitochondria, and cell death. IUBMB Life. 2001, 52: 189-95.

    CAS  PubMed  Google Scholar 

  146. Gomes A, Fernandes E, Lima J: Use of fluorescence probes for detection of reactive nitrogen species: A review. J Fluorescence. 2006, 16: 119-139.

    CAS  Google Scholar 

  147. Yokoyama H, Kuroiwa H, Yano R, Araki T: Targeting reactive oxygen species, reactive nitrogen species and inflammation in MPTP neurotoxicity and Parkinson's disease. Neurol Sci. 2008, 29: 293-301.

    PubMed  Google Scholar 

  148. Moncada S, Palmer RMJ, Higgs EA: Nitric-Oxide – Physiology, Pathophysiology, and Pharmacology. Pharmacological Reviews. 1991, 43: 109-142.

    CAS  PubMed  Google Scholar 

  149. Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA: Apparent hydroxyl radical production by peroxynitrite: implications for endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci USA. 1990, 87: 1620-4.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Radi R, Cassina A, Hodara R, Quijano C, Castro L: Peroxynitrite reactions and formation in mitochondria. Free Radic Biol Med. 2002, 33: 1451-64.

    CAS  PubMed  Google Scholar 

  151. Rubbo H, O'Donnell V: Nitric oxide, peroxynitrite and lipoxygenase in atherogenesis: mechanistic insights. Toxicology. 2005, 208: 305-17.

    CAS  PubMed  Google Scholar 

  152. Zimmet JM, Hare JM: Nitroso-redox interactions in the cardiovascular system. Circulation. 2006, 114: 1531-44.

    PubMed  Google Scholar 

  153. Pacher P, Beckman JS, Liaudet L: Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007, 87: 315-424.

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Pavlovic R, Santaniello E: Peroxynitrite and nitrosoperoxycarbonate, a tightly connected oxidizing-nitrating couple in the reactive nitrogen-oxygen species family: new perspectives for protection from radical-promoted injury by flavonoids. J Pharm Pharmacol. 2007, 59: 1687-95.

    CAS  PubMed  Google Scholar 

  155. Aslan M, Ryan TM, Adler B, Townes TM, Parks DA, Thompson JA, Tousson A, Gladwin MT, Patel RP, Tarpey MM, Batinic-Haberle I, White CR, Freeman BA: Oxygen radical inhibition of nitric oxide-dependent vascular function in sickle cell disease. Proc Natl Acad Sci. 2001, 98: 15215-15220.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. O'Donnell VB, Freeman BA: Interactions between nitric oxide and lipid oxidation pathways – Implications for vascular disease. Circulation Research. 2001, 88: 12-21.

    PubMed  Google Scholar 

  157. Lancaster JR: Protein cysteine thiol nitrosation: maker or marker of reactive nitrogen species-induced nonerythroid cellular signaling?. Nitric Oxide. 2008, 19: 68-72.

    CAS  PubMed  Google Scholar 

  158. Vaz SM, Augusto O: Inhibition of myeloperoxidase-mediated protein nitration by tempol: Kinetics, mechanism, and implications. Proc Natl Acad Sci USA. 2008, 105: 8191-6.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Brown GC, Bal-Price A: Inflammatory neurodegeneration mediated by nitric oxide, glutamate, and mitochondria. Mol Neurobiol. 2003, 27: 325-55.

    CAS  PubMed  Google Scholar 

  160. Kim YM, Chung HT, Simmons RL, Billiar TR: Cellular non-heme iron content is a determinant of nitric oxide-mediated apoptosis, necrosis, and caspase inhibition. J Biol Chem. 2000, 275: 10954-61.

    CAS  PubMed  Google Scholar 

  161. Galli SJ, Tsai M, Piliponsky AM: The development of allergic inflammation. Nature. 2008, 454: 445-54.

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Finkel T: Radical medicine: treating ageing to cure disease. Nat Rev Mol Cell Biol. 2005, 6: 971-6.

    CAS  PubMed  Google Scholar 

  163. Finch CE: The biology of human longevity: inflammation, nutrition and aging in the evolution of lifespans. 2007, Academic Press, Amsterdam

    Google Scholar 

  164. Serrano M, Blasco MA: Cancer and ageing: convergent and divergent mechanisms. Nat Rev Mol Cell Biol. 2007, 8: 715-22.

    CAS  PubMed  Google Scholar 

  165. Brown GM: The living end: the future of death, aging and immortality. 2008, Macmillan, London

    Google Scholar 

  166. Renshaw JC, Robson GD, Trinci APJ, Wiebe MG, Livens FR, Collison D, Taylor RJ: Fungal siderophores: structures, functions and applications. Mycol Res. 2002, 106: 1123-1142.

    CAS  Google Scholar 

  167. Haas H: Molecular genetics of fungal siderophore biosynthesis and uptake: the role of siderophores in iron uptake and storage. Applied Microbiology and Biotechnology. 2003, 62: 316-330.

    CAS  PubMed  Google Scholar 

  168. Haas H, Eisendle M, Turgeon BG: Siderophores in fungal physiology and virulence. Annu Rev Phytopathol. 2008, 46: 149-87.

    CAS  PubMed  Google Scholar 

  169. Johnson L: Iron and siderophores in fungal-host interactions. Mycol Res. 2008, 112: 170-83.

    CAS  PubMed  Google Scholar 

  170. Raymond KN, Muller G, Matzanke BF: Complexation of iron by siderophores – a review of their solution and structural chemistry and biological function. Top Curr Chem. 1984, 123: 49-102.

    CAS  Google Scholar 

  171. Guerinot ML: Microbial iron transport. Annu Rev Microbiol. 1994, 48: 743-72.

    CAS  PubMed  Google Scholar 

  172. Jurado RL: Iron, infections, and anemia of inflammation. Clin Infect Dis. 1997, 25: 888-95.

    CAS  PubMed  Google Scholar 

  173. Drechsel H, Jung G: Peptide siderophores. Journal of Peptide Science. 1998, 4: 147-181.

    CAS  PubMed  Google Scholar 

  174. Ratledge C, Dover LG: Iron metabolism in pathogenic bacteria. Annu Rev Microbiol. 2000, 54: 881-941.

    CAS  PubMed  Google Scholar 

  175. Boukhalfa H, Crumbliss AL: Chemical aspects of siderophore mediated iron transport. Biometals. 2002, 15: 325-339.

    CAS  PubMed  Google Scholar 

  176. Braun V, Braun M: Active transport of iron and siderophore antibiotics. Curr Opin Microbiol. 2002, 5: 194-201.

    CAS  PubMed  Google Scholar 

  177. Brown JS, Holden DW: Iron acquisition by Gram-positive bacterial pathogens. Microbes Infect. 2002, 4: 1149-56.

    CAS  PubMed  Google Scholar 

  178. Cornelis P, Matthijs S: Diversity of siderophore-mediated iron uptake systems in fluorescent pseudomonads: not only pyoverdines. Environ Microbiol. 2002, 4: 787-798.

    CAS  PubMed  Google Scholar 

  179. Crosa JH, Walsh CT: Genetics and assembly line enzymology of siderophore biosynthesis in bacteria. Microbiology and Molecular Biology Reviews. 2002, 66: 223.

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Marx JJ: Iron and infection: competition between host and microbes for a precious element. Best Pract Res Clin Haematol. 2002, 15: 411-26.

    CAS  PubMed  Google Scholar 

  181. Winkelmann G: Microbial siderophore-mediated transport. Biochem Soc Trans. 2002, 30: 691-6.

    CAS  PubMed  Google Scholar 

  182. Andrews SC, Robinson AK, Rodriguez-Quinones F: Bacterial iron homeostasis. FEMS Microbiol Rev. 2003, 27: 215-37.

    CAS  PubMed  Google Scholar 

  183. Faraldo-Gómez JD, Sansom MSP: Acquisition of siderophores in Gram-negative bacteria. Nat Rev Mol Cell Biol. 2003, 4: 105-116.

    PubMed  Google Scholar 

  184. Raymond KN, Dertz EA, Kim SS: Enterobactin: an archetype for microbial iron transport. Proc Natl Acad Sci USA. 2003, 100: 3584-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Wandersman C, Delepelaire P: Bacterial iron sources: From siderophores to hemophores. Annual Review of Microbiology. 2004, 58: 611-647.

    CAS  PubMed  Google Scholar 

  186. Abergel RJ, Wilson MK, Arceneaux JE, Hoette TM, Strong RK, Byers BR, Raymond KN: Anthrax pathogen evades the mammalian immune system through stealth siderophore production. Proc Natl Acad Sci USA. 2006, 103: 18499-503.

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Fischbach MA, Lin HN, Liu DR, Walsh CT: How pathogenic bacteria evade mammalian sabotage in the battle for iron. Nature Chemical Biology. 2006, 2: 132-138.

    CAS  PubMed  Google Scholar 

  188. Grass G: Iron transport in Escherichia coli: All has not been said and done. Biometals. 2006, 19: 159-172.

    CAS  PubMed  Google Scholar 

  189. Miethke M, Marahiel MA: Siderophore-based iron acquisition and pathogen control. Microbiol Mol Biol Rev. 2007, 71: 413-51.

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Genco CA, Dixon DW: Emerging strategies in microbial haem capture. Molecular Microbiology. 2001, 39: 1-11.

    CAS  PubMed  Google Scholar 

  191. Loomis LD, Raymond KN: Solution equilibria of enterobactin and metal enterobactin complexes. Inorganic Chemistry. 1991, 30: 906-911.

    CAS  Google Scholar 

  192. Stintzi A, Barnes C, Xu J, Raymond KN: Microbial iron transport via a siderophore shuttle: a membrane ion transport paradigm. Proc Natl Acad Sci USA. 2000, 97: 10691-6.

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Braun V: Iron uptake mechanisms and their regulation in pathogenic bacteria. Int J Med Microbiol. 2001, 291: 67-79.

    CAS  PubMed  Google Scholar 

  194. Cendrowski S, MacArthur W, Hanna P: Bacillus anthracis requires siderophore biosynthesis for growth in macrophages and mouse virulence. Mol Microbiol. 2004, 51: 407-417.

    CAS  PubMed  Google Scholar 

  195. Schaible ME, Kaufmann SHE: Iron and microbial infection. Nat Rev Microbiol. 2004, 2: 946-953.

    CAS  PubMed  Google Scholar 

  196. Ong ST, Ho JZS, Ho B, Ding JL: Iron-withholding strategy in innate immunity. Immunobiology. 2006, 211: 295-314.

    CAS  PubMed  Google Scholar 

  197. Challis GL, Ravel J: Coelichelin, a new peptide siderophore encoded by the Streptomyces coelicolor genome: structure prediction from the sequence of its non-ribosomal peptide synthetase. FEMS Microbiol Lett. 2000, 187: 111-4.

    CAS  PubMed  Google Scholar 

  198. Fernandez-Pol JA: Isolation and characterization of a siderophore-like growth factor from mutants of SV40-transformed cells adapted to picolinic acid. Cell. 1978, 14: 489-99.

    CAS  PubMed  Google Scholar 

  199. Jones RL, Peterson CM, Grady RW, Cerami A: Low molecular weight iron-binding factor from mammalian tissue that potentiates bacterial growth. J Exp Med. 1980, 151: 418-28.

    CAS  PubMed  Google Scholar 

  200. Primosigh JV, Thomas ED: Studies on the partition of iron in bone marrow cells. J Clin Invest. 1968, 47: 1473-82.

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Jacobs A: Low molecular weight intracellular iron transport compounds. Blood. 1977, 50: 433-9.

    CAS  PubMed  Google Scholar 

  202. Hershko C, Graham G, Bates GW, Rachmilewitz EA: Non-specific serum iron in thalassaemia: an abnormal serum iron fraction of potential toxicity. Br J Haematol. 1978, 40: 255-63.

    CAS  PubMed  Google Scholar 

  203. Graham G, Bates GW, Rachmilewitz EA, Hershko C: Nonspecific serum iron in thalassemia: quantitation and chemical reactivity. Am J Hematol. 1979, 6: 207-17.

    CAS  PubMed  Google Scholar 

  204. Craven CM, Alexander J, Eldridge M, Kushner JP, Bernstein S, Kaplan J: Tissue distribution and clearance kinetics of non-transferrin-bound iron in the hypotransferrinemic mouse: a rodent model for hemochromatosis. Proc Natl Acad Sci. 1987, 84: 3457-61.

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Fontecave M, Pierre JL: Iron metabolism – the low molecular mass iron pool. Biology of Metals. 1991, 4: 133-135.

    CAS  PubMed  Google Scholar 

  206. Kaplan J: Mechanisms of cellular iron acquisition: another iron in the fire. Cell. 2002, 111: 603-6.

    CAS  PubMed  Google Scholar 

  207. Ganz T: Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation. Blood. 2003, 102: 783-788.

    CAS  PubMed  Google Scholar 

  208. Finch C: Regulators of iron balance in humans. Blood. 1994, 84: 1697-702.

    CAS  PubMed  Google Scholar 

  209. Kohgo Y, Ikuta K, Ohtake T, Torimoto Y, Kato J: Body iron metabolism and pathophysiology of iron overload. Int J Hematol. 2008, 88: 7-15.

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Crichton RR, Ward RJ: Iron metabolism–new perspectives in view. Biochemistry. 1992, 31: 11255-64.

    CAS  PubMed  Google Scholar 

  211. Andrews NC: Medical progress: Disorders of iron metabolism. New England Journal of Medicine. 1999, 341: 1986-1995.

    CAS  PubMed  Google Scholar 

  212. Beard JL, Dawson H, Piñero DJ: Iron metabolism: a comprehensive review. Nutr Rev. 1996, 54: 295-317.

    CAS  PubMed  Google Scholar 

  213. Lieu PT, Heiskala M, Peterson PA, Yang Y: The roles of iron in health and disease. Mol Aspects Med. 2001, 22: 1-87.

    CAS  PubMed  Google Scholar 

  214. Chung JY, Wessling-Resnick M: Molecular mechanisms and regulation of iron transport. Crit Rev Clin Lab Sci. 2003, 40: 151-182.

    CAS  PubMed  Google Scholar 

  215. Frazer DM, Anderson GJ: The orchestration of body iron intake: how and where do enterocytes receive their cues?. Blood Cells Molecules and Diseases. 2003, 30: 288-297.

    CAS  Google Scholar 

  216. Knutson M, Wessling-Resnick M: Iron metabolism in the reticuloendothelial system. Crit Rev Biochem Mol Biol. 2003, 38: 61-88.

    CAS  PubMed  Google Scholar 

  217. McArdle HJ, Danzeisen R, Fosset C, Gambling L: The role of the placenta in iron transfer from mother to fetus and the relationship between iron status and fetal outcome. Biometals. 2003, 16: 161-7.

    CAS  PubMed  Google Scholar 

  218. Hentze MW, Muckenthaler MU, Andrews NC: Balancing acts: molecular control of mammalian iron metabolism. Cell. 2004, 117: 285-97.

    CAS  PubMed  Google Scholar 

  219. Andrews NC: Molecular control of iron metabolism. Best Pract Res Clin Haematol. 2005, 18: 159-69.

    CAS  PubMed  Google Scholar 

  220. Papanikolaou G, Pantopoulos K: Iron metabolism and toxicity. Toxicol Appl Pharmacol. 2005, 202: 199-211.

    CAS  PubMed  Google Scholar 

  221. Taketani S: Aquisition, mobilization and utilization of cellular iron and heme: Endless findings and growing evidence of tight regulation. Tohoku J Exp Med. 2005, 205: 297-318.

    CAS  PubMed  Google Scholar 

  222. Donovan A, Roy CN, Andrews NC: The ins and outs of iron homeostasis. Physiology (Bethesda). 2006, 21: 115-23.

    CAS  Google Scholar 

  223. Ganz T, Nemeth E: Regulation of iron acquisition and iron distribution in mammals. Biochim Biophys Acta. 2006, 1763: 690-699.

    CAS  PubMed  Google Scholar 

  224. Huang XP, O'Brien PJ, Templeton DM: Mitochondrial involvement in genetically determined transition metal toxicity I. Iron toxicity. Chemico-Biological Interactions. 2006, 163: 68-76.

    CAS  PubMed  Google Scholar 

  225. Nairz M, Weiss G: Molecular and clinical aspects of iron homeostasis: from anemia to hemochromatosis. Wiener Klinische Wochenschrift. 2006, 118: 442-462.

    CAS  PubMed  Google Scholar 

  226. Anderson GJ: Mechanisms of iron loading and toxicity. Am J Hematol. 2007, 82: 1128-31.

    CAS  PubMed  Google Scholar 

  227. Andrews NC, Schmidt PJ: Iron homeostasis. Annu Rev Physiol. 2007, 69: 69-85.

    CAS  PubMed  Google Scholar 

  228. Harrison-Findik DD: Role of alcohol in the regulation of iron metabolism. World J Gastroenterol. 2007, 13: 4925-30.

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Jones BC, Beard JL, Gibson JN, Unger EL, Allen RP, McCarthy KA, Earley CJ: Systems genetic analysis of peripheral iron parameters in the mouse. Am J Physiol Regul Integr Comp Physiol. 2007, 293: R116-24.

    CAS  PubMed  Google Scholar 

  230. Mithen R: Effect of genotype on micronutrient absorption and metabolism: a review of iron, copper, iodine and selenium, and folates. Int J Vitam Nutr Res. 2007, 77: 205-16.

    CAS  PubMed  Google Scholar 

  231. Vallerio LG: Mammalian iron metabolism. Toxicology Mechanisms and Methods. 2007, 17: 497-517.

    Google Scholar 

  232. Andrews NC: Forging a field: the golden age of iron biology. Blood. 2008, 112: 219-30.

    CAS  PubMed  PubMed Central  Google Scholar 

  233. Beard J, Han O: Systemic iron status. Biochim Biophys Acta. 2008.

    Google Scholar 

  234. Deugnier Y, Brissot P, Loréal O: Iron and the liver: update 2008. J Hepatol. 2008, 48 (Suppl 1): S113-23.

    CAS  PubMed  Google Scholar 

  235. Ganz T: Iron homeostasis: fitting the puzzle pieces together. Cell Metab. 2008, 7: 288-90.

    CAS  PubMed  Google Scholar 

  236. Handelman GJ, Levin NW: Iron and anemia in human biology: a review of mechanisms. Heart Fail Rev. 2008, 13: 393-404.

    PubMed  Google Scholar 

  237. Hower V, Mendes P, Torti FM, Laubenbacher R, Akman S, Shulaev V, Torti SV: A general map of iron metabolism and tissue-specific subnetworks. Mol Biosyst. 2008

    Google Scholar 

  238. Kemna EH, Tjalsma H, Willems HL, Swinkels DW: Hepcidin: from discovery to differential diagnosis. Haematologica. 2008, 93: 90-7.

    CAS  PubMed  Google Scholar 

  239. Kong W, Duan X, Shi Z, Chang Y: Iron metabolism in the mononuclear phagocyte system. Progr Nat Sci. 2008, 18: 1197-1202.

    CAS  Google Scholar 

  240. Levi S, Rovida E: The role of iron in mitochondrial function. Biochim Biophys Acta. 2008.

    Google Scholar 

  241. Wrighting DM, Andrews NC: Iron homeostasis and erythropoiesis. Red Cell Development. 2008, 82: 141-167.

    CAS  Google Scholar 

  242. Frazer DM, Anderson GJ: Iron Imports. I. Intestinal iron absorption and its regulation. Amer J Physiol. 2005, 289: G631-G635.

    CAS  Google Scholar 

  243. Mackenzie B, Garrick MD: Iron Imports. II. Iron uptake at the apical membrane in the intestine. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2005, 289: G981-G986.

    CAS  PubMed  Google Scholar 

  244. Wessling-Resnick M: Iron Imports. III. Transfer of iron from the mucosa into circulation. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2006, 290: G1-G6.

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Ganz T, Nemeth E: Iron imports. IV. Hepcidin and regulation of body iron metabolism. Amer J Physiol. 2006, 290: G199-G203.

    CAS  Google Scholar 

  246. Fleming RE, Britton RS: Iron Imports. VI. HFE and regulation of intestinal iron absorption. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2006, 290: G590-G594.

    CAS  PubMed  Google Scholar 

  247. Ma YX, Yeh M, Yeh KY, Glass J: Iron imports. V. Transport of iron through the intestinal epithelium. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2006, 290: G417-G422.

    CAS  PubMed  Google Scholar 

  248. De Domenico I, Ward DM, Kaplan J: Regulation of iron acquisition and storage: consequences for iron-linked disorders. Nature Reviews Molecular Cell Biology. 2008, 9: 72-81.

    CAS  PubMed  Google Scholar 

  249. Gunshin H, Mackenzie B, Berger UV, Gunshin Y, Romero MF, Boron WF, Nussberger S, Gollan JL, Hediger MA: Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature. 1997, 388: 482-8.

    CAS  PubMed  Google Scholar 

  250. Forbes JR, Gros P: Divalent-metal transport by NRAMP proteins at the interface of host-pathogen interactions. Trends Microbiol. 2001, 9: 397-403.

    CAS  PubMed  Google Scholar 

  251. Shayeghi M, Latunde-Dada GO, Oakhill JS, Laftah AH, Takeuchi K, Halliday N, Khan Y, Warley A, McCann FE, Hider RC, Frazer DM, Anderson GJ, Vulpe CD, Simpson RJ, McKie AT: Identification of an intestinal heme transporter. Cell. 2005, 122: 789-801.

    CAS  PubMed  Google Scholar 

  252. Ryter SW, Alam J, Choi AM: Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiol Rev. 2006, 86: 583-650.

    CAS  PubMed  Google Scholar 

  253. Vítek L, Schwertner HA: The heme catabolic pathway and its protective effects on oxidative stress-mediated diseases. Adv Clin Chem. 2007, 43: 1-57.

    PubMed  Google Scholar 

  254. Abraham NG, Kappas A: Pharmacological and clinical aspects of heme oxygenase. Pharmacol Rev. 2008, 60: 79-127.

    CAS  PubMed  Google Scholar 

  255. Ponka P: Tissue-specific regulation of iron metabolism and heme synthesis: Distinct control mechanisms in erythroid cells. Blood. 1997, 89: 1-25.

    CAS  PubMed  Google Scholar 

  256. Linder MC, Mortya M, Whon A, Kassa A, Gilley C: Vesicular transport of Fe and interaction with other metal ions in polarized Caco2 Cell monolayers. Biological Research. 2006, 39: 143-156.

    CAS  PubMed  Google Scholar 

  257. Moriya M, Linder MC: Vesicular transport and apotransferrin in intestinal iron absorption, as shown in the Caco-2 cell model. Amer J Physiol. 2006, 290: G301-G309.

    CAS  Google Scholar 

  258. Petrat F, de Groot H, Sustmann R, Rauen U: The chelatable iron pool in living cells: A methodically defined quantity. Biological Chemistry. 2002, 383: 489-502.

    CAS  PubMed  Google Scholar 

  259. Tenopoulou M, Kurz T, Doulias PT, Galaris D, Brunk UT: Does the calcein-AM method assay the total cellular 'labile iron pool' or only a fraction of it?. Biochem J. 2007, 403: 261-6.

    CAS  PubMed  PubMed Central  Google Scholar 

  260. Shvartsman M, Kikkeri R, Shanzer A, Cabantchik ZI: Non-transferrin-bound iron reaches mitochondria by a chelator-inaccessible mechanism: biological and clinical implications. Am J Physiol Cell Physiol. 2007, 293: C1383-94.

    CAS  PubMed  Google Scholar 

  261. Breuer W, Shvartsman M, Cabantchik ZI: Intracellular labile iron. Int J Biochem Cell Biol. 2008, 40: 350-4.

    CAS  PubMed  Google Scholar 

  262. Fakih S, Podinovskaia M, Kong X, Collins HL, Schaible UE, Hider RC: Targeting the lysosome: fluorescent iron(III) chelators to selectively monitor endosomal/lysosomal labile iron pools. J Med Chem. 2008, 51: 4539-52.

    CAS  PubMed  Google Scholar 

  263. Arosio P, Ingrassia R, Cavadini P: Ferritins: A family of molecules for iron storage, antioxidation and more. Biochim Biophys Acta. 2008

    Google Scholar 

  264. Shi H, Bencze KZ, Stemmler TL, Philpott CC: A cytosolic iron chaperone that delivers iron to ferritin. Science. 2008, 320: 1207-10.

    CAS  PubMed  PubMed Central  Google Scholar 

  265. Hallberg L, Hulthen L: Perspectives on iron absorption. Blood Cells Molecules and Diseases. 2002, 29: 562-573.

    Google Scholar 

  266. Arosio P, Levi S: Ferritin, iron homeostasis, and oxidative damage. Free Radic Biol Med. 2002, 33: 457-63.

    CAS  PubMed  Google Scholar 

  267. Lee DH, Zacharski LR, Jacobs DR: Comparison of the serum ferritin and percentage of transferrin saturation as exposure markers of iron-driven oxidative stress-related disease outcomes. Am Heart J. 2006, 151: 1247 e1-7.

    PubMed  Google Scholar 

  268. McKie AT, Barlow DJ: The SLC40 basolateral iron transporter family (IREG1/ferroportin/MTP1). Pflugers Arch. 2004, 447: 801-6.

    CAS  PubMed  Google Scholar 

  269. Donovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, Robine S, Andrews NC: The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metabolism. 2005, 1: 191-200.

    CAS  PubMed  Google Scholar 

  270. Ganz T: Cellular iron: ferroportin is the only way out. Cell Metab. 2005, 1: 155-7.

    CAS  PubMed  Google Scholar 

  271. Oates PS: The role of hepcidin and ferroportin in iron absorption. Histol Histopathol. 2007, 22: 791-804.

    CAS  PubMed  Google Scholar 

  272. Vulpe CD, Kuo YM, Murphy TL, Cowley L, Askwith C, Libina N, Gitschier J, Anderson GJ: Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse. Nature Genetics. 1999, 21: 195-199.

    CAS  PubMed  Google Scholar 

  273. Worwood M: Serum transferrin receptor assays and their application. Annals of Clinical Biochemistry. 2002, 39: 221-230.

    CAS  PubMed  Google Scholar 

  274. Mense SM, Zhang L: Herne: a versatile signaling molecule controlling the activities of diverse regulators ranging from transcription factors to MAP kinases. Cell Res. 2006, 16: 681-692.

    CAS  PubMed  Google Scholar 

  275. Balla J, Vercellotti GM, Nath K, Yachie A, Nagy E, Eaton JW, Balla G: Haem, haem oxygenase and ferritin in vascular endothelial cell injury. Nephrology Dialysis Transplantation. 2003, 18: 8-12.

    Google Scholar 

  276. Choi AM, Alam J: Heme oxygenase-1: function, regulation, and implication of a novel stress-inducible protein in oxidant-induced lung injury. Am J Respir Cell Mol Biol. 1996, 15: 9-19.

    CAS  PubMed  Google Scholar 

  277. Foresti R, Green CJ, Motterlini R: Generation of bile pigments by haem oxygenase: a refined cellular strategy in response to stressful insults. Biochem Soc Symp. 2004, 71: 177-192.

    CAS  PubMed  Google Scholar 

  278. Poon HF, Calabrese V, Scapagnini G, Butterfield DA: Free radicals: key to brain aging and heme oxygenase as a cellular response to oxidative stress. J Gerontol A Biol Sci Med Sci. 2004, 59: 478-93.

    PubMed  Google Scholar 

  279. Akagi R, Takahashi T, Sassa S: Cytoprotective effects of heme oxygenase in acute renal failure. Cellular Stress Responses in Renal Diseases, Contrib Nephrol. 2005, 148: 70-85.

    CAS  Google Scholar 

  280. Fredenburgh LE, Perrella MA, Mitsialis SA: The role of heme oxygenase-1 in pulmonary disease. Amer J Resp Cell Mol Biol. 2007, 36: 158-165.

    CAS  Google Scholar 

  281. Gueler F, Park JK, Rong S, Kirsch T, Lindschau C, Zheng W, Elger M, Fiebeler A, Fliser D, Luft FC, Haller H: Statins attenuate ischemia-reperfusion injury by inducing heme oxygenase-1 in infiltrating macrophages. Am J Pathol. 2007, 170: 1192-1199.

    CAS  PubMed  PubMed Central  Google Scholar 

  282. Koizumi S: Human heme oxygenase-1 deficiency: a lesson on serendipity in the discovery of the novel disease. Pediatr Int. 2007, 49: 125-32.

    CAS  PubMed  Google Scholar 

  283. Horváth I, Donnelly LE, Kiss A, Paredi P, Kharitonov SA, Barnes PJ: Raised levels of exhaled carbon monoxide are associated with an increased expression of heme oxygenase-1 in airway macrophages in asthma: a new marker of oxidative stress. Thorax. 1998, 53: 668-72.

    PubMed  PubMed Central  Google Scholar 

  284. Uhlen M, Bjorling E, Agaton C, Szigyarto CA, Amini B, Andersen E, Andersson AC, Angelidou P, Asplund A, Asplund C, Berglund L, Bergstrom K, Brumer H, Cerjan D, Ekstrom M, Elobeid A, Eriksson C, Fagerberg L, Falk R, Fall J, Forsberg M, Bjorklund MG, Gumbel K, Halimi A, Hallin I, Hamsten C, Hansson M, Hedhammar M, Hercules G, Kampf C, Larsson K, Lindskog M, Lodewyckx W, Lund J, Lundeberg J, Magnusson K, Malm E, Nilsson P, Odling J, Oksvold P, Olsson I, Oster E, Ottosson J, Paavilainen L, Persson A, Rimini R, Rockberg J, Runeson M, Sivertsson A, Skollermo A, Steen J, Stenvall M, Sterky F, Stromberg S, Sundberg M, Tegel H, Tourle S, Wahlund E, Walden A, Wan J, Wernerus H, Westberg J, Wester K, Wrethagen U, Xu LL, Hober S, Ponten F: A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol Cell Proteomics. 2005, 4: 1920-32.

    CAS  PubMed  Google Scholar 

  285. Lee DW, Andersen JK: Role of HIF-1 in iron regulation: potential therapeutic strategy for neurodegenerative disorders. Curr Mol Med. 2006, 6: 883-893.

    CAS  PubMed  Google Scholar 

  286. Rouault TA: The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nature Chem Biol. 2006, 2: 406-414.

    CAS  Google Scholar 

  287. Wallander ML, Leibold EA, Eisenstein RS: Molecular control of vertebrate iron homeostasis by iron regulatory proteins. Biochim Biophys Acta. 2006, 1763: 668-689.

    CAS  PubMed  PubMed Central  Google Scholar 

  288. Lee K, Roth RA, LaPres JJ: Hypoxia, drug therapy and toxicity. Pharmacology & Therapeutics. 2007, 113: 229-246.

    CAS  Google Scholar 

  289. Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P, Loreal O: A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem. 2001, 276: 7811-7819.

    CAS  PubMed  Google Scholar 

  290. Fleming RE, Sly WS: Hepcidin: A putative iron-regulatory hormone relevant to hereditary hemochromatosis and the anemia of chronic disease. Proc Nal Acad Sci. 2001, 98: 8160-8162.

    CAS  Google Scholar 

  291. Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B, Kahn A, Vaulont S: Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad Sci. 2001, 98: 8780-8785.

    CAS  PubMed  PubMed Central  Google Scholar 

  292. Weinstein DA, Roy CN, Fleming MD, Loda MF, Wolfsdorf JI, Andrews NC: Inappropriate expression of hepcidin is associated with iron refractory anemia: implications for the anemia of chronic disease. Blood. 2002, 100: 3776-81.

    CAS  PubMed  Google Scholar 

  293. Bridle KR, Frazer DM, Wilkins SJ, Dixon JL, Purdie DM, Crawford DHG, Subramaniam VN, Powell LW, Anderson GJ, Ramm GA: Disrupted hepcidin regulation in HFE-associated haemochromatosis and the liver as a regulator of body iron homoeostasis. Lancet. 2003, 361: 669-673.

    CAS  PubMed  Google Scholar 

  294. Nicolas G, Viatte L, Lou DQ, Bennoun M, Beaumont C, Kahn A, Andrews NC, Vaulont S: Constitutive hepcidin expression prevents iron overload in a mouse model of hemochromatosis. Nature Genetics. 2003, 34: 97-101.

    CAS  PubMed  Google Scholar 

  295. Roetto A, Papanikolaou G, Politou M, Alberti F, Girelli D, Christakis J, Loukopoulos D, Camaschella C: Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis. Nature Genetics. 2003, 33: 21-22.

    CAS  PubMed  Google Scholar 

  296. Deicher R, Horl WH: Hepcidin: a molecular link between inflammation and anaemia. Nephrol Dial Transplant. 2004, 19: 521-4.

    CAS  PubMed  Google Scholar 

  297. Leong WI, Lönnerdal B: Hepcidin, the recently identified peptide that appears to regulate iron absorption. J Nutr. 2004, 134: 1-4.

    CAS  PubMed  Google Scholar 

  298. Robson KJ: Hepcidin and its role in iron absorption. Gut. 2004, 53: 617-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  299. Camaschella C: Understanding iron homeostasis through genetic analysis of hemochromatosis and related disorders. Blood. 2005, 106: 3710-7.

    CAS  PubMed  Google Scholar 

  300. Loréal O, Haziza-Pigeon C, Troadec MB, Detivaud L, Turlin B, Courselaud B, Ilyin G, Brissot P: Hepcidin in iron metabolism. Curr Protein Pept Sci. 2005, 6: 279-91.

    PubMed  Google Scholar 

  301. Roetto A, Camaschella C: New insights into iron homeostasis through the study of non-HFE hereditary haemochromatosis. Best Pract Res Clin Haematol. 2005, 18: 235-50.

    CAS  PubMed  Google Scholar 

  302. Roy CN, Andrews NC: Anemia of inflammation: the hepcidin link. Curr Opin Hematol. 2005, 12: 107-11.

    CAS  PubMed  Google Scholar 

  303. Beutler E: Hemochromatosis: Genetics and pathophysiology. Annu Rev Med. 2006, 57: 331-347.

    CAS  PubMed  Google Scholar 

  304. Deicher R, Hörl WH: New insights into the regulation of iron homeostasis. Eur J Clin Invest. 2006, 36: 301-309.

    CAS  PubMed  Google Scholar 

  305. Farnaud S, Patel A, Evans RW: Modelling of a metal-containing hepcidin. Biometals. 2006, 19: 527-533.

    CAS  PubMed  Google Scholar 

  306. Ganz T: Hepcidin – A peptide hormone at the interface of innate immunity and iron metabolism. Antimicrobial Peptides and Human Disease, Current Topics in Microbiology and Immunology. 2006, 306: 183-198.

    CAS  Google Scholar 

  307. Harrison-Findik DD, Schafer D, Klein E, Timchenko NA, Kulaksiz H, Clemens D, Fein E, Andriopoulos B, Pantopoulos K, Gollan J: Alcohol metabolism-mediated oxidative stress down-regulates hepcidin transcription and leads to increased duodenal iron transporter expression. J Biol Chem. 2006, 281: 22974-22982.

    CAS  PubMed  Google Scholar 

  308. Hugman A: Hepcidin: an important new regulator of iron homeostasis. Clin Lab Haematol. 2006, 28: 75-83.

    CAS  PubMed  Google Scholar 

  309. Mena NP, Esparza AL, Núñez MT: Regulation of transepithelial transport of iron by hepcidin. Biol Res. 2006, 39: 191-193.

    CAS  PubMed  Google Scholar 

  310. Mok H, Mlodnicka AE, Hentze MW, Muckenthaler M, Schumacher A: The molecular circuitry regulating the switch between iron deficiency and overload in mice. J Biol Chem. 2006, 281: 7946-51.

    CAS  PubMed  Google Scholar 

  311. Nemeth E, Ganz T: Regulation of iron metabolism by hepcidin. Annual Review of Nutrition. 2006, 26: 323-342.

    CAS  PubMed  Google Scholar 

  312. Atanasiu V, Manolescu B, Stoian I: Hepcidin – central regulator of iron metabolism. European Journal of Haematology. 2007, 78: 1-10.

    CAS  PubMed  Google Scholar 

  313. Malyszko J, Mysliwiec M: Hepcidin in anemia and inflammation in chronic kidney disease. Kidney & Blood Pressure Research. 2007, 30: 15-30.

    CAS  Google Scholar 

  314. Oates PS, Ahmed U: Molecular regulation of hepatic expression of iron regulatory hormone hepcidin. J Gastroenterol Hepatol. 2007, 22: 1378-87.

    CAS  PubMed  Google Scholar 

  315. Barisani D, Pelucchi S, Mariani R, Galimberti S, Trombini P, Fumagalli D, Meneveri R, Nemeth E, Ganz T, Piperno A: Hepcidin and iron-related gene expression in subjects with Dysmetabolic Hepatic Iron Overload. J Hepatol. 2008, 49: 123-33.

    CAS  PubMed  Google Scholar 

  316. Camaschella C, Silvestri L: New and old players in the hepcidin pathway. Haematologica. 2008, 93: 1441-4.

    CAS  PubMed  Google Scholar 

  317. De Domenico I, Nemeth E, Nelson JM, Phillips JD, Ajioka RS, Kay MS, Kushner JP, Ganz T, Ward DM, Kaplan J: The hepcidin-binding site on ferroportin is evolutionarily conserved. Cell Metab. 2008, 8: 146-56.

    CAS  PubMed  PubMed Central  Google Scholar 

  318. Farnaud S, Rapisarda C, Bui T, Drake A, Cammack R, Evans RW: Identification of an iron-hepcidin complex. Biochem J. 2008, 413: 553-7.

    CAS  PubMed  Google Scholar 

  319. Kartikasari AE, Roelofs R, Schaeps RM, Kemna EH, Peters WH, Swinkels DW, Tjalsma H: Secretion of bioactive hepcidin-25 by liver cells correlates with its gene transcription and points towards synergism between iron and inflammation signaling pathways. Biochim Biophys Acta. 2008, 1784: 2029-37.

    CAS  PubMed  Google Scholar 

  320. Muckenthaler MU, Galy B, Hentze MW: Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu Rev Nutr. 2008, 28: 197-213.

    CAS  PubMed  Google Scholar 

  321. Nemeth E: Iron regulation and erythropoiesis. Curr Opin Hematol. 2008, 15: 169-75.

    CAS  PubMed  Google Scholar 

  322. Nishina S, Hino K, Korenaga M, Vecchi C, Pietrangelo A, Mizukami Y, Furutani T, Sakai A, Okuda M, Hidaka I, Okita K, Sakaida I: Hepatitis C virus-induced reactive oxygen species raise hepatic iron level in mice by reducing hepcidin transcription. Gastroenterology. 2008, 134: 226-38.

    CAS  PubMed  Google Scholar 

  323. Peeling P, Dawson B, Goodman C, Landers G, Trinder D: Athletic induced iron deficiency: new insights into the role of inflammation, cytokines and hormones. Eur J Appl Physiol. 2008, 103: 381-91.

    CAS  PubMed  Google Scholar 

  324. Swinkels DW, Girelli D, Laarakkers C, Kroot J, Campostrini N, Kemna EH, Tjalsma H: Advances in quantitative hepcidin measurements by time-of-flight mass spectrometry. PLoS ONE. 2008, 3: e2706.

    PubMed  PubMed Central  Google Scholar 

  325. Swinkels DW, Wetzels JF: Hepcidin: a new tool in the management of anaemia in patients with chronic kidney disease?. Nephrol Dial Transplant. 2008, 23: 2450-3.

    CAS  PubMed  Google Scholar 

  326. Toblli JE, Cao G, Rivas C, Kulaksiz H: Heart and iron deficiency anaemia in rats with renal insufficiency: The role of hepcidin. Nephrology (Carlton). 2008, 13: 636-645.

    CAS  Google Scholar 

  327. Trinder D, Ayonrinde OT, Olynyk JK: HCV, iron, and oxidative stress: the new choreography of hepcidin. Gastroenterology. 2008, 134: 348-51.

    CAS  PubMed  Google Scholar 

  328. Knutson MD, Oukka M, Koss LM, Aydemir F, Wessling-Resnick M: Iron release from macrophages after erythrophagocytosis is up-regulated by ferroportin 1 overexpression and down-regulated by hepcidin. Proc Natl Acad Sci USA. 2005, 102: 1324-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  329. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, Ganz T, Kaplan J: Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004, 306: 2090-2093.

    CAS  PubMed  Google Scholar 

  330. Delaby C, Pilard N, Goncalves AS, Beaumont C, Canonne-Hergaux F: Presence of the iron exporter ferroportin at the plasma membrane of macrophages is enhanced by iron loading and down-regulated by hepcidin. Blood. 2005, 106: 3979-84.

    CAS  PubMed  Google Scholar 

  331. Ganz T: Hepcidin – a regulator of intestinal iron absorption and iron recycling by macrophages. Best Practice & Research Clinical Haematology. 2005, 18: 171-182.

    CAS  Google Scholar 

  332. Papanikolaou G, Tzilianos M, Christakis JI, Bogdanos D, Tsimirika K, MacFarlane J, Goldberg YP, Sakellaropoulos N, Ganz T, Nemeth E: Hepcidin in iron overload disorders. Blood. 2005, 105: 4103-4105.

    CAS  PubMed  PubMed Central  Google Scholar 

  333. Lesbordes-Brion JC, Viatte L, Bennoun M, Lou DQ, Ramey G, Houbron C, Hamard G, Kahn A, Vaulont S: Targeted disruption of the hepcidin 1 gene results in severe hemochromatosis. Blood. 2006, 108: 1402-1405.

    CAS  PubMed  Google Scholar 

  334. Anderson GJ, Frazer DM: Iron metabolism meets signal transduction. Nature Genetics. 2006, 38: 503-504.

    CAS  PubMed  Google Scholar 

  335. Kulaksiz H, Gehrke SG, Janetzko A, Rost D, Bruckner T, Kallinowski B, Stremmel W: Pro-hepcidin: expression and cell specific localisation in the liver and its regulation in hereditary haemochromatosis, chronic renal insufficiency, and renal anaemia. Gut. 2004, 53: 735-743.

    CAS  PubMed  PubMed Central  Google Scholar 

  336. Kulaksiz H, Theilig F, Bachmann S, Gehrke SG, Rost D, Janetzko A, Cetin Y, Stremmel W: The iron-regulatory peptide hormone hepcidin: expression and cellular localization in the mammalian kidney. Journal of Endocrinology. 2005, 184: 361-370.

    CAS  PubMed  Google Scholar 

  337. Tiker F, Celik B, Tarcan A, Kilicdag H, Ozbek N, Gurakan B: Serum pro-hepcidin levels and relationships with iron parameters in healthy preterm and term newborns. Pediatr Hematol Oncol. 2006, 23: 293-297.

    CAS  PubMed  Google Scholar 

  338. Roe MA, Spinks C, Heath ALM, Harvey LJ, Foxall R, Wimperis J, Wolf C, Fairweather-Tait SJ: Serum prohepcidin concentration: no association with iron absorption in healthy men; and no relationship with iron status in men carrying. HFE mutations, hereditary haemochromatosis patients undergoing phlebotomy treatment, or pregnant women. Br J Nutr. 2007, 97: 544-549.

    CAS  PubMed  Google Scholar 

  339. Nicolas G, Chauvet C, Viatte L, Danan JL, Bigard X, Devaux I, Beaumont C, Kahn A, Vaulont S: The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin Invest. 2002, 110: 1037-1044.

    CAS  PubMed  PubMed Central  Google Scholar 

  340. Pak M, Lopez MA, Gabayan V, Ganz T, Rivera S: Suppression of hepcidin during anemia requires erythropoietic activity. Blood. 2006, 108: 3730-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  341. Nemeth E, Valore EV, Territo M, Schiller G, Lichtenstein A, Ganz T: Hepcidin, a putative mediator of anemia of inflammation, is a type II acute-phase protein. Blood. 2003, 101: 2461-2463.

    CAS  PubMed  Google Scholar 

  342. Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK, Ganz T: IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest. 2004, 113: 1271-1276.

    CAS  PubMed  PubMed Central  Google Scholar 

  343. Kemna E, Pickkers P, Nemeth E, Hoeven van der H, Swinkels D: Time-course analysis of hepcidin, serum iron, and plasma cytokine levels in humans injected with LPS. Blood. 2005, 106: 1864-1866.

    CAS  PubMed  Google Scholar 

  344. Fleming RE: Iron and inflammation: cross-talk between pathways regulating hepcidin. J Mol Med. 2008, 86: 491-4.

    PubMed  Google Scholar 

  345. Malyszko J, Malyszko JS, Pawlak K, Drozdowska-Rams L, Brzosko S, Mysliwiec M: Hepcidin is linked to anemia and inflammation in peritoneal dialysis patients. Perit Dial Int. 2008, 28: 418-21.

    CAS  PubMed  Google Scholar 

  346. Wang RH, Li CL, Xu XL, Zheng Y, Xiao CY, Zerfas P, Cooperman S, Eckhaus M, Rouault T, Mishra L, Deng CX: A role of SMAD4 in iron metabolism through the positive regulation of hepicidin expression. Cell Metabolism. 2005, 2: 399-409.

    CAS  PubMed  Google Scholar 

  347. Falzacappa MVV, Spasic MV, Kessler R, Stolte J, Hentze MW, Muckenthaler MU: STAT3 mediates hepatic hepcidin expression and its inflammatory stimulation. Blood. 2007, 109: 353-358.

    CAS  Google Scholar 

  348. Pietrangelo A, Dierssen U, Valli L, Garuti C, Rump A, Corradini E, Ernst M, Klein C, Trautwein C: STAT3 is required for IL-6-gp130-dependent activation of hepcidin in vivo. Gastroenterol. 2007, 132: 294-300.

    CAS  Google Scholar 

  349. Choi SO, Cho YS, Kim HL, Park JW: ROS mediate the hypoxic repression of the hepcidin gene by inhibiting C/EBP alpha and STAT-3. Biochem Biophys Res Comm. 2007, 356: 312-317.

    CAS  PubMed  Google Scholar 

  350. Flanagan JM, Truksa J, Peng HF, Lee P, Beutler E: In vivo imaging of hepcidin promoter stimulation by iron and inflammation. Blood Cells Mols Dis. 2007, 38: 253-257.

    CAS  Google Scholar 

  351. Détivaud L, Nemeth E, Boudjema K, Turlin B, Troadec MB, Leroyer P, Ropert M, Jacquelinet S, Courselaud B, Ganz T, Brissot P, Loréal O: Hepcidin levels in humans are correlated with hepatic iron stores, hemoglobin levels, and hepatic function. Blood. 2005, 106: 746-748.

    PubMed  Google Scholar 

  352. Weizer-Stern O, Adamsky K, Amariglio N, Rachmilewitz E, Breda L, Rivella S, Rechavi G: mRNA expression of iron regulatory genes in beta-thalassemia intermedia and beta-thalassemia major mouse models. Amer J Hematol. 2006, 81: 479-483.

    CAS  Google Scholar 

  353. Weizer-Stern O, Adamsky K, Amariglio N, Levin C, Koren A, Breuer W, Rachmilewitz E, Breda L, Rivella S, Cabantchik ZI, Rechavi G: Downregulation of hepcidin and haemojuvelin expression in the hepatocyte cell-line HepG2 induced by thalassaemic sera. Br J Haematol. 2006, 135: 129-138.

    CAS  PubMed  Google Scholar 

  354. Weizer-Stern O, Adamsky K, Margalit O, Ashur-Fabian O, Givol D, Amariglio N, Rechavi G: Hepcidin, a key regulator of iron metabolism, is transcriptionally activated by p53. Br J Haematol. 2007, 138: 253-62.

    CAS  PubMed  Google Scholar 

  355. Sullivan JL: Macrophage iron, hepcidin, and atherosclerotic plaque stability. Exp Biol Med (Maywood). 2007, 232: 1014-20.

    CAS  Google Scholar 

  356. Adamsky K, Weizer O, Amariglio N, Breda L, Harmelin A, Rivella S, Rachmilewitz E, Rechavi G: Decreased hepcidin mRNA expression in thalassemic mice. Br J Haematol. 2004, 124: 123-124.

    CAS  PubMed  Google Scholar 

  357. Flower DR, North AC, Sansom CE: The lipocalin protein family: structural and sequence overview. Biochim Biophys Acta. 2000, 1482: 9-24.

    CAS  PubMed  Google Scholar 

  358. Kjeldsen L, Johnsen AH, Sengelov H, Borregaard N: Isolation and primary structure of NGAL, a novel protein associated with human neutrophil gelatinase. J Biol Chem. 1993, 268: 10425-32.

    CAS  PubMed  Google Scholar 

  359. Stoesz SP, Gould MN: Overexpression of neu-related lipocalin (NRL) in neu-initiated but not ras or chemically initiated rat mammary carcinomas. Oncogene. 1995, 11: 2233-41.

    CAS  PubMed  Google Scholar 

  360. Kjeldsen L, Cowland JB, Borregaard N: Human neutrophil gelatinase-associated lipocalin and homologous proteins in rat and mouse. Biochim Biophys Acta. 2000, 1482: 272-83.

    CAS  PubMed  Google Scholar 

  361. Liu Q, Ryon J, Nilsen-Hamilton M: Uterocalin: a mouse acute phase protein expressed in the uterus around birth. Mol Reprod Dev. 1997, 46: 507-14.

    CAS  PubMed  Google Scholar 

  362. Goetz DH, Holmes MA, Borregaard N, Bluhm ME, Raymond KN, Strong RK: The neutrophil lipocalin NGAL is a bacteriostatic agent that interferes with siderophore-mediated iron acquisition. Mol Cell. 2002, 10: 1033-43.

    CAS  PubMed  Google Scholar 

  363. Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, Akira S, Aderem A: Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature. 2004, 432: 917-21.

    CAS  PubMed  Google Scholar 

  364. Fluckinger M, Haas H, Merschak P, Glasgow BJ, Redl B: Human tear lipocalin exhibits antimicrobial activity by scavenging microbial siderophores. Antimicrob Agents Chemother. 2004, 48: 3367-72.

    CAS  PubMed  PubMed Central  Google Scholar 

  365. Holmes MA, Paulsene W, Jide X, Ratledge C, Strong RK: Siderocalin (Lcn 2) also binds carboxymycobactins, potentially defending against mycobacterial infections through iron sequestration. Structure. 2005, 13: 29-41.

    CAS  PubMed  Google Scholar 

  366. Berger T, Togawa A, Duncan GS, Elia AJ, You-Ten A, Wakeham A, Fong HE, Cheung CC, Mak TW: Lipocalin 2-deficient mice exhibit increased sensitivity to Escherichia coli infection but not to ischemia-reperfusion injury. Proc Natl Acad Sci USA. 2006, 103: 1834-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  367. Borregaard N, Cowland JB: Neutrophil gelatinase-associated lipocalin, a siderophore-binding eukaryotic protein. Biometals. 2006, 19: 211-215.

    CAS  PubMed  Google Scholar 

  368. Yang J, Goetz D, Li JY, Wang W, Mori K, Setlik D, Du T, Erdjument-Bromage H, Tempst P, Strong R, Barasch J: An iron delivery pathway mediated by a lipocalin. Mol Cell. 2002, 10: 1045-56.

    CAS  PubMed  Google Scholar 

  369. Coles M, Diercks T, Muehlenweg B, Bartsch S, Zolzer V, Tschesche H, Kessler H: The solution structure and dynamics of human neutrophil gelatinase-associated lipocalin. J Mol Biol. 1999, 289: 139-57.

    CAS  PubMed  Google Scholar 

  370. Tschesche H, Zolzer V, Triebel S, Bartsch S: The human neutrophil lipocalin supports the allosteric activation of matrix metalloproteinases. Eur J Biochem. 2001, 268: 1918-1928.

    CAS  PubMed  Google Scholar 

  371. Yan L, Borregaard N, Kjeldsen L, Moses MA: The high molecular weight urinary matrix metalloproteinase (MMP) activity is a complex of gelatinase B/MMP-9 and neutrophil gelatinase-associated lipocalin (NGAL). Modulation of MMP-9 activity by NGAL. J Biol Chem. 2001, 276: 37258-65.

    CAS  PubMed  Google Scholar 

  372. Devarajan P, Mishra J, Supavekin S, Patterson LT, Steven Potter S: Gene expression in early ischemic renal injury: clues towards pathogenesis, biomarker discovery, and novel therapeutics. Mol Genet Metab. 2003, 80: 365-76.

    CAS  PubMed  Google Scholar 

  373. Mishra J, Ma Q, Prada A, Mitsnefes M, Zahedi K, Yang J, Barasch J, Devarajan P: Identification of neutrophil gelatinase-associated lipocalin as a novel early urinary biomarker for ischemic renal injury. J Am Soc Nephrol. 2003, 14: 2534-43.

    CAS  PubMed  Google Scholar 

  374. Mishra J, Dent C, Tarabishi R, Mitsnefes MM, Ma Q, Kelly C, Ruff SM, Zahedi K, Shao M, Bean J, Mori K, Barasch J, Devarajan P: Neutrophil gelatinase-associated lipocalin (NGAL) as a biomarker for acute renal injury after cardiac surgery. Lancet. 2005, 365: 1231-8.

    CAS  PubMed  Google Scholar 

  375. Mori K, Lee HT, Rapoport D, Drexler IR, Foster K, Yang J, Schmidt-Ott KM, Chen X, Li JY, Weiss S, Mishra J, Cheema FH, Markowitz G, Suganami T, Sawai K, Mukoyama M, Kunis C, D'Agati V, Devarajan P, Barasch J: Endocytic delivery of lipoccalin-siderophore-iron complex rescues the kidney from ischemia-reperfusion injury. Journal of Clinical Investigation. 2005, 115: 610-621.

    CAS  PubMed  PubMed Central  Google Scholar 

  376. Herget-Rosenthal S: One step forward in the early detection of acute renal failure. Lancet. 2005, 365: 1205-1206.

    PubMed  Google Scholar 

  377. Trachtman H, Christen E, Cnaan A, Patrick J, Mai V, Mishra J, Jain A, Bullington N, Devarajan P: Urinary neutrophil gelatinase-associated lipocalcin in D+HUS: a novel marker of renal injury. Pediatric Nephrology. 2006, 21: 989-994.

    PubMed  Google Scholar 

  378. Dent CL, Ma Q, Dastrala S, Bennett M, Mitsnefes MM, Barasch J, Devarajan P: Plasma neutrophil gelatinase-associated lipocalin predicts acute kidney injury, morbidity and mortality after pediatric cardiac surgery: a prospective uncontrolled cohort study. Critical Care. 2007, 11: R127.

    PubMed  PubMed Central  Google Scholar 

  379. Devarajan P: Update on mechanisms of ischemic acute kidney injury. Journal of the American Society of Nephrology. 2006, 17: 1503-1520.

    CAS  PubMed  Google Scholar 

  380. Schmidt-Ott KM, Mori K, Kalandadze A, Li JY, Paragas N, Nicholas T, Devarajan P, Barasch J: Neutrophil gelatinase-associated lipocalin-mediated iron traffic in kidney epithelia. Curr Opin Nephrol Hypertens. 2006, 15: 442-9.

    CAS  PubMed  Google Scholar 

  381. Schmidt-Ott KM, Mori K, Li JY, Kalandadze A, Cohen DJ, Devarajan P, Barasch J: Dual action of neutrophil gelatinase-associated lipocalin. J Am Soc Nephrol. 2007, 18: 407-13.

    CAS  PubMed  Google Scholar 

  382. Mori K, Nakao K: Neutrophil gelatinase-associated lipocalin as the real-time indicator of active kidney damage. Kidney Int. 2007, 71: 967-70.

    CAS  PubMed  Google Scholar 

  383. Mitsnefes MM, Kathman TS, Mishra J, Kartal J, Khoury PR, Nickolas TL, Barasch J, Devarajan P: Serum neutrophil gelatinase-associated lipocalin as a marker of renal function in children with chronic kidney disease. Pediatr Nephrol. 2007, 22: 101-8.

    PubMed  Google Scholar 

  384. Zappitelli M, Washburn KK, Arikan AA, Loftis L, Ma Q, Devarajan P, Parikh CR, Goldstein SL: Urine neutrophil gelatinase-associated lipocalin is an early marker of acute kidney injury in critically ill children: a prospective cohort study. Critical Care. 2007, 11.

    Google Scholar 

  385. Molitoris BA, Melnikov VY, Okusa MD, Himmelfarb J: Technology Insight: biomarker development in acute kidney injury–what can we anticipate?. Nat Clin Pract Nephrol. 2008, 4: 154-65.

    CAS  PubMed  Google Scholar 

  386. Nickolas TL, O'Rourke MJ, Yang J, Sise ME, Canetta PA, Barasch N, Buchen C, Khan F, Mori K, Giglio J, Devarajan P, Barasch J: Sensitivity and specificity of a single emergency department measurement of urinary neutrophil gelatinase-associated lipocalin for diagnosing acute kidney injury. Ann Intern Med. 2008, 148: 810-9.

    PubMed  PubMed Central  Google Scholar 

  387. Malyszko J, Bachorzewska-Gajewska H, Sitniewska E, Malyszko JS, Poniatowski B, Dobrzycki S: Serum neutrophil gelatinase-associated lipocalin as a marker of renal function in non-diabetic patients with stage 2–4 chronic kidney disease. Ren Fail. 2008, 30: 625-8.

    CAS  PubMed  Google Scholar 

  388. Devarajan P: NGAL in acute kidney injury: from serendipity to utility. Am J Kidney Dis. 2008, 52: 395-9.

    PubMed  Google Scholar 

  389. Devarajan P: Neutrophil gelatinase-associated lipocalin (NGAL): a new marker of kidney disease. Scand J Clin Lab Invest Suppl. 2008, 241: 89-94.

    PubMed  PubMed Central  Google Scholar 

  390. Devireddy LR, Gazin C, Zhu X, Green MR: A cell-surface receptor for lipocalin 24p3 selectively mediates apoptosis and iron uptake. Cell. 2005, 123: 1293-305.

    CAS  PubMed  Google Scholar 

  391. Richardson DR: 24p3 and its receptor: dawn of a new iron age?. Cell. 2005, 123: 1175-7.

    CAS  PubMed  Google Scholar 

  392. Hvidberg V, Jacobsen C, Strong RK, Cowland JB, Moestrup SK, Borregaard N: The endocytic receptor megalin binds the iron transporting neutrophil-gelatinase-associated lipocalin with high affinity and mediates its cellular uptake. FEBS Lett. 2005, 579: 773-7.

    CAS  PubMed  Google Scholar 

  393. Roudkenar MH, Kuwahara Y, Baba T, Roushandeh AM, Ebishima S, Abe S, Ohkubo Y, Fukumoto M: Oxidative stress induced lipocalin 2 gene expression: Addressing its expression under the harmful conditions. Journal of Radiation Research. 2007, 48: 39-44.

    CAS  PubMed  Google Scholar 

  394. Roudkenar MH, Halabian R, Ghasemipour Z, Roushandeh AM, Rouhbakhsh M, Nekogoftar M, Kuwahara Y, Fukumoto M, Shokrgozar MA: Neutrophil Gelatinase-associated Lipocalin acts as a protective factor against H2O2 toxicity. Arch Med Res. 2008, 39: 560-6.

    CAS  PubMed  Google Scholar 

  395. Gueler F, Gwinner W, Schwarz A, Haller H: Long-term effects of acute ischemia and reperfusion injury. Kidney International. 2004, 66: 523-527.

    PubMed  Google Scholar 

  396. Sikorski EM, Hock T, Hill-Kapturczak N, Agarwal A: The story so far: molecular regulation of the heme oxygenase-1 gene in renal injury. Am J Physiol. 2004, 286: F425-F441.

    CAS  Google Scholar 

  397. Shah S: Role of iron in progressive renal disease. Am J Kidney Dis. 2001, 37: S30-3.

    CAS  PubMed  Google Scholar 

  398. Aigner F, Maier HT, Schwelberger HG, Wallnofer EA, Amberger A, Obrist P, Berger T, Mak TW, Maglione M, Margreiter R, Schneeberger S, Troppmair J: Lipocalin-2 regulates the inflammatory response during ischemia and reperfusion of the transplanted heart. Amer J Transplantation. 2007, 7: 779-788.

    CAS  Google Scholar 

  399. Abergel RJ, Moore EG, Strong RK, Raymond KN: Microbial evasion of the immune system: structural modifications of enterobactin impair siderocalin recognition. J Am Chem Soc. 2006, 128: 10998-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  400. Fischbach MA, Lin H, Zhou L, Yu Y, Abergel RJ, Liu DR, Raymond KN, Wanner BL, Strong RK, Walsh CT, Aderem A, Smith KD: The pathogen-associated iroA gene cluster mediates bacterial evasion of lipocalin 2. Proc Natl Acad Sci USA. 2006, 103: 16502-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  401. Abergel RJ, Clifton MC, Pizarro JC, Warner JA, Shuh DK, Strong RK, Raymond KN: The siderocalin/enterobactin interaction: a link between mammalian immunity and bacterial iron transport. J Am Chem Soc. 2008, 130: 11524-34.

    CAS  PubMed  PubMed Central  Google Scholar 

  402. Cowland JB, Borregaard N: Molecular characterization and pattern of tissue expression of the gene for neutrophil gelatinase-associated lipocalin from humans. Genomics. 1997, 45: 17-23.

    CAS  PubMed  Google Scholar 

  403. Xu S, Venge P: Lipocalins as biochemical markers of disease. Biochim Biophys Acta. 2000, 1482: 298-307.

    CAS  PubMed  Google Scholar 

  404. Cowland JB, Sorensen OE, Sehested M, Borregaard N: Neutrophil gelatinase-associated lipocalin is up-regulated in human epithelial cells by IL-1 beta, but not by TNF-alpha. J Immunol. 2003, 171: 6630-9.

    CAS  PubMed  Google Scholar 

  405. Cowland JB, Muta T, Borregaard N: IL-1beta-specific up-regulation of neutrophil gelatinase-associated lipocalin is controlled by IkappaB-zeta. J Immunol. 2006, 176: 5559-66.

    CAS  PubMed  Google Scholar 

  406. Elneihoum AM, Falke P, Axelsson L, Lundberg E, Lindgarde F, Ohlsson K: Leukocyte activation detected by increased plasma levels of inflammatory mediators in patients with ischemic cerebrovascular diseases. Stroke. 1996, 27: 1734-8.

    CAS  PubMed  Google Scholar 

  407. Elneihoum AM, Falke P, Hedblad B, Lindgarde F, Ohlsson K: Leukocyte activation in atherosclerosis: correlation with risk factors. Atherosclerosis. 1997, 131: 79-84.

    CAS  PubMed  Google Scholar 

  408. Falke P, Elneihoum AM, Ohlsson K: Leukocyte activation: relation to cardiovascular mortality after cerebrovascular ischemia. Cerebrovasc Dis. 2000, 10: 97-101.

    CAS  PubMed  Google Scholar 

  409. Hemdahl AL, Gabrielsen A, Zhu C, Eriksson P, Hedin U, Kastrup J, Thoren P, Hansson GK: Expression of neutrophil gelatinase-associated lipocalin in atherosclerosis and myocardial infarction. Arterioscler Thromb Vasc Biol. 2006, 26: 136-42.

    CAS  PubMed  Google Scholar 

  410. Stoesz SP, Friedl A, Haag JD, Lindstrom MJ, Clark GM, Gould MN: Heterogeneous expression of the lipocalin NGAL in primary breast cancers. Int J Cancer. 1998, 79: 565-72.

    CAS  PubMed  Google Scholar 

  411. Tong Z, Wu X, Ovcharenko D, Zhu J, Chen CS, Kehrer JP: Neutrophil gelatinase-associated lipocalin as a survival factor. Biochem J. 2005, 391: 441-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  412. Tong Z, Kunnumakkara AB, Wang H, Matsuo Y, Diagaradjane P, Harikumar KB, Ramachandran V, Sung B, Chakraborty A, Bresalier RS, Logsdon C, Aggarwal BB, Krishnan S, Guha S: Neutrophil gelatinase-associated lipocalin: a novel suppressor of invasion and angiogenesis in pancreatic cancer. Cancer Res. 2008, 68: 6100-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  413. Weinberg ED: Iron out-of-balance: a risk factor for acute and chronic diseases. Hemoglobin. 2008, 32: 117-22.

    CAS  PubMed  Google Scholar 

  414. CESDI: Confidential Enquiry into Stillbirths and Deaths in Infancy. 5th Annual Report. 1998, Maternal and Child Health Research Consortium

    Google Scholar 

  415. LaMarca BD, Gilbert J, Granger JP: Recent progress toward the understanding of the pathophysiology of hypertension during preeclampsia. Hypertension. 2008, 51: 982-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  416. Redman CW, Sargent IL: Placental debris, oxidative stress and pre-eclampsia. Placenta. 2000, 21: 597-602.

    CAS  PubMed  Google Scholar 

  417. DiFederico E, Genbacev O, Fisher SJ: Preeclampsia is associated with widespread apoptosis of placental cytotrophoblasts within the uterine wall. Am J Pathol. 1999, 155: 293-301.

    CAS  PubMed  PubMed Central  Google Scholar 

  418. Leung DN, Smith SC, To KF, Sahota DS, Baker PN: Increased placental apoptosis in pregnancies complicated by preeclampsia. Am J Obstet Gynecol. 2001, 184: 1249-50.

    CAS  PubMed  Google Scholar 

  419. Huppertz B, Kingdom JC: Apoptosis in the trophoblast–role of apoptosis in placental morphogenesis. J Soc Gynecol Investig. 2004, 11: 353-62.

    CAS  PubMed  Google Scholar 

  420. Harris LK, Keogh RJ, Wareing M, Baker PN, Cartwright JE, Aplin JD, Whitley GS: Invasive trophoblasts stimulate vascular smooth muscle cell apoptosis by a fas ligand-dependent mechanism. Am J Pathol. 2006, 169: 1863-74.

    CAS  PubMed  PubMed Central  Google Scholar 

  421. Heazell AE, Moll SJ, Jones CJ, Baker PN, Crocker IP: Formation of syncytial knots is increased by hyperoxia, hypoxia and reactive oxygen species. Placenta. 2007, 28 (Suppl A): S33-40.

    PubMed  Google Scholar 

  422. Keogh RJ, Harris LK, Freeman A, Baker PN, Aplin JD, Whitley GS, Cartwright JE: Fetal-derived trophoblast use the apoptotic cytokine tumor necrosis factor-alpha-related apoptosis-inducing ligand to induce smooth muscle cell death. Circ Res. 2007, 100: 834-41.

    CAS  PubMed  Google Scholar 

  423. Heazell AE, Lacey HA, Jones CJ, Huppertz B, Baker PN, Crocker IP: Effects of oxygen on cell turnover and expression of regulators of apoptosis in human placental trophoblast. Placenta. 2008, 29: 175-86.

    CAS  PubMed  Google Scholar 

  424. Combs CA, Katz MA, Kitzmiller JL, Brescia RJ: Experimental preeclampsia produced by chronic constriction of the lower aorta: validation with longitudinal blood pressure measurements in conscious rhesus monkeys. Am J Obstet Gynecol. 1993, 169: 215-23.

    CAS  PubMed  Google Scholar 

  425. Wisdom SJ, Wilson R, McKillop JH, Walker JJ: Antioxidant systems in normal pregnancy and in pregnancy-induced hypertension. Am J Obs Gynecol. 1991, 165: 1701-1704.

    CAS  Google Scholar 

  426. Genbacev O, Joslin R, Damsky CH, Polliotti BM, Fisher SJ: Hypoxia alters early gestation human cytotrophoblast differentiation/invasion in vitro and models the placental defects that occur in preeclampsia. J Clin Invest. 1996, 97: 540-50.

    CAS  PubMed  PubMed Central  Google Scholar 

  427. Benyo DF, Miles TM, Conrad KP: Hypoxia stimulates cytokine production by villous explants from the human placenta. J Clin Endocrinol Metab. 1997, 82: 1582-8.

    CAS  PubMed  Google Scholar 

  428. Conrad KP, Benyo DF: Placental cytokines and the pathogenesis of preeclampsia. Am J Reprod Immunol. 1997, 37: 240-9.

    CAS  PubMed  Google Scholar 

  429. Lefevre G, Berkane N, Uzan S, Etienne J: Preeclampsia and oxygen free radicals. Annales De Biologie Clinique. 1997, 55: 443-450.

    CAS  PubMed  Google Scholar 

  430. Conrad KP, Miles TM, Benyo DF: Circulating levels of immunoreactive cytokines in women with preeclampsia. Am J Reprod Immunol. 1998, 40: 102-11.

    CAS  PubMed  Google Scholar 

  431. Davidge ST: Oxidative stress and altered endothelial cell function in preeclampsia. Semin Reprod Endocrinol. 1998, 16: 65-73.

    CAS  PubMed  Google Scholar 

  432. Mise H, Sagawa N, Matsumoto T, Yura S, Nanno H, Itoh H, Mori T, Masuzaki H, Hosoda K, Ogawa Y, Nakao K: Augmented placental production of leptin in preeclampsia: possible involvement of placental hypoxia. J Clin Endocrinol Metab. 1998, 83: 3225-9.

    CAS  PubMed  Google Scholar 

  433. Walker JJ: Antioxidants and inflammatory cell response in preeclampsia. Semin Reprod Endocrinol. 1998, 16: 47-55.

    CAS  PubMed  Google Scholar 

  434. Walsh SW: Maternal-placental interactions of oxidative stress and antioxidants in preeclampsia. Seminars in Reproductive Endocrinology. 1998, 16: 93-104.

    CAS  PubMed  Google Scholar 

  435. Caniggia I, Grisaru-Gravnosky S, Kuliszewsky M, Post M, Lye SJ: Inhibition of TGF-beta 3 restores the invasive capability of extravillous trophoblasts in preeclamptic pregnancies. J Clin Invest. 1999, 103: 1641-50.

    CAS  PubMed  PubMed Central  Google Scholar 

  436. Hubel CA: Oxidative stress in the pathogenesis of preeclampsia. Proc Soc Exp Biol Med. 1999, 222: 222-35.

    CAS  PubMed  Google Scholar 

  437. Rinehart BK, Terrone DA, Lagoo-Deenadayalan S, Barber WH, Hale EA, Martin JN, Bennett WA: Expression of the placental cytokines tumor necrosis factor alpha, interleukin 1beta, and interleukin 10 is increased in preeclampsia. Am J Obstet Gynecol. 1999, 181: 915-20.

    CAS  PubMed  Google Scholar 

  438. Caniggia I, Mostachfi H, Winter J, Gassmann M, Lye SJ, Kuliszewski M, Post M: Hypoxia-inducible factor-1 mediates the biological effects of oxygen on human trophoblast differentiation through TGFbeta(3). J Clin Invest. 2000, 105: 577-87.

    CAS  PubMed  PubMed Central  Google Scholar 

  439. Kharb S: Total free radical trapping antioxidant potential in pre-eclampsia. Int J Gynaecol Obstet. 2000, 69: 23-6.

    CAS  PubMed  Google Scholar 

  440. Tálosi G, Endreffy E, Túri S, Németh I: Molecular and genetic aspects of preeclampsia: State of the art. Mol Genet Metab. 2000, 71: 565-572.

    PubMed  Google Scholar 

  441. Walsh SW, Vaughan JE, Wang Y, Roberts LJ: Placental isoprostane is significantly increased in preeclampsia. FASEB J. 2000, 14: 1289-96.

    CAS  PubMed  Google Scholar 

  442. Wolf M, Kettyle E, Sandler L, Ecker JL, Roberts J, Thadhani R: Obesity and preeclampsia: the potential role of inflammation. Obstet Gynecol. 2001, 98: 757-62.

    CAS  PubMed  Google Scholar 

  443. Benyo DF, Smarason A, Redman CW, Sims C, Conrad KP: Expression of inflammatory cytokines in placentas from women with preeclampsia. J Clin Endocrinol Metab. 2001, 86: 2505-12.

    CAS  PubMed  Google Scholar 

  444. Hung TH, Skepper JN, Burton GJ: In vitro ischemia-reperfusion injury in term human placenta as a model for oxidative stress in pathological pregnancies. Am J Pathol. 2001, 159: 1031-43.

    CAS  PubMed  PubMed Central  Google Scholar 

  445. López-Jaramillo P, Casas JP, Serrano N: Preeclampsia: from epidemiological observations to molecular mechanisms. Braz J Med Biol Res. 2001, 34: 1227-35.

    PubMed  Google Scholar 

  446. Malek A, Sager R, Schneider H: Effect of hypoxia, oxidative stress and lipopolysaccharides on the release of prostaglandins and cytokines from human term placental explants. Placenta. 2001, 22 (Suppl A): S45-50.

    PubMed  Google Scholar 

  447. Gitto E, Reiter RJ, Karbownik M, Tan DX, Gitto P, Barberi S, Barberi I: Causes of oxidative stress in the pre- and perinatal period. Biology of the Neonate. 2002, 81: 146-157.

    CAS  PubMed  Google Scholar 

  448. Granger JP, Alexander BT, Llinas MT, Bennett WA, Khalil RA: Pathophysiology of preeclampsia: linking placental ischemia/hypoxia with microvascular dysfunction. Microcirculation. 2002, 9: 147-60.

    CAS  PubMed  Google Scholar 

  449. Roberts JM, Lain KY: Recent Insights into the pathogenesis of pre-eclampsia. Placenta. 2002, 23: 359-72.

    CAS  PubMed  Google Scholar 

  450. Vaughan JE, Walsh SW: Oxidative stress reproduces placental abnormalities of preeclampsia. Hypertens Preg. 2002, 21: 205-223.

    CAS  Google Scholar 

  451. Burton GJ, Jauniaux E: Placental oxidative stress: from miscarriage to preeclampsia. J Soc Gynecol Investig. 2004, 11: 342-352.

    CAS  PubMed  Google Scholar 

  452. Connors N, Merrill D: Antioxidants for prevention of preterm delivery. Clin Obs Gynecol. 2004, 47: 822-832.

    Google Scholar 

  453. Raijmakers MT, Peters WH, Steegers EA, Poston L: Amino thiols, detoxification and oxidative stress in pre-eclampsia and other disorders of pregnancy. Curr Pharm Des. 2005, 11: 711-34.

    CAS  PubMed  Google Scholar 

  454. Redman CW, Sargent IL: Pre-eclampsia, the placenta and the maternal systemic inflammatory response–a review. Placenta. 2003, 24 (Suppl A): S21-7.

    PubMed  Google Scholar 

  455. Downing JW, Ramasubramanian R, Johnson RF, Minzter BH, Paschall RL, Sundell HW, Engelhardt B, Lewis R: Hypothesis: selective phosphodiesterase-5 inhibition improves outcome in preeclampsia. Med Hypotheses. 2004, 63: 1057-64.

    CAS  PubMed  Google Scholar 

  456. Myatt L, Cui XL: Oxidative stress in the placenta. Histochem Cell Biol. 2004, 122: 369-382.

    CAS  PubMed  Google Scholar 

  457. Touyz RM, Schiffrin EL: Reactive oxygen species in vascular biology: implications in hypertension. Histochem Cell Biol. 2004, 122: 339-52.

    CAS  PubMed  Google Scholar 

  458. Touyz RM: Reactive oxygen species, vascular oxidative stress, and redox signaling in hypertension: what is the clinical significance?. Hypertension. 2004, 44: 248-52.

    CAS  PubMed  Google Scholar 

  459. Tsukahara H, Ohta N, Sato S, Hiraoka M, Shukunami K, Uchiyama M, Kawakami H, Sekine K, Mayumi M: Concentrations of pentosidine, an advanced glycation end-product, in umbilical cord blood. Free Radic Res. 2004, 38: 691-5.

    CAS  PubMed  Google Scholar 

  460. Bdolah Y, Karumanchi SA, Sachs BP: Recent advances in understanding of preeclampsia. Croat Med J. 2005, 46: 728-36.

    PubMed  Google Scholar 

  461. Biondi C, Pavan B, Lunghi L, Florini S, Vesce F: The role and modulation of the oxidative balance in pregnancy. Curr Pharmaceut Design. 2005, 11: 2075-2089.

    CAS  Google Scholar 

  462. Engin-Üstün Y, Üstün Y, Kamaci M, Şekeroğlu R: Maternal serum ceruloplasmin in preectampsia. Int J Gynecol Obs. 2005, 89: 51-52.

    Google Scholar 

  463. Gupta S, Agarwal A, Sharma RK: The role of placental oxidative stress and lipid peroxidation in preeclampsia. Obstet Gynecol Surv. 2005, 60: 807-16.

    PubMed  Google Scholar 

  464. Noris M, Perico N, Remuzzi G: Mechanisms of disease: pre-eclampsia. Nature Clin Pract Nephrol. 2005, 1: 98-114.

    CAS  Google Scholar 

  465. Redman CW, Sargent IL: Latest advances in understanding preeclampsia. Science. 2005, 308: 1592-1594.

    CAS  PubMed  Google Scholar 

  466. Roberts JM, Gammill HS: Preeclampsia: recent insights. Hypertension. 2005, 46: 1243-9.

    CAS  PubMed  Google Scholar 

  467. Rodrigo R, Parra M, Bosco C, Fernandez V, Barja P, Guajardo J, Messina R: Pathophysiological basis for the prophylaxis of preeclampsia through early supplementation with antioxidant vitamins. Pharmacology & Therapeutics. 2005, 107: 177-197.

    CAS  Google Scholar 

  468. Scholl TO, Leskiw M, Chen X, Sims M, Stein TP: Oxidative stress, diet, and the etiology of preeclampsia. Am J Clin Nutr. 2005, 81: 1390-6.

    CAS  PubMed  Google Scholar 

  469. Soleymanlou N, Jurisica I, Nevo O, Ietta F, Zhang X, Zamudio S, Post M, Caniggia I: Molecular evidence of placental hypoxia in preeclampsia. J Clin Endocrinol Metab. 2005, 90: 4299-4308.

    CAS  PubMed  Google Scholar 

  470. Spinnato JA, Livingston JC: Prevention of preeclampsia with antioxidants: Evidence from randomized trials. Clinical Obstetrics and Gynecology. 2005, 48: 416-429.

    PubMed  Google Scholar 

  471. Blackburn S: Free radicals in perinatal and neonatal care, part 2 – Oxidative stress during the perinatal and neonatal period. Journal of Perinatal & Neonatal Nursing. 2006, 20: 125-127.

    Google Scholar 

  472. Borzychowski AM, Sargent IL, Redman CW: Inflammation and pre-eclampsia. Semin Fetal Neonatal Med. 2006, 11: 309-16.

    CAS  PubMed  Google Scholar 

  473. Hauguel-de Mouzon S, Guerre-Millo M: The placenta cytokine network and inflammatory signals. Placenta. 2006, 27: 794-8.

    CAS  PubMed  Google Scholar 

  474. Perkins AV: Endogenous anti-oxidants in pregnancy and preeclampsia. Aus New Zeal J Obs Gynaecol. 2006, 46: 77-83.

    Google Scholar 

  475. Rogers MS, Wang CC, Tam WH, Li CY, Chu KO, Chu CY: Oxidative stress in midpregnancy as a predictor of gestational hypertension and pre-eclampsia. BJOG. 2006, 113: 1053-9.

    CAS  PubMed  Google Scholar 

  476. Sankaralingam S, Arenas IA, Lalu MM, Davidge ST: Preeclampsia: current understanding of the molecular basis of vascular dysfunction. Expert Rev Mol Med. 2006, 8: 1-20.

    PubMed  Google Scholar 

  477. Sargent IL, Borzychowski AM, Redman CW: Immunoregulation in normal pregnancy and pre-eclampsia: an overview. Reprod Biomed Online. 2006, 13: 680-6.

    CAS  PubMed  Google Scholar 

  478. Sharma JB, Sharma A, Bahadur A, Vimala N, Satyam A, Mittal S: Oxidative stress markers and antioxidant levels in normal pregnancy and pre-eclampsia. Int J Gynecol Obs. 2006, 94: 23-27.

    CAS  Google Scholar 

  479. Webster RP, Brockman D, Myatt L: Nitration of p38 MAPK in the placenta: association of nitration with reduced catalytic activity of p38 MAPK in pre-eclampsia. Molecular Human Reproduction. 2006, 12: 677-685.

    CAS  PubMed  Google Scholar 

  480. Burdon C, Mann C, Cindrova-Davies T, Ferguson-Smith AC, Burton GJ: Oxidative stress and the induction of cyclooxygenase enzymes and apoptosis in the murine placenta. Placenta. 2007, 28: 724-33.

    CAS  PubMed  PubMed Central  Google Scholar 

  481. Crocker I: Gabor Than Award Lecture 2006: pre-eclampsia and villous trophoblast turnover: perspectives and possibilities. Placenta. 2007, 28 (Suppl A): S4-13.

    PubMed  Google Scholar 

  482. Keelan JA, Mitchell MD: Placental cytokines and preeclampsia. Front Biosci. 2007, 12: 2706-27.

    CAS  PubMed  Google Scholar 

  483. Milczarek R, Sokolowska E, Hallmann A, Klimek J: The NADPH- and iron-dependent lipid peroxidation in human placental microsomes. Molecular and Cellular Biochemistry. 2007, 295: 105-111.

    CAS  PubMed  Google Scholar 

  484. Schiessl B: Inflammatory response in preeclampsia. Mol Aspects Med. 2007, 28: 210-9.

    CAS  PubMed  Google Scholar 

  485. Shah TJ, Walsh SW: Activation of NF-kappaB and expression of COX-2 in association with neutrophil infiltration in systemic vascular tissue of women with preeclampsia. Am J Obstet Gynecol. 2007, 196: 48 e1-8.

    PubMed  Google Scholar 

  486. Walsh SW: Obesity: a risk factor for preeclampsia. Trends Endocrinol Metab. 2007, 18: 365-70.

    CAS  PubMed  Google Scholar 

  487. Zamudio S: High-altitude hypoxia and preeclampsia. Frontiers in Bioscience. 2007, 12: 2967-2977.

    CAS  PubMed  Google Scholar 

  488. Gilbert JS, Ryan MJ, LaMarca BB, Sedeek M, Murphy SR, Granger JP: Pathophysiology of hypertension during preeclampsia: linking placental ischemia with endothelial dysfunction. Am J Physiol Heart Circ Physiol. 2008, 294: H541-50.

    CAS  PubMed  Google Scholar 

  489. Robinson NJ, Wareing M, Hudson NK, Blankley RT, Baker PN, Aplin JD, Crocker IP: Oxygen and the liberation of placental factors responsible for vascular compromise. Lab Invest. 2008

    Google Scholar 

  490. Entman SS, Richardson LD, Killam AP: Elevated serum ferritin in the altered ferrokinetics of toxemia of pregnancy. AmJ Obs Gynecol. 1982, 144: 418-422.

    CAS  Google Scholar 

  491. Entman SS, Richardson LD: Clinical applications of the altered iron kinetics of toxemia of pregnancy. Am J Obs Gynecol. 1983, 146: 568-574.

    CAS  Google Scholar 

  492. Entman SS, Richardson LD, Killam AP: Altered ferrokinetics in toxemia of pregnancy – a possible indicator of decreased red cell survival. Clin Exp Hypertens B. 1983, 2: 171-178.

    CAS  PubMed  Google Scholar 

  493. Entman SS, Kambam JR, Bradley CA, Cousar JB: Increased levels of carboxyhemoglobin and serum iron as an indicator of increased red cell turnover in preeclampsia. Am J Obstet Gynecol. 1987, 156: 1169-73.

    CAS  PubMed  Google Scholar 

  494. Samuels P, Main EK, Mennuti MT, Gabbe SG: The origin of increased serum iron in pregnancy-induced hypertension. Am J Obstet Gynecol. 1987, 157: 721-5.

    CAS  PubMed  Google Scholar 

  495. Lindheimer MD, Katz AI: Preeclampsia – Patho-physiology, diagnosis, and management. Annual Review of Medicine. 1989, 40: 233-250.

    CAS  PubMed  Google Scholar 

  496. Raman L, Pawashe AB, Yasodhara P: Hyperferritinemia in pregnancy induced hypertension and eclampsia. J Postgrad Med. 1992, 38: 65-7.

    CAS  PubMed  Google Scholar 

  497. Das SS, Dhall GI, Dhall K, Dash S: Significance of serum iron levels as a biochemical marker in pregnancy-induced hypertension. Int J Gynecol Obs. 1994, 45: 3-9.

    CAS  Google Scholar 

  498. Hubel CA, Kozlov AV, Kagan VE, Evans RW, Davidge ST, McLaughlin MK, Roberts JM: Decreased transferrin and increased transferrin saturation in sera of women with preeclampsia: implications for oxidative stress. Am J Obstet Gynecol. 1996, 175: 692-700.

    CAS  PubMed  Google Scholar 

  499. Sizoo BB, Paarlberg MM, Bouman AA, Dekker GA: The role of serum iron levels in diagnostic hypertensive disorders in pregnancy. Hypertension in Pregnancy. 1997, 16: 425-433.

    Google Scholar 

  500. Vitoratos N, Salamalekis E, Dalamaga N, Kassanos D, Creatsas G: Defective antioxidant mechanisms via changes in serum ceruloplasmin and total iron binding capacity of serum in women with pre-eclampsia. Eur J Obstet Gynecol Reprod Biol. 1999, 84: 63-7.

    CAS  PubMed  Google Scholar 

  501. Lao TT, Tam KF, Chan LY: Third trimester iron status and pregnancy outcome in non-anaemic women; pregnancy unfavourably affected by maternal iron excess. Hum Reprod. 2000, 15: 1843-8.

    CAS  PubMed  Google Scholar 

  502. Waterstone M, Bewley S, Wolfe C: Incidence and predictors of severe obstetric morbidity: case-control study. BMJ. 2001, 322: 1089-93.

    CAS  PubMed  PubMed Central  Google Scholar 

  503. Mello G, Parretti E, Cioni R, Lagozio C, Mealli F, Pratesi M: Individual longitudinal patterns in biochemical and hematological markers for the early prediction of pre-eclampsia. J Matern Fetal Neonatal Med. 2002, 11: 93-9.

    CAS  PubMed  Google Scholar 

  504. Casanueva E, Viteri FE: Iron and oxidative stress in pregnancy. J Nutr. 2003, 133: 1700S-1708S.

    CAS  PubMed  Google Scholar 

  505. Hubel CA, Bodnar LM, Many A, Harger G, Ness RB, Roberts JM: Nonglycosylated ferritin predominates in the circulation of women with preeclampsia but not intrauterine growth restriction. Clin Chem. 2004, 50: 948-951.

    CAS  PubMed  Google Scholar 

  506. Norwitz ER, Tsen LC, Park JS, Fitzpatrick PA, Dorfman DM, Saade GR, Buhimschi CS, Buhimschi IA: Discriminatory proteomic biomarker analysis identifies free hemoglobin in the cerebrospinal fluid of women with severe preeclampsia. Amer J Obs Gynecol. 2005, 193: 957-964.

    CAS  Google Scholar 

  507. Scholl TO: Iron status during pregnancy: setting the stage for mother and infant. Am J Clin Nutr. 2005, 81: 1218S-1222S.

    CAS  PubMed  Google Scholar 

  508. Serdar Z, Gür E, Develioğlu O: Serum iron and copper status and oxidative stress in severe and mild preeclampsia. Cell Biochem Funct. 2006, 24: 209-15.

    CAS  PubMed  Google Scholar 

  509. Smith TG, Robbins PA: Iron, pre-eclampsia and hypoxia-inducible factor. Bjog. 2007, 114: 1581-2.

    CAS  PubMed  Google Scholar 

  510. Ziaei S, Norrozi M, Faghihzadeh S, Jafarbegloo E: A randomised placebo-controlled trial to determine the effect of iron supplementation on pregnancy outcome in pregnant women with haemoglobin > or = 13.2 g/dl. BJOG. 2007, 114: 684-8.

    CAS  PubMed  Google Scholar 

  511. Guller S, Buhimschi CS, Ma YY, Huang ST, Yang L, Kuczynski E, Zambrano E, Lockwood CJ, Buhimschi IA: Placental expression of ceruloplasmin in pregnancies complicated by severe preeclampsia. Lab Invest. 2008, 88: 1057-67.

    CAS  PubMed  PubMed Central  Google Scholar 

  512. Gambling L, Danzeisen R, Gair S, Lea RG, Charania Z, Solanky N, Joory KD, Srai SKS, McArdle HJ: Effect of iron deficiency on placental transfer of iron and expression of iron transport proteins in vivo and in vitro. Biochem J. 2001, 356: 883-889.

    CAS  PubMed  PubMed Central  Google Scholar 

  513. Freeman DJ, McManus F, Brown EA, Cherry L, Norrie J, Ramsay JE, Clark P, Walker ID, Sattar N, Greer IA: Short- and long-term changes in plasma inflammatory markers associated with preeclampsia. Hypertension. 2004, 44: 708-14.

    CAS  PubMed  Google Scholar 

  514. Bodnar LM, Ness RB, Harger GF, Roberts JM: Inflammation and triglycerides partially mediate the effect of prepregnancy body mass index on the risk of preeclampsia. Am J Epidemiol. 2005, 162: 1198-206.

    PubMed  Google Scholar 

  515. Braekke K, Holthe MR, Harsem NK, Fagerhol MK, Staff AC: Calprotectin, a marker of inflammation, is elevated in the maternal but not in the fetal circulation in preeclampsia. Am J Obstet Gynecol. 2005, 193: 227-33.

    CAS  PubMed  Google Scholar 

  516. Hu W, Wang H, Wang Z, Huang H, Dong M: Elevated serum levels of interleukin-15 and interleukin-16 in preeclampsia. J Reprod Immunol. 2007, 73: 166-71.

    CAS  PubMed  Google Scholar 

  517. Keelan JA, Mitchell MD: Cytokines, hypoxia, and preeclampsia. J Soc Gynecol Investig. 2005, 12: 385-7.

    PubMed  Google Scholar 

  518. Elovitz MA: Anti-inflammatory interventions in pregnancy: now and the future. Semin Fetal Neonatal Med. 2006, 11: 327-32.

    PubMed  Google Scholar 

  519. Paternoster DM, Fantinato S, Stella A, Nanhorngue KN, Milani M, Plebani M, Nicolini U, Girolami A: C-reactive protein in hypertensive disorders in pregnancy. Clin Appl Thromb Hemost. 2006, 12: 330-7.

    PubMed  Google Scholar 

  520. Dorrepaal CA, Berger HM, Benders M, vanZoerenGrobben D, VanDeBor M, VanBel F: Nonprotein-bound iron in postasphyxial reperfusion injury of the newborn. Pediatrics. 1996, 98: 883-889.

    CAS  PubMed  Google Scholar 

  521. Goldenberg RL, Tamura T, DuBard M, Johnston KE, Copper RL, Neggers Y: Plasma ferritin and pregnancy outcome. Am J Obs Gynecol. 1996, 175: 1356-1359.

    CAS  Google Scholar 

  522. Tamura T, Goldenberg RL, Johnston KE, Cliver SP, Hickey CA: Serum ferritin: A predictor of early spontaneous preterm delivery. Obs Gynecol. 1996, 87: 360-365.

    CAS  Google Scholar 

  523. Buhimschi IA, Buhimschi CS, Weiner CP: Protective effect of N-acetylcysteine against fetal death and preterm labor induced by maternal inflammation. American Journal of Obstetrics and Gynecology. 2003, 188: 203-208.

    CAS  PubMed  Google Scholar 

  524. Ciccoli L, Rossi V, Leoncini S, Signorini C, Paffetti P, Bracci R, Buonocore G, Comporti M: Iron release in erythrocytes and plasma non protein-bound iron in hypoxic and non hypoxic newborns. Free Radical Research. 2003, 37: 51-58.

    CAS  PubMed  Google Scholar 

  525. Buhimschi IA, Saade GR, Chwalisz K, Garfield RE: The nitric oxide pathway in pre-eclampsia: pathophysiological implications. Human Reproduction Update. 1998, 4: 25-42.

    CAS  PubMed  Google Scholar 

  526. Milman N: Iron and pregnancy – a delicate balance. Annals of Hematology. 2006, 85: 559-565.

    CAS  PubMed  Google Scholar 

  527. Milman N: Iron prophylaxis in pregnancy-general or individual and in which dose?. Annals of Hematology. 2006, 85: 821-828.

    CAS  PubMed  Google Scholar 

  528. Lachili B, Hininger I, Faure H, Arnaud J, Richard MJ, Favier A, Roussel AM: Increased lipid peroxidation in pregnant women after iron and vitamin C supplementation. Biol Trace Elem Res. 2001, 83: 103-10.

    CAS  PubMed  Google Scholar 

  529. Devrim E, Tarhan I, Ergüder IB, Durak I: Oxidant/antioxidant status of placenta, blood, and cord blood samples from pregnant women supplemented with iron. J Soc Gynecol Investig. 2006, 13: 502-5.

    CAS  PubMed  Google Scholar 

  530. Keyes LE, Armaza JF, Niermeyer S, Vargas E, Young DA, Moore LG: Intrauterine growth restriction, preeclampsia, and intrauterine mortality at high altitude in Bolivia. Pediatric Research. 2003, 54: 20-25.

    PubMed  Google Scholar 

  531. Teramo KA, Hiilesmaa VK, Schwartz R, Clemons GK, Widness JA: Amniotic fluid and cord plasma erythropoietin levels in pregnancies complicated by preeclampsia, pregnancy-induced hypertension and chronic hypertension. J Perinat Med. 2004, 32: 240-7.

    CAS  PubMed  Google Scholar 

  532. Liang ST, Wong VC, So WW, Ma HK, Chan V, Todd D: Homozygous alpha-thalassaemia: clinical presentation, diagnosis and management. A review of 46 cases. Br J Obstet Gynaecol. 1985, 92: 680-4.

    CAS  PubMed  Google Scholar 

  533. Tungwiwat W, Fucharoen S, Fucharoen G, Ratanasiri T, Sanchaisuriya K: Development and application of a real-time quantitative PCR for prenatal detection of fetal alpha(0)-thalassemia from maternal plasma. Ann N Y Acad Sci. 2006, 1075: 103-7.

    CAS  PubMed  Google Scholar 

  534. Senden IP, de Groot CJ, Steegers EA, Bertina RM, Swinkels DW: Preeclampsia and the C282Y mutation in the hemochromatosis (HFE) gene. Clin Chem. 2004, 50: 973-4.

    CAS  PubMed  Google Scholar 

  535. Quek L, Thein SL: Molecular therapies in beta-thalassaemia. British Journal of Haematology. 2007, 136: 353-365.

    CAS  PubMed  Google Scholar 

  536. Knox KS, Baker JC: Genome-wide expression profiling of placentas in the p57Kip2 model of pre-eclampsia. Mol Hum Reprod. 2007, 13: 251-63.

    CAS  PubMed  Google Scholar 

  537. Suire S, Stewart F, Beauchamp J, Kennedy MW: Uterocalin, a lipocalin provisioning the preattachment equine conceptus: fatty acid and retinol binding properties, and structural characterization. Biochem J. 2001, 356: 369-76.

    CAS  PubMed  PubMed Central  Google Scholar 

  538. Ryon J, Bendickson L, Nilsen-Hamilton M: High expression in involuting reproductive tissues of uterocalin/24p3, a lipocalin and acute phase protein. Biochem J. 2002, 367: 271-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  539. Nilsen-Hamilton M, Liu Q, Ryon J, Bendickson L, Lepont P, Chang Q: Tissue involution and the acute phase response. Ann N Y Acad Sci. 2003, 995: 94-108.

    CAS  PubMed  Google Scholar 

  540. D'Anna R, Baviera G, Giordano D, Todarello G, Corrado F, Buemi M: Second trimester neutrophil gelatinase-associated lipocalin as a potential prediagnostic marker of preeclampsia. Acta Obstet Gynecol Scand. 2008, 1-4.

    Google Scholar 

  541. Coughlan MT, Permezel M, Georgiou HM, Rice GE: Repression of oxidant-induced nuclear factor-kappaB activity mediates placental cytokine responses in gestational diabetes. J Clin Endocrinol Metab. 2004, 89: 3585-94.

    CAS  PubMed  Google Scholar 

  542. Coughlan MT, Vervaart PP, Permezel M, Georgiou HM, Rice GE: Altered placental oxidative stress status in gestational diabetes mellitus. Placenta. 2004, 25: 78-84.

    CAS  PubMed  Google Scholar 

  543. King JC: Maternal obesity, metabolism and pregnancy outcomes. Annu Rev Nutrition. 2006, 26: 271-291.

    CAS  PubMed  Google Scholar 

  544. Mazar RM, Srinivas SK, Sammel MD, Andrela CM, Elovitz MA: Metabolic score as a novel approach to assessing preeclampsia risk. Am J Obstet Gynecol. 2007, 197: 411 e1-5.

    PubMed  Google Scholar 

  545. Rademacher TW, Gumaa K, Scioscia M: Preeclampsia, insulin signalling and immunological dysfunction: a fetal, maternal or placental disorder?. J Reprod Immunol. 2007, 76: 78-84.

    CAS  PubMed  Google Scholar 

  546. Hales CN, Ozanne SE: For Debate: Fetal and early postnatal growth restriction lead to diabetes, the metabolic syndrome and renal failure. Diabetologia. 2003, 46: 1013-1019.

    CAS  PubMed  Google Scholar 

  547. McMillen IC, Robinson JS: Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev. 2005, 85: 571-633.

    CAS  PubMed  Google Scholar 

  548. Williams PJ, Gumaa K, Scioscia M, Redman CW, Rademacher TW: Inositol phosphoglycan P-type in preeclampsia: a novel marker?. Hypertension. 2007, 49: 84-9.

    CAS  PubMed  Google Scholar 

  549. West IC: Radicals and oxidative stress in diabetes. Diabet Med. 2000, 17: 171-80.

    CAS  PubMed  Google Scholar 

  550. Lipinski B: Pathophysiology of oxidative stress in diabetes mellitus. J Diabetes Complications. 2001, 15: 203-10.

    CAS  PubMed  Google Scholar 

  551. Maddux BA, See W, Lawrence JC, Goldfine AL, Goldfine ID, Evans JL: Protection against oxidative stress-induced insulin resistance in rat L6 muscle cells by mircomolar concentrations of alpha-lipoic acid. Diabetes. 2001, 50: 404-10.

    CAS  PubMed  Google Scholar 

  552. Evans JL, Maddux BA, Goldfine ID: The molecular basis for oxidative stress-induced insulin resistance. Antioxid Redox Signal. 2005, 7: 1040-52.

    CAS  PubMed  Google Scholar 

  553. Houstis N, Rosen ED, Lander ES: Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature. 2006, 440: 944-8.

    CAS  PubMed  Google Scholar 

  554. Meigs JB, Larson MG, Fox CS, Keaney JF, Vasan RS, Benjamin EJ: Association of oxidative stress, insulin resistance, and diabetes risk phenotypes: the Framingham Offspring Study. Diabetes Care. 2007, 30: 2529-35.

    CAS  PubMed  Google Scholar 

  555. Shah S, Iqbal M, Karam J, Salifu M, McFarlane SI: Oxidative stress, glucose metabolism, and the prevention of type 2 diabetes: Pathophysiological insights. Antioxid Redox Signal. 2007, 9: 911-929.

    CAS  PubMed  Google Scholar 

  556. Choi SW, Benzie IF, Ma SW, Strain JJ, Hannigan BM: Acute hyperglycemia and oxidative stress: direct cause and effect?. Free Radic Biol Med. 2008, 44: 1217-31.

    CAS  PubMed  Google Scholar 

  557. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP, Giardino I, Brownlee M: Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature. 2000, 404: 787-90.

    CAS  PubMed  Google Scholar 

  558. Evans JL, Goldfine ID, Maddux BA, Grodsky GM: Oxidative stress and stress-activated signaling pathways: A unifying hypothesis of type 2 diabetes. Endocrine Reviews. 2002, 23: 599-622.

    CAS  PubMed  Google Scholar 

  559. Niedowicz DM, Daleke DL: The role of oxidative stress in diabetic complications. Cell Biochem Biophys. 2005, 43: 289-330.

    CAS  PubMed  Google Scholar 

  560. Kelley DE, He J, Menshikova EV, Ritov VB: Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. 2002, 51: 2944-50.

    CAS  PubMed  Google Scholar 

  561. Inoue I, Katayama S, Takahashi K, Negishi K, Miyazaki T, Sonoda M, Komoda T: Troglitazone has a scavenging effect on reactive oxygen species. Biochem Biophys Res Commun. 1997, 235: 113-6.

    CAS  PubMed  Google Scholar 

  562. Fukui T, Noma T, Mizushige K, Aki Y, Kimura S, Abe Y: Dietary troglitazone decreases oxidative stress in early stage type II diabetic rats. Life Sci. 2000, 66: 2043-9.

    CAS  PubMed  Google Scholar 

  563. Bao Y, Jia RH, Yuan J, Li J: Rosiglitazone ameliorates diabetic nephropathy by inhibiting reactive oxygen species and its downstream-signaling pathways. Pharmacology. 2007, 80: 57-64.

    CAS  PubMed  Google Scholar 

  564. Jung TW, Lee JY, Shim WS, Kang ES, Kim SK, Ahn CW, Lee HC, Cha BS: Rosiglitazone protects human neuroblastoma SH-SY5Y cells against MPP+ induced cytotoxicity via inhibition of mitochondrial dysfunction and ROS production. J Neurol Sci. 2007, 253: 53-60.

    CAS  PubMed  Google Scholar 

  565. Jung Y, Song S, Choi C: Peroxisome proliferator activated receptor gamma agonists suppress TNFalpha-induced ICAM-1 expression by endothelial cells in a manner potentially dependent on inhibition of reactive oxygen species. Immunol Lett. 2008, 117: 63-9.

    CAS  PubMed  Google Scholar 

  566. Allahtavakoli M, Shabanzadeh AP, Sadr SS, Parviz M, Djahanguiri B: Rosiglitazone, a peroxisome proliferator-activated receptor-gamma ligand, reduces infarction volume and neurological deficits in an embolic model of stroke. Clin Exp Pharmacol Physiol. 2006, 33: 1052-8.

    CAS  PubMed  Google Scholar 

  567. Collino M, Aragno M, Mastrocola R, Benetti E, Gallicchio M, Dianzani C, Danni O, Thiemermann C, Fantozzi R: Oxidative stress and inflammatory response evoked by transient cerebral ischemia/reperfusion: effects of the PPAR-alpha agonist WY14643. Free Radic Biol Med. 2006, 41: 579-89.

    CAS  PubMed  Google Scholar 

  568. Collino M, Aragno M, Mastrocola R, Gallicchio M, Rosa AC, Dianzani C, Danni O, Thiemermann C, Fantozzi R: Modulation of the oxidative stress and inflammatory response by PPAR-gamma agonists in the hippocampus of rats exposed to cerebral ischemia/reperfusion. Eur J Pharmacol. 2006, 530: 70-80.

    CAS  PubMed  Google Scholar 

  569. Allahtavakoli M, Shabanzadeh A, Roohbakhsh A, Pourshanazari A: Combination therapy of rosiglitazone, a peroxisome proliferator-activated receptor-gamma ligand, and NMDA receptor antagonist (MK-801) on experimental embolic stroke in rats. Basic Clin Pharmacol Toxicol. 2007, 101: 309-14.

    CAS  PubMed  Google Scholar 

  570. Wessel J, Moratorio G, Rao F, Mahata M, Zhang L, Greene W, Rana BK, Kennedy BP, Khandrika S, Huang P, Lillie EO, Shih PA, Smith DW, Wen G, Hamilton BA, Ziegler MG, Witztum JL, Schork NJ, Schmid-Schonbein GW, O'Connor DT: C-reactive protein, an 'intermediate phenotype' for inflammation: human twin studies reveal heritability, association with blood pressure and the metabolic syndrome, and the influence of common polymorphism at catecholaminergic/beta-adrenergic pathway loci. J Hypertens. 2007, 25: 329-43.

    CAS  PubMed  Google Scholar 

  571. Baynes JW, Thorpe SR: Role of oxidative stress in diabetic complications: a new perspective on an old paradigm. Diabetes. 1999, 48: 1-9.

    CAS  PubMed  Google Scholar 

  572. Hu FB, Meigs JB, Li TY, Rifai N, Manson JE: Inflammatory markers and risk of developing type 2 diabetes in women. Diabetes. 2004, 53: 693-700.

    CAS  PubMed  Google Scholar 

  573. Pickup JC: Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care. 2004, 27: 813-23.

    PubMed  Google Scholar 

  574. Kolb H, Mandrup-Poulsen T: An immune origin of type 2 diabetes?. Diabetologia. 2005, 48: 1038-50.

    CAS  PubMed  Google Scholar 

  575. Lee YH, Pratley RE: The evolving role of inflammation in obesity and the metabolic syndrome. Curr Diab Rep. 2005, 5: 70-5.

    CAS  PubMed  Google Scholar 

  576. Tataranni PA, Ortega E: A burning question: does an adipokine-induced activation of the immune system mediate the effect of overnutrition on type 2 diabetes?. Diabetes. 2005, 54: 917-27.

    CAS  PubMed  Google Scholar 

  577. Wellen KE, Hotamisligil GS: Inflammation, stress, and diabetes. J Clin Invest. 2005, 115: 1111-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  578. Alexandraki K, Piperi C, Kalofoutis C, Singh J, Alaveras A, Kalofoutis A: Inflammatory process in type 2 diabetes: The role of cytokines. Ann N Y Acad Sci. 2006, 1084: 89-117.

    CAS  PubMed  Google Scholar 

  579. Chen H: Cellular inflammatory responses: novel insights for obesity and insulin resistance. Pharmacol Res. 2006, 53: 469-77.

    CAS  PubMed  Google Scholar 

  580. Herder C, Peltonen M, Koenig W, Kraft I, Muller-Scholze S, Martin S, Lakka T, Ilanne-Parikka P, Eriksson JG, Hamalainen H, Keinanen-Kiukaanniemi S, Valle TT, Uusitupa M, Lindstrom J, Kolb H, Tuomilehto J: Systemic immune mediators and lifestyle changes in the prevention of type 2 diabetes: results from the Finnish Diabetes Prevention Study. Diabetes. 2006, 55: 2340-6.

    CAS  PubMed  Google Scholar 

  581. Hotamisligil GS: Inflammation and metabolic disorders. Nature. 2006, 444: 860-7.

    CAS  PubMed  Google Scholar 

  582. Kempf K, Rose B, Herder C, Kleophas U, Martin S, Kolb H: Inflammation in metabolic syndrome and type 2 diabetes: Impact of dietary glucose. Ann N Y Acad Sci. 2006, 1084: 30-48.

    CAS  PubMed  Google Scholar 

  583. Neels JG, Olefsky JM: Inflamed fat: what starts the fire?. J Clin Invest. 2006, 116: 33-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  584. Shoelson SE, Lee J, Goldfine AB: Inflammation and insulin resistance. J Clin Invest. 2006, 116: 1793-801.

    CAS  PubMed  PubMed Central  Google Scholar 

  585. Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, Charo I, Leibel RL, Ferrante AW: CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest. 2006, 116: 115-24.

    CAS  PubMed  Google Scholar 

  586. Zozulinska D, Wierusz-Wysocka B: Type 2 diabetes mellitus as inflammatory disease. Diabetes Research and Clinical Practice. 2006, 74: S12-S16.

    CAS  Google Scholar 

  587. Pradhan A: Obesity, metabolic syndrome, and type 2 diabetes: inflammatory basis of glucose metabolic disorders. Nutr Rev. 2007, 65: S152-6.

    PubMed  Google Scholar 

  588. Shoelson SE, Herrero L, Naaz A: Obesity, inflammation, and insulin resistance. Gastroenterology. 2007, 132: 2169-2180.

    CAS  PubMed  Google Scholar 

  589. Thorand B, Baumert J, Kolb H, Meisinger C, Chambless L, Koenig W, Herder C: Sex differences in the prediction of type 2 diabetes by inflammatory markers: results from the MONICA/KORA Augsburg case-cohort study, 1984–2002. Diabetes Care. 2007, 30: 854-60.

    CAS  PubMed  Google Scholar 

  590. Wei Y, Chen K, Whaley-Connell AT, Stump CS, Ibdah JA, Sowers JR: Skeletal muscle insulin resistance: role of inflammatory cytokines and reactive oxygen species. Am J Physiol Regul Integr Comp Physiol. 2008, 294: R673-80.

    CAS  PubMed  Google Scholar 

  591. Lao TT, Chan PL, Tam KF: Gestational diabetes mellitus in the last trimester – a feature of maternal iron excess?. Diabet Med. 2001, 18: 218-23.

    CAS  PubMed  Google Scholar 

  592. Lao TT, Tse KY, Chan LY, Tam KF, Ho LF: NBsAg carrier status and the association between gestational diabetes with increased serum ferritin concentration in Chinese women. Diabetes Care. 2003, 26: 3011-3016.

    PubMed  Google Scholar 

  593. Chen X, Scholl TO, Stein TP: Association of elevated serum ferritin levels and the risk of gestational diabetes mellitus in pregnant women: The Camden study. Diabetes Care. 2006, 29: 1077-82.

    CAS  PubMed  Google Scholar 

  594. Oberley LW: Free radicals and diabetes. Free Radic Biol Med. 1988, 5: 113-24.

    CAS  PubMed  Google Scholar 

  595. Wolff SP: Diabetes mellitus and free radicals. Free radicals, transition metals and oxidative stress in the aetiology of diabetes mellitus and complications. Br Med Bull. 1993, 49: 642-52.

    CAS  PubMed  Google Scholar 

  596. Tuomainen TP, Nyyssonen K, Salonen R, Tervahauta A, Korpela H, Lakka T, Kaplan GA, Salonen JT: Body iron stores are associated with serum insulin and blood glucose concentrations. Population study in 1,013 eastern Finnish men. Diabetes Care. 1997, 20: 426-8.

    CAS  PubMed  Google Scholar 

  597. Salonen JT, Tuomainen TP, Nyyssonen K, Lakka HM, Punnonen K: Relation between iron stores and non-insulin dependent diabetes in men: case-control study. BMJ. 1998, 317: 727.

    CAS  PubMed  PubMed Central  Google Scholar 

  598. Fernández-Real JM, López-Bermejo A, Ricart W: Cross-talk between iron metabolism and diabetes. Diabetes. 2002, 51: 2348-54.

    PubMed  Google Scholar 

  599. Fernández-Real JM, Ricart W: Insulin resistance and chronic cardiovascular inflammatory syndrome. Endocr Rev. 2003, 24: 278-301.

    PubMed  Google Scholar 

  600. Wilson JG, Lindquist JH, Grambow SC, Crook ED, Maher JF: Potential role of increased iron stores in diabetes. Am J Med Sci. 2003, 325: 332-9.

    PubMed  Google Scholar 

  601. Lee DH, Folsom AR, Jacobs DR: Dietary iron intake and Type 2 diabetes incidence in postmenopausal women: the Iowa Women's Health Study. Diabetologia. 2004, 47: 185-94.

    PubMed  Google Scholar 

  602. Kazi TG, Afridi HI, Kazi N, Jamali MK, Arain MB, Jalbani N, Kandhro GA: Copper, chromium, manganese, iron, nickel, and zinc levels in biological samples of diabetes mellitus patients. Biol Trace Elem Res. 2008, 122: 1-18.

    CAS  PubMed  Google Scholar 

  603. Rajpathak SN, Crandall JP, Wylie-Rosett J, Kabat GC, Rohan TE, Hu FB: The role of iron in type 2 diabetes in humans. Biochim Biophys Acta. 2008.

    Google Scholar 

  604. Adams PC, Kertesz AE, Valberg LS: Clinical presentation of hemochromatosis: a changing scene. Am J Med. 1991, 90: 445-9.

    CAS  PubMed  Google Scholar 

  605. Kaye TB, Guay AT, Simonson DC: Non-insulin-dependent diabetes mellitus and elevated serum ferritin level. J Diabetes Complic. 1993, 7: 246-249.

    CAS  Google Scholar 

  606. Fernández-Real JM, Ricart-Engel W, Arroyo E, Balanca R, Casamitjana-Abella R, Cabrero D, Fernandez-Castaner M, Soler J: Serum ferritin as a component of the insulin resistance syndrome. Diabetes Care. 1998, 21: 62-68.

    PubMed  Google Scholar 

  607. Ford ES, Cogswell ME: Diabetes and serum ferritin concentration among U.S. adults. Diabetes Care. 1999, 22: 1978-83.

    CAS  PubMed  Google Scholar 

  608. Jiang R, Manson JE, Meigs JB, Ma J, Rifai N, Hu FB: Body iron stores in relation to risk of type 2 diabetes in apparently healthy women. JAMA. 2004, 291: 711-7.

    CAS  PubMed  Google Scholar 

  609. Mojiminiyi OA, Marouf R, Abdella NA: Body iron stores in relation to the metabolic syndrome, glycemic control and complications in female patients with type 2 diabetes. Nutr Metab Cardiovasc Dis. 2008, 18: 559-66.

    CAS  PubMed  Google Scholar 

  610. Jehn ML, Guallar E, Clark JM, Couper D, Duncan BB, Ballantyne CM, Hoogeveen RC, Harris ZL, Pankow JS: A prospective study of plasma ferritin level and incident diabetes: the Atherosclerosis Risk in Communities (ARIC) Study. Am J Epidemiol. 2007, 165: 1047-54.

    PubMed  Google Scholar 

  611. Fernández-Real JM, Moreno JM, Chico B, Lopez-Bermejo A, Ricart W: Circulating visfatin is associated with parameters of iron metabolism in subjects with altered glucose tolerance. Diabetes Care. 2007, 30: 616-21.

    PubMed  Google Scholar 

  612. Wang Y, Lam KSL, Kraegen EW, Sweeney G, Zhang JL, Tso AWK, Chow WS, Wat NMS, Xu JY, Hoo RLC, Xu AM: Lipocalin-2 is an inflammatory marker closely associated with obesity, insulin resistance, and hyperglycemia in humans. Clinical Chemistry. 2007, 53: 34-41.

    CAS  PubMed  Google Scholar 

  613. van Dam RM, Hu FB: Lipocalins and insulin resistance: Etiological role of retinol-binding protein 4 and lipocalin-2?. Clinical Chemistry. 2007, 53: 5-7.

    CAS  PubMed  Google Scholar 

  614. Hua NW, Stoohs RA, Facchini FS: Low iron status and enhanced insulin sensitivity in lacto-ovo vegetarians. Br J Nutr. 2001, 86: 515-9.

    CAS  PubMed  Google Scholar 

  615. Liang Q, Carlson EC, Donthi RV, Kralik PM, Shen X, Epstein PN: Overexpression of metallothionein reduces diabetic cardiomyopathy. Diabetes. 2002, 51: 174-81.

    CAS  PubMed  Google Scholar 

  616. Ye G, Metreveli NS, Ren J, Epstein PN: Metallothionein prevents diabetes-induced deficits in cardiomyocytes by inhibiting reactive oxygen species production. Diabetes. 2003, 52: 777-83.

    CAS  PubMed  Google Scholar 

  617. Cai L, Wang J, Li Y, Sun X, Wang L, Zhou Z, Kang YJ: Inhibition of superoxide generation and associated nitrosative damage is involved in metallothionein prevention of diabetic cardiomyopathy. Diabetes. 2005, 54: 1829-37.

    CAS  PubMed  Google Scholar 

  618. Islam MS, Loots du T: Diabetes, metallothionein, and zinc interactions: a review. Biofactors. 2007, 29: 203-12.

    CAS  PubMed  Google Scholar 

  619. Li X, Cai L, Feng W: Diabetes and metallothionein. Mini Rev Med Chem. 2007, 7: 761-8.

    CAS  PubMed  Google Scholar 

  620. Zheng Y, Li XK, Wang Y, Cai L: The role of zinc, copper and iron in the pathogenesis of diabetes and diabetic complications: therapeutic effects by chelators. Hemoglobin. 2008, 32: 135-45.

    CAS  PubMed  Google Scholar 

  621. Lee DH, Liu DY, Jacobs DR, Shin HR, Song K, Lee IK, Kim B, Hider RC: Common presence of non-transferrin-bound iron among patients with type 2 diabetes. Diabetes Care. 2006, 29: 1090-5.

    CAS  PubMed  Google Scholar 

  622. Chittum HS, Champney WS: Erythromycin inhibits the assembly of the large ribosomal subunit in growing Escherichia coli cells. Curr Microbiol. 1995, 30: 273-9.

    CAS  PubMed  Google Scholar 

  623. Facchini FS, Hua NW, Stoohs RA: Effect of iron depletion in carbohydrate-intolerant patients with clinical evidence of nonalcoholic fatty liver disease. Gastroenterology. 2002, 122: 931-9.

    CAS  PubMed  Google Scholar 

  624. Machado M, Cortez-Pinto H: Nash, insulin resistance and iron. Liver International. 2006, 26: 1159-1162.

    CAS  PubMed  Google Scholar 

  625. Allerson CR, Cazzola M, Rouault TA: Clinical severity and thermodynamic effects of iron-responsive element mutations in hereditary hyperferritinemia-cataract syndrome. Journal of Biological Chemistry. 1999, 274: 26439-26447.

    CAS  PubMed  Google Scholar 

  626. Beaumont C, Leneuve P, Devaux I, Scoazec JY, Berthier M, Loiseau MN, Grandchamp B, Bonneau D: Mutation in the Iron-Responsive Element of the L-Ferritin Messenger-Rna in a Family with Dominant Hyperferritinemia and Cataract. Nature Genetics. 1995, 11: 444-446.

    CAS  PubMed  Google Scholar 

  627. Dandona P, Aljada A, Chaudhuri A, Mohanty P, Garg R: Metabolic syndrome – A comprehensive perspective based on interactions between obesity, diabetes, and inflammation. Circulation. 2005, 111: 1448-1454.

    PubMed  Google Scholar 

  628. Eckel RH, Grundy SM, Zimmet PZ: The metabolic syndrome. Lancet. 2005, 365: 1415-28.

    CAS  PubMed  Google Scholar 

  629. Roberts CK, Barnard RJ: Effects of exercise and diet on chronic disease. J Appl Physiol. 2005, 98: 3-30.

    PubMed  Google Scholar 

  630. Nicolson GL: Metabolic syndrome and mitochondrial function: Molecular replacement and antioxidant supplements to prevent membrane peroxidation and restore mitochondrial function. Journal of Cellular Biochemistry. 2007, 100: 1352-1369.

    CAS  PubMed  Google Scholar 

  631. Yudkin JS: Insulin resistance and the metabolic syndrome–or the pitfalls of epidemiology. Diabetologia. 2007, 50: 1576-86.

    CAS  PubMed  Google Scholar 

  632. Jehn M, Clark JM, Guallar E: Serum ferritin and risk of the metabolic syndrome in U.S. adults. Diabetes Care. 2004, 27: 2422-8.

    PubMed  Google Scholar 

  633. Wrede CE, Buettner R, Bollheimer LC, Scholmerich J, Palitzsch KD, Hellerbrand C: Association between serum ferritin and the insulin resistance syndrome in a representative population. Eur J Endocrinol. 2006, 154: 333-340.

    CAS  PubMed  Google Scholar 

  634. Bozzini C, Girelli D, Olivieri O, Martinelli N, Bassi A, De Matteis G, Tenuti I, Lotto V, Friso S, Pizzolo F, Corrocher R: Prevalence of body iron excess in the metabolic syndrome. Diabetes Care. 2005, 28: 2061-3.

    PubMed  Google Scholar 

  635. Choi KM, Lee KW, Kim HY, Seo JA, Kim SG, Kim NH, Choi DS, Baik SH: Association among serum ferritin, alanine aminotransferase levels, and metabolic syndrome in Korean postmenopausal women. Metabolism. 2005, 54: 1510-4.

    CAS  PubMed  Google Scholar 

  636. Kurella M, Lo JC, Chertow GM: Metabolic syndrome and the risk for chronic kidney disease among nondiabetic adults. J Am Soc Nephrol. 2005, 16: 2134-2140.

    PubMed  Google Scholar 

  637. den Boer M, Voshol PJ, Kuipers F, Havekes LM, Romijn JA: Hepatic steatosis: a mediator of the metabolic syndrome. Lessons from animal models. Arterioscler Thromb Vasc Biol. 2004, 24: 644-9.

    CAS  PubMed  Google Scholar 

  638. Mittra S, Bansal VS, Bhatnagar PK: From a glucocentric to a lipocentric approach towards metabolic syndrome. Drug Discov Today. 2008, 13: 211-8.

    CAS  PubMed  Google Scholar 

  639. Roberts CK, Barnard RJ, Sindhu RK, Jurczak M, Ehdaie A, Vaziri ND: Oxidative stress and dysregulation of NAD(P)H oxidase and antioxidant enzymes in diet-induced metabolic syndrome. Metabolism. 2006, 55: 928-34.

    CAS  PubMed  Google Scholar 

  640. Booth FW, Gordon SE, Carlson CJ, Hamilton MT: Waging war on modern chronic diseases: primary prevention through exercise biology. J Appl Physiol. 2000, 88: 774-87.

    CAS  PubMed  Google Scholar 

  641. Wild S, Roglic G, Green A, Sicree R, King H: Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004, 27: 1047-53.

    PubMed  Google Scholar 

  642. Kahn SE, Hull RL, Utzschneider KM: Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006, 444: 840-6.

    CAS  PubMed  Google Scholar 

  643. Walker CG, Zariwala MG, Holness MJ, Sugden MC: Diet, obesity and diabetes: a current update. Clin Sci (Lond). 2007, 112: 93-111.

    CAS  Google Scholar 

  644. Lecube A, Hernandez C, Pelegri D, Simo R: Factors accounting for high ferritin levels in obesity. Int J Obes (Lond). 2008

    Google Scholar 

  645. Fricker J, Lemoel G, Apfelbaum M: Obesity and iron status in menstruating women. Am J Clin Nutrition. 1990, 52: 863-866.

    CAS  Google Scholar 

  646. Farahani P, Chiu S, Bowlus CL, Boffelli D, Lee E, Fisler JS, Krauss RM, Warden CH: Obesity in BSB mice is correlated with expression of genes for iron homeostasis and leptin. Obesity Research. 2004, 12: 191-204.

    CAS  PubMed  Google Scholar 

  647. Das UN: Is obesity an inflammatory condition?. Nutrition. 2001, 17: 953-66.

    CAS  PubMed  Google Scholar 

  648. Hall JE, Crook ED, Jones DW, Wofford MR, Dubbert PM: Mechanisms of obesity-associated cardiovascular and renal disease. Am J Med Sci. 2002, 324: 127-37.

    PubMed  Google Scholar 

  649. Trayhurn P, Wood IS: Adipokines: inflammation and the pleiotropic role of white adipose tissue. Br J Nutr. 2004, 92: 347-55.

    CAS  PubMed  Google Scholar 

  650. Berg AH, Scherer PE: Adipose tissue, inflammation, and cardiovascular disease. Circ Res. 2005, 96: 939-49.

    CAS  PubMed  Google Scholar 

  651. Hukshorn CJ, Lindeman JH, Toet KH, Saris WH, Eilers PH, Westerterp-Plantenga MS, Kooistra T: Leptin and the proinflammatory state associated with human obesity. J Clin Endocrinol Metab. 2004, 89: 1773-8.

    CAS  PubMed  Google Scholar 

  652. Toni R, Malaguti A, Castorina S, Roti E, Lechan RM: New paradigms in neuroendocrinology: relationships between obesity, systemic inflammation and the neuroendocrine system. J Endocrinol Invest. 2004, 27: 182-6.

    CAS  PubMed  Google Scholar 

  653. Koh KK, Han SH, Quon MJ: Inflammatory markers and the metabolic syndrome: insights from therapeutic interventions. J Am Coll Cardiol. 2005, 46: 1978-85.

    CAS  PubMed  Google Scholar 

  654. Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H, Capeau J, Feve B: Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw. 2006, 17: 4-12.

    CAS  PubMed  Google Scholar 

  655. Kapiotis S, Holzer G, Schaller G, Haumer M, Widhalm H, Weghuber D, Jilma B, Roggla G, Wolzt M, Widhalm K, Wagner OF: A proinflammatory state is detectable in obese children and is accompanied by functional and morphological vascular changes. Arterioscler Thromb Vasc Biol. 2006, 26: 2541-6.

    CAS  PubMed  Google Scholar 

  656. Tilg H, Moschen AR: Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 2006, 6: 772-83.

    CAS  PubMed  Google Scholar 

  657. Vincent HK, Taylor AG: Biomarkers and potential mechanisms of obesity-induced oxidant stress in humans. Int J Obes (Lond). 2006, 30: 400-18.

    CAS  Google Scholar 

  658. Wang B, Trayhurn P: Acute and prolonged effects of TNF-alpha on the expression and secretion of inflammation-related adipokines by human adipocytes differentiated in culture. Pflugers Arch. 2006, 452: 418-27.

    CAS  PubMed  Google Scholar 

  659. Chandalia M, Abate N: Metabolic complications of obesity: inflated or inflamed?. J Diabetes Complications. 2007, 21: 128-36.

    PubMed  Google Scholar 

  660. Lumeng CN, Bodzin JL, Saltiel AR: Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007, 117: 175-84.

    CAS  PubMed  PubMed Central  Google Scholar 

  661. Chen Y, Zhu J, Lum PY, Yang X, Pinto S, Macneil DJ, Zhang C, Lamb J, Edwards S, Sieberts SK, Leonardson A, Castellini LW, Wang S, Champy MF, Zhang B, Emilsson V, Doss S, Ghazalpour A, Horvath S, Drake TA, Lusis AJ, Schadt EE: Variations in DNA elucidate molecular networks that cause disease. Nature. 2008, 452: 429-435.

    CAS  PubMed  PubMed Central  Google Scholar 

  662. Chung J, Nguyen AK, Henstridge DC, Holmes AG, Chan MH, Mesa JL, Lancaster GI, Southgate RJ, Bruce CR, Duffy SJ, Horvath I, Mestril R, Watt MJ, Hooper PL, Kingwell BA, Vigh L, Hevener A, Febbraio MA: HSP72 protects against obesity-induced insulin resistance. Proc Natl Acad Sci USA. 2008, 105: 1739-44.

    CAS  PubMed  PubMed Central  Google Scholar 

  663. Emilsson V, Thorleifsson G, Zhang B, Leonardson AS, Zink F, Zhu J, Carlson S, Helgason A, Walters GB, Gunnarsdottir S, Mouy M, Steinthorsdottir V, Eiriksdottir GH, Bjornsdottir G, Reynisdottir I, Gudbjartsson D, Helgadottir A, Jonasdottir A, Jonasdottir A, Styrkarsdottir U, Gretarsdottir S, Magnusson KP, Stefansson H, Fossdal R, Kristjansson K, Gislason HG, Stefansson T, Leifsson BG, Thorsteinsdottir U, Lamb JR, Gulcher JR, Reitman ML, Kong A, Schadt EE, Stefansson K: Genetics of gene expression and its effect on disease. Nature. 2008

    Google Scholar 

  664. Wärnberg J, Marcos A: Low-grade inflammation and the metabolic syndrome in children and adolescents. Curr Opin Lipidol. 2008, 19: 11-5.

    PubMed  Google Scholar 

  665. Rajala MW, Scherer PE: Minireview: The adipocyte–at the crossroads of energy homeostasis, inflammation, and atherosclerosis. Endocrinology. 2003, 144: 3765-73.

    CAS  PubMed  Google Scholar 

  666. Vendrell J, Broch M, Vilarrasa N, Molina A, Gomez JM, Gutierrez C, Simon I, Soler J, Richart C: Resistin, adiponectin, ghrelin, leptin, and proinflammatory cytokines: relationships in obesity. Obes Res. 2004, 12: 962-71.

    CAS  PubMed  Google Scholar 

  667. Arner P: Insulin resistance in type 2 diabetes – role of the adipokines. Curr Mol Med. 2005, 5: 333-9.

    CAS  PubMed  Google Scholar 

  668. Hutley L, Prins JB: Fat as an endocrine organ: relationship to the metabolic syndrome. Am J Med Sci. 2005, 330: 280-9.

    PubMed  Google Scholar 

  669. Kougias P, Chai H, Lin PH, Yao Q, Lumsden AB, Chen C: Effects of adipocyte-derived cytokines on endothelial functions: implication of vascular disease. J Surg Res. 2005, 126: 121-9.

    CAS  PubMed  Google Scholar 

  670. Lau DC, Dhillon B, Yan H, Szmitko PE, Verma S: Adipokines: molecular links between obesity and atheroslcerosis. Am J Physiol Heart Circ Physiol. 2005, 288: H2031-41.

    CAS  PubMed  Google Scholar 

  671. Rosen ED, Spiegelman BM: Adipocytes as regulators of energy balance and glucose homeostasis. Nature. 2006, 444: 847-53.

    CAS  PubMed  PubMed Central  Google Scholar 

  672. Kim F, Pham M, Luttrell I, Bannerman DD, Tupper J, Thaler J, Hawn TR, Raines EW, Schwartz MW: Toll-like receptor-4 mediates vascular inflammation and insulin resistance in diet-induced obesity. Circ Res. 2007, 100: 1589-96.

    CAS  PubMed  Google Scholar 

  673. Shah PK: Innate immune pathway links obesity to insulin resistance. Circ Res. 2007, 100: 1531-3.

    CAS  PubMed  Google Scholar 

  674. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I: Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004, 114: 1752-61.

    CAS  PubMed  PubMed Central  Google Scholar 

  675. Keaney JF, Larson MG, Vasan RS, Wilson PW, Lipinska I, Corey D, Massaro JM, Sutherland P, Vita JA, Benjamin EJ: Obesity and systemic oxidative stress: clinical correlates of oxidative stress in the Framingham Study. Arterioscler Thromb Vasc Biol. 2003, 23: 434-9.

    CAS  PubMed  Google Scholar 

  676. Higdon JV, Frei B: Obesity and oxidative stress: a direct link to CVD?. Arterioscler Thromb Vasc Biol. 2003, 23: 365-7.

    CAS  PubMed  Google Scholar 

  677. Whitworth JA: 2003 World Health Organization (WHO)/International Society of Hypertension (ISH) statement on management of hypertension. J Hypertens. 2003, 21: 1983-92.

    PubMed  Google Scholar 

  678. Tuder RM, Voelkel NF: Pulmonary hypertension and inflammation. J Lab Clin Med. 1998, 132: 16-24.

    CAS  PubMed  Google Scholar 

  679. Intengan HD, Schiffrin EL: Vascular remodeling in hypertension: roles of apoptosis, inflammation, and fibrosis. Hypertension. 2001, 38: 581-7.

    CAS  PubMed  Google Scholar 

  680. Grundy SM: Inflammation, hypertension, and the metabolic syndrome. JAMA. 2003, 290: 3000-2.

    CAS  PubMed  Google Scholar 

  681. Virdis A, Schiffrin EL: Vascular inflammation: a role in vascular disease in hypertension?. Curr Opin Nephrol Hypertens. 2003, 12: 181-7.

    CAS  PubMed  Google Scholar 

  682. Boos CJ, Lip GY: Elevated high-sensitive C-reactive protein, large arterial stiffness and atherosclerosis: a relationship between inflammation and hypertension?. J Hum Hypertens. 2005, 19: 511-3.

    CAS  PubMed  Google Scholar 

  683. Varughese GI, Lip GY: Hypertension in patients with type-II diabetes: relation to urinary albumin excretion, endothelial function and inflammation. J Hum Hypertens. 2005, 19: 421-4.

    CAS  PubMed  Google Scholar 

  684. Li JJ: Inflammation in hypertension: primary evidence. Chin Med J (Engl). 2006, 119: 1215-21.

    CAS  Google Scholar 

  685. Savoia C, Schiffrin EL: Inflammation in hypertension. Curr Opin Nephrol Hypertens. 2006, 15: 152-8.

    CAS  PubMed  Google Scholar 

  686. Savoia C, Schiffrin EL: Vascular inflammation in hypertension and diabetes: molecular mechanisms and therapeutic interventions. Clin Sci (Lond). 2007, 112: 375-84.

    CAS  Google Scholar 

  687. Kannel WB, Brand N, Skinner JJ, Dawber TR, McNamara PM: The relation of adiposity to blood pressure and development of hypertension. The Framingham study. Ann Intern Med. 1967, 67: 48-59.

    CAS  PubMed  Google Scholar 

  688. Mark AL, Correia M, Morgan DA, Shaffer RA, Haynes WG: State-of-the-art-lecture: Obesity-induced hypertension: new concepts from the emerging biology of obesity. Hypertension. 1999, 33: 537-41.

    CAS  PubMed  Google Scholar 

  689. Aizawa-Abe M, Ogawa Y, Masuzaki H, Ebihara K, Satoh N, Iwai H, Matsuoka N, Hayashi T, Hosoda K, Inoue G, Yoshimasa Y, Nakao K: Pathophysiological role of leptin in obesity-related hypertension. J Clin Invest. 2000, 105: 1243-52.

    CAS  PubMed  PubMed Central  Google Scholar 

  690. Hall JE: The kidney, hypertension, and obesity. Hypertension. 2003, 41: 625-33.

    PubMed  Google Scholar 

  691. Yanai H, Tomono Y, Ito K, Furutani N, Yoshida H, Tada N: The underlying mechanisms for development of hypertension in the metabolic syndrome. Nutr J. 2008, 7: 10.

    PubMed  PubMed Central  Google Scholar 

  692. Vaziri ND, Rodriguez-Iturbe B: Mechanisms of disease: oxidative stress and inflammation in the pathogenesis of hypertension. Nat Clin Pract Nephrol. 2006, 2: 582-93.

    CAS  PubMed  Google Scholar 

  693. Yakobson MG, Antonov AR, Golovatyuk AV, Efremov AV, Markel AL, Yakobson GS: Iron content and parameters of blood antioxidant activity in rats with hereditary arterial hypertension during experimental myocardial infarction. Bull Exp Biol Med. 2001, 132: 1041-4.

    CAS  PubMed  Google Scholar 

  694. Peterson JR, Sharma RV, Davisson RL: Reactive oxygen species in the neuropathogenesis of hypertension. Curr Hypertens Rep. 2006, 8: 232-41.

    CAS  PubMed  Google Scholar 

  695. Sullivan JL: Iron and the sex difference in heart-disease risk. Lancet. 1981, 1: 1293-1294.

    CAS  PubMed  Google Scholar 

  696. Sullivan JL: Misconceptions in the debate on the iron hypothesis. Journal of Nutritional Biochemistry. 2001, 12: 33-37.

    CAS  PubMed  Google Scholar 

  697. Sullivan JL: Stored iron and vascular reactivity. Arteriosclerosis Thrombosis and Vascular Biology. 2005, 25: 1532-1535.

    CAS  Google Scholar 

  698. Sullivan JL: The big idea: the coxib crisis iron, aspirin and heart disease risk revisited. J R Soc Med. 2007, 100: 346-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  699. Meyers DG: The iron hypothesis: does iron play a role in atherosclerosis?. Transfusion. 2000, 40: 1023-1029.

    CAS  PubMed  Google Scholar 

  700. Yuan XM, Li W: The iron hypothesis of atherosclerosis and its clinical impact. Ann Med. 2003, 35: 578-91.

    CAS  PubMed  Google Scholar 

  701. Zheng H, Dimayuga C, Hudaihed A, Katz SD: Effect of dexrazoxane on homocysteine-induced endothelial dysfunction in normal subjects. Arterioscler Thromb Vasc Biol. 2002, 22: E15-8.

    PubMed  Google Scholar 

  702. Zheng H, Huang X, Zhang Q, Katz SD: Iron sucrose augments homocysteine-induced endothelial dysfunction in normal subjects. Kidney Int. 2006, 69: 679-84.

    CAS  PubMed  PubMed Central  Google Scholar 

  703. Sullivan JL: Is homocysteine an iron-dependent cardiovascular risk factor?. Kidney Int. 2006, 69: 642-4.

    CAS  PubMed  Google Scholar 

  704. Clarke R: Homocysteine-lowering trials for prevention of heart disease and stroke. Semin Vasc Med. 2005, 5: 215-22.

    PubMed  Google Scholar 

  705. Tuomainen TP, Punnonen K, Nyyssonen K, Salonen JT: Association between body iron stores and the risk of acute myocardial infarction in men. Circulation. 1998, 97: 1461-6.

    CAS  PubMed  Google Scholar 

  706. Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL, McCullough PA, Kasiske BL, Kelepouris E, Klag MJ, Parfrey P, Pfeffer M, Raij L, Spinosa DJ, Wilson PW: Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation. 2003, 108: 2154-69.

    PubMed  Google Scholar 

  707. Oberg BP, McMenamin E, Lucas FL, McMonagle E, Morrow J, Ikizler TA, Himmelfarb J: Increased prevalence of oxidant stress and inflammation in patients with moderate to severe chronic kidney disease. Kidney Int. 2004, 65: 1009-16.

    PubMed  Google Scholar 

  708. Vaziri ND: Roles of oxidative stress and antioxidant therapy in chronic kidney disease and hypertension. Curr Opin Nephrol Hypertens. 2004, 13: 93-9.

    CAS  PubMed  Google Scholar 

  709. Abramson JL, Jurkovitz CT, Vaccarino V, Weintraub WS, McClellan W: Chronic kidney disease, anemia, and incident stroke in a middle-aged, community-based population: the ARIC Study. Kidney Int. 2003, 64: 610-5.

    PubMed  Google Scholar 

  710. Smith JK, Carden DL, Grisham MB, Granger DN, Korthuis RJ: Role of iron in postischemic microvascular injury. Am J Physiol. 1989, 256: H1472-7.

    CAS  PubMed  Google Scholar 

  711. Kukreja RC, Hess ML: The oxygen free radical system: from equations through membrane protein interactions to cardiovascular injury and protection. Cardiovasc Res. 1992, 26: 641-655.

    CAS  PubMed  Google Scholar 

  712. Lesnefsky E, Ye J: Exogenous intracellular, but not extracellular, iron augments myocardial reperfusion injury. Amer J Physiol. 1994, 266: H384-H392.

    CAS  PubMed  Google Scholar 

  713. Berenshtein E, Mayer B, Goldberg C, Kitrossky N, Chevion M: Patterns of mobilization of copper and iron following myocardial ischemia: Possible predictive criteria for tissue injury. J Mol Cell Cardiol. 1997, 29: 3025-3034.

    CAS  PubMed  Google Scholar 

  714. Horwitz LD, Sherman NA, Kong YN, Pike AW, Gobin J, Fennessey PV, Horwitz MA: Lipophilic siderophores Mycobacterium tuberculosis prevent cardiac reperfusion injury. Proc Natl Acad Sci. 1998, 95: 5263-5268.

    CAS  PubMed  PubMed Central  Google Scholar 

  715. Horwitz LD, Rosenthal EA: Iron-mediated cardiovascular injury. Vasc Med. 1999, 4: 93-9.

    CAS  PubMed  Google Scholar 

  716. Pucheu S, Coudray C, Tresallet N, Favier A, Deleiris J: Effect of iron overload in the isolated ischemic and reperfused rat heart. Cardiovascular Drugs and Therapy. 1993, 7: 701-711.

    CAS  PubMed  Google Scholar 

  717. Amersi F, Dulkanchainun T, Nelson SK, Farmer DG, Kato H, Zaky J, Melinek J, Shaw GD, Kupiec-Weglinski JW, Horwitz LD, Horwitz MA, Busuttil RW: Novel iron chelator in combination with a P-selectin antagonist prevents ischemia/reperfusion injury in a rat liver model. Transplantation. 2001, 71: 112-118.

    CAS  PubMed  Google Scholar 

  718. Mamtani M, Kulkarni H: Influence of iron chelators on myocardial iron and cardiac function in transfusion-dependent thalassaemia: a systematic review and meta-analysis. Br J Haematol. 2008, 141: 882-90.

    CAS  PubMed  Google Scholar 

  719. Ravati A, Ahlemeyer B, Becker A, Klumpp S, Krieglstein J: Preconditioning-induced neuroprotection is mediated by reactive oxygen species and activation of the transcription factor nuclear factor-kappa B. Journal of Neurochemistry. 2001, 78: 909-919.

    CAS  PubMed  Google Scholar 

  720. Berenshtein E, Vaisman B, Goldberg-Langerman C, Kitrossky N, Konijn AM, Chevion M: Roles of ferritin and iron in ischemic preconditioning of the heart. Mol Cell Biochem. 2002, 234: 283-292.

    PubMed  Google Scholar 

  721. Calvillo L, Latini R, Kajstura J, Leri A, Anversa P, Ghezzi P, Salio M, Cerami A, Brines M: Recombinant human erythropoietin protects the myocardium from ischemia-reperfusion injury and promotes beneficial remodeling. ProcNatl Acad Sci. 2003, 100: 4802-4806.

    CAS  Google Scholar 

  722. Parsa CJ, Matsumoto A, Kim J, Riel RU, Pascal LS, Walton GB, Thompson RB, Petrofski JA, Annex BH, Stamler JS, Koch WJ: A novel protective effect of erythropoietin in the infarcted heart. J Clin Invest. 2003, 112: 999-1007.

    CAS  PubMed  PubMed Central  Google Scholar 

  723. Engberding N, Spiekermann S, Schaefer A, Heineke A, Wiencke A, Muller M, Fuchs M, Hilfiker-Kleiner D, Hornig B, Drexler H, Landmesser U: Allopurinol attenuates left ventricular remodeling and dysfunction after experimental myocardial infarction: a new action for an old drug?. Circulation. 2004, 110: 2175-9.

    CAS  PubMed  Google Scholar 

  724. Reyes AJ: Cardiovascular drugs and serum uric acid. Cardiovasc Drugs Ther. 2003, 17: 397-414.

    CAS  PubMed  Google Scholar 

  725. Dunn WB, Broadhurst DI, Sasalu D, Buch M, McDowell G, Spasic I, Ellis DI, Brooks N, Kell DB, Neyses L: Serum metabolomics reveals many novel metabolic markers of heart failure, including pseudouridine and 2-oxoglutarate. Metabolomics. 2007, 3: 413-426.

    CAS  Google Scholar 

  726. Hokamaki J, Kawano H, Yoshimura M, Soejima H, Miyamoto S, Kajiwara I, Kojima S, Sakamoto T, Sugiyama S, Hirai N, Shimomura H, Nagayoshi Y, Tsujita K, Shioji I, Sasaki S, Ogawa H: Urinary biopyrrins levels are elevated in relation to severity of heart failure. J Am Coll Cardiol. 2004, 43: 1880-5.

    CAS  PubMed  Google Scholar 

  727. Mozaffarian D, Nye R, Levy WC: Anemia predicts mortality in severe heart failure: the prospective randomized amlodipine survival evaluation (PRAISE). J Am Coll Cardiol. 2003, 41: 1933-9.

    PubMed  Google Scholar 

  728. Felker GM, Adams KF, Gattis WA, O'Connor CM: Anemia as a risk factor and therapeutic target in heart failure. J Am Coll Cardiol. 2004, 44: 959-66.

    PubMed  Google Scholar 

  729. Bolger AP, Bartlett FR, Penston HS, O'Leary J, Pollock N, Kaprielian R, Chapman CM: Intravenous iron alone for the treatment of anemia in patients with chronic heart failure. J Am Coll Cardiol. 2006, 48: 1225-7.

    CAS  PubMed  Google Scholar 

  730. Weiss G, Goodnough LT: Anemia of chronic disease. N Engl J Med. 2005, 352: 1011-23.

    CAS  PubMed  Google Scholar 

  731. Kunsch C, Medford RM: Oxidative stress as a regulator of gene expression in the vasculature. Circ Res. 1999, 85: 753-66.

    CAS  PubMed  Google Scholar 

  732. Ross R: Atherosclerosis: an inflammatory disease. N Engl J Med. 1999, 340: 115-26.

    CAS  PubMed  Google Scholar 

  733. Blake GJ, Ridker PM: Novel clinical markers of vascular wall inflammation. Circ Res. 2001, 89: 763-71.

    CAS  PubMed  Google Scholar 

  734. Hansson GK: Immune mechanisms in atherosclerosis. Arterioscler Thromb Vasc Biol. 2001, 21: 1876-90.

    CAS  PubMed  Google Scholar 

  735. Forrester JS: Prevention of plaque rupture: a new paradigm of therapy. Ann Intern Med. 2002, 137: 823-33.

    PubMed  Google Scholar 

  736. Himmelfarb J, Stenvinkel P, Ikizler TA, Hakim RM: The elephant in uremia: oxidant stress as a unifying concept of cardiovascular disease in uremia. Kidney Int. 2002, 62: 1524-38.

    CAS  PubMed  Google Scholar 

  737. Libby P: Inflammation in atherosclerosis. Nature. 2002, 420: 868-74.

    CAS  PubMed  Google Scholar 

  738. Libby P, Ridker PM, Maseri A: Inflammation and atherosclerosis. Circulation. 2002, 105: 1135-43.

    CAS  PubMed  Google Scholar 

  739. Young JL, Libby P, Schonbeck U: Cytokines in the pathogenesis of atherosclerosis. Thromb Haemost. 2002, 88: 554-67.

    CAS  PubMed  Google Scholar 

  740. Binder CJ, Chang MK, Shaw PX, Miller YI, Hartvigsen K, Dewan A, Witztum JL: Innate and acquired immunity in atherogenesis. Nat Med. 2002, 8: 1218-26.

    CAS  PubMed  Google Scholar 

  741. Altman R: Risk factors in coronary atherosclerosis athero-inflammation: the meeting point. Thromb J. 2003, 1: 4.

    PubMed  PubMed Central  Google Scholar 

  742. Dwyer JH, Allayee H, Dwyer KM, Fan J, Wu H, Mar R, Lusis AJ, Mehrabian M: Arachidonate 5-lipoxygenase promoter genotype, dietary arachidonic acid, and atherosclerosis. N Engl J Med. 2004, 350: 29-37.

    CAS  PubMed  Google Scholar 

  743. Forrester JS: Common ancestors: chronic progressive diseases have the same pathogenesis. Clin Cardiol. 2004, 27: 186-90.

    PubMed  Google Scholar 

  744. Paoletti R, Gotto AM, Hajjar DP: Inflammation in atherosclerosis and implications for therapy. Circulation. 2004, 109: III20-6.

    PubMed  Google Scholar 

  745. Ridker PM, Brown NJ, Vaughan DE, Harrison DG, Mehta JL: Established and emerging plasma biomarkers in the prediction of first atherothrombotic events. Circulation. 2004, 109 (Suppl 1): 6-19.

    Google Scholar 

  746. van Oostrom AJ, van Wijk J, Cabezas MC: Lipaemia, inflammation and atherosclerosis: novel opportunities in the understanding and treatment of atherosclerosis. Drugs. 2004, 64 (Suppl 2): 19-41.

    CAS  PubMed  Google Scholar 

  747. Willerson JT, Ridker PM: Inflammation as a cardiovascular risk factor. Circulation. 2004, 109: II2-10.

    PubMed  Google Scholar 

  748. Hansson GK: Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005, 352: 1685-95.

    CAS  PubMed  Google Scholar 

  749. Madamanchi NR, Hakim ZS, Runge MS: Oxidative stress in atherogenesis and arterial thrombosis: the disconnect between cellular studies and clinical outcomes. J Thromb Haemost. 2005, 3: 254-67.

    CAS  PubMed  Google Scholar 

  750. Madamanchi NR, Vendrov A, Runge MS: Oxidative stress and vascular disease. Arterioscler Thromb Vasc Biol. 2005, 25: 29-38.

    CAS  PubMed  Google Scholar 

  751. Mullenix PS, Andersen CA, Starnes BW: Atherosclerosis as inflammation. Ann Vasc Surg. 2005, 19: 130-8.

    PubMed  Google Scholar 

  752. Nigro J, Osman N, Dart AM, Little PJ: Insulin resistance and atherosclerosis. Endocr Rev. 2006, 27: 242-59.

    CAS  PubMed  Google Scholar 

  753. Taqueti VR, Mitchell RN, Lichtman AH: Protecting the pump: controlling myocardial inflammatory responses. Annu Rev Physiol. 2006, 68: 67-95.

    CAS  PubMed  Google Scholar 

  754. Tedgui A, Mallat Z: Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev. 2006, 86: 515-81.

    CAS  PubMed  Google Scholar 

  755. Grainger DJ: TGF-beta and atherosclerosis in man. Cardiovasc Res. 2007, 74: 213-22.

    CAS  PubMed  Google Scholar 

  756. Popa C, Netea MG, van Riel PL, Meer van der JW, Stalenhoef AF: The role of TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk. J Lipid Res. 2007, 48: 751-62.

    CAS  PubMed  Google Scholar 

  757. Schleicher E, Friess U: Oxidative stress, AGE, and atherosclerosis. Kidney Int Suppl. 2007, S17-26.

    Google Scholar 

  758. Kibel A, Belovari T, Drenjančević-Perić I: The role of transferrin in atherosclerosis. Med Hypotheses. 2008, 70: 793-7.

    CAS  PubMed  Google Scholar 

  759. Packard RR, Libby P: Inflammation in atherosclerosis: from vascular biology to biomarker discovery and risk prediction. Clin Chem. 2008, 54: 24-38.

    CAS  PubMed  Google Scholar 

  760. Rader DJ, Daugherty A: Translating molecular discoveries into new therapies for atherosclerosis. Nature. 2008, 451: 904-13.

    CAS  PubMed  Google Scholar 

  761. Tan KT, Lip GY: Imaging of the unstable plaque. Int J Cardiol. 2008, 127: 157-65.

    PubMed  Google Scholar 

  762. van Leuven SI, Franssen R, Kastelein JJ, Levi M, Stroes ES, Tak PP: Systemic inflammation as a risk factor for atherothrombosis. Rheumatology (Oxford). 2008, 47: 3-7.

    CAS  Google Scholar 

  763. Lusis AJ: Atherosclerosis. Nature. 2000, 407: 233-241.

    CAS  PubMed  PubMed Central  Google Scholar 

  764. Stocker R, Keaney JF: Role of oxidative modifications in atherosclerosis. Physiol Rev. 2004, 84: 1381-1478.

    CAS  PubMed  Google Scholar 

  765. Smith C, Mitchinson MJ, Aruoma OI, Halliwell B: Stimulation of lipid peroxidation and hydroxyl radical generation by the contents of human atherosclerotic lesions. Biochem J. 1992, 286: 901-905.

    CAS  PubMed  PubMed Central  Google Scholar 

  766. Ramakrishna G, Rooke TW, Cooper LT: Iron and peripheral arterial disease: revisiting the iron hypothesis in a different light. Vasc Med. 2003, 8: 203-210.

    PubMed  Google Scholar 

  767. Stadler N, Lindner RA, Davies MJ: Direct detection and quantification of transition metal ions in human atherosclerotic plaques: evidence for the presence of elevated levels of iron and copper. Arterioscler Thromb Vasc Biol. 2004, 24: 949-54.

    CAS  PubMed  Google Scholar 

  768. Wolff B, Volzke H, Ludemann J, Robinson D, Vogelgesang D, Staudt A, Kessler C, Dahm JB, John U, Felix SB: Association between high serum ferritin levels and carotid atherosclerosis in the study of health in Pomerania (SHIP). Stroke. 2004, 35: 453-7.

    CAS  PubMed  Google Scholar 

  769. Roijers RB, Dutta RK, Mutsaers PHA, Gijbels MJJ, de Winther MPJ, de Goeij JJM, Vusse van der GJ: Spatial correlation of trace elements with morphological features of atherosclerotic plaques. Nuclear Instruments & Methods in Physics Research Section B-Beam Interactions with Materials and Atoms. 2005, 231: 239-244.

    CAS  Google Scholar 

  770. Stocker R, Keaney JF: New insights on oxidative stress in the artery wall. Journal of Thrombosis and Haemostasis. 2005, 3: 1825-1834.

    CAS  PubMed  Google Scholar 

  771. Stanley N, Stadler N, Woods AA, Bannon PG, Davies MJ: Concentrations of iron correlate with the extent of protein, but not lipid, oxidation in advanced human atherosclerotic lesions. Free Radical Biology and Medicine. 2006, 40: 1636-1643.

    CAS  PubMed  Google Scholar 

  772. Watt F, Rajendran R, Ren MQ, Tan BKH, Halliwell B: A nuclear microscopy study of trace elements Ca, Fe, Zn and Cu in atherosclerosis. Nucl Instr Meth Phys Res B. 2006, 249: 646-652.

    CAS  Google Scholar 

  773. Fernandes de Godoy M, Takakura IT, Machado RD, Grassi LV, Nogueira PR: Serum ferritin and obstructive coronary artery disease: Angiographic correlation. Arquivos Brasileiros De Cardiologia. 2007, 88: 430-433.

    CAS  Google Scholar 

  774. Rajendran R, Ren M, Ning P, Tan Kwong Huat B, Halliwell B, Watt F: Promotion of atherogenesis by copper or iron–which is more likely?. Biochem Biophys Res Commun. 2007, 353: 6-10.

    CAS  PubMed  Google Scholar 

  775. Kazi TG, Afridi HI, Kazi N, Jamali MK, Arain MB, Sarfraz RA, Jalbani N, Ansari R, Shah AQ, Memon AU, Khandhro GA: Distribution of zinc, copper and iron in biological samples of Pakistani myocardial infarction (1st, 2nd and 3rd heart attack) patients and controls. Clin Chim Acta. 2008, 389: 114-9.

    CAS  PubMed  Google Scholar 

  776. Sullivan JL: Iron in arterial plaque: A modifiable risk factor for atherosclerosis. Biochim Biophys Acta. 2008.

    Google Scholar 

  777. Yuan XM, Li W: Iron involvement in multiple signaling pathways of atherosclerosis: a revisited hypothesis. Curr Med Chem. 2008, 15: 2157-72.

    CAS  PubMed  Google Scholar 

  778. Matthews AJ, Vercellotti GM, Menchaca HJ, Bloch PH, Michalek VN, Marker PH, Murar J, Buchwald H: Iron and atherosclerosis: inhibition by the iron chelator deferiprone (L1). J Surg Res. 1997, 73: 35-40.

    CAS  PubMed  Google Scholar 

  779. Ponraj D, Makjanic J, Thong PSP, Tan BKH, Watt F: The onset of atherosclerotic lesion formation in hypercholesterolemic rabbits is delayed by iron depletion. FEBS Lett. 1999, 459: 218-222.

    CAS  PubMed  Google Scholar 

  780. Lee TS, Shiao MS, Pan CC, Chau LY: Iron-deficient diet reduces atherosclerotic lesions in ApoE-deficient mice. Circulation. 1999, 99: 1222-1229.

    CAS  PubMed  Google Scholar 

  781. Ren MQ, Rajendran R, Pan N, Tan BKH, Ong WY, Watt F, Halliwell B: The iron chelator desferrioxamine inhibits atherosclerotic lesion development and decreases lesion iron concentrations in the cholesterol-fed rabbit. Free Radical Biology and Medicine. 2005, 38: 1206-1211.

    Google Scholar 

  782. Ferrara DE, Taylor WR: Iron chelation and vascular function: in search of the mechanisms. Arterioscler Thromb Vasc Biol. 2005, 25: 2235-7.

    CAS  PubMed  Google Scholar 

  783. Kiechl S, Willeit J, Egger G, Poewe W, Oberhollenzer F: Body iron stores and the risk of carotid atherosclerosis: prospective results from the Bruneck study. Circulation. 1997, 96: 3300-7.

    CAS  PubMed  Google Scholar 

  784. de Valk B, Marx JJM: Iron, atherosclerosis, and ischemic heart disease. Arch Internal Med. 1999, 159: 1542-1548.

    CAS  Google Scholar 

  785. Zacharski LR, Chow B, Lavori PW, Howes PS, Bell MR, DiTommaso MA, Carnegie NM, Bech F, Amidi M, Muluk S: The Iron (Fe) and Atherosclerosis Study (FeAST): A pilot study of reduction of body iron stores in atherosclerotic peripheral vascular disease. American Heart Journal. 2000, 139: 337-345.

    CAS  PubMed  Google Scholar 

  786. Day SM, Duquaine D, Mundada LV, Menon RG, Khan BV, Rajagopalan S, Fay WP: Chronic iron administration increases vascular oxidative stress and accelerates arterial thrombosis. Circulation. 2003, 107: 2601-2606.

    CAS  PubMed  Google Scholar 

  787. Ren MQ, Watt F, Huat BTK, Halliwell B: Trace elemental distributions in induced atherosclerotic lesions using nuclear microscopy. Nuclear Instr Meth Phys Res B. 2003, 210: 336-342.

    Google Scholar 

  788. Minqin R, Watt F, Huat BTK, Halliwell B: Correlation of iron and zinc levels with lesion depth in newly formed atherosclerotic lesions. Free Radical Biology and Medicine. 2003, 34: 746-752.

    PubMed  Google Scholar 

  789. You SA, Archacki SR, Angheloiu G, Moravec CS, Rao S, Kinter M, Topol EJ, Wang Q: Proteomic approach to coronary atherosclerosis shows ferritin light chain as a significant marker: evidence consistent with iron hypothesis in atherosclerosis. Physiol Genomics. 2003, 13: 25-30.

    CAS  PubMed  Google Scholar 

  790. Zacharski LR, Gerhard GS: Atherosclerosis: a manifestation of chronic iron toxicity?. Vascular Medicine. 2003, 8: 153-155.

    PubMed  Google Scholar 

  791. Kallianpur AR: Iron and oxidative injury – A commentary on "Fatty acid-mediated iron translocation: A synergistic mechanism of oxidative injury" by D. Yao et al. Free Radical Biology and Medicine. 2005, 39: 1305-1309.

    CAS  PubMed  Google Scholar 

  792. You SA, Wang Q: Ferritin in atherosclerosis. Clin Chim Acta. 2005, 357: 1-16.

    CAS  PubMed  Google Scholar 

  793. Lapenna D, Pierdomenico SD, Ciofani G, Ucchino S, Neri M, Giamberardino MA, Cuccurullo F: Association of body iron stores with low molecular weight iron and oxidant damage of human atherosclerotic plaques. Free Rad Biol Med. 2007, 42: 492-498.

    CAS  PubMed  Google Scholar 

  794. Marx JJ, Kartikasari AE, Georgiou NA: Can iron chelators influence the progression of atherosclerosis?. Hemoglobin. 2008, 32: 123-34.

    CAS  PubMed  Google Scholar 

  795. Paulsson J, Dadfar E, Held C, Jacobson SH, Lundahl J: Activation of peripheral and in vivo transmigrated neutrophils in patients with stable coronary artery disease. Atherosclerosis. 2007, 192: 328-34.

    CAS  PubMed  Google Scholar 

  796. Rooyakkers TM, Stroes ES, Kooistra MP, van Faassen EE, Hider RC, Rabelink TJ, Marx JJ: Ferric saccharate induces oxygen radical stress and endothelial dysfunction in vivo. Eur J Clin Invest. 2002, 32 (Suppl 1): 9-16.

    CAS  PubMed  Google Scholar 

  797. Duffy SJ, Biegelsen ES, Holbrook M, Russell JD, Gokce N, Keaney JF, Vita JA: Iron chelation improves endothelial function in patients with coronary artery disease. Circulation. 2001, 103: 2799-804.

    CAS  PubMed  Google Scholar 

  798. Ishizaka N, Saito K, Mori I, Matsuzaki G, Ohno M, Nagai R: Iron chelation suppresses ferritin upregulation and attenuates vascular dysfunction in the aorta of angiotensin II-infused rats. Arterioscler Thromb Vasc Biol. 2005, 25: 2282-8.

    CAS  PubMed  Google Scholar 

  799. Saito K, Ishizaka N, Aizawa T, Sata M, Iso-o N, Noiri E, Mori I, Ohno M, Nagai R: Iron chelation and a free radical scavenger suppress angiotensin II-induced upregulation of TGF-beta1 in the heart. Am J Physiol Heart Circ Physiol. 2005, 288: H1836-43.

    CAS  PubMed  Google Scholar 

  800. Thomas SR, Schulz E, Keaney JF: Hydrogen peroxide restrains endothelium-derived nitric oxide bioactivity – Role for iron-dependent oxidative stress. Free Radical Biology and Medicine. 2006, 41: 681-688.

    CAS  PubMed  Google Scholar 

  801. Zacharski LR, Chow BK, Howes PS, Shamayeva G, Baron JA, Dalman RL, Malenka DJ, Ozaki CK, Lavori PW: Reduction of iron stores and cardiovascular outcomes in patients with peripheral arterial disease – A randomized controlled trial. JAMA. 2007, 297: 603-610.

    CAS  PubMed  Google Scholar 

  802. Shiesh SC, Chen CY, Lin XZ, Liu ZA, Tsao HC: Melatonin prevents pigment gallstone formation induced by bile duct ligation in guinea pigs. Hepatology. 2000, 32: 455-60.

    CAS  PubMed  Google Scholar 

  803. Koppisetti S, Jenigiri B, Terron MP, Tengattini S, Tamura H, Flores LJ, Tan DX, Reiter RJ: Reactive oxygen species and the hypomotility of the gall bladder as targets for the treatment of gallstones with melatonin: a review. Dig Dis Sci. 2008, 53: 2592-603.

    CAS  PubMed  Google Scholar 

  804. Robinson JG: Models for describing relations among the various statin drugs, low-density lipoprotein cholesterol lowering, pleiotropic effects, and cardiovascular risk. Am J Cardiol. 2008, 101: 1009-15.

    CAS  PubMed  Google Scholar 

  805. Prentice RL, Langer RD, Stefanick ML, Howard BV, Pettinger M, Anderson GL, Barad D, Curb JD, Kotchen J, Kuller L, Limacher M, Wactawski-Wende J: Combined analysis of Women's Health Initiative observational and clinical trial data on postmenopausal hormone treatment and cardiovascular disease. Am J Epidemiol. 2006, 163: 589-99.

    PubMed  Google Scholar 

  806. Couzin J: Cholesterol veers off script. Science. 2008, 322: 220-3.

    CAS  PubMed  Google Scholar 

  807. Peterson RT: Chemical biology and the limits of reductionism. Nat Chem Biol. 2008, 4: 635-8.

    CAS  PubMed  Google Scholar 

  808. Weitz-Schmidt G, Welzenbach K, Brinkmann V, Kamata T, Kallen J, Bruns C, Cottens S, Takada Y, Hommel U: Statins selectively inhibit leukocyte function antigen-1 by binding to a novel regulatory integrin site. Nat Med. 2001, 7: 687-92.

    CAS  PubMed  Google Scholar 

  809. Arkin MR, Wells JA: Small-molecule inhibitors of protein-protein interactions: progressing towards the dream. Nat Rev Drug Discov. 2004, 3: 301-17.

    CAS  PubMed  Google Scholar 

  810. Bellosta S, Ferri N, Arnaboldi L, Bernini F, Paoletti R, Corsini A: Pleiotropic effects of statins in atherosclerosis and diabetes. Diabetes Care. 2000, 23: B72-U5.

    PubMed  Google Scholar 

  811. Kwak B, Mulhaupt F, Myit S, Mach F: Statins as a newly recognized type of immunomodulator. Nat Med. 2000, 6: 1399-402.

    CAS  PubMed  Google Scholar 

  812. Albert MA, Danielson E, Rifai N, Ridker PM: Effect of statin therapy on C-reactive protein levels: the pravastatin inflammation/CRP evaluation (PRINCE): a randomized trial and cohort study. JAMA. 2001, 286: 64-70.

    CAS  PubMed  Google Scholar 

  813. LaRosa JC: Pleiotropic effects of statins and their clinical significance. American Journal of Cardiology. 2001, 88: 291.

    CAS  PubMed  Google Scholar 

  814. Takemoto M, Node K, Nakagami H, Liao YL, Grimm M, Takemoto Y, Kitakaze M, Liao JK: Statins as antioxidant therapy for preventing cardiac myocyte hypertrophy. Journal of Clinical Investigation. 2001, 108: 1429-1437.

    CAS  PubMed  PubMed Central  Google Scholar 

  815. Liao JK: Beyond lipid lowering: the role of statins in vascular protection. Int J Cardiol. 2002, 86: 5-18.

    PubMed  Google Scholar 

  816. McFarlane SI, Muniyappa R, Francisco R, Sowers JR: Clinical review 145 – Pleiotropic effects of statins: Lipid reduction and beyond. J Clin Endocrinol Metab. 2002, 87: 1451-1458.

    CAS  PubMed  Google Scholar 

  817. Undas A, Brozek J, Musial J: Anti-inflammatory and antithrombotic effects of statins in the management of coronary artery disease. Clin Lab. 2002, 48: 287-96.

    CAS  PubMed  Google Scholar 

  818. Bonetti PO, Lerman LO, Napoli C, Lerman A: Statin effects beyond lipid lowering–are they clinically relevant?. Eur Heart J. 2003, 24: 225-48.

    CAS  PubMed  Google Scholar 

  819. Dichtl W, Dulak J, Frick M, Alber HF, Schwarzacher SP, Ares MPS, Nilsson J, Pachinger O, Weidinger F: HMG-CoA reductase inhibitors regulate inflammatory transcription factors in human endothelial and vascular smooth muscle cells. Arteriosclerosis Thrombosis and Vascular Biology. 2003, 23: 58-63.

    CAS  Google Scholar 

  820. Mulhaupt F, Matter CM, Kwak BR, Pelli G, Veillard NR, Burger F, Graber P, Luscher TF, Mach F: Statins (HMG-CoA reductase inhibitors) reduce CD40 expression in human vascular cells. Cardiovasc Res. 2003, 59: 755-66.

    CAS  PubMed  Google Scholar 

  821. Wierzbicki AS, Poston R, Ferro A: The lipid and non-lipid effects of statins. Pharmacol Ther. 2003, 99: 95-112.

    CAS  PubMed  Google Scholar 

  822. Davignon J: Beneficial cardiovascular pleiotropic effects of statins. Circulation. 2004, 109: 39-43.

    Google Scholar 

  823. Halcox JP, Deanfield JE: Beyond the laboratory: clinical implications for statin pleiotropy. Circulation. 2004, 109: II42-8.

    PubMed  Google Scholar 

  824. Liao JK: Statins: potent vascular anti-inflammatory agents. Int J Clin Pract Suppl. 2004, 41-8.

    Google Scholar 

  825. Schönbeck U, Libby P: Inflammation, immunity, and HMG-CoA reductase inhibitors – Statins as antiinflammatory agents?. Circulation. 2004, 109: 18-26.

    Google Scholar 

  826. Liao JK, Laufs U: Pleiotropic effects of statins. Annu Rev Pharmacol Toxicol. 2005, 45: 89-118.

    CAS  PubMed  PubMed Central  Google Scholar 

  827. Elrod JW, Lefer DJ: The effects of statins on endothelium, inflammation and cardioprotection. Drug News Perspect. 2005, 18: 229-36.

    CAS  PubMed  Google Scholar 

  828. Robinson JG, Smith B, Maheshwari N, Schrott H: Pleiotropic effects of statins: Benefit beyond cholesterol reduction? A meta-regression analysis. J Amer Coll Cardiol. 2005, 46: 1855-1862.

    CAS  Google Scholar 

  829. Tsubouchi H, Inoguchi T, Sonta T, Sato N, Sekiguchi N, Kobayashi K, Sumimoto H, Utsumi H, Nawata H: Statin attenuates high glucose-induced and diabetes-induced oxidative stress in vitro and in vivo evaluated by electron spin resonance measurement. Free Radical Biology and Medicine. 2005, 39: 444-452.

    CAS  PubMed  Google Scholar 

  830. Beckman JA, Creager MA: The nonlipid effects of statins on endothelial function. Trends Cardiovasc Med. 2006, 16: 156-62.

    CAS  PubMed  Google Scholar 

  831. Carloni S, Mazzoni E, Cimino M, De Simoni MG, Perego C, Scopa C, Balduini W: Simvastatin reduces caspase-3 activation and inflammatory markers induced by hypoxia-ischemia in the newborn rat. Neurobiol Dis. 2006, 21: 119-26.

    CAS  PubMed  Google Scholar 

  832. Grimes DS: Are statins analogues of vitamin D?. Lancet. 2006, 368: 83-6.

    CAS  PubMed  Google Scholar 

  833. Paumelle R, Blanquart C, Briand O, Barbier O, Duhem C, Woerly G, Percevault F, Fruchart JC, Dombrowicz D, Glineur C, Staels B: Acute antiinflammatory properties of statins involve peroxisome proliferator-activated receptor-alpha via inhibition of the protein kinase C signaling pathway. Circ Res. 2006, 98: 361-9.

    CAS  PubMed  Google Scholar 

  834. Alegret M, Silvestre JS: Pleiotropic effects of statins and related pharmacological experimental approaches. Timely Top Med Cardiovasc Dis. 2007, 11: E10.

    CAS  PubMed  Google Scholar 

  835. Farooqui AA, Ong WY, Horrocks LA, Chen P, Farooqui T: Comparison of biochemical effects of statins and fish oil in brain: the battle of the titans. Brain Res Rev. 2007, 56: 443-71.

    CAS  PubMed  Google Scholar 

  836. Kuipers HF, Elsen van den PJ: Immunomodulation by statins: inhibition of cholesterol vs. isoprenoid biosynthesis. Biomed Pharmacother. 2007, 61: 400-7.

    CAS  PubMed  Google Scholar 

  837. Lahera V, Goicoechea M, de Vinuesa SG, Miana M, de las Heras N, Cachofeiro V, Luno J: Endothelial dysfunction, oxidative stress and inflammation in atherosclerosis: beneficial effects of statins. Curr Med Chem. 2007, 14: 243-8.

    CAS  PubMed  Google Scholar 

  838. Paraskevas KI, Tzovaras AA, Briana DD, Mikhailidis DP: Emerging indications for statins: a pluripotent family of agents with several potential applications. Curr Pharm Des. 2007, 13: 3622-36.

    CAS  PubMed  Google Scholar 

  839. Terblanche M, Almog Y, Rosenson R, Smith TS, Hackam DG: Statins and sepsis: multiple modifications at multiple levels. Lancet Infectious Diseases. 2007, 7: 358-368.

    CAS  PubMed  Google Scholar 

  840. Wang CY, Liu PY, Liao JK: Pleiotropic effects of statin therapy: molecular mechanisms and clinical results. Trends Mol Med. 2008, 14: 37-44.

    CAS  PubMed  PubMed Central  Google Scholar 

  841. Wagner BK, Kitami T, Gilbert TJ, Peck D, Ramanathan A, Schreiber SL, Golub TR, Mootha VK: Large-scale chemical dissection of mitochondrial function. Nat Biotechnol. 2008, 26: 343-351.

    CAS  PubMed  PubMed Central  Google Scholar 

  842. Libby P, Aikawa M: Stabilization of atherosclerotic plaques: new mechanisms and clinical targets. Nat Med. 2002, 8: 1257-62.

    CAS  PubMed  Google Scholar 

  843. Blake GJ, Ridker PM: Are statins anti-inflammatory?. Curr Control Trials Cardiovasc Med. 2000, 1: 161-165.

    CAS  PubMed  PubMed Central  Google Scholar 

  844. Diomede L, Albani D, Sottocorno M, Donati MB, Bianchi M, Fruscella P, Salmona M: In vivo anti-inflammatory effect of statins is mediated by nonsterol mevalonate products. Arterioscler Thromb Vasc Biol. 2001, 21: 1327-32.

    CAS  PubMed  Google Scholar 

  845. Laufs U, Gertz K, Dirnagl U, Bohm M, Nickenig G, Endres M: Rosuvastatin, a new HMG-CoA reductase inhibitor, upregulates endothelial nitric oxide synthase and protects from ischemic stroke in mice. Brain Res. 2002, 942: 23-30.

    CAS  PubMed  Google Scholar 

  846. Neuhaus O, Strasser-Fuchs S, Fazekas F, Kieseier BC, Niederwieser G, Hartung HP, Archelos JJ: Statins as immunomodulators: comparison with interferon-beta 1b in MS. Neurology. 2002, 59: 990-7.

    CAS  PubMed  Google Scholar 

  847. Rasmussen LM, Hansen PR, Nabipour MT, Olesen P, Kristiansen MT, Ledet T: Diverse effects of inhibition of 3-hydroxy-3-methylglutaryl-CoA reductase on the expression of VCAM-1 and E-selectin in endothelial cells. Biochem J. 2001, 360: 363-70.

    CAS  PubMed  PubMed Central  Google Scholar 

  848. Takemoto M, Liao JK: Pleiotropic effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors. Arterioscler Thromb Vasc Biol. 2001, 21: 1712-9.

    CAS  PubMed  Google Scholar 

  849. Sukhova GK, Williams JK, Libby P: Statins reduce inflammation in atheroma of nonhuman primates independent of effects on serum cholesterol. Arterioscler Thromb Vasc Biol. 2002, 22: 1452-8.

    CAS  PubMed  Google Scholar 

  850. Scalia R, Stalker TJ: Microcirculation as a target for the anti-inflammatory properties of statins. Microcirculation. 2002, 9: 431-42.

    CAS  PubMed  Google Scholar 

  851. Weitz-Schmidt G: Statins as anti-inflammatory agents. Trends Pharmacol Sci. 2002, 23: 482-6.

    CAS  PubMed  Google Scholar 

  852. Youssef S, Stuve O, Patarroyo JC, Ruiz PJ, Radosevich JL, Hur EM, Bravo M, Mitchell DJ, Sobel RA, Steinman L, Zamvil SS: The HMG-CoA reductase inhibitor, atorvastatin, promotes a Th2 bias and reverses paralysis in central nervous system autoimmune disease. Nature. 2002, 420: 78-84.

    CAS  PubMed  Google Scholar 

  853. Balk EM, Lau J, Goudas LC, Jordan HS, Kupelnick B, Kim LU, Karas RH: Effects of statins on nonlipid serum markers associated with cardiovascular disease: a systematic review. Ann Intern Med. 2003, 139: 670-82.

    CAS  PubMed  Google Scholar 

  854. Blanco-Colio LM, Tuñon J, Martin-Ventura JL, Egido J: Anti-inflammatory and immunomodulatory effects of statins. Kidney Int. 2003, 63: 12-23.

    CAS  PubMed  Google Scholar 

  855. Kwak BR, Mulhaupt F, Mach F: Atherosclerosis: anti-inflammatory and immunomodulatory activities of statins. Autoimmun Rev. 2003, 2: 332-8.

    CAS  PubMed  Google Scholar 

  856. Mason JC: Statins and their role in vascular protection. Clin Sci (Lond). 2003, 105: 251-66.

    CAS  Google Scholar 

  857. Naidu BV, Woolley SM, Farivar AS, Thomas R, Fraga C, Mulligan MS: Simvastatin ameliorates injury in an experimental model of lung ischemia-reperfusion. J Thorac Cardiovasc Surg. 2003, 126: 482-9.

    CAS  PubMed  Google Scholar 

  858. Pate GE, Tahir MN, Murphy RT, Foley JB: Anti-inflammatory effects of statins in patients with aortic stenosis. J Cardiovasc Pharmacol Ther. 2003, 8: 201-6.

    CAS  PubMed  Google Scholar 

  859. Shishehbor MH, Aviles RJ, Brennan ML, Fu XM, Goormastic M, Pearce GL, Gokce N, Keaney JF, Penn MS, Sprecher DL, Vita JA, Hazen SL: Association of nitrotyrosine levels with cardiovascular disease and modulation by statin therapy. JAMA. 2003, 289: 1675-1680.

    CAS  PubMed  Google Scholar 

  860. Stüve O, Youssef S, Dunn S, Slavin AJ, Steinman L, Zamvil SS: The potential therapeutic role of statins in central nervous system autoimmune disorders. Cell Mol Life Sci. 2003, 60: 2483-91.

    PubMed  Google Scholar 

  861. Stüve O, Prod'homme T, Slavin A, Youssef S, Dunn S, Steinman L, Zamvil SS: Statins and their potential targets in multiple sclerosis therapy. Expert Opin Ther Targets. 2003, 7: 613-22.

    PubMed  Google Scholar 

  862. Stüve O, Youssef S, Steinman L, Zamvil SS: Statins as potential therapeutic agents in neuroinflammatory disorders. Curr Opin Neurol. 2003, 16: 393-401.

    PubMed  Google Scholar 

  863. Durant R, Klouche K, Delbosc S, Morena M, Amigues L, Beraud JJ, Canaud B, Cristol JP: Superoxide anion overproduction in sepsis: effects of vitamin e and simvastatin. Shock. 2004, 22: 34-9.

    PubMed  Google Scholar 

  864. Endres M, Laufs U: Effects of statins on endothelium and signaling mechanisms. Stroke. 2004, 35: 2708-11.

    CAS  PubMed  Google Scholar 

  865. Kunkel EJ, Dea M, Ebens A, Hytopoulos E, Melrose J, Nguyen D, Ota KS, Plavec I, Wang Y, Watson SR, Butcher EC, Berg EL: An integrative biology approach for analysis of drug action in models of human vascular inflammation. FASEB J. 2004, 18: 1279-81.

    CAS  PubMed  Google Scholar 

  866. McCarey DW, McInnes IB, Madhok R, Hampson R, Scherbakov O, Ford I, Capell HA, Sattar N: Trial of Atorvastatin in Rheumatoid Arthritis (TARA): double-blind, randomised placebo-controlled trial. Lancet. 2004, 363: 2015-21.

    CAS  PubMed  Google Scholar 

  867. Rosenson RS: Statins in atherosclerosis: lipid-lowering agents with antioxidant capabilities. Atherosclerosis. 2004, 173: 1-12.

    CAS  PubMed  Google Scholar 

  868. McInnes IB, McCarey DW, Sattar N: Do statins offer therapeutic potential in inflammatory arthritis?. Ann Rheum Dis. 2004, 63: 1535-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  869. Steffens S, Mach F: Anti-inflammatory properties of statins. Semin Vasc Med. 2004, 4: 417-22.

    PubMed  Google Scholar 

  870. Stoll LL, McCormick ML, Denning GM, Weintraub NL: Antioxidant effects of statins. Drugs Today (Barc). 2004, 40: 975-90.

    CAS  Google Scholar 

  871. Arnaud C, Braunersreuther V, Mach F: Toward immunomodulatory and anti-inflammatory properties of statins. Trends Cardiovasc Med. 2005, 15: 202-6.

    CAS  PubMed  Google Scholar 

  872. Endres M: Statins and stroke. J Cereb Blood Flow Metab. 2005, 25: 1093-110.

    CAS  PubMed  Google Scholar 

  873. Gaugler MH, Vereycken-Holler V, Squiban C, Vandamme M, Vozenin-Brotons MC, Benderitter M: Pravastatin limits endothelial activation after irradiation and decreases the resulting inflammatory and thrombotic responses. Radiat Res. 2005, 163: 479-87.

    CAS  PubMed  Google Scholar 

  874. Jain MK, Ridker PM: Anti-inflammatory effects of statins: clinical evidence and basic mechanisms. Nat Rev Drug Discov. 2005, 4: 977-87.

    CAS  PubMed  Google Scholar 

  875. Macin SM, Perna ER, Farias EF, Franciosi V, Cialzeta JR, Brizuela M, Medina F, Tajer C, Doval H, Badaracco R: Atorvastatin has an important acute anti-inflammatory effect in patients with acute coronary syndrome: results of a randomized, double-blind, placebo-controlled study. Am Heart J. 2005, 149: 451-7.

    CAS  PubMed  Google Scholar 

  876. Methe H, Kim JO, Kofler S, Nabauer M, Weis M: Statins decrease Toll-like receptor 4 expression and downstream signaling in human CD14+ monocytes. Arterioscler Thromb Vasc Biol. 2005, 25: 1439-45.

    CAS  PubMed  Google Scholar 

  877. Schultz Johansen J, Harris AK, Rychly DJ, Ergul A: Oxidative stress and the use of antioxidants in diabetes: linking basic science to clinical practice. Cardiovasc Diabetol. 2005, 4: 5.

    Google Scholar 

  878. Tonelli M, Sacks F, Pfeffer M, Jhangri GS, Curhan G: Biomarkers of inflammation and progression of chronic kidney disease. Kidney Int. 2005, 68: 237-45.

    CAS  PubMed  Google Scholar 

  879. Abeles AM, Pillinger MH: Statins as antiinflammatory and immunomodulatory agents: a future in rheumatologic therapy?. Arthritis Rheum. 2006, 54: 393-407.

    CAS  PubMed  Google Scholar 

  880. Endres M: Statins: potential new indications in inflammatory conditions. Atheroscler Suppl. 2006, 7: 31-5.

    CAS  PubMed  Google Scholar 

  881. Yang J, Li XP, Zhao SP, Li J, Li JD, Xie XM: The effect of different doses of fluvastatin on inflammatory markers in the early phase of acute coronary syndrome. Clin Chim Acta. 2006, 368: 183-7.

    CAS  PubMed  Google Scholar 

  882. Ahn KS, Sethi G, Aggarwal BB: Simvastatin potentiates TNF-alpha-induced apoptosis through the down-regulation of NF-kappaB-dependent antiapoptotic gene products: role of IkappaBalpha kinase and TGF-beta-activated kinase-1. J Immunol. 2007, 178: 2507-16.

    CAS  PubMed  Google Scholar 

  883. Becker RC: Off-target properties of pharmacotherapy and the importance of mechanistic investigations in early clinical phase drug development. J Thromb Thrombolysis. 2007, 23: 159-61.

    PubMed  Google Scholar 

  884. Greenwood J, Mason JC: Statins and the vascular endothelial inflammatory response. Trends Immunol. 2007, 28: 88-98.

    CAS  PubMed  Google Scholar 

  885. Li JJ, Zheng X, Li J: Statins may be beneficial for patients with slow coronary flow syndrome due to its anti-inflammatory property. Med Hypotheses. 2007, 69: 333-7.

    CAS  PubMed  Google Scholar 

  886. Sandek A, Utchill S, Rauchhaus M: The endotoxin-lipoprotein hypothesis – an update. Archives of Medical Science. 2007, 3: S81-S90.

    CAS  Google Scholar 

  887. Siffrin V, Brandt AU, Herz J, Zipp F: New insights into adaptive immunity in chronic neuroinflammation. Adv Immunol. 2007, 96: 1-40.

    CAS  PubMed  Google Scholar 

  888. Souza-Costa DC, Sandrim VC, Lopes LF, Gerlach RF, Rego EM, Tanus-Santos JE: Anti-inflammatory effects of atorvastatin: modulation by the T-786C polymorphism in the endothelial nitric oxide synthase gene. Atherosclerosis. 2007, 193: 438-44.

    CAS  PubMed  Google Scholar 

  889. Hernández-Romero MdC, Argüelles S, Villaran RF, de Pablos RM, Delgado-Cortés MJ, Santiago M, Herrera AJ, Cano J, Machado A: Simvastatin prevents the inflammatory process and the dopaminergic degeneration induced by the intranigral injection of lipopolysaccharide. J Neurochem. 2008, 105: 445-59.

    PubMed  Google Scholar 

  890. Paraskevas KI: Statin treatment for rheumatoid arthritis: a promising novel indication. Clin Rheumatol. 2008, 27: 281-7.

    PubMed  Google Scholar 

  891. Prinz V, Laufs U, Gertz K, Kronenberg G, Balkaya M, Leithner C, Lindauer U, Endres M: Intravenous rosuvastatin for acute stroke treatment: an animal study. Stroke. 2008, 39: 433-8.

    CAS  PubMed  Google Scholar 

  892. Xu SZ, Zhong W, Watson NM, Dickerson E, Wake JD, Lindow SW, Newton CJ, Atkin SL: Fluvastatin reduces oxidative damage in human vascular endothelial cells by upregulating Bcl-2. J Thromb Haemost. 2008, 6: 692-700.

    CAS  PubMed  Google Scholar 

  893. Dombrecht EJ, De Tollenaere CB, Aerts K, Cos P, Schuerwegh AJ, Bridts CH, Van Offel JF, Ebo DG, Stevens WJ, De Clerck LS: Antioxidant effect of bisphosphonates and simvastatin on chondrocyte lipid peroxidation. Biochem Biophys Res Commun. 2006, 348: 459-64.

    CAS  PubMed  Google Scholar 

  894. Lee DH, Zacharski LR, Jacobs DR: Comparison of the serum ferritin and percentage of transferrin saturation as exposure markers of iron-driven oxidative stress-related disease outcomes. American Heart Journal. 2006, 151.

    Google Scholar 

  895. Anderson LJ, Holden S, Davis B, Prescott E, Charrier CC, Bunce NH, Firmin DN, Wonke B, Porter J, Walker JM, Pennell DJ: Cardiovascular T2-star (T2*) magnetic resonance for the early diagnosis of myocardial iron overload. Eur Heart J. 2001, 22: 2171-2179.

    CAS  PubMed  Google Scholar 

  896. Yeoh-Ellerton S, Stacey MC: Iron and 8-isoprostane levels in acute and chronic wounds. J Invest Dermatol. 2003, 121: 918-925.

    PubMed  Google Scholar 

  897. Bergan JJ, Schmid-Schonbein GW, Smith PDC, Nicolaides AN, Boisseau MR, Eklof B: Mechanisms of disease: Chronic venous disease. New England Journal of Medicine. 2006, 355: 488-498.

    CAS  PubMed  Google Scholar 

  898. Zamboni P, Izzo M, Fogato L, Carandina S, Lanzara V: Urine hemosiderin: a novel marker to assess the severity of chronic venous disease. J Vasc Surg. 2003, 37: 132-6.

    PubMed  Google Scholar 

  899. Zamboni P: The Big Idea: Iron-dependent inflammation in venous disease and proposed parallels in multiple sclerosis. J R Soc Med. 2006, 99: 589-593.

    PubMed  PubMed Central  Google Scholar 

  900. Lucas SM, Rothwell NJ, Gibson RM: The role of inflammation in CNS injury and disease. Br J Pharmacol. 2006, 147 (Suppl 1): S232-40.

    CAS  PubMed  PubMed Central  Google Scholar 

  901. Lewén A, Matz P, Chan PH: Free radical pathways in CNS injury. J Neurotrauma. 2000, 17: 871-90.

    PubMed  Google Scholar 

  902. Que EL, Domaille DW, Chang CJ: Metals in neurobiology: probing their chemistry and biology with molecular imaging. Chem Rev. 2008, 108: 1517-49.

    CAS  PubMed  Google Scholar 

  903. Kent TA, Soukup VM, Fabian RH: Heterogeneity affecting outcome from acute stroke therapy: making reperfusion worse. Stroke. 2001, 32: 2318-27.

    CAS  PubMed  Google Scholar 

  904. Emsley HC, Tyrrell PJ: Inflammation and infection in clinical stroke. J Cereb Blood Flow Metab. 2002, 22: 1399-419.

    CAS  PubMed  Google Scholar 

  905. Patt A, Horesh IR, Berger EM, Harken AH, Repine JE: Iron depletion or chelation reduces ischemia/reperfusion-induced edema in gerbil brains. J Pediatr Surg. 1990, 25: 224-7.

    CAS  PubMed  Google Scholar 

  906. Dávalos A, Fernandezreal JM, Ricart W, Soler S, Molins A, Planas E, Genis D: Iron-related damage in acute ischemic stroke. Stroke. 1994, 25: 1543-1546.

    PubMed  Google Scholar 

  907. Bishop GM, Robinson SR: Quantitative analysis of cell death and ferritin expression in response to cortical iron: implications for hypoxia-ischemia and stroke. Brain Research. 2001, 907: 175-187.

    CAS  PubMed  Google Scholar 

  908. Demougeot C, Van Hoecke M, Bertrand N, Prigent-Tessier A, Mossiat C, Beley A, Marie C: Cytoprotective efficacy and mechanisms of the liposoluble iron chelator 2,2 '-dipyridyl in the rat photothrombotic ischemic stroke model. J Pharm Exp Therapeut. 2004, 311: 1080-1087.

    CAS  Google Scholar 

  909. Mehta SH, Webb RC, Ergul A, Tawak A, Dorrance AM: Neuroprotection by tempol in a model of iron-induced oxidative stress in acute ischemic stroke. Am J Physiol. 2004, 286: R283-R288.

    CAS  Google Scholar 

  910. Selim MH, Ratan RR: The role of iron neurotoxicity in ischemic stroke. Ageing Research Reviews. 2004, 3: 345-353.

    CAS  PubMed  Google Scholar 

  911. van der ADL, Grobbee DE, Roest M, Marx JJM, Voorbij HA, Schouw van der YT: Serum ferritin is a risk factor for stroke in postmenopausal women. Stroke. 2005, 36: 1637-1641.

    Google Scholar 

  912. Hua Y, Keep RF, Hoff JT, Xi G: Brain injury after intracerebral hemorrhage: the role of thrombin and iron. Stroke. 2007, 38: 759-62.

    CAS  PubMed  Google Scholar 

  913. Millan M, Sobrino T, Castellanos M, Nombela F, Arenillas JF, Riva E, Cristobo I, Garcia MM, Vivancos J, Serena J, Moro MA, Castillo J, Dávalos A: Increased body iron stores are associated with poor outcome after thrombolytic treatment in acute stroke. Stroke. 2007, 38: 90-95.

    PubMed  Google Scholar 

  914. Slemmer JE, Shacka JJ, Sweeney MI, Weber JT: Antioxidants and free radical scavengers for the treatment of stroke, traumatic brain injury and aging. Curr Med Chem. 2008, 15: 404-14.

    CAS  PubMed  Google Scholar 

  915. Baker K, Marcus CB, Huffman K, Kruk H, Malfroy B, Doctrow SR: Synthetic combined superoxide dismutase/catalase mimetics are protective as a delayed treatment in a rat stroke model: a key role for reactive oxygen species in ischemic brain injury. J Pharmacol Exp Ther. 1998, 284: 215-21.

    CAS  PubMed  Google Scholar 

  916. Sun AY, Chen YM: Oxidative stress and neurodegenerative disorders. Journal of Biomedical Science. 1998, 5: 401-414.

    CAS  PubMed  Google Scholar 

  917. Pong K: Oxidative stress in neurodegenerative diseases: therapeutic implications for superoxide dismutase mimetics. Expert Opinion on Biological Therapy. 2003, 3: 127-139.

    CAS  PubMed  Google Scholar 

  918. Bastianetto S, Quirion R: Natural antioxidants and neurodegenerative diseases. Frontiers in Bioscience. 2004, 9: 3447-3452.

    CAS  PubMed  Google Scholar 

  919. Warner DS, Sheng HX, Batinic-Haberle I: Oxidants, antioxidants and the ischemic brain. J Exp Biol. 2004, 207: 3221-3231.

    CAS  PubMed  Google Scholar 

  920. Craft JM, Watterson DM, Van Eldik LJ: Neuroinflammation: a potential therapeutic target. Expert Opin Ther Targets. 2005, 9: 887-900.

    CAS  PubMed  Google Scholar 

  921. Margaill I, Plotkine M, Lerouet D: Antioxidant strategies in the treatment of stroke. Free Radical Biology and Medicine. 2005, 39: 429-443.

    CAS  PubMed  Google Scholar 

  922. Zhao BL: Natural antioxidants for neurodegenerative diseases. Mol Neurobiol. 2005, 31: 283-293.

    CAS  PubMed  Google Scholar 

  923. Obrenovitch TP: Molecular physiology of preconditioning-induced brain tolerance to ischemia. Physiol Rev. 2008, 88: 211-47.

    CAS  PubMed  Google Scholar 

  924. Sirén AL, Fratelli M, Brines M, Goemans C, Casagrande S, Lewczuk P, Keenan S, Gleiter C, Pasquali C, Capobianco A, Mennini T, Heumann R, Cerami A, Ehrenreich H, Ghezzi P: Erythropoietin prevents neuronal apoptosis after cerebral ischemia and metabolic stress. Proc Natl Acad Sci USA. 2001, 98: 4044-9.

    PubMed  PubMed Central  Google Scholar 

  925. Brines ML, Ghezzi P, Keenan S, Agnello D, de Lanerolle NC, Cerami C, Itri LM, Cerami A: Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proc Natl Acad Sci USA. 2000, 97: 10526-31.

    CAS  PubMed  PubMed Central  Google Scholar 

  926. Digicaylioglu M, Lipton SA: Erythropoietin-mediated neuroprotection involves cross-talk between Jak2 and NF-kappaB signalling cascades. Nature. 2001, 412: 641-7.

    CAS  PubMed  Google Scholar 

  927. Dawson TM: Preconditioning-mediated neuroprotection through erythropoietin?. Lancet. 2002, 359: 96-7.

    PubMed  Google Scholar 

  928. Ehrenreich H, Hasselblatt M, Dembowski C, Cepek L, Lewczuk P, Stiefel M, Rustenbeck HH, Breiter N, Jacob S, Knerlich F, Bohn M, Poser W, Ruther E, Kochen M, Gefeller O, Gleiter C, Wessel TC, De Ryck M, Itri L, Prange H, Cerami A, Brines M, Sirén AL: Erythropoietin therapy for acute stroke is both safe and beneficial. Mol Med. 2002, 8: 495-505.

    CAS  PubMed  PubMed Central  Google Scholar 

  929. Leker RR, Shohami E: Cerebral ischemia and trauma-different etiologies yet similar mechanisms: neuroprotective opportunities. Brain Res Rev. 2002, 39: 55-73.

    PubMed  Google Scholar 

  930. Aydin A, Genc K, Akhisaroglu M, Yorukoglu K, Gokmen N, Gonullu E: Erythropoietin exerts neuroprotective effect in neonatal rat model of hypoxic-ischemic brain injury. Brain Dev. 2003, 25: 494-8.

    PubMed  Google Scholar 

  931. Kumral A, Ozer E, Yilmaz O, Akhisaroglu M, Gokmen N, Duman N, Ulukus C, Genc S, Ozkan H: Neuroprotective effect of erythropoietin on hypoxic-ischemic brain injury in neonatal rats. Biol Neonate. 2003, 83: 224-8.

    CAS  PubMed  Google Scholar 

  932. Matsushita H, Johnston MV, Lange MS, Wilson MA: Protective effect of erythropoietin in neonatal hypoxic ischemia in mice. Neuroreport. 2003, 14: 1757-61.

    CAS  PubMed  Google Scholar 

  933. Grasso G, Sfacteria A, Cerami A, Brines M: Erythropoietin as a tissue-protective cytokine in brain injury: what do we know and where do we go?. Neuroscientist. 2004, 10: 93-8.

    CAS  PubMed  Google Scholar 

  934. Sun Y, Zhou C, Polk P, Nanda A, Zhang JH: Mechanisms of erythropoietin-induced brain protection in neonatal hypoxia-ischemia rat model. J Cereb Blood Flow Metab. 2004, 24: 259-70.

    CAS  PubMed  Google Scholar 

  935. Wang CH, Liang CL, Huang LT, Liu JK, Hung PH, Sun A, Hung KS: Single intravenous injection of naked plasmid DNA encoding erythropoietin provides neuroprotection in hypoxia-ischemia rats. Biochem Biophys Res Commun. 2004, 314: 1064-71.

    CAS  PubMed  Google Scholar 

  936. Bailey DM, Robach P, Thomsen JJ, Lundby C: Erythropoietin depletes iron stores: antioxidant neuroprotection for ischemic stroke?. Stroke. 2006, 37: 2453.

    PubMed  Google Scholar 

  937. Grasso G, Sfacteria A, Meli F, Passalacqua M, Fodale V, Buemi M, Giambartino F, Iacopino DG, Tomasello F: The role of erythropoietin in neuroprotection: therapeutic perspectives. Drug News Perspect. 2007, 20: 315-20.

    CAS  PubMed  Google Scholar 

  938. McPherson RJ, Juul SE: Recent trends in erythropoietin-mediated neuroprotection. Int J Dev Neurosci. 2008, 26: 103-11.

    CAS  PubMed  Google Scholar 

  939. Sola A, Peng H, Rogido M, Wen TC: Animal models of neonatal stroke and response to erythropoietin and cardiotrophin-1. Int J Dev Neurosci. 2008, 26: 27-35.

    CAS  PubMed  Google Scholar 

  940. Erbayraktar S, Yilmaz O, Gokmen N, Brines M: Erythropoietin is a multifunctional tissue-protective cytokine. Curr Hematol Rep. 2003, 2: 465-70.

    PubMed  Google Scholar 

  941. Leist M, Ghezzi P, Grasso G, Bianchi R, Villa P, Fratelli M, Savino C, Bianchi M, Nielsen J, Gerwien J, Kallunki P, Larsen AK, Helboe L, Christensen S, Pedersen LO, Nielsen M, Torup L, Sager T, Sfacteria A, Erbayraktar S, Erbayraktar Z, Gokmen N, Yilmaz O, Cerami-Hand C, Xie QW, Coleman T, Cerami A, Brines M: Derivatives of erythropoietin that are tissue protective but not erythropoietic. Science. 2004, 305: 239-42.

    CAS  PubMed  Google Scholar 

  942. Arcasoy MO: The non-haematopoietic biological effects of erythropoietin. Br J Haematol. 2008, 141: 14-31.

    CAS  PubMed  Google Scholar 

  943. Agnello D, Bigini P, Villa P, Mennini T, Cerami A, Brines ML, Ghezzi P: Erythropoietin exerts an anti-inflammatory effect on the CNS in a model of experimental autoimmune encephalomyelitis. Brain Res. 2002, 952: 128-34.

    CAS  PubMed  Google Scholar 

  944. Fahn S, Cohen G: The oxidant stress hypothesis in Parkinson's disease: evidence supporting it. Ann Neurol. 1992, 32: 804-12.

    CAS  PubMed  Google Scholar 

  945. Halliwell B: Reactive oxygen species and the central nervous system. J Neurochem. 1992, 59: 1609-1623.

    CAS  PubMed  Google Scholar 

  946. Coyle JT, Puttfarcken P: Oxidative stress, glutamate, and neurodegenerative disorders. Science. 1993, 262: 689-95.

    CAS  PubMed  Google Scholar 

  947. Behl C, Davis JB, Lesley R, Schubert D: Hydrogen peroxide mediates amyloid beta protein toxicity. Cell. 1994, 77: 817-27.

    CAS  PubMed  Google Scholar 

  948. Good PF, Werner P, Hsu A, Olanow CW, Perl DP: Evidence of neuronal oxidative damage in Alzheimer's disease. Am J Pathol. 1996, 149: 21-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  949. Jenner P, Olanow CW: Oxidative stress and the pathogenesis of Parkinson's disease. Neurology. 1996, 47: S161-70.

    CAS  PubMed  Google Scholar 

  950. Simonian NA, Coyle JT: Oxidative stress in neurodegenerative diseases. Annu Rev Pharmacol Toxicol. 1996, 36: 83-106.

    CAS  PubMed  Google Scholar 

  951. Yoritaka A, Hattori N, Uchida K, Tanaka M, Stadtman ER, Mizuno Y: Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease. Proc Natl Acad Sci USA. 1996, 93: 2696-701.

    CAS  PubMed  PubMed Central  Google Scholar 

  952. Packer L, Tritschler HJ, Wessel K: Neuroprotection by the metabolic antioxidant alpha-lipoic acid. Free Radic Biol Med. 1997, 22: 359-78.

    CAS  PubMed  Google Scholar 

  953. Gao HM, Liu B, Zhang W, Hong JS: Novel anti-inflammatory therapy for Parkinson's disease. Trends Pharmacol Sci. 2003, 24: 395-401.

    CAS  PubMed  Google Scholar 

  954. Mariani E, Polidori MC, Cherubini A, Mecocci P: Oxidative stress in brain aging, neurodegenerative and vascular diseases: an overview. J Chromatogr B Analyt Technol Biomed Life Sci. 2005, 827: 65-75.

    CAS  PubMed  Google Scholar 

  955. Zeevalk GD, Bernard LP, Song C, Gluck M, Ehrhart J: Mitochondrial inhibition and oxidative stress: reciprocating players in neurodegeneration. Antioxid Redox Signal. 2005, 7: 1117-39.

    CAS  PubMed  Google Scholar 

  956. Lin MT, Beal MF: Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006, 443: 787-95.

    CAS  PubMed  Google Scholar 

  957. Trushina E, McMurray CT: Oxidative stress and mitochondrial dysfunction in neurodegenerative diseases. Neuroscience. 2007, 145: 1233-48.

    CAS  PubMed  Google Scholar 

  958. Benzi G, Moretti A: Are reactive oxygen species involved in Alzheimer's disease?. Neurobiol Aging. 1995, 16: 661-74.

    CAS  PubMed  Google Scholar 

  959. Hensley K, Hall N, Subramaniam R, Cole P, Harris M, Aksenov M, Aksenova M, Gabbita SP, Wu JF, Carney JM, et al: Brain regional correspondence between Alzheimer's disease histopathology and biomarkers of protein oxidation. J Neurochem. 1995, 65: 2146-56.

    CAS  PubMed  Google Scholar 

  960. Markesbery WR: Oxidative stress hypothesis in Alzheimer's disease. Free Radic Biol Med. 1997, 23: 134-47.

    CAS  PubMed  Google Scholar 

  961. Markesbery WR, Lovell MA: 4-hydroxynonenal, a product of lipid peroxidation, is increased in the brain in Alzheimer's disease. Neurobiol Aging. 1998, 19: 33-6.

    CAS  PubMed  Google Scholar 

  962. Cassarino DS, Bennett JP: An evaluation of the role of mitochondria in neurodegenerative diseases: mitochondrial mutations and oxidative pathology, protective nuclear responses, and cell death in neurodegeneration. Brain Res Brain Res Rev. 1999, 29: 1-25.

    CAS  PubMed  Google Scholar 

  963. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O'Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T: Inflammation and Alzheimer's disease. Neurobiol Aging. 2000, 21: 383-421.

    CAS  PubMed  PubMed Central  Google Scholar 

  964. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS, Petersen RB, Smith MA: Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001, 60: 759-767.

    CAS  PubMed  Google Scholar 

  965. McGeer PL, McGeer EG: Inflammation, autotoxicity and Alzheimer disease. Neurobiol Aging. 2001, 22: 799-809.

    CAS  PubMed  Google Scholar 

  966. Butterfield DA, Lauderback CM: Lipid peroxidation and protein oxidation in Alzheimer's disease brain: potential causes and consequences involving amyloid beta-peptide-associated free radical oxidative stress. Free Radic Biol Med. 2002, 32: 1050-60.

    CAS  PubMed  Google Scholar 

  967. Eikelenboom P, Bate C, Van Gool WA, Hoozemans JJ, Rozemuller JM, Veerhuis R, Williams A: Neuroinflammation in Alzheimer's disease and prion disease. Glia. 2002, 40: 232-9.

    CAS  PubMed  Google Scholar 

  968. Suh YH, Checler F: Amyloid precursor protein, presenilins, and alpha-synuclein: molecular pathogenesis and pharmacological applications in Alzheimer's disease. Pharmacol Rev. 2002, 54: 469-525.

    CAS  PubMed  Google Scholar 

  969. McGeer EG, McGeer PL: Inflammatory processes in Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry. 2003, 27: 741-9.

    CAS  PubMed  Google Scholar 

  970. Butterfield DA, Perluigi M, Sultana R: Oxidative stress in Alzheimer's disease brain: new insights from redox proteomics. Eur J Pharmacol. 2006, 545: 39-50.

    CAS  PubMed  Google Scholar 

  971. Jenner P: Oxidative stress in Parkinson's disease. Ann Neurol. 2003, 53 (Suppl 3): S26-36.

    CAS  PubMed  Google Scholar 

  972. Nunomura A, Castellani RJ, Zhu X, Moreira PI, Perry G, Smith MA: Involvement of oxidative stress in Alzheimer disease. J Neuropathol Exp Neurol. 2006, 65: 631-41.

    CAS  PubMed  Google Scholar 

  973. Licastro F, Porcellini E, Caruso C, Lio D, Corder EH: Genetic risk profiles for Alzheimer's disease: integration of APOE genotype and variants that up-regulate inflammation. Neurobiol Aging. 2007, 28: 1637-43.

    CAS  PubMed  Google Scholar 

  974. Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia-Alloza M, de Calignon A, Rozkalne A, Koenigsknecht-Talboo J, Holtzman DM, Bacskai BJ, Hyman BT: Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer's disease. Nature. 2008, 451: 720-4.

    CAS  PubMed  PubMed Central  Google Scholar 

  975. Goodman L: Alzheimer's disease; a clinico-pathologic analysis of twenty-three cases with a theory on pathogenesis. J Nerv Ment Dis. 1953, 118: 97-130.

    CAS  PubMed  Google Scholar 

  976. Dexter DT, Wells FR, Lees AJ, Agid F, Agid Y, Jenner P, Marsden CD: Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinsons disease. J Neurochem. 1989, 52: 1830-1836.

    CAS  PubMed  Google Scholar 

  977. Riederer P, Sofic E, Rausch WD, Schmidt B, Reynolds GP, Jellinger K, Youdim MB: Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J Neurochem. 1989, 52: 515-20.

    CAS  PubMed  Google Scholar 

  978. Jellinger K, Paulus W, Grundke-Iqbal I, Riederer P, Youdim MB: Brain iron and ferritin in Parkinson's and Alzheimer's diseases. J Neural Transm Park Dis Dement Sect. 1990, 2: 327-40.

    CAS  PubMed  Google Scholar 

  979. O'Brien PJ: Molecular mechanisms of quinone cytotoxicity. Chemico-Biol Interact. 1991, 80: 1-41.

    Google Scholar 

  980. Connor JR, Snyder BS, Beard JL, Fine RE, Mufson EJ: Regional distribution of iron and iron-regulatory proteins in the brain in aging and Alzheimers disease. J Neuroscie Res. 1992, 31: 327-335.

    CAS  Google Scholar 

  981. Good PF, Perl DP, Bierer LM, Schmeidler J: Selective accumulation of aluminum and iron in the neurofibrillary tangles of Alzheimer's disease: a laser microprobe (LAMMA) study. Ann Neurol. 1992, 31: 286-92.

    CAS  PubMed  Google Scholar 

  982. Montgomery EB: Heavy metals and the etiology of Parkinson's disease and other movement disorders. Toxicology. 1995, 97: 3-9.

    CAS  PubMed  Google Scholar 

  983. Koeppen AH: The history of iron in the brain. J Neurol Sci. 1995, 134 (Suppl): 1-9.

    CAS  PubMed  Google Scholar 

  984. Deibel MA, Ehmann WD, Markesbery WR: Copper, iron, and zinc imbalances in severely degenerated brain regions in Alzheimer's disease: possible relation to oxidative stress. J Neurol Sci. 1996, 143: 137-42.

    CAS  PubMed  Google Scholar 

  985. Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Kortsha GX, Brown GG, Richardson RJ: Occupational exposures to metals as risk factors for Parkinson's disease. Neurology. 1997, 48: 650-8.

    CAS  PubMed  Google Scholar 

  986. Koeppen AH: A brief history of brain iron research. J Neurol Sci. 2003, 207: 95-7.

    PubMed  Google Scholar 

  987. Youdim MBH, Benshachar D, Riederer P: The possible role of iron in the etiopathology of Parkinsons disease. Movement Disorders. 1993, 8: 1-12.

    CAS  PubMed  Google Scholar 

  988. Gerlach M, Benshachar D, Riederer P, Youdim MBH: Altered brain metabolism of iron as a cause of neurodegenerative diseases. J Neurochem. 1994, 63: 793-807.

    CAS  PubMed  Google Scholar 

  989. Götz ME, Künig G, Riederer P, Youdim MBH: Oxidative stress – free radical production in neural degeneration. Pharmacol Therapeut. 1994, 63: 37-122.

    Google Scholar 

  990. Smith MA, Perry G, Richey PL, Sayre LM, Anderson VE, Beal MF, Kowall N: Oxidative damage in Alzheimer's. Nature. 1996, 382: 120-121.

    CAS  PubMed  Google Scholar 

  991. LeVine SM: Iron deposits in multiple sclerosis and Alzheimer's disease brains. Brain Research. 1997, 760: 298-303.

    CAS  PubMed  Google Scholar 

  992. Smith MA, Harris PLR, Sayre LM, Perry G: Iron accumulation in Alzheimer disease is a source of redox-generated free radicals. Proc Natl Acad Sci. 1997, 94: 9866-9868.

    CAS  PubMed  PubMed Central  Google Scholar 

  993. Lovell MA, Robertson JD, Teesdale WJ, Campbell JL, Markesbery WR: Copper, iron and zinc in Alzheimer's disease senile plaques. J Neurol Sci. 1998, 158: 47-52.

    CAS  PubMed  Google Scholar 

  994. Bartzokis G, Cummings J, Perlman S, Hance DB, Mintz J: Increased basal ganglia iron levels in Huntington disease. Arch Neurol. 1999, 56: 569-74.

    CAS  PubMed  Google Scholar 

  995. Huang XD, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JDA, Hanson GR, Stokes KC, Leopold M, Multhaup G, Goldstein LE, Scarpa RC, Saunders AJ, Lim J, Moir RD, Glabe C, Bowden EF, Masters CL, Fairlie DP, Tanzi RE, Bush AI: Cu(II) potentiation of Alzheimer A beta neurotoxicity – Correlation with cell-free hydrogen peroxide production and metal reduction. J Biol Chem. 1999, 274: 37111-37116.

    CAS  PubMed  Google Scholar 

  996. Jellinger KA: The role of iron in neurodegeneration – Prospects for pharmacotherapy of Parkinson's disease. Drugs & Aging. 1999, 14: 115-140.

    CAS  Google Scholar 

  997. Olanow CW, Tatton WG: Etiology and pathogenesis of Parkinson's disease. Annu Rev Neurosci. 1999, 22: 123-144.

    CAS  PubMed  Google Scholar 

  998. Sayre LM, Perry G, Smith MA: Redox metals and neurodegenerative disease. Current Opinion in Chemical Biology. 1999, 3: 220-225.

    CAS  PubMed  Google Scholar 

  999. Bush AI: Metals and neuroscience. Curr Opin Chem Biol. 2000, 4: 184-91.

    CAS  PubMed  Google Scholar 

  1000. Chiueh CC, Andoh T, Lai AR, Lai E, Krishna G: Neuroprotective strategies in Parkinson's disease: protection against progressive nigral damage induced by free radicals. Neurotox Res. 2000, 2: 293-310.

    CAS  PubMed  Google Scholar 

  1001. Christen Y: Oxidative stress and Alzheimer disease. American Journal of Clinical Nutrition. 2000, 71: 621S-629S.

    CAS  PubMed  Google Scholar 

  1002. Floor E: Iron as a vulnerability factor in nigrostriatal degeneration in aging and Parkinson's disease. Cell Mol Biol (Noisy-le-grand). 2000, 46: 709-20.

    CAS  Google Scholar 

  1003. Smith MA, Rottkamp CA, Nunomura A, Raina AK, Perry G: Oxidative stress in Alzheimer's disease. Biochim Biophys Acta. 2000, 1502: 139-44.

    CAS  PubMed  Google Scholar 

  1004. Varadarajan S, Yatin S, Aksenova M, Butterfield DA: Alzheimer's amyloid beta-peptide-associated free radical oxidative stress and neurotoxicity. J Struct Biol. 2000, 130: 184-208.

    CAS  PubMed  Google Scholar 

  1005. Berg D, Gerlach M, Youdim MBH, Double KL, Zecca L, Riederer P, Becker G: Brain iron pathways and their relevance to Parkinson's disease. J Neurochem. 2001, 79: 225-236.

    CAS  PubMed  Google Scholar 

  1006. Blum D, Torch S, Lambeng N, Nissou MF, Benabid AL, Sadoul R, Verna JM: Molecular pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: contribution to the apoptotic theory in Parkinson's disease. Progress in Neurobiology. 2001, 65: 135-172.

    CAS  PubMed  Google Scholar 

  1007. Campbell A, Smith MA, Sayre LM, Bondy SC, Perry G: Mechanisms by which metals promote events connected to neurodegenerative diseases. Brain Research Bulletin. 2001, 55: 125-132.

    CAS  PubMed  Google Scholar 

  1008. Halliwell B: Role of free radicals in the neurodegenerative diseases – Therapeutic implications for antioxidant treatment. Drugs & Aging. 2001, 18: 685-716.

    CAS  Google Scholar 

  1009. HaMai D, Bondy SC, Becaria A, Campbell A: The chemistry of transition metals in relation to their potential role in neurodegenerative processes. Curr Top Med Chem. 2001, 1: 541-51.

    CAS  PubMed  Google Scholar 

  1010. Rottkamp CA, Raina AK, Zhu XW, Gaier E, Bush AI, Atwood CS, Chevion M, Perry G, Smith MA: Redox-active iron mediates amyloid-beta toxicity. Free Rad Biol Med. 2001, 30: 447-450.

    CAS  PubMed  Google Scholar 

  1011. Sayre LM, Smith MA, Perry G: Chemistry and biochemistry of oxidative stress in neurodegenerative disease. Current Medicinal Chemistry. 2001, 8: 721-738.

    CAS  PubMed  Google Scholar 

  1012. Bharath S, Hsu M, Kaur D, Rajagopalan S, Andersen JK: Glutathione, iron and Parkinson's disease. Biochem Pharmacol. 2002, 64: 1037-48.

    CAS  PubMed  Google Scholar 

  1013. Bishop GM, Robinson SR, Liu Q, Perry G, Atwood CS, Smith MA: Iron: a pathological mediator of Alzheimer disease?. Dev Neurosci. 2002, 24: 184-7.

    CAS  PubMed  Google Scholar 

  1014. Giasson BI, Ischiropoulos H, Lee VM, Trojanowski JQ: The relationship between oxidative/nitrative stress and pathological inclusions in Alzheimer's and Parkinson's diseases. Free Radic Biol Med. 2002, 32: 1264-75.

    CAS  PubMed  Google Scholar 

  1015. Gnjec A, Fonte JA, Atwood C, Martins RN: Transition metal chelator therapy – A potential treatment for Alzheimer's disease?. Frontiers in Bioscience. 2002, 7: D1016-D1023.

    CAS  PubMed  Google Scholar 

  1016. Perry G, Sayre LM, Atwood CS, Castellani RJ, Cash AD, Rottkamp CA, Smith MA: The role of iron and copper in the aetiology of neurodegenerative disorders – Therapeutic implications. CNS Drugs. 2002, 16: 339-352.

    CAS  PubMed  Google Scholar 

  1017. Rao AV, Balachandran B: Role of oxidative stress and antioxidants in neurodegenerative diseases. Nutritional Neuroscience. 2002, 5: 291-309.

    CAS  PubMed  Google Scholar 

  1018. Burdo JR, Connor JR: Brain iron uptake and homeostatic mechanisms: An overview. Biometals. 2003, 16: 63-75.

    CAS  PubMed  Google Scholar 

  1019. Bush AI: The metallobiology of Alzheimer's disease. Trends Neurosci. 2003, 26: 207-214.

    CAS  PubMed  Google Scholar 

  1020. Finefrock AE, Bush AI, Doraiswamy PM: Current status of metals as therapeutic targets in Alzheimer's disease. J Am Geriatr Soc. 2003, 51: 1143-8.

    PubMed  Google Scholar 

  1021. Ke Y, Qian ZM: Iron misregulation in the brain: a primary cause of neurodegenerative disorders. Lancet Neurol. 2003, 2: 246-253.

    CAS  PubMed  Google Scholar 

  1022. Barnham KJ, Masters CL, Bush AI: Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov. 2004, 3: 205-14.

    CAS  PubMed  Google Scholar 

  1023. Bondy SC, Lahiri DK, Perreau VM, Sharman KZ, Campbell A, Zhou J, Sharman EH: Retardation of brain aging by chronic treatment with melatonin. Ann N Y Acad Sci. 2004, 1035: 197-215.

    CAS  PubMed  Google Scholar 

  1024. Casadesus G, Smith MA, Zhu X, Aliev G, Cash AD, Honda K, Petersen RB, Perry G: Alzheimer disease: evidence for a central pathogenic role of iron-mediated reactive oxygen species. J Alzheimers Dis. 2004, 6: 165-9.

    CAS  PubMed  Google Scholar 

  1025. Doraiswamy PM, Finefrock AE: Metals in our minds: therapeutic implications for neurodegenerative disorders. Lancet Neurology. 2004, 3: 431-434.

    CAS  PubMed  Google Scholar 

  1026. Gotz ME, Double K, Gerlach M, Youdim MBH, Riederer P: The relevance of iron in the pathogenesis of Parkinson's disease. Redox-Active Metals in Neurological Disorders, Annals of the New York Academy of Sciences. 2004, 1012: 193-208.

    Google Scholar 

  1027. Huang X, Moir RD, Tanzi RE, Bush AI, Rogers JT: Redox-active metals, oxidative stress, and Alzheimer's disease pathology. Ann N Y Acad Sci. 2004, 1012: 153-63.

    CAS  PubMed  Google Scholar 

  1028. Kalivendi SV, Cunningham S, Kotamraju S, Joseph J, Hillard CJ, Kalyanaraman B: alpha-synuclein up-regulation and aggregation during MPP+-induced apoptosis in neuroblastoma cells – Intermediacy of transferrin receptor iron and hydrogen peroxide. Journal of Biological Chemistry. 2004, 279: 15240-15247.

    CAS  PubMed  Google Scholar 

  1029. Kaur D, Andersen J: Does cellular iron dysregulation play a causative role in Parkinson's disease?. Ageing Res Rev. 2004, 3: 327-343.

    CAS  PubMed  Google Scholar 

  1030. Mattson MP: Metal-catalyzed disruption of membrane protein and lipid signaling in the pathogenesis of neurodegenerative disorders. Ann N Y Acad Sci. 2004, 1012: 37-50.

    CAS  PubMed  Google Scholar 

  1031. Mattson MP: Pathways towards and away from Alzheimer's disease. Nature. 2004, 430: 631-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  1032. Milton NGN: Role of hydrogen peroxide in the aetiology of Alzheimer's disease – Implications for treatment. Drugs & Aging. 2004, 21: 81-100.

    CAS  Google Scholar 

  1033. Moos T, Morgan EH: The metabolism of neuronal iron and its pathogenic role in neurological disease – Review. Ann NY Acad Sci. 2004, 1012: 14-26.

    CAS  PubMed  Google Scholar 

  1034. Poon HF, Calabrese V, Scapagnini G, Butterfield DA: Free radicals and brain aging. Clin Geriatr Med. 2004, 20: 329-59.

    PubMed  Google Scholar 

  1035. Richardson DR: Novel chelators for central nervous system disorders that involve alterations in the metabolism of iron and other metal ions. Ann N Y Acad Sci. 2004, 1012: 326-41.

    CAS  PubMed  Google Scholar 

  1036. Warner DS, Sheng H, Batinić-Haberle I: Oxidants, antioxidants and the ischemic brain. J Exp Biol. 2004, 207: 3221-31.

    CAS  PubMed  Google Scholar 

  1037. Youdim MBH, Fridkin M, Zheng H: Novel bifunctional drugs targeting monoamine oxidase inhibition and iron chelation as an approach to neuroprotection in Parkinson's disease and other neurodegenerative diseases. J Neural Transmission. 2004, 111: 1455-1471.

    CAS  Google Scholar 

  1038. Zecca L, Youdim MBH, Riederer P, Connor JR, Crichton RR: Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci. 2004, 5: 863-873.

    CAS  PubMed  Google Scholar 

  1039. Zhu X, Raina AK, Lee HG, Casadesus G, Smith MA, Perry G: Oxidative stress signalling in Alzheimer's disease. Brain Res. 2004, 1000: 32-9.

    CAS  PubMed  Google Scholar 

  1040. Calabrese V, Lodi R, Tonon C, D'Agata V, Sapienza M, Scapagnini G, Mangiameli A, Pennisi G, Stella AMG, Butterfield DA: Oxidative stress, mitochondrial dysfunction and cellular stress response in Friedreich's ataxia. Journal of the Neurological Sciences. 2005, 233: 145-162.

    CAS  PubMed  Google Scholar 

  1041. Gaeta A, Hider RC: The crucial role of metal ions in neurodegeneration: the basis for a promising therapeutic strategy. Br J Pharmacol. 2005, 146: 1041-59.

    CAS  PubMed  PubMed Central  Google Scholar 

  1042. Hald A, Lotharius J: Oxidative stress and inflammation in Parkinson's disease: Is there a causal link?. Exp Neurol. 2005, 193: 279-290.

    CAS  PubMed  Google Scholar 

  1043. Honda K, Smith MA, Zhu X, Baus D, Merrick WC, Tartakoff AM, Hattier T, Harris PL, Siedlak SL, Fujioka H, Liu Q, Moreira PI, Miller FP, Nunomura A, Shimohama S, Perry G: Ribosomal RNA in Alzheimer disease is oxidized by bound redox-active iron. J Biol Chem. 2005, 280: 20978-86.

    CAS  PubMed  Google Scholar 

  1044. Ide-Ektessabi A, Rabionet M: The role of trace metallic elements in neurodegenerative disorders: quantitative analysis using XRF and XANES spectroscopy. Anal Sci. 2005, 21: 885-92.

    CAS  PubMed  Google Scholar 

  1045. Liu G, Garrett MR, Men P, Zhu XW, Perry G, Smith MA: Nanoparticle and other metal chelation therapeutics in Alzheimer disease. Biochimica Et Biophysica Acta-Molecular Basis of Disease. 2005, 1741: 246-252.

    CAS  Google Scholar 

  1046. Maynard CJ, Bush AI, Masters CL, Cappai R, Li QX: Metals and amyloid-beta in Alzheimer's disease. Int J Exp Pathol. 2005, 86: 147-159.

    CAS  PubMed  PubMed Central  Google Scholar 

  1047. Ong WY, Farooqui AA: Iron, neuroinflammation, and Alzheimer's disease. J Alzheimers Dis. 2005, 8: 183-200.

    CAS  PubMed  Google Scholar 

  1048. Adlard PA, Bush AI: Metals and Alzheimer's disease. J Alzheimers Dis. 2006, 10: 145-163.

    PubMed  Google Scholar 

  1049. Aracena P, Aguirre P, Munoz P, Nunez MT: Iron and glutathione at the crossroad of redox metabolism in neurons. Biol Res. 2006, 39: 157-65.

    CAS  PubMed  Google Scholar 

  1050. Berg D, Hochstrasser H: Iron metabolism in parkinsonian syndromes. Movement Disorders. 2006, 21: 1299-1310.

    PubMed  Google Scholar 

  1051. Collingwood J, Dobson J: Mapping and characterization of iron compounds in Alzheimer's tissue. J Alzheimers Dis. 2006, 10: 215-222.

    PubMed  Google Scholar 

  1052. Fasano M, Bergamasco B, Lopiano L: Modifications of the iron-neuromelanin system in Parkinson's disease. J Neurochem. 2006, 96: 909-916.

    CAS  PubMed  Google Scholar 

  1053. Gaggelli E, Kozlowski H, Valensin D, Valensin G: Copper homeostasis and neurodegenerative disorders (Alzheimer's, prion, and Parkinson's diseases and amyotrophic lateral sclerosis). Chem Rev. 2006, 106: 1995-2044.

    CAS  PubMed  Google Scholar 

  1054. Gerlach M, Double KL, Youdim MBH, Riederer P: Potential sources of increased iron in the substantia nigra of parkinsonian patients. J Neural Transmission – Suppl. 2006, 133-142.

    Google Scholar 

  1055. Halliwell B: Oxidative stress and neurodegeneration: where are we now?. Journal of Neurochemistry. 2006, 97: 1634-1658.

    CAS  PubMed  Google Scholar 

  1056. Huber A, Stuchbury G, Burkle A, Burnell J, Munch G: Neuroprotective therapies for Alzheimer's disease. Curr Pharmaceut Des. 2006, 12: 705-717.

    CAS  Google Scholar 

  1057. Liu GJ, Huang WD, Moir RD, Vanderburg CR, Lai B, Peng ZC, Tanzi RE, Rogers JT, Huang XD: Metal exposure and Alzheimer's pathogenesis. J Struct Biol. 2006, 155: 45-51.

    CAS  PubMed  Google Scholar 

  1058. Lee DW, Andersen JK, Kaur D: Iron dysregulation and neurodegeneration – The molecular connection. Molecular Interventions. 2006, 6: 89-97.

    CAS  PubMed  Google Scholar 

  1059. Mancuso M, Coppede F, Migliore L, Siciliano G, Murri L: Mitochondrial dysfunction, oxidative stress and neurodegeneration. Journal of Alzheimers Disease. 2006, 10: 59-73.

    CAS  Google Scholar 

  1060. Mosley RL, Benner EJ, Kadiu I, Thomas M, Boska MD, Hasan K, Laurie C, Gendelman HE: Neuroinflammation, oxidative stress, and the pathogenesis of Parkinson's disease. Clinical Neuroscience Research. 2006, 6: 261-281.

    CAS  PubMed  PubMed Central  Google Scholar 

  1061. Quintana C, Bellefqih S, Laval JY, Guerquin-Kern JL, Wu TD, Avila J, Ferrer I, Arranz R, Patino C: Study of the localization of iron, ferritin, and hemosiderin in Alzheimer's disease hippocampus by analytical microscopy at the subcellular level. J Struct Biol. 2006, 153: 42-54.

    CAS  PubMed  Google Scholar 

  1062. Whitnall M, Richardson DR: Iron: a new target for pharmacological intervention in neurodegenerative diseases. Semin Pediatr Neurol. 2006, 13: 186-97.

    PubMed  Google Scholar 

  1063. Zhang HY: Same causes, same cures. Biochem Biophys Res Commun. 2006, 351: 578-581.

    CAS  PubMed  Google Scholar 

  1064. Bartzokis G, Tishler TA, Lu PH, Villablanca P, Altshuler LL, Carter M, Huang D, Edwards N, Mintz J: Brain ferritin iron may influence age- and gender-related risks of neurodegeneration. Neurobiol Aging. 2007, 28: 414-23.

    CAS  PubMed  Google Scholar 

  1065. Bondy SC, Sharman EH: Melatonin and the aging brain. Neurochem Int. 2007, 50: 571-80.

    CAS  PubMed  Google Scholar 

  1066. Cass WA, Grondin R, Andersen AH, Zhang Z, Hardy PA, Hussey-Andersen LK, Rayens WS, Gerhardt GA, Gash DM: Iron accumulation in the striatum predicts aging-related decline in motor function in rhesus monkeys. Neurobiol Aging. 2007, 28: 258-71.

    CAS  PubMed  Google Scholar 

  1067. Castellani RJ, Moreira PI, Liu G, Dobson J, Perry G, Smith MA, Zhu X: Iron: the Redox-active center of oxidative stress in Alzheimer disease. Neurochem Res. 2007, 32: 1640-5.

    CAS  PubMed  Google Scholar 

  1068. Crouch PJ, White AR, Bush AI: The modulation of metal bio-availability as a therapeutic strategy for the treatment of Alzheimer's disease. FEBS J. 2007, 274: 3775-83.

    CAS  PubMed  Google Scholar 

  1069. Donnelly PS, Xiao Z, Wedd AG: Copper and Alzheimer's disease. Curr Opin Chem Biol. 2007, 11: 128-33.

    CAS  PubMed  Google Scholar 

  1070. Gaasch JA, Lockman PR, Geldenhuys WJ, Allen DD, Schyf Van der CJ: Brain iron toxicity: differential responses of astrocytes, neurons, and endothelial cells. Neurochem Res. 2007, 32: 1196-208.

    CAS  PubMed  Google Scholar 

  1071. Ke Y, Qian ZM: Brain iron metabolism: neurobiology and neurochemistry. Prog Neurobiol. 2007, 83: 149-73.

    CAS  PubMed  Google Scholar 

  1072. Mancuso C, Scapagnini G, Curro D, Stella AMG, De Marco C, Butterfield DA, Calabrese V: Mitochondrial dysfunction, free radical generation and cellular stress response in neurodegenerative disorders. Frontiers in Bioscience. 2007, 12: 1107-1123.

    CAS  PubMed  Google Scholar 

  1073. Mandel S, Amit T, Bar-Am O, Youdim MB: Iron dysregulation in Alzheimer's disease: multimodal brain permeable iron chelating drugs, possessing neuroprotective-neurorescue and amyloid precursor protein-processing regulatory activities as therapeutic agents. Prog Neurobiol. 2007, 82: 348-60.

    CAS  PubMed  Google Scholar 

  1074. Molina-Holgado F, Hider RC, Gaeta A, Williams R, Francis P: Metals ions and neurodegeneration. Biometals. 2007, 20: 639-54.

    CAS  PubMed  Google Scholar 

  1075. Petersen RB, Nunomura A, Lee HG, Casadesus G, Perry G, Smith MA, Zhu X: Signal transduction cascades associated with oxidative stress in Alzheimer's disease. J Alzheimers Dis. 2007, 11: 143-52.

    CAS  PubMed  Google Scholar 

  1076. Reynolds A, Laurie C, Mosley RL, Gendelman HE: Oxidative stress and the pathogenesis of neurodegenerative disorders. Int Rev Neurobiol. 2007, 82: 297-325.

    CAS  PubMed  Google Scholar 

  1077. Singh C, Ahmad I, Kumar A: Pesticides and metals induced Parkinson's disease: involvement of free radicals and oxidative stress. Cell Mol Biol (Noisy-le-grand). 2007, 53: 19-28.

    CAS  Google Scholar 

  1078. Stankiewicz J, Panter SS, Neema M, Arora A, Batt CE, Bakshi R: Iron in chronic brain disorders: imaging and neurotherapeutic implications. Neurotherapeutics. 2007, 4: 371-86.

    CAS  PubMed  PubMed Central  Google Scholar 

  1079. Zhu X, Su B, Wang X, Smith MA, Perry G: Causes of oxidative stress in Alzheimer disease. Cell Mol Life Sci. 2007, 64: 2202-10.

    CAS  PubMed  Google Scholar 

  1080. Amit T, Avramovich-Tirosh Y, Youdim MB, Mandel S: Targeting multiple Alzheimer's disease etiologies with multimodal neuroprotective and neurorestorative iron chelators. FASEB J. 2008, 22: 1296-305.

    CAS  PubMed  Google Scholar 

  1081. Bush AI, Curtain CC: Twenty years of metallo-neurobiology: where to now?. Eur Biophys J. 2008, 37: 241-5.

    CAS  PubMed  Google Scholar 

  1082. Drechsel DA, Patel M: Role of reactive oxygen species in the neurotoxicity of environmental agents implicated in Parkinson's disease. Free Radic Biol Med. 2008, 44: 1873-86.

    CAS  PubMed  PubMed Central  Google Scholar 

  1083. Pankhurst Q, Hautot D, Khan N, Dobson J: Increased levels of magnetic iron compounds in Alzheimer's disease. J Alzheimers Dis. 2008, 13: 49-52.

    CAS  PubMed  Google Scholar 

  1084. Rogers JT, Bush AI, Cho HH, Smith DH, Thomson AM, Friedlich AL, Lahiri DK, Leedman PJ, Huang X, Cahill CM: Iron and the translation of the amyloid precursor protein (APP) and ferritin mRNAs: riboregulation against neural oxidative damage in Alzheimer's disease. Biochem Soc Trans. 2008, 36: 1282-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  1085. Silvestri L, Camaschella C: A potential pathogenetic role of iron in Alzheimer's Disease. J Cell Mol Med. 2008, 12: 1548-1550.

    CAS  PubMed  PubMed Central  Google Scholar 

  1086. Snyder AM, Connor JR: Iron, the substantia nigra and related neurological disorders. Biochim Biophys Acta. 2008.

    Google Scholar 

  1087. Szczerbowska-Boruchowska M: X-ray fluorescence spectrometry, an analytical tool in neurochemical research. X-Ray Spectrometry. 2008, 37: 21-31.

    CAS  Google Scholar 

  1088. Sayre LM, Perry G, Harris PLR, Liu YH, Schubert KA, Smith MA: In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer's disease: A central role for bound transition metals. Journal of Neurochemistry. 2000, 74: 270-279.

    CAS  PubMed  Google Scholar 

  1089. Vélez-Pardo C, Del Río MJ, Verschueren H, Ebinger G, Vauquelin G: Dopamine and iron induce apoptosis in PC12 cells. Pharmacol Toxicol. 1997, 80: 76-84.

    PubMed  Google Scholar 

  1090. Kostoff RN, Briggs MB: Literature-Related Discovery (LRD): potential treatments for Parkinson's disease. Technol. Forecast. Soc. Change. 2007

    Google Scholar 

  1091. LeVine SM, Chakrabarty A: The role of iron in the pathogenesis of experimental allergic encephalomyelitis and multiple sclerosis. Redox-Active Metals in Neurological Disorders, Annals of the New York Academy of Sciences. 2004, 1012: 252-266.

    CAS  Google Scholar 

  1092. Kotze MJ, de Villiers JNP, Rooney RN, Grobbelaar JJ, Mansvelt EPG, Bouwens CSH, Carr J, Stander I, du Plessis L: Analysis of the NRAMP1 gene implicated in iron transport: Association with multiple sclerosis and age effects. Blood Cells Molecules and Diseases. 2001, 27: 44-53.

    CAS  Google Scholar 

  1093. LeVine SM, Lynch SG, Ou CN, Wulser MJ, Tam E, Boo N: Ferritin, transferrin and iron concentrations in the cerebrospinal fluid of multiple sclerosis patients. Brain Research. 1999, 821: 511-515.

    CAS  PubMed  Google Scholar 

  1094. Valberg LS, Flanagan PR, Kertesz A, Ebers GC: Abnormalities in Iron-Metabolism in Multiple-Sclerosis. Canadian Journal of Neurological Sciences. 1989, 16: 184-186.

    CAS  PubMed  Google Scholar 

  1095. Adams CWM: Perivascular Iron Deposition and Other Vascular Damage in Multiple-Sclerosis. Journal of Neurology Neurosurgery and Psychiatry. 1988, 51: 260-265.

    CAS  Google Scholar 

  1096. Drayer B, Burger P, Hurwitz B, Dawson D, Cain J: Reduced Signal Intensity on Mr Images of Thalamus and Putamen in Multiple-Sclerosis – Increased Iron Content. American Journal of Neuroradiology. 1987, 8: 413-419.

    Google Scholar 

  1097. Craelius W, Migdal MW, Luessenhop CP, Sugar A, Mihalakis I: Iron Deposits Surrounding Multiple-Sclerosis Plaques. Archives of Pathology & Laboratory Medicine. 1982, 106: 397-399.

    CAS  Google Scholar 

  1098. Ge Y, Jensen JH, Lu H, Helpern JA, Miles L, Inglese M, Babb JS, Herbert J, Grossman RI: Quantitative assessment of iron accumulation in the deep gray matter of multiple sclerosis by magnetic field correlation imaging. AJNR Am J Neuroradiol. 2007, 28: 1639-44.

    CAS  PubMed  Google Scholar 

  1099. Kim NH, Choi JK, Jeong BH, Kim JI, Kwon MS, Carp RI, Kim YS: Effect of transition metals (Mn, Cu, Fe) and deoxycholic acid (DA) on the conversion of PrPC to PrPres. FASEB J. 2005, 19: 783-5.

    CAS  PubMed  Google Scholar 

  1100. Basu S, Mohan ML, Luo X, Kundu B, Kong Q, Singh N: Modulation of proteinase K-resistant prion protein in cells and infectious brain homogenate by redox iron: implications for prion replication and disease pathogenesis. Mol Biol Cell. 2007, 18: 3302-12.

    CAS  PubMed  PubMed Central  Google Scholar 

  1101. Lill R, Muhlenhoff U: Maturation of iron-sulfur proteins in eukaryotes: mechanisms, connected processes, and diseases. Annu Rev Biochem. 2008, 77: 669-700.

    CAS  PubMed  Google Scholar 

  1102. Cavadini P, O'Neill HA, Benada O, Isaya G: Assembly and iron-binding properties of human frataxin, the protein deficient in Friedreich ataxia. Hum Mol Genet. 2002, 11: 217-27.

    CAS  PubMed  Google Scholar 

  1103. Napier I, Ponka P, Richardson DR: Iron trafficking in the mitochondrion: novel pathways revealed by disease. Blood. 2005, 105: 1867-74.

    CAS  PubMed  Google Scholar 

  1104. O'Neill HA, Gakh O, Park S, Cui J, Mooney SM, Sampson M, Ferreira GC, Isaya G: Assembly of human frataxin is a mechanism for detoxifying redox-active iron. Biochemistry. 2005, 44: 537-45.

    PubMed  Google Scholar 

  1105. Gakh O, Park S, Liu G, Macomber L, Imlay JA, Ferreira GC, Isaya G: Mitochondrial iron detoxification is a primary function of frataxin that limits oxidative damage and preserves cell longevity. Human Molecular Genetics. 2006, 15: 467-479.

    CAS  PubMed  Google Scholar 

  1106. Napoli E, Taroni F, Cortopassi GA: Frataxin, iron-sulfur clusters, heme, ROS, and aging. Antioxid Redox Signal. 2006, 8: 506-16.

    CAS  PubMed  PubMed Central  Google Scholar 

  1107. Babady NE, Carelle N, Wells RD, Rouault TA, Hirano M, Lynch DR, Delatycki MB, Wilson RB, Isaya G, Puccio H: Advancements in the pathophysiology of Friedreich's Ataxia and new prospects for treatments. Mol Genet Metab. 2007, 92: 23-35.

    CAS  PubMed  PubMed Central  Google Scholar 

  1108. Lu C, Cortopassi G: Frataxin knockdown causes loss of cytoplasmic iron-sulfur cluster functions, redox alterations and induction of heme transcripts. Arch Biochem Biophys. 2007, 457: 111-22.

    CAS  PubMed  Google Scholar 

  1109. Zanella I, Derosas M, Corrado M, Cocco E, Cavadini P, Biasiotto G, Poli M, Verardi R, Arosio P: The effects of frataxin silencing in HeLa cells are rescued by the expression of human mitochondrial ferritin. Biochim Biophys Acta. 2008, 1782: 90-8.

    CAS  PubMed  Google Scholar 

  1110. Anderson PR, Kirby K, Hilliker AJ, Phillips JP: RNAi-mediated suppression of the mitochondrial iron chaperone, frataxin, in Drosophila. Hum Mol Genet. 2005, 14: 3397-405.

    CAS  PubMed  Google Scholar 

  1111. Jo WJ, Loguinov A, Chang M, Wintz H, Nislow C, Arkin AP, Giaever G, Vulpe CD: Identification of genes involved in the toxic response of Saccharomyces cerevisiae against iron and copper overload by parallel analysis of deletion mutants. Toxicol Sci. 2008, 101: 140-51.

    CAS  PubMed  Google Scholar 

  1112. Gibson TJ, Koonin EV, Musco G, Pastore A, Bork P: Friedreich's ataxia protein: phylogenetic evidence for mitochondrial dysfunction. Trends Neurosci. 1996, 19: 465-8.

    CAS  PubMed  Google Scholar 

  1113. Koutnikova H, Campuzano V, Foury F, Dolle P, Cazzalini O, Koenig M: Studies of human, mouse and yeast homologues indicate a mitochondrial function for frataxin. Nat Genet. 1997, 16: 345-51.

    CAS  PubMed  Google Scholar 

  1114. Foury F: Human genetic diseases: A cross-talk between man and yeast. Gene. 1997, 195: 1-10.

    CAS  PubMed  Google Scholar 

  1115. Foury F, Cazzalini O: Deletion of the yeast homologue of the human gene associated with Friedreich's ataxia elicits iron accumulation in mitochondria. FEBS Lett. 1997, 411: 373-377.

    CAS  PubMed  Google Scholar 

  1116. Cavadini P, Gellera C, Patel PI, Isaya G: Human frataxin maintains mitochondrial iron homeostasis in Saccharomyces cerevisiae. Hum Mol Genet. 2000, 9: 2523-30.

    CAS  PubMed  Google Scholar 

  1117. Becker E, Richardson DR: Frataxin: its role in iron metabolism and the pathogenesis of Friedreich's ataxia. Int J Biochem Cell Biol. 2001, 33: 1-10.

    CAS  PubMed  Google Scholar 

  1118. Foury F, Talibi D: Mitochondrial control of iron homeostasis. A genome wide analysis of gene expression in a yeast frataxin-deficient strain. J Biol Chem. 2001, 276: 7762-8.

    CAS  PubMed  Google Scholar 

  1119. De Freitas J, Wintz H, Kim JH, Poynton H, Fox T, Vulpe C: Yeast, a model organism for iron and copper metabolism studies. Biometals. 2003, 16: 185-197.

    CAS  PubMed  Google Scholar 

  1120. González-Cabo P, Vázquez-Manrique RP, García-Gimeno MA, Sanz P, Palau F: Frataxin interacts functionally with mitochondrial electron transport chain proteins. Hum Mol Genet. 2005, 14: 2091-8.

    PubMed  Google Scholar 

  1121. Irazusta V, Cabiscol E, Reverter-Branchat G, Ros J, Tamarit J: Manganese is the link between frataxin and iron-sulfur deficiency in the yeast model of Friedreich ataxia. J Biol Chem. 2006, 281: 12227-32.

    CAS  PubMed  Google Scholar 

  1122. Vázquez-Manrique RP, González-Cabo P, Ros S, Aziz H, Baylis HA, Palau F: Reduction of Caenorhabditis elegans frataxin increases sensitivity to oxidative stress, reduces lifespan, and causes lethality in a mitochondrial complex II mutant. Faseb J. 2006, 20: 172-4.

    PubMed  Google Scholar 

  1123. Irazusta V, Moreno-Cermeno A, Cabiscol E, Ros J, Tamarit J: Major targets of iron-induced protein oxidative damage in frataxin-deficient yeasts are magnesium-binding proteins. Free Radic Biol Med. 2008

    Google Scholar 

  1124. Campuzano V, Montermini L, Molto MD, Pianese L, Cossee M, Cavalcanti F, Monros E, Rodius F, Duclos F, Monticelli A, Zara F, Canizares J, Koutnikova H, Bidichandani SI, Gellera C, Brice A, Trouillas P, De Michele G, Filla A, De Frutos R, Palau F, Patel PI, Di Donato S, Mandel JL, Cocozza S, Koenig M, Pandolfo M: Friedreich's ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science. 1996, 271: 1423-7.

    CAS  PubMed  Google Scholar 

  1125. Palau F: Friedreich's ataxia and frataxin: molecular genetics, evolution and pathogenesis (Review). Int J Mol Med. 2001, 7: 581-9.

    CAS  PubMed  Google Scholar 

  1126. Patel PI, Isaya G: Friedreich ataxia: from GAA triplet-repeat expansion to frataxin deficiency. Am J Hum Genet. 2001, 69: 15-24.

    CAS  PubMed  PubMed Central  Google Scholar 

  1127. Wilson RB: Frataxin and frataxin deficiency in Friedreich's ataxia. J Neurol Sci. 2003, 207: 103-5.

    PubMed  Google Scholar 

  1128. Michael S, Petrocine SV, Qian J, Lamarche JB, Knutson MD, Garrick MD, Koeppen AH: Iron and iron-responsive proteins in the cardiomyopathy of Friedreich's ataxia. Cerebellum. 2006, 5: 257-67.

    CAS  PubMed  Google Scholar 

  1129. Rötig A, Sidi D, Munnich A, Rustin P: Molecular insights into Friedreich's ataxia and antioxidant-based therapies. Trends Mol Med. 2002, 8: 221-4.

    PubMed  Google Scholar 

  1130. Boddaert N, Le Quan Sang KH, Rotig A, Leroy-Willig A, Gallet S, Brunelle F, Sidi D, Thalabard JC, Munnich A, Cabantchik ZI: Selective iron chelation in Friedreich ataxia: biologic and clinical implications. Blood. 2007, 110: 401-8.

    CAS  PubMed  Google Scholar 

  1131. Sohn YS, Breuer W, Munnich A, Cabantchik ZI: Redistribution of accumulated cell iron: a modality of chelation with therapeutic implications. Blood. 2008, 111: 1690-9.

    CAS  PubMed  Google Scholar 

  1132. Besarab A, Bolton WK, Browne JK, Egrie JC, Nissenson AR, Okamoto DM, Schwab SJ, Goodkin DA: The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. N Engl J Med. 1998, 339: 584-90.

    CAS  PubMed  Google Scholar 

  1133. Campuzano V, Montermini L, Lutz Y, Cova L, Hindelang C, Jiralerspong S, Trottier Y, Kish SJ, Faucheux B, Trouillas P, Authier FJ, Durr A, Mandel JL, Vescovi A, Pandolfo M, Koenig M: Frataxin is reduced in Friedreich ataxia patients and is associated with mitochondrial membranes. Hum Mol Genet. 1997, 6: 1771-80.

    CAS  PubMed  Google Scholar 

  1134. Wong A, Yang J, Cavadini P, Gellera C, Lonnerdal B, Taroni F, Cortopassi G: The Friedreich's ataxia mutation confers cellular sensitivity to oxidant stress which is rescued by chelators of iron and calcium and inhibitors of apoptosis. Hum Mol Genet. 1999, 8: 425-30.

    CAS  PubMed  Google Scholar 

  1135. Park S, Gakh O, Mooney SM, Isaya G: The ferroxidase activity of yeast frataxin. J Biol Chem. 2002, 277: 38589-95.

    CAS  PubMed  PubMed Central  Google Scholar 

  1136. Sturm B, Bistrich U, Schranzhofer M, Sarsero JP, Rauen U, Scheiber-Mojdehkar B, de Groot H, Ioannou P, Petrat F: Friedreich's ataxia, no changes in mitochondrial labile iron in human lymphoblasts and fibroblasts: a decrease in antioxidative capacity?. J Biol Chem. 2005, 280: 6701-8.

    CAS  PubMed  Google Scholar 

  1137. Anderson PR, Kirby K, Orr WC, Hilliker AJ, Phillips JP: Hydrogen peroxide scavenging rescues frataxin deficiency in a Drosophila model of Friedreich's ataxia. Proc Natl Acad Sci USA. 2008, 105: 611-6.

    CAS  PubMed  PubMed Central  Google Scholar 

  1138. Wilson RB, Roof DM: Respiratory deficiency due to loss of mitochondrial DNA in yeast lacking the frataxin homologue. Nat Genet. 1997, 16: 352-7.

    CAS  PubMed  Google Scholar 

  1139. Seznec H, Simon D, Bouton C, Reutenauer L, Hertzog A, Golik P, Procaccio V, Patel M, Drapier JC, Koenig M, Puccio H: Friedreich ataxia: the oxidative stress paradox. Hum Mol Genet. 2005, 14: 463-74.

    CAS  PubMed  Google Scholar 

  1140. Visapää I, Fellman V, Vesa J, Dasvarma A, Hutton JL, Kumar V, Payne GS, Makarow M, Van Coster R, Taylor RW, Turnbull DM, Suomalainen A, Peltonen L: GRACILE syndrome, a lethal metabolic disorder with iron overload, is caused by a point mutation in BCS1L. Am J Hum Genet. 2002, 71: 863-76.

    PubMed  PubMed Central  Google Scholar 

  1141. Fellman V, Lemmela S, Sajantila A, Pihko H, Jarvela I: Screening of BCS1L mutations in severe neonatal disorders suspicious for mitochondrial cause. J Hum Genet. 2008, 53: 554-8.

    CAS  PubMed  Google Scholar 

  1142. McGeer PL, McGeer EG: Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve. 2002, 26: 459-70.

    CAS  PubMed  Google Scholar 

  1143. Garber K: The elusive ALS genes. Science. 2008, 319: 20.

    PubMed  Google Scholar 

  1144. Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O'Regan JP, Deng HX, et al: Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993, 362: 59-62.

    CAS  PubMed  Google Scholar 

  1145. Kruman II, Pedersen WA, Springer JE, Mattson MP: ALS-linked Cu/Zn-SOD mutation increases vulnerability of motor neurons to excitotoxicity by a mechanism involving increased oxidative stress and perturbed calcium homeostasis. Exp Neurol. 1999, 160: 28-39.

    CAS  PubMed  Google Scholar 

  1146. Bruijn LI, Miller TM, Cleveland DW: Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annu Rev Neurosci. 2004, 27: 723-49.

    CAS  PubMed  Google Scholar 

  1147. Kasarskis EJ, Tandon L, Lovell MA, Ehmann WD: Aluminum, calcium, and iron in the spinal cord of patients with sporadic amyotrophic lateral sclerosis using laser microprobe mass spectroscopy: a preliminary study. J Neurol Sci. 1995, 130: 203-8.

    CAS  PubMed  Google Scholar 

  1148. Carri MT, Ferri A, Cozzolino M, Calabrese L, Rotilio G: Neurodegeneration in amyotrophic lateral sclerosis: the role of oxidative stress and altered homeostasis of metals. Brain Res Bull. 2003, 61: 365-74.

    CAS  PubMed  Google Scholar 

  1149. Carri MT, Grignaschi G, Bendotti C: Targets in ALS: designing multidrug therapies. Trends Pharmacol Sci. 2006, 27: 267-73.

    CAS  PubMed  Google Scholar 

  1150. Hughes KA, Reynolds RM: Evolutionary and mechanistic theories of aging. Annual Review of Entomology. 2005, 50: 421-445.

    CAS  PubMed  Google Scholar 

  1151. Zacharski LR, Ornstein DL, Woloshin S, Schwartz LM: Association of age, sex, and race with body iron stores in adults: analysis of NHANES III data. Am Heart J. 2000, 140: 98-104.

    CAS  PubMed  Google Scholar 

  1152. Ritchie RF, Palomaki GE, Neveux LM, Navolotskaia O, Ledue TB, Craig WY: Reference distributions for serum iron and transferrin saturation: a comparison of a large cohort to the world's literature. J Clin Lab Anal. 2002, 16: 246-52.

    CAS  PubMed  Google Scholar 

  1153. Ritchie RF, Palomaki GE, Neveux LM, Navolotskaia O, Ledue TB, Craig WY: Reference distributions for serum iron and transferrin saturation: a practical, simple, and clinically relevant approach in a large cohort. J Clin Lab Anal. 2002, 16: 237-45.

    CAS  PubMed  Google Scholar 

  1154. Liu JM, Hankinson SE, Stampfer MJ, Rifai N, Willett WC, Ma J: Body iron stores and their determinants in healthy postmenopausal US women. Am J Clin Nutr. 2003, 78: 1160-7.

    CAS  PubMed  Google Scholar 

  1155. Doulias PT, Vlachou C, Boudouri C, Kanavaros P, Siamopoulos KC, Galaris D: Flow cytometric estimation of 'labile iron pool' in human white blood cells reveals a positive association with ageing. Free Radic Res. 2008, 42: 253-9.

    CAS  PubMed  Google Scholar 

  1156. Fleming DJ, Tucker KL, Jacques PF, Dallal GE, Wilson PW, Wood RJ: Dietary factors associated with the risk of high iron stores in the elderly Framingham Heart Study cohort. Am J Clin Nutr. 2002, 76: 1375-84.

    CAS  PubMed  Google Scholar 

  1157. Beard J: Dietary iron intakes and elevated iron stores in the elderly: is it time to abandon the set-point hypothesis of regulation of iron absorption?. Am J Clin Nutr. 2002, 76: 1189-90.

    CAS  PubMed  Google Scholar 

  1158. Cade JE, Moreton JA, O'Hara B, Greenwood DC, Moor J, Burley VJ, Kukalizch K, Bishop DT, Worwood M: Diet and genetic factors associated with iron status in middle-aged women. Am J Clin Nutr. 2005, 82: 813-20.

    CAS  PubMed  Google Scholar 

  1159. Guralnik JM, Eisenstaedt RS, Ferrucci L, Klein HG, Woodman RC: Prevalence of anemia in persons 65 years and older in the United States: evidence for a high rate of unexplained anemia. Blood. 2004, 104: 2263-8.

    CAS  PubMed  Google Scholar 

  1160. Price EA: Aging and erythropoiesis: current state of knowledge. Blood Cells Mol Dis. 2008, 41: 158-65.

    CAS  PubMed  Google Scholar 

  1161. Sharman EH, Bondy SC, Sharman KG, Lahiri D, Cotman CW, Perreau VM: Effects of melatonin and age on gene expression in mouse CNS using microarray analysis. Neurochemistry International. 2007, 50: 336-344.

    CAS  PubMed  Google Scholar 

  1162. Kirkwood TBL: Evolution of Aging. Nature. 1977, 270: 301-304.

    CAS  PubMed  Google Scholar 

  1163. Beckman KB, Ames BN: The free radical theory of aging matures. Physiol Rev. 1998, 78: 547-81.

    CAS  PubMed  Google Scholar 

  1164. Finkel T, Holbrook NJ: Oxidants, oxidative stress and the biology of ageing. Nature. 2000, 408: 239-47.

    CAS  PubMed  Google Scholar 

  1165. Sohal RS, Mockett RJ, Orr WC: Current issues concerning the role of oxidative stress in aging: a perspective. Results Probl Cell Differ. 2000, 29: 45-66.

    CAS  PubMed  Google Scholar 

  1166. Cutler RG, Rodriguez H: Critical reviews of oxidative stress and aging: advances in basic science, diagnostics and intervention. World Scientific, Singapore. 2002

    Google Scholar 

  1167. Herndon LA, Schmeissner PJ, Dudaronek JM, Brown PA, Listner KM, Sakano Y, Paupard MC, Hall DH, Driscoll M: Stochastic and genetic factors influence tissue-specific decline in ageing C. elegans. Nature. 2002, 419: 808-14.

    CAS  PubMed  Google Scholar 

  1168. Sohal RS, Mockett RJ, Orr WC: Mechanisms of aging: an appraisal of the oxidative stress hypothesis. Free Radic Biol Med. 2002, 33: 575-86.

    CAS  PubMed  Google Scholar 

  1169. Partridge L, Gems D: Mechanisms of ageing: public or private?. Nat Rev Genet. 2002, 3: 165-75.

    CAS  PubMed  Google Scholar 

  1170. Hekimi S, Guarente L: Genetics and the specificity of the aging process. Science. 2003, 299: 1351-4.

    CAS  PubMed  Google Scholar 

  1171. Krabbe KS, Pedersen M, Bruunsgaard H: Inflammatory mediators in the elderly. Exp Gerontol. 2004, 39: 687-99.

    CAS  PubMed  Google Scholar 

  1172. Longo VD, Finch CE: Evolutionary medicine: from dwarf model systems to healthy centenarians?. Science. 2003, 299: 1342-6.

    PubMed  Google Scholar 

  1173. McGeer PL, McGeer EG: Inflammation and the degenerative diseases of aging. Ann N Y Acad Sci. 2004, 1035: 104-16.

    CAS  PubMed  Google Scholar 

  1174. Afanas'ev IB: Free radical mechanisms of aging processes under physiological conditions. Biogerontology. 2005, 6: 283-90.

    PubMed  Google Scholar 

  1175. Kirkwood TB: Understanding the odd science of aging. Cell. 2005, 120: 437-47.

    CAS  PubMed  Google Scholar 

  1176. Tuppo EE, Arias HR: The role of inflammation in Alzheimer's disease. Int J Biochem Cell Biol. 2005, 37: 289-305.

    CAS  PubMed  Google Scholar 

  1177. Partridge L, Gems D: Beyond the evolutionary theory of ageing, from functional genomics to evo-gero. Trends Ecol Evol. 2006, 21: 334-40.

    PubMed  Google Scholar 

  1178. Salvioli S, Capri M, Valensin S, Tieri P, Monti D, Ottaviani E, Franceschi C: Inflamm-aging, cytokines and aging: state of the art, new hypotheses on the role of mitochondria and new perspectives from systems biology. Curr Pharm Des. 2006, 12: 3161-71.

    CAS  PubMed  Google Scholar 

  1179. Franco OH, Kirkwood TB, Powell JR, Catt M, Goodwin J, Ordovas JM, Ouderaa van der F: Ten commandments for the future of ageing research in the UK: a vision for action. BMC Geriatr. 2007, 7: 10.

    PubMed  PubMed Central  Google Scholar 

  1180. Kirkwood TB: A systematic look at an old problem. Nature. 2008, 451: 644-7.

    CAS  PubMed  Google Scholar 

  1181. Harman D: Aging: a theory based on free radical and radiation chemistry. J Gerontol. 1956, 11: 298-300.

    CAS  PubMed  Google Scholar 

  1182. Ames BN, Shigenaga MK, Hagen TM: Oxidants, antioxidants, and the degenerative diseases of aging. Proc Natl Acad Sci USA. 1993, 90: 7915-22.

    CAS  PubMed  PubMed Central  Google Scholar 

  1183. Martin GM, Austad SN, Johnson TE: Genetic analysis of ageing: role of oxidative damage and environmental stresses. Nat Genet. 1996, 13: 25-34.

    CAS  PubMed  Google Scholar 

  1184. Ishii N, Fujii M, Hartman PS, Tsuda M, Yasuda K, Senoo-Matsuda N, Yanase S, Ayusawa D, Suzuki K: A mutation in succinate dehydrogenase cytochrome b causes oxidative stress and ageing in nematodes. Nature. 1998, 394: 694-7.

    CAS  PubMed  Google Scholar 

  1185. Fukagawa NK: Aging: is oxidative stress a marker or is it causal?. Proc Soc Exp Biol Med. 1999, 222: 293-8.

    CAS  PubMed  Google Scholar 

  1186. Johnson FB, Sinclair DA, Guarente L: Molecular biology of aging. Cell. 1999, 96: 291-302.

    CAS  PubMed  Google Scholar 

  1187. Melov S, Ravenscroft J, Malik S, Gill MS, Walker DW, Clayton PE, Wallace DC, Malfroy B, Doctrow SR, Lithgow GJ: Extension of life-span with superoxide dismutase/catalase mimetics. Science. 2000, 289: 1567-9.

    CAS  PubMed  Google Scholar 

  1188. Hamilton ML, Van Remmen H, Drake JA, Yang H, Guo ZM, Kewitt K, Walter CA, Richardson A: Does oxidative damage to DNA increase with age?. Proc Natl Acad Sci USA. 2001, 98: 10469-74.

    CAS  PubMed  PubMed Central  Google Scholar 

  1189. von Zglinicki T, Bürkle A, Kirkwood TBL: Stress, DNA damage and ageing – an integrative approach. Exp Gerontol. 2001, 36: 1049-1062.

    CAS  PubMed  Google Scholar 

  1190. Barja G: Rate of generation of oxidative stress-related damage and animal longevity. Free Radical Biology and Medicine. 2002, 33: 1167-1172.

    CAS  PubMed  Google Scholar 

  1191. Bonilla E, Medina-Leendertz S, Diaz S: Extension of life span and stress resistance of Drosophila melanogaster by long-term supplementation with melatonin. Exp Gerontol. 2002, 37: 629-38.

    CAS  PubMed  Google Scholar 

  1192. Golden TR, Hinerfeld DA, Melov S: Oxidative stress and aging: beyond correlation. Aging Cell. 2002, 1: 117-23.

    CAS  PubMed  Google Scholar 

  1193. Hagen TM: Oxidative stress, redox imbalance, and the aging process. Antioxidants & Redox Signaling. 2003, 5: 503-506.

    CAS  Google Scholar 

  1194. Karanjawala ZE, Murphy N, Hinton DR, Hsieh CL, Lieber MR: Oxygen metabolism causes chromosome breaks and is associated with the neuronal apoptosis observed in DNA double-strand break repair mutants. Curr Biol. 2002, 12: 397-402.

    CAS  PubMed  Google Scholar 

  1195. Melov S: Animal models of oxidative stress, aging, and therapeutic antioxidant interventions. Int J Biochem Cell Biol. 2002, 34: 1395-400.

    CAS  PubMed  Google Scholar 

  1196. Barros MH, Bandy B, Tahara EB, Kowaltowski AJ: Higher respiratory activity decreases mitochondrial reactive oxygen release and increases life span in Saccharomyces cerevisiae. Journal of Biological Chemistry. 2004, 279: 49883-49888.

    CAS  PubMed  Google Scholar 

  1197. Gredilla R, Barja G: Minireview: the role of oxidative stress in relation to caloric restriction and longevity. Endocrinology. 2005, 146: 3713-7.

    CAS  PubMed  Google Scholar 

  1198. Blagosklonny MV: Aging and immortality: quasi-programmed senescence and its pharmacologic inhibition. Cell Cycle. 2006, 5: 2087-102.

    CAS  PubMed  Google Scholar 

  1199. Chung HY, Sung B, Jung KJ, Zou Y, Yu BP: The molecular inflammatory process in aging. Antioxid Redox Signal. 2006, 8: 572-81.

    CAS  PubMed  Google Scholar 

  1200. Sedensky MM, Morgan PG: Mitochondrial respiration and reactive oxygen species in C. elegans. Exp Gerontol. 2006, 41: 957-67.

    CAS  PubMed  Google Scholar 

  1201. Terman A, Brunk UT: Oxidative stress, accumulation of biological 'garbage', and aging. Antioxidants & Redox Signaling. 2006, 8: 197-204.

    CAS  Google Scholar 

  1202. Yu BP, Chung HY: Adaptive mechanisms to oxidative stress during aging. Mechanisms of Ageing and Development. 2006, 127: 436-443.

    CAS  PubMed  Google Scholar 

  1203. Chakravarti B, Chakravarti DN: Oxidative modification of proteins: Age-related changes. Gerontology. 2007, 53: 128-139.

    CAS  PubMed  Google Scholar 

  1204. Kim SK: Common aging pathways in worms, flies, mice and humans. J Exp Biol. 2007, 210: 1607-12.

    CAS  PubMed  Google Scholar 

  1205. Kregel KC, Zhang HJ: An integrated view of oxidative stress in aging: basic mechanisms, functional effects, and pathological considerations. Am J Physiol Regul Integr Comp Physiol. 2007, 292: R18-36.

    CAS  PubMed  Google Scholar 

  1206. Nijnik A, Woodbine L, Marchetti C, Dawson S, Lambe T, Liu C, Rodrigues NP, Crockford TL, Cabuy E, Vindigni A, Enver T, Bell JI, Slijepcevic P, Goodnow CC, Jeggo PA, Cornall RJ: DNA repair is limiting for haematopoietic stem cells during ageing. Nature. 2007, 447: 686-90.

    CAS  PubMed  Google Scholar 

  1207. Sohal RS, Weindruch R: Oxidative stress, caloric restriction, and aging. Science. 1996, 273: 59-63.

    CAS  PubMed  PubMed Central  Google Scholar 

  1208. Pardon MC: Stress and ageing interactions: a paradox in the context of shared etiological and physiopathological processes. Brain Res Rev. 2007, 54: 251-73.

    PubMed  Google Scholar 

  1209. Killilea DW, Atamna H, Liao C, Ames BN: Iron accumulation during cellular senescence in human fibroblasts in vitro. Antioxid Redox Signal. 2003, 5: 507-16.

    CAS  PubMed  PubMed Central  Google Scholar 

  1210. Hamerman D: Toward an understanding of frailty. Ann Int Med. 1999, 130: 945-950.

    CAS  PubMed  Google Scholar 

  1211. Docherty JR: Cardiovascular responses in aging – a review. Pharmacol Rev. 1990, 42: 103-125.

    CAS  PubMed  Google Scholar 

  1212. Dröge W, Schipper HM: Oxidative stress and aberrant signaling in aging and cognitive decline. Aging Cell. 2007, 6: 361-370.

    PubMed  PubMed Central  Google Scholar 

  1213. Massie HR, Aiello VR, Banziger V: Iron accumulation and lipid peroxidation in aging C57bl/6j mice. Exp Gerontol. 1983, 18: 277-285.

    CAS  PubMed  Google Scholar 

  1214. Ershler WB: Biological interactions of aging and anemia: A focus on cytokines. J Amer Geriatr Soc. 2003, 51: S18-S21.

    Google Scholar 

  1215. Massie HR, Aiello VR, Williams TR: Inhibition of iron absorption prolongs the lifespan of Drosophila. Mech Ageing Dev. 1993, 67: 227-237.

    CAS  PubMed  Google Scholar 

  1216. Rattan SIS: Aging, anti-aging, and hormesis. Mechanisms of Ageing and Development. 2004, 125: 285-289.

    CAS  PubMed  Google Scholar 

  1217. Kenyon C: The plasticity of aging: insights from long-lived mutants. Cell. 2005, 120: 449-60.

    CAS  PubMed  Google Scholar 

  1218. Lee CK, Klopp RG, Weindruch R, Prolla TA: Gene expression profile of aging and its retardation by caloric restriction. Science. 1999, 285: 1390-3.

    CAS  PubMed  Google Scholar 

  1219. Bordone L, Guarente L: Calorie restriction, SIRT1 and metabolism: understanding longevity. Nat Rev Mol Cell Biol. 2005, 6: 298-305.

    CAS  PubMed  Google Scholar 

  1220. Taguchi A, Wartschow LM, White MF: Brain IRS2 signaling coordinates life span and nutrient homeostasis. Science. 2007, 317: 369-72.

    CAS  PubMed  Google Scholar 

  1221. Hulbert AJ, Pamplona R, Buffenstein R, Buttemer WA: Life and death: metabolic rate, membrane composition, and life span of animals. Physiol Rev. 2007, 87: 1175-213.

    CAS  PubMed  Google Scholar 

  1222. Kennedy BK, Steffen KK, Kaeberlein M: Ruminations on dietary restriction and aging. Cell Mol Life Sci. 2007, 64: 1323-8.

    CAS  PubMed  Google Scholar 

  1223. Mair W, Dillin A: Aging and survival: the genetics of life span extension by dietary restriction. Annu Rev Biochem. 2008, 77: 727-54.

    CAS  PubMed  Google Scholar 

  1224. Piper MD, Partridge L: Dietary restriction in Drosophila: delayed aging or experimental artefact?. PLoS Genet. 2007, 3: e57.

    PubMed  PubMed Central  Google Scholar 

  1225. Weindruch R, Sohal RS: Caloric intake and aging. New England Journal of Medicine. 1997, 337: 986-994.

    CAS  PubMed  PubMed Central  Google Scholar 

  1226. Lass A, Sohal BH, Weindruch R, Forster MJ, Sohal RS: Caloric restriction prevents age-associated accrual of oxidative damage to mouse skeletal muscle mitochondria. Free Radical Biology and Medicine. 1998, 25: 1089-1097.

    CAS  PubMed  PubMed Central  Google Scholar 

  1227. Masoro EJ: Caloric restriction and aging: an update. Exp Gerontol. 2000, 35: 299-305.

    CAS  PubMed  Google Scholar 

  1228. Thrasivoulou C, Soubeyre V, Ridha H, Giuliani D, Giaroni C, Michael GJ, Saffrey MJ, Cowen T: Reactive oxygen species, dietary restriction and neurotrophic factors in age-related loss of myenteric neurons. Aging Cell. 2006, 5: 247-57.

    CAS  PubMed  Google Scholar 

  1229. Walz D: Thermodynamics of oxidation-reduction reactions and its application to bioenergetics. Biochim Biophys Acta. 1979, 505: 279-353.

    CAS  PubMed  Google Scholar 

  1230. Cook CI, Yu BP: Iron accumulation in aging: modulation by dietary restriction. Mechanisms of Ageing and Development. 1998, 102: 1-13.

    CAS  PubMed  Google Scholar 

  1231. Reverter-Branchat G, Cabiscol E, Tamarit J, Ros J: Oxidative damage to specific proteins in replicative and chronological-aged Saccharomyces cerevisiae – Common targets and prevention by calorie restriction. J Biol Chem. 2004, 279: 31983-31989.

    CAS  PubMed  Google Scholar 

  1232. Raffaghello L, Lee C, Safdie FM, Wei M, Madia F, Bianchi G, Longo VD: Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc Natl Acad Sci USA. 2008, 105: 8215-20.

    CAS  PubMed  PubMed Central  Google Scholar 

  1233. Anisimov VN: Effects of exogenous melatonin–a review. Toxicol Pathol. 2003, 31: 589-603.

    CAS  PubMed  Google Scholar 

  1234. Poeggeler B: Melatonin, aging, and age-related diseases: perspectives for prevention, intervention, and therapy. Endocrine. 2005, 27: 201-12.

    CAS  PubMed  Google Scholar 

  1235. Bonilla E, Medina-Leendertz S, Villalobos V, Molero L, Bohorquez A: Paraquat-induced oxidative stress in Drosophila melanogaster: Effects of melatonin, glutathione, serotonin, minocycline, lipoic acid and ascorbic acid. Neurochemical Research. 2006, 31: 1425-1432.

    CAS  PubMed  Google Scholar 

  1236. Salvemini D, Riley DP, Cuzzocrea S: SOD mimetics are coming of age. Nature Reviews Drug Discovery. 2002, 1: 367-374.

    CAS  PubMed  Google Scholar 

  1237. Lithgow GJ, Gill MS, Olsen A, Sampayo JN: Pharmacological intervention in invertebrate aging. Ageing Res Rev. 2005, 27: 213-223.

    CAS  Google Scholar 

  1238. Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS, Ahringer J, Li H, Kenyon C: Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature. 2003, 424: 277-83.

    CAS  PubMed  Google Scholar 

  1239. Dong MQ, Venable JD, Au N, Xu T, Park SK, Cociorva D, Johnson JR, Dillin A, Yates JR: Quantitative mass spectrometry identifies insulin signaling targets in C. elegans. Science. 2007, 317: 660-3.

    CAS  PubMed  Google Scholar 

  1240. Honda Y, Honda S: The daf-2 gene network for longevity regulates oxidative stress resistance and Mn-superoxide dismutase gene expression in Caenorhabditis elegans. FASEB J. 1999, 13: 1385-93.

    CAS  PubMed  Google Scholar 

  1241. Bauerova K, Bezek S: Role of reactive oxygen and nitrogen species in etiopathogenesis of rheumatoid arthritis. General Physiology and Biophysics. 1999, 18: 15-20.

    CAS  PubMed  Google Scholar 

  1242. Aruoma OI: Free radicals, oxidative stress, and antioxidants in human health and disease. J Amer Oil Chemists Soc. 1998, 75: 199-212.

    CAS  Google Scholar 

  1243. Mapp PI, Grootveld MC, Blake DR: Hypoxia, oxidative stress and rheumatoid arthritis. British Medical Bulletin. 1995, 51: 419-436.

    CAS  PubMed  Google Scholar 

  1244. Gracy RW, Talent JM, Kong Y, Conrad CC: Reactive oxygen species: the unavoidable environmental insult?. Mut Res. 1999, 428: 17-22.

    CAS  Google Scholar 

  1245. Henrotin YE, Bruckner P, Pujol JPL: The role of reactive oxygen species in homeostasis and degradation of cartilage. Osteoarthritis and Cartilage. 2003, 11: 747-755.

    CAS  PubMed  Google Scholar 

  1246. Griffiths HR: ROS as signalling molecules in T cells – evidence for abnormal redox signalling in the autoimmune disease, rheumatoid arthritis. Redox Report. 2005, 10: 273-280.

    CAS  PubMed  Google Scholar 

  1247. Weinstein IM: A Correlative Study of the Erythrokinetics and Disturbances in Iron Metabolism Associated with the Anemia of Rheumatoid Arthritis. Blood. 1959, 14: 950-966.

    CAS  PubMed  Google Scholar 

  1248. Bentley DP, Williams P: Serum ferritin concentration as an index of storage iron in rheumatoid arthritis. J Clin Pathol. 1974, 27: 786-788.

    CAS  PubMed  PubMed Central  Google Scholar 

  1249. Hansen TM, Hansen NE, Birgens HS, Holund B, Lorenzen I: Serum Ferritin and the Assessment of Iron-Deficiency in Rheumatoid-Arthritis. Scandinavian Journal of Rheumatology. 1983, 12: 353-359.

    CAS  PubMed  Google Scholar 

  1250. Weber J, Werre JM, Julius HW, Marx JJ: Decreased iron absorption in patients with active rheumatoid arthritis, with and without iron deficiency. Ann Rheum Dis. 1988, 47: 404-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  1251. Vreugdenhil G, Wognum AW, Vaneijk HG, Swaak AJG: Anemia in Rheumatoid-Arthritis – the Role of Iron, Vitamin-B12, and Folic-Acid Deficiency, and Erythropoietin Responsiveness. Annals of the Rheumatic Diseases. 1990, 49: 93-98.

    CAS  PubMed  PubMed Central  Google Scholar 

  1252. Muirden KD, Senator GB: Iron in synovial membrane in rheumatoid arthritis and other joint diseases. Ann Rheumat Dis. 1968, 27: 38.

    CAS  PubMed  PubMed Central  Google Scholar 

  1253. Rowley D, Gutteridge JMC, Blake D, Farr M, Halliwell B: Lipid peroxidation in rheumatoid arthritis – thiobarbituric acid-reactive material and catalytic iron salts in synovial fluid from rheumatoid patients. Clin Sci. 1984, 66: 691-695.

    CAS  PubMed  Google Scholar 

  1254. Biemond P, Swaak AJG, Vaneijk HG, Koster JF: Intraarticular ferritin-bound iron in rheumatoid arthritis – a factor that increases oxygen free radical-induced tissue destruction. Arthritis and Rheumatism. 1986, 29: 1187-1193.

    CAS  PubMed  Google Scholar 

  1255. Lipiński P, Drapier JC: Interplay between ferritin metabolism, reactive oxygen species and nitric oxide. J Biol Inorg Chem. 1997, 2: 559-566.

    Google Scholar 

  1256. Biemond P, Swaak AJG, Vaneijk HG, Koster JF: Superoxide Dependent Iron Release from Ferritin in Inflammatory Diseases. Free Radical Biology and Medicine. 1988, 4: 185-198.

    CAS  PubMed  Google Scholar 

  1257. Biemond P, Swaak AJG, Beindorff CM, Koster JF: Superoxide-Dependent and Superoxide-Independent Mechanisms of Iron Mobilization from Ferritin by Xanthine-Oxidase – Implications for Oxygen-Free-Radical-Induced Tissue Destruction During Ischemia and Inflammation. Biochemical Journal. 1986, 239: 169-173.

    CAS  PubMed  PubMed Central  Google Scholar 

  1258. Biemond P, Swaak AJG, Penders JMA, Beindorff CM, Koster JF: Superoxide Production by Polymorphonuclear Leukocytes in Rheumatoid-Arthritis and Osteoarthritis – Invivo Inhibition by the Antirheumatic Drug Piroxicam Due to Interference with the Activation of the Nadph-Oxidase. Annals of the Rheumatic Diseases. 1986, 45: 249-255.

    CAS  PubMed  PubMed Central  Google Scholar 

  1259. Biemond P, Vaneijk HG, Swaak AJG, Koster JF: Iron Mobilization from Ferritin by Superoxide Derived from Stimulated Polymorphonuclear Leukocytes – Possible Mechanism in Inflammation Diseases. Journal of Clinical Investigation. 1984, 73: 1576-1579.

    CAS  PubMed  PubMed Central  Google Scholar 

  1260. Mladĕnka P, Šimůnek T, Hübl M, Hrdina R: The role of reactive oxygen and nitrogen species in cellular iron metabolism.". Free Radic Res. 2006, 40: 263-72.

    PubMed  Google Scholar 

  1261. Harris LR, Cake MH, Macey DJ: Iron release from ferritin and its sensitivity to superoxide ions differs among vertebrates. Biochem J. 1994, 301: 385-389.

    CAS  PubMed  PubMed Central  Google Scholar 

  1262. Fairburn K, Grootveld M, Ward RJ, Abiuka C, Kus M, Williams RB, Winyard PG, Blake DR: Alpha-Tocopherol, Lipids and Lipoproteins in Knee-Joint Synovial-Fluid and Serum from Patients with Inflammatory Joint Disease. Clinical Science. 1992, 83: 657-664.

    CAS  PubMed  Google Scholar 

  1263. Dombrecht EJ, Cos P, Berghe Vanden D, Van Offel JF, Schuerwegh AJ, Bridts CH, Stevens WJ, De Clerck LS: Selective in vitro antioxidant properties of bisphosphonates. Biochem Biophys Res Commun. 2004, 314: 675-80.

    CAS  PubMed  Google Scholar 

  1264. Ostrakhovitch EA, Afanas'ev IB: Oxidative stress in rheumatoid arthritis leukocytes: suppression by rutin and other antioxidants and chelators. Biochem Pharmacol. 2001, 62: 743-746.

    CAS  PubMed  Google Scholar 

  1265. Hazes JMW, Dijkmans BAC, Vandenbroucke JP, Devries RRP, Cats A: Pregnancy and the risk of developing rheumatoid arthritis. Arthritis and Rheumatism. 1990, 33: 1770-1775.

    CAS  PubMed  Google Scholar 

  1266. Østensen M, Husby G: A prospective clinical study of the effect of pregnancy on rheumatoid arthritis and ankylosing spondylitis. Arthritis and Rheumatism. 1983, 26: 1155-1159.

    PubMed  Google Scholar 

  1267. Da Silva JAP, Spector TD: The role of pregnancy in the course and etiology of rheumatoid arthritis. Clin Rheumat. 1992, 11: 189-194.

    CAS  Google Scholar 

  1268. O'Keeffe ST: Restless legs syndrome – A review. Archives of Internal Medicine. 1996, 156: 243-248.

    PubMed  Google Scholar 

  1269. Ondo W, Tan EK, Mansoor J: Rheumatologic serologies in secondary restless legs syndrome. Movement Disorders. 2000, 15: 321-323.

    CAS  PubMed  Google Scholar 

  1270. Trenkwalder C, Paulus W, Walters AS: The restless legs syndrome. Lancet Neurology. 2005, 4: 465-475.

    CAS  PubMed  Google Scholar 

  1271. Feelders RA, Kuiper-Kramer EPA, van Eijk HG: Structure, function and clinical significance of transferrin receptors. Clinical Chemistry and Laboratory Medicine. 1999, 37: 1-10.

    CAS  PubMed  Google Scholar 

  1272. Ahluwalia N: Diagnostic utility of serum transferrin receptors measurement in assessing iron status. Nutrition Reviews. 1998, 56: 133-141.

    CAS  PubMed  Google Scholar 

  1273. Frostegård J: SLE, atherosclerosis and cardiovascular disease. J Intern Med. 2005, 257: 485-95.

    PubMed  Google Scholar 

  1274. Mascitelli L, Pezzetta F: High iron stores and cardiovascular disease in systemic lupus erythematosus. J Intern Med. 2005, 258: 584.

    CAS  PubMed  Google Scholar 

  1275. Harvey AM, Shulman LE, Tumulty PA, Conley CL, Schoenrich EH: Systemic lupus erythematosus: review of the literature and clinical analysis of 138 cases. Medicine (Baltimore). 1954, 33: 291-437.

    CAS  Google Scholar 

  1276. Johnson RT, Richardson EP: The neurological manifestations of systemic lupus erythematosus. Medicine (Baltimore). 1968, 47: 337-69.

    CAS  Google Scholar 

  1277. Feng PH, Cheah PS, Lee YK: Mortality in systemic lupus erythematosus: a 10-year review. Br Med J. 1973, 4: 772-4.

    CAS  PubMed  PubMed Central  Google Scholar 

  1278. Matthay RA, Schwarz MI, Petty TL, Stanford RE, Gupta RC, Sahn SA, Steigerwald JC: Pulmonary manifestations of systemic lupus erythematosus: review of twelve cases of acute lupus pneumonitis. Medicine (Baltimore). 1975, 54: 397-409.

    CAS  Google Scholar 

  1279. O'Loughlin S, Schroeter AL, Jordon RE: Chronic urticaria-like lesions in systemic lupus erythematosus. A review of 12 cases. Arch Dermatol. 1978, 114: 879-83.

    PubMed  Google Scholar 

  1280. Hoffman BI, Katz WA: The gastrointestinal manifestations of systemic lupus erythematosus: a review of the literature. Semin Arthritis Rheum. 1980, 9: 237-47.

    CAS  PubMed  Google Scholar 

  1281. Reynolds JC, Inman RD, Kimberly RP, Chuong JH, Kovacs JE, Walsh MB: Acute pancreatitis in systemic lupus erythematosus: report of twenty cases and a review of the literature. Medicine (Baltimore). 1982, 61: 25-32.

    CAS  Google Scholar 

  1282. Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, Schaller JG, Talal N, Winchester RJ: The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1982, 25: 1271-7.

    CAS  PubMed  Google Scholar 

  1283. Levine SR, Joseph R, D'Andrea G, Welch KM: Migraine and the lupus anticoagulant. Case reports and review of the literature. Cephalalgia. 1987, 7: 93-9.

    CAS  PubMed  Google Scholar 

  1284. Crowson AN, Magro C: The cutaneous pathology of lupus erythematosus: a review. J Cutan Pathol. 2001, 28: 1-23.

    CAS  PubMed  Google Scholar 

  1285. Karrar A, Sequeira W, Block JA: Coronary artery disease in systemic lupus erythematosus: A review of the literature. Semin Arthritis Rheum. 2001, 30: 436-43.

    CAS  PubMed  Google Scholar 

  1286. Ad hoc committee on systemic lupus erythematosus response criteria for fatigue: Measurement of fatigue in systemic lupus erythematosus: a systematic review. Arthritis Rheum. 2007, 57: 1348-57.

    Google Scholar 

  1287. Zhang X, Jin M, Wu H, Nadasdy T, Nadasdy G, Harris N, Green-Church K, Nagaraja H, Birmingham DJ, Yu CY, Hebert LA, Rovin BH: Biomarkers of lupus nephritis determined by serial urine proteomics. Kidney Int. 2008, 74: 799-807.

    CAS  PubMed  PubMed Central  Google Scholar 

  1288. Lim MK, Lee CK, Ju YS, Cho YS, Lee MS, Yoo B, Moon HB: Serum ferritin as a serologic marker of activity in systemic lupus erythematosus. Rheumatol Int. 2001, 20: 89-93.

    CAS  PubMed  Google Scholar 

  1289. Abou-Raya A, el-Hallous D, Fayed H: 8-Isoprostaglandin F2 alpha: a potential index of lipid peroxidation in systemic lupus erythematosus. Clin Invest Med. 2004, 27: 306-11.

    CAS  PubMed  Google Scholar 

  1290. Cutolo M, Otsa K: Review: vitamin D, immunity and lupus. Lupus. 2008, 17: 6-10.

    CAS  PubMed  Google Scholar 

  1291. Li HF: Are statins analogues of vitamin D?. Lancet. 2006, 368: 1233-4.

    PubMed  Google Scholar 

  1292. Wu-Wong JR, Nakane M, Ma J, Ju TR: Statins do not directly activate vitamin D receptor. J Thromb Haemost. 2007, 5: 415-6.

    CAS  PubMed  Google Scholar 

  1293. Lawman S, Mauri C, Jury EC, Cook HT, Ehrenstein MR: Atorvastatin inhibits autoreactive B cell activation and delays lupus development in New Zealand black/white F1 mice. J Immunol. 2004, 173: 7641-6.

    CAS  PubMed  Google Scholar 

  1294. Jury EC, Isenberg DA, Mauri C, Ehrenstein MR: Atorvastatin restores Lck expression and lipid raft-associated signaling in T cells from patients with systemic lupus erythematosus. J Immunol. 2006, 177: 7416-22.

    CAS  PubMed  Google Scholar 

  1295. Swanson DR: Undiscovered public knowledge. Library Quarterly. 1986, 56: 103-118.

    Google Scholar 

  1296. Swanson DR, Smalheiser NR, Torvik VI: Ranking indirect connections in literature-based discovery: The role of medical subject headings. J Amer Soc Inf Sci Technol. 2006, 57: 1427-1439.

    CAS  Google Scholar 

  1297. Andreadis AA, Hazen SL, Comhair SAA, Erzurum SC: Oxidative and nitrosative events in asthma. Free Radical Biology and Medicine. 2003, 35: 213-225.

    CAS  PubMed  Google Scholar 

  1298. Greene LS: Asthma and oxidant stress – nutritional, environmental, and genetic risk factors. J Amer Coll Nutrition. 1995, 14: 317-324.

    CAS  Google Scholar 

  1299. Han JY, Takeshita K, Utsumi H: Noninvasive detection of hydroxyl radical generation in lung by diesel exhaust particles. Free Radic Biol Med. 2001, 30: 516-25.

    CAS  PubMed  Google Scholar 

  1300. Kocyigit A, Armutcu F, Gurel A, Ermis B: Alterations in plasma essential trace elements selenium, manganese, zinc, copper, and iron concentrations and the possible role of these elements on oxidative status in patients with childhood asthma. Biological Trace Element Research. 2004, 97: 31-41.

    CAS  PubMed  Google Scholar 

  1301. Lih-Brody L, Powell SR, Collier KP, Reddy GM, Cerchia R, Kahn E, Weissman GS, Katz S, Floyd RA, McKinley MJ, Fisher SE, Mullin GE: Increased oxidative stress and decreased antioxidant defenses in mucosa of inflammatory bowel disease. Dig Dis Sci. 1996, 41: 2078-86.

    CAS  PubMed  Google Scholar 

  1302. Kruidenier L, Verspaget HW: Review article: oxidative stress as a pathogenic factor in inflammatory bowel disease–radicals or ridiculous?. Aliment Pharmacol Ther. 2002, 16: 1997-2015.

    CAS  PubMed  Google Scholar 

  1303. Kruidenier L, Kuiper I, Lamers CB, Verspaget HW: Intestinal oxidative damage in inflammatory bowel disease: semi-quantification, localization, and association with mucosal antioxidants. J Pathol. 2003, 201: 28-36.

    CAS  PubMed  Google Scholar 

  1304. Martin AR, Villegas I, La Casa C, de la Lastra CA: Resveratrol, a polyphenol found in grapes, suppresses oxidative damage and stimulates apoptosis during early colonic inflammation in rats. Biochem Pharmacol. 2004, 67: 1399-410.

    CAS  PubMed  Google Scholar 

  1305. Seril DN, Liao J, Yang GY, Yang CS: Oxidative stress and ulcerative colitis-associated carcinogenesis: studies in humans and animal models. Carcinogenesis. 2003, 24: 353-62.

    CAS  PubMed  Google Scholar 

  1306. Gisbert JP, Gomollón F: Common misconceptions in the diagnosis and management of anemia in inflammatory bowel disease. Am J Gastroenterol. 2008, 103: 1299-307.

    PubMed  Google Scholar 

  1307. Semrin G, Fishman DS, Bousvaros A, Zholudev A, Saunders AC, Correia CE, Nemeth E, Grand RJ, Weinstein DA: Impaired intestinal iron absorption in Crohn's disease correlates with disease activity and markers of inflammation. Inflamm Bowel Dis. 2006, 12: 1101-6.

    PubMed  PubMed Central  Google Scholar 

  1308. Zaahl MG, Winter TA, Warnich L, Kotze MJ: The -237C-->T promoter polymorphism of the SLC11A1 gene is associated with a protective effect in relation to inflammatory bowel disease in the South African population. Int J Colorectal Dis. 2006, 21: 402-8.

    PubMed  Google Scholar 

  1309. Bartels U, Pedersen NS, Jarnum S: Iron absorption and serum ferritin in chronic inflammatory bowel disease. Scand J Gastroenterol. 1978, 13: 649-56.

    CAS  PubMed  Google Scholar 

  1310. Thomson AB, Brust R, Ali MA, Mant MJ, Valberg LS: Iron deficiency in inflammatory bowel disease. Diagnostic efficacy of serum ferritin. Am J Dig Dis. 1978, 23: 705-9.

    CAS  PubMed  Google Scholar 

  1311. Oldenburg B, Koningsberger JC, Van Berge Henegouwen GP, Van Asbeck BS, Marx JJ: Iron and inflammatory bowel disease. Aliment Pharmacol Ther. 2001, 15: 429-38.

    CAS  PubMed  Google Scholar 

  1312. Gasche C, Lomer MC, Cavill I, Weiss G: Iron, anaemia, and inflammatory bowel diseases. Gut. 2004, 53: 1190-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  1313. de Silva AD, Tsironi E, Feakins RM, Rampton DS: Efficacy and tolerability of oral iron therapy in inflammatory bowel disease: a prospective, comparative trial. Aliment Pharmacol Ther. 2005, 22: 1097-105.

    CAS  PubMed  Google Scholar 

  1314. Guagnozzi D, Severi C, Ialongo P, Viscido A, Patrizi F, Testino G, Vannella L, Labriola R, Strom R, Caprilli R: Ferritin as a simple indicator of iron deficiency in anemic IBD patients. Inflamm Bowel Dis. 2006, 12: 150-1.

    PubMed  Google Scholar 

  1315. Gasche C, Berstad A, Befrits R, Beglinger C, Dignass A, Erichsen K, Gomollon F, Hjortswang H, Koutroubakis I, Kulnigg S, Oldenburg B, Rampton D, Schroeder O, Stein J, Travis S, Van Assche G: Guidelines on the diagnosis and management of iron deficiency and anemia in inflammatory bowel diseases. Inflamm Bowel Dis. 2007, 13: 1545-53.

    PubMed  Google Scholar 

  1316. Rattner A, Nathans J: Macular degeneration: recent advances and therapeutic opportunities. Nat Rev Neurosci. 2006, 7: 860-72.

    CAS  PubMed  Google Scholar 

  1317. Tielsch JM, Javitt JC, Coleman A, Katz J, Sommer A: The prevalence of blindness and visual impairment among nursing home residents in Baltimore. N Engl J Med. 1995, 332: 1205-9.

    CAS  PubMed  Google Scholar 

  1318. Harvey PT: Common eye diseases of elderly people: identifying and treating causes of vision loss. Gerontology. 2003, 49: 1-11.

    PubMed  Google Scholar 

  1319. Fine SL: Age-related macular degeneration 1969–2004: a 35-year personal perspective. Am J Ophthalmol. 2005, 139: 405-20.

    PubMed  Google Scholar 

  1320. Lotery A, Trump D: Progress in defining the molecular biology of age related macular degeneration. Hum Genet. 2007, 122: 219-36.

    PubMed  Google Scholar 

  1321. Anderson DH, Mullins RF, Hageman GS, Johnson LV: A role for local inflammation in the formation of drusen in the aging eye. Am J Ophthalmol. 2002, 134: 411-31.

    CAS  PubMed  Google Scholar 

  1322. Hahn P, Ying GS, Beard J, Dunaief JL: Iron levels in human retina: sex difference and increase with age. Neuroreport. 2006, 17: 1803-6.

    CAS  PubMed  Google Scholar 

  1323. Cai J, Nelson KC, Wu M, Sternberg P, Jones DP: Oxidative damage and protection of the RPE. Prog Retin Eye Res. 2000, 19: 205-21.

    CAS  PubMed  Google Scholar 

  1324. Richer S, Rudy D, Statkute L, Karofty K, Frankowski J: Serum iron, transferrin saturation, ferritin, and dietary data in age-related macular degeneration. Am J Ther. 2002, 9: 25-8.

    PubMed  Google Scholar 

  1325. Hahn P, Milam AH, Dunaief JL: Maculas affected by age-related macular degeneration contain increased chelatable iron in the retinal pigment epithelium and Bruch's membrane. Arch Ophthalmol. 2003, 121: 1099-105.

    PubMed  Google Scholar 

  1326. Hahn P, Qian Y, Dentchev T, Chen L, Beard J, Harris ZL, Dunaief JL: Disruption of ceruloplasmin and hephaestin in mice causes retinal iron overload and retinal degeneration with features of age-related macular degeneration. Proc Natl Acad Sci USA. 2004, 101: 13850-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  1327. Dunaief JL: Iron induced oxidative damage as a potential factor in age-related macular degeneration: The Cogan Lecture. Invest Ophthalmol Vis Sci. 2006, 47: 4660-4664.

    PubMed  Google Scholar 

  1328. He X, Hahn P, Iacovelli J, Wong R, King C, Bhisitkul R, Massaro-Giordano M, Dunaief JL: Iron homeostasis and toxicity in retinal degeneration. Prog Retin Eye Res. 2007, 26: 649-73.

    CAS  PubMed  PubMed Central  Google Scholar 

  1329. Wong RW, Richa DC, Hahn P, Green WR, Dunaief JL: Iron toxicity as a potential factor in AMD. Retina. 2007, 27: 997-1003.

    PubMed  Google Scholar 

  1330. Voloboueva LA, Killilea DW, Atamna H, Ames BN: N-tert-butyl hydroxylamine, a mitochondrial antioxidant, protects human retinal pigment epithelial cells from iron overload: relevance to macular degeneration. FASEB J. 2007, 21: 4077-86.

    CAS  PubMed  PubMed Central  Google Scholar 

  1331. Tan JS, Wang JJ, Flood V, Rochtchina E, Smith W, Mitchell P: Dietary antioxidants and the long-term incidence of age-related macular degeneration: the Blue Mountains Eye Study. Ophthalmology. 2008, 115: 334-41.

    PubMed  Google Scholar 

  1332. Wilson DS, Szostak JW: In vitro selection of functional nucleic acids. Annu Rev Biochem. 1999, 68: 611-647.

    CAS  PubMed  Google Scholar 

  1333. Nimjee SM, Rusconi CP, Sullenger BA: Aptamers: an emerging class of therapeutics. Annu Rev Med. 2005, 56: 555-83.

    CAS  PubMed  Google Scholar 

  1334. Famulok M, Hartig JS, Mayer G: Functional aptamers and aptazymes in biotechnology, diagnostics, and therapy. Chem Rev. 2007, 107: 3715-43.

    CAS  PubMed  Google Scholar 

  1335. Chapman JA, Beckey C: Pegaptanib: a novel approach to ocular neovascularization. Ann Pharmacother. 2006, 40: 1322-6.

    CAS  PubMed  Google Scholar 

  1336. Kourlas H, Schiller DS: Pegaptanib sodium for the treatment of neovascular age-related macular degeneration: a review. Clin Ther. 2006, 28: 36-44.

    CAS  PubMed  Google Scholar 

  1337. Zhou B, Wang B: Pegaptanib for the treatment of age-related macular degeneration. Exp Eye Res. 2006, 83: 615-9.

    CAS  PubMed  Google Scholar 

  1338. Virgili G, Do DV, Bressler NM, Menchini U: New therapies for neovascular age-related macular degeneration: critical appraisal of the current evidence. Acta Ophthalmol Scand. 2007, 85: 6-20.

    CAS  PubMed  Google Scholar 

  1339. Takeda AL, Colquitt J, Clegg AJ, Jones J: Pegaptanib and ranibizumab for neovascular age-related macular degeneration: a systematic review. Br J Ophthalmol. 2007, 91: 1177-82.

    CAS  PubMed  PubMed Central  Google Scholar 

  1340. Apte RS: Pegaptanib sodium for the treatment of age-related macular degeneration. Expert Opin Pharmacother. 2008, 9: 499-508.

    CAS  PubMed  Google Scholar 

  1341. Trenam CW, Dabbagh AJ, Morris CJ, Blake DR: Skin inflammation induced by reactive oxygen species (ROS): an in vivo model. Br J Dermatol. 1991, 125: 325-9.

    CAS  PubMed  Google Scholar 

  1342. Trouba KJ, Hamadeh HK, Amin RP, Germolec DR: Oxidative stress and its role in skin disease. Antioxidants & Redox Signaling. 2002, 4: 665-673.

    CAS  Google Scholar 

  1343. Wojas-Pelc A, Marcinkiewicz J: What is a role of haeme oxygenase-1 in psoriasis? Current concepts of pathogenesis. Int J Exp Pathol. 2007, 88: 95-102.

    CAS  PubMed  PubMed Central  Google Scholar 

  1344. Trenam CW, Blake DR, Morris CJ: Skin inflammation – reactive oxygen species and the role of iron. J Invest Dermatol. 1992, 99: 675-682.

    CAS  PubMed  Google Scholar 

  1345. Trenam CW, Dabbagh AJ, Blake DR, Morris CJ: The role of iron in an acute model of skin inflammation induced by reactive oxygen species (ROS). Br J Dermatol. 1992, 126: 250-256.

    CAS  PubMed  Google Scholar 

  1346. Hider RC, Singh S, Porter JB: Iron chelating agents with clinical potential. Proc R Soc Ed B. 1992, 99: 137-168.

    CAS  Google Scholar 

  1347. Kim KY, Schumacher HR, Hunsche E, Wertheimer AI, Kong SX: A literature review of the epidemiology and treatment of acute gout. Clinical Therapeutics. 2003, 25: 1593-1617.

    PubMed  Google Scholar 

  1348. Davies KJ, Sevanian A, Muakkassah-Kelly SF, Hochstein P: Uric acid-iron ion complexes. A new aspect of the antioxidant functions of uric acid. Biochem J. 1986, 235: 747-54.

    CAS  PubMed  PubMed Central  Google Scholar 

  1349. Ghio AJ, Kennedy TP, Rao G, Cooke CL, Miller MJ, Hoidal JR: Complexation of iron cation by sodium urate crystals and gouty inflammation. Arch Biochem Biophys. 1994, 313: 215-221.

    CAS  PubMed  Google Scholar 

  1350. Ghio AJ, Ford ES, Kennedy TP, Hoidal JR: The association between serum ferritin and uric acid in humans. Free Radical Research. 2005, 39: 337-342.

    CAS  PubMed  Google Scholar 

  1351. Facchini FS: Near-iron deficiency-induced remission of gouty arthritis. Rheumatology. 2003, 42: 1550-1555.

    CAS  PubMed  Google Scholar 

  1352. Poli R, Langdon WB: On the search properties of different crossover operators in genetic programming. Proc. 3rd Conf Genetic Programming GP'98. 1998, Morgan Kaufmann, Madison, WI, 293-301.

    Google Scholar 

  1353. Brittenham GM: Iron chelators and iron toxicity. Alcohol. 2003, 30: 151-8.

    CAS  PubMed  Google Scholar 

  1354. Tung BY, Emond MJ, Bronner MP, Raaka SD, Cotler SJ, Kowdley KV: Hepatitis C, iron status, and disease severity: relationship with HFE mutations. Gastroenterology. 2003, 124: 318-26.

    CAS  PubMed  Google Scholar 

  1355. Corradini E, Ferrara F, Pietrangelo A: Iron and the liver. Pediatr Endocrinol Rev. 2004, 2 (Suppl 2): 245-8.

    PubMed  Google Scholar 

  1356. Rigamonti C, Andorno S, Maduli E, Capelli F, Boldorini R, Sartori M: Gender and liver fibrosis in chronic hepatitis: the role of iron status. Aliment Pharmacol Ther. 2005, 21: 1445-51.

    CAS  PubMed  Google Scholar 

  1357. Philippe MA, Ruddell RG, Ramm GA: Role of iron in hepatic fibrosis: one piece in the puzzle. World J Gastroenterol. 2007, 13: 4746-54.

    CAS  PubMed  PubMed Central  Google Scholar 

  1358. Kohgo Y, Ohtake T, Ikuta K, Suzuki Y, Torimoto Y, Kato J: Dysregulation of systemic iron metabolism in alcoholic liver diseases. J Gastroenterol Hepatol. 2008, 23 (Suppl 1): S78-81.

    CAS  PubMed  Google Scholar 

  1359. Ramm GA, Ruddell RG: Hepatotoxicity of iron overload: Mechanisms of iron-induced hepatic fibrogenesis. Seminars in Liver Disease. 2005, 25: 433-449.

    CAS  PubMed  Google Scholar 

  1360. McCullough KD, Bartfay WJ: The dose-dependent effects of chronic iron overload on the production of oxygen free radicals and vitamin E concentrations in the liver of a murine model. Biol Res Nurs. 2007, 8: 300-4.

    CAS  PubMed  Google Scholar 

  1361. Petrak J, Myslivcova D, Man P, Cmejla R, Cmejlova J, Vyoral D, Elleder M, Vulpe CD: Proteomic analysis of hepatic iron overload in mice suggests dysregulation of urea cycle, impairment of fatty acid oxidation, and changes in the methylation cycle. Am J Physiol Gastrointest Liver Physiol. 2007, 292: G1490-8.

    CAS  PubMed  Google Scholar 

  1362. Aigner E, Theurl I, Theurl M, Lederer D, Haufe H, Dietze O, Strasser M, Datz C, Weiss G: Pathways underlying iron accumulation in human nonalcoholic fatty liver disease. Am J Clin Nutr. 2008, 87: 1374-83.

    CAS  PubMed  Google Scholar 

  1363. Chen CJ, Yu MW, Liaw YF: Epidemiological characteristics and risk factors of hepatocellular carcinoma. J Gastroenterol Hepatol. 1997, 12: S294-308.

    CAS  PubMed  Google Scholar 

  1364. Asare GA, Mossanda KS, Kew MC, Paterson AC, Kahler-Venter CP, Siziba K: Hepatocellular carcinoma caused by iron overload: A possible mechanism of direct hepatocarcinogenicity. Toxicology. 2006, 219: 41-52.

    CAS  PubMed  Google Scholar 

  1365. Kato J, Kobune M, Nakamura T, Kuroiwa G, Takada K, Takimoto R, Sato Y, Fujikawa K, Takahashi M, Takayama T, Ikeda T, Niitsu Y: Normalization of elevated hepatic 8-hydroxy-2'-deoxyguanosine levels in chronic hepatitis C patients by phlebotomy and low iron diet. Cancer Res. 2001, 61: 8697-702.

    CAS  PubMed  Google Scholar 

  1366. Ritter C, Reinke A, Andrades M, Martins MR, Rocha J, Menna-Barreto S, Quevedo J, Moreira JC, Dal-Pizzol F: Protective effect of N-acetylcysteine and deferoxamine on carbon tetrachloride-induced acute hepatic failure in rats. Crit Care Med. 2004, 32: 2079-83.

    CAS  PubMed  Google Scholar 

  1367. Bridle K, Cheung TK, Murphy T, Walters M, Anderson G, Crawford DG, Fletcher LM: Hepcidin is down-regulated in alcoholic liver injury: implications for the pathogenesis of alcoholic liver disease. Alcohol Clin Exp Res. 2006, 30: 106-12.

    CAS  PubMed  Google Scholar 

  1368. Antó JM, Vermeire P, Vestbo J, Sunyer J: Epidemiology of chronic obstructive pulmonary disease. European Respiratory Journal. 2001, 17: 982-994.

    PubMed  Google Scholar 

  1369. White AJ, Gompertz S, Stockley RA: Chronic obstructive pulmonary disease .6: The aetiology of exacerbations of chronic obstructive pulmonary disease. Thorax. 2003, 58: 73-80.

    CAS  PubMed  PubMed Central  Google Scholar 

  1370. Tsoumakidou M, Siafakas NM: Novel insights into the aetiology and pathophysiology of increased airway inflammation during COPD exacerbations. Respiratory Research. 2006, 7.

    Google Scholar 

  1371. Sapey E, Stockley RA: COPD exacerbations 2: Aetiology. Thorax. 2006, 61: 250-258.

    CAS  PubMed  PubMed Central  Google Scholar 

  1372. Rodriguez-Roisin R: Toward a consensus definition for COPD exacerbations. Chest. 2000, 117: 398S-401S.

    CAS  PubMed  Google Scholar 

  1373. Weinberg ED: Tobacco smoke iron: an initiator/promoter of multiple diseases. Biometals. 2008.

    Google Scholar 

  1374. Rahman I, van Schadewijk AA, Crowther AJ, Hiemstra PS, Stolk J, MacNee W, De Boer WI: 4-Hydroxy-2-nonenal, a specific lipid peroxidation product, is elevated in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002, 166: 490-5.

    PubMed  Google Scholar 

  1375. Langen RCJ, Korn SH, Wouters EFM: ROS in the local and systemic pathogenesis of COPD. Free Rad Biol Med. 2003, 35: 226-235.

    CAS  PubMed  Google Scholar 

  1376. Barnes PJ: Mediators of chronic obstructive pulmonary disease. Pharmacol Rev. 2004, 56: 515-548.

    CAS  PubMed  Google Scholar 

  1377. Santos MC, Oliveira AL, Viegas-Crespo AM, Vicente L, Barreiros A, Monteiro P, Pinheiro T, De Almeida AB: Systemic markers of the redox balance in chronic obstructive pulmonary disease. Biomarkers. 2004, 9: 461-469.

    CAS  PubMed  Google Scholar 

  1378. Kinnula VL: Focus on antioxidant enzymes and antioxidant strategies in smoking related airway diseases. Thorax. 2005, 60: 693-700.

    CAS  PubMed  PubMed Central  Google Scholar 

  1379. Rytila P, Rehn T, Ilumets H, Rouhos A, Sovijarvi A, Myllarniemi M, Kinnula VL: Increased oxidative stress in asymptomatic current chronic smokers and GOLD stage 0 COPD. Respiratory Research. 2006, 7.

    Google Scholar 

  1380. Stevenson CS, Koch LG, Britton SL: Aerobic capacity, oxidant stress, and chronic obstructive pulmonary disease – A new take on an old hypothesis. Pharmacol Therapeut. 2006, 110: 71-82.

    CAS  Google Scholar 

  1381. Kinnula VL, Ilumets H, Myllarnlemi M, Sovijarvi A, Rytila P: 8-Isoprostane as a marker of oxidative stress in nonsymptomatic cigarette smokers and COPD. European Respiratory Journal. 2007, 29: 51-55.

    CAS  PubMed  Google Scholar 

  1382. Huang X, Li WH, Attfield MD, Nadas A, Frenkel K, Finkelman RB: Mapping and prediction of coal workers' pneumoconiosis with bioavailable iron content in the bituminous coals. Environmental Health Perspectives. 2005, 113: 964-968.

    CAS  PubMed  PubMed Central  Google Scholar 

  1383. Mutti A, Corradi M, Goldoni M, Vettori MV, Bernard A, Apostoli P: Exhaled metallic elements and serum pneumoproteins in asymptomatic smokers and patients with COPD or asthma. Chest. 2006, 129: 1288-1297.

    CAS  PubMed  PubMed Central  Google Scholar 

  1384. Carter JD, Ghio AJ, Samet JM, Devlin RB: Cytokine production by human airway epithelial cells after exposure to an air pollution particle is metal-dependent. Toxicol Appl Pharmacol. 1997, 146: 180-8.

    CAS  PubMed  Google Scholar 

  1385. Corhay JL, Weber G, Bury T, Mariz S, Roelandts I, Radermecker MF: Iron content in human alveolar macrophages. Eur Resp J. 1992, 5: 804-809.

    CAS  Google Scholar 

  1386. Stites SW, Nelson ME, Wesselius LJ: Transferrin concentrations in serum and lower respiratory tract fluid of mechanically ventilated patients with COPD or ARDS. Chest. 1995, 107: 1681-1685.

    CAS  PubMed  Google Scholar 

  1387. Southorn PA, Powis G: Free radicals in medicine .2. involvement in human disease. Mayo Clinic Proceedings. 1988, 63: 390-408.

    CAS  PubMed  Google Scholar 

  1388. Quinlan GJ, Lamb NJ, Tilley R, Evans TW, Gutteridge JMC: Plasma hypoxanthine levels in ARDS: Implications for oxidative stress, morbidity, and mortality. Amer J Resp Crit Care Med. 1997, 155: 479-484.

    CAS  Google Scholar 

  1389. Quinlan GJ, Evans TW, Gutteridge JMC: Oxidative damage to plasma proteins in adult respiratory distress syndrome. Free Radical Research. 1994, 20: 289-298.

    CAS  PubMed  Google Scholar 

  1390. Gutteridge JMC, Quinlan GJ: Antioxidant protection against organic and inorganic oxygen radicals by normal human plasma – the important primary role for iron-binding and iron-oxidizing proteins. Biochim Biophys Acta. 1993, 1156: 144-150.

    CAS  PubMed  Google Scholar 

  1391. Reid DW, Lam QT, Schneider H, Walters EH: Airway iron and iron-regulatory cytokines in cystic fibrosis. European Respiratory Journal. 2004, 24: 286-291.

    CAS  PubMed  Google Scholar 

  1392. Doll R, Hill AB: Smoking and carcinoma of the lung; preliminary report. Br Med J. 1950, 2: 739-48.

    CAS  PubMed  PubMed Central  Google Scholar 

  1393. Peto R, Darby S, Deo H, Silcocks P, Whitley E, Doll R: Smoking, smoking cessation, and lung cancer in the UK since 1950: combination of national statistics with two case-control studies. BMJ. 2000, 321: 323-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  1394. Doll R, Hill AB: The mortality of doctors in relation to their smoking habits: a preliminary report. 1954. BMJ. 2004, 328: 1529-33.

    PubMed  PubMed Central  Google Scholar 

  1395. Doll R, Peto R, Boreham J, Sutherland I: Mortality from cancer in relation to smoking: 50 years observations on British doctors. Br J Cancer. 2005, 92: 426-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  1396. Thompson AB, Bohling T, Heires A, Linder J, Rennard SI: Lower respiratory tract iron burden is increased in association with cigarette smoking. J Lab Clin Med. 1991, 117: 493-9.

    CAS  PubMed  Google Scholar 

  1397. Dreher KL, Jaskot RH, Lehmann JR, Richards JH, McGee JK, Ghio AJ, Costa DL: Soluble transition metals mediate residual oil fly ash induced acute lung injury. J Toxicol Environ Health. 1997, 50: 285-305.

    CAS  PubMed  Google Scholar 

  1398. Mateos F, Brock JH, Perez-Arellano JL: Iron metabolism in the lower respiratory tract. Thorax. 1998, 53: 594-600.

    CAS  PubMed  PubMed Central  Google Scholar 

  1399. Boelaert JR, Gomes MS, Gordeuk VR: Smoking, iron, and tuberculosis. Lancet. 2003, 362: 1243-4.

    PubMed  Google Scholar 

  1400. Ghio AJ, Carter JD, Richards JH, Richer LD, Grissom CK, Elstad MR: Iron and iron-related proteins in the lower respiratory tract of patients with acute respiratory distress syndrome. Crit Care Med. 2003, 31: 395-400.

    CAS  PubMed  Google Scholar 

  1401. Ernst P: Review article: the role of inflammation in the pathogenesis of gastric cancer. Aliment Pharmacol Ther. 1999, 13 (Suppl 1): 13-18.

    PubMed  Google Scholar 

  1402. Kelloff GJ, Crowell JA, Steele VE, Lubet RA, Malone WA, Boone CW, Kopelovich L, Hawk ET, Lieberman R, Lawrence JA, Ali I, Viner JL, Sigman CC: Progress in cancer chemoprevention: development of diet-derived chemopreventive agents. J Nutr. 2000, 130: 467S-471S.

    CAS  PubMed  Google Scholar 

  1403. Balkwill F, Mantovani A: Inflammation and cancer: back to Virchow?. Lancet. 2001, 357: 539-45.

    CAS  PubMed  Google Scholar 

  1404. Coussens LM, Werb Z: Inflammation and cancer. Nature. 2002, 420: 860-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  1405. Aggarwal BB: Nuclear factor-kappaB: the enemy within. Cancer Cell. 2004, 6: 203-8.

    CAS  PubMed  Google Scholar 

  1406. Vakkila J, Lotze MT: Inflammation and necrosis promote tumour growth. Nat Rev Immunol. 2004, 4: 641-8.

    CAS  PubMed  Google Scholar 

  1407. Balkwill F, Charles KA, Mantovani A: Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell. 2005, 7: 211-7.

    CAS  PubMed  Google Scholar 

  1408. Karin M, Greten FR: NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 2005, 5: 749-59.

    CAS  PubMed  Google Scholar 

  1409. Li Q, Withoff S, Verma IM: Inflammation-associated cancer: NF-kappaB is the lynchpin. Trends Immunol. 2005, 26: 318-25.

    PubMed  Google Scholar 

  1410. Steele VE, Kelloff GJ: Development of cancer chemopreventive drugs based on mechanistic approaches. Mutat Res. 2005, 591: 16-23.

    CAS  PubMed  Google Scholar 

  1411. Aggarwal BB, Shishodia S, Sandur SK, Pandey MK, Sethi G: Inflammation and cancer: how hot is the link?. Biochem Pharmacol. 2006, 72: 1605-21.

    CAS  PubMed  Google Scholar 

  1412. Karin M: NF-kappaB and cancer: mechanisms and targets. Mol Carcinog. 2006, 45: 355-61.

    CAS  PubMed  Google Scholar 

  1413. Kelloff GJ, Lippman SM, Dannenberg AJ, Sigman CC, Pearce HL, Reid BJ, Szabo E, Jordan VC, Spitz MR, Mills GB, Papadimitrakopoulou VA, Lotan R, Aggarwal BB, Bresalier RS, Kim J, Arun B, Lu KH, Thomas ME, Rhodes HE, Brewer MA, Follen M, Shin DM, Parnes HL, Siegfried JM, Evans AA, Blot WJ, Chow WH, Blount PL, Maley CC, Wang KK, Lam S, Lee JJ, Dubinett SM, Engstrom PF, Meyskens FL, O'Shaughnessy J, Hawk ET, Levin B, Nelson WG, Hong WK: Progress in chemoprevention drug development: the promise of molecular biomarkers for prevention of intraepithelial neoplasia and cancer–a plan to move forward. Clin Cancer Res. 2006, 12: 3661-97.

    CAS  PubMed  Google Scholar 

  1414. Rakoff-Nahoum S: Why cancer and inflammation?. Yale J Biol Med. 2006, 79: 123-30.

    CAS  PubMed  Google Scholar 

  1415. Yoshimura A: Signal transduction of inflammatory cytokines and tumor development. Cancer Sci. 2006, 97: 439-47.

    CAS  PubMed  Google Scholar 

  1416. Hussain SP, Harris CC: Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007, 121: 2373-80.

    CAS  PubMed  Google Scholar 

  1417. Allavena P, Garlanda C, Borrello MG, Sica A, Mantovani A: Pathways connecting inflammation and cancer. Curr Opin Genet Dev. 2008, 18: 3-10.

    CAS  PubMed  Google Scholar 

  1418. Borrello MG, Degl'Innocenti D, Pierotti MA: Inflammation and cancer: the oncogene-driven connection. Cancer Lett. 2008, 267: 262-70.

    CAS  PubMed  Google Scholar 

  1419. Haverkamp J, Charbonneau B, Ratliff TL: Prostate inflammation and its potential impact on prostate cancer: a current review. J Cell Biochem. 2008, 103: 1344-53.

    CAS  PubMed  Google Scholar 

  1420. Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yamaguchi A, Imanishi H, Nakada K, Honma Y, Hayashi J: ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science. 2008, 320: 661-4.

    CAS  PubMed  Google Scholar 

  1421. Karin M: The IkappaB kinase – a bridge between inflammation and cancer. Cell Res. 2008, 18: 334-42.

    CAS  PubMed  Google Scholar 

  1422. Maeda S, Omata M: Inflammation and cancer: role of nuclear factor-kappaB activation. Cancer Sci. 2008, 99: 836-42.

    CAS  PubMed  Google Scholar 

  1423. Mantovani A, Allavena P, Sica A, Balkwill F: Cancer-related inflammation. Nature. 2008, 454: 436-44.

    CAS  PubMed  Google Scholar 

  1424. Sica A, Allavena P, Mantovani A: Cancer related inflammation: the macrophage connection. Cancer Lett. 2008, 267: 204-15.

    CAS  PubMed  Google Scholar 

  1425. Vasto S, Carruba G, Lio D, Colonna-Romano G, Di Bona D, Candore G, Caruso C: Inflammation, ageing and cancer. Mech Ageing Dev. 2008

    Google Scholar 

  1426. Baron JA, Sandler RS: Nonsteroidal anti-inflammatory drugs and cancer prevention. Annu Rev Med. 2000, 51: 511-23.

    CAS  PubMed  Google Scholar 

  1427. Rostom A, Dube C, Lewin G, Tsertsvadze A, Barrowman N, Code C, Sampson M, Moher D: Nonsteroidal anti-inflammatory drugs and cyclooxygenase-2 inhibitors for primary prevention of colorectal cancer: a systematic review prepared for the U.S. Preventive Services Task Force. Ann Intern Med. 2007, 146: 376-89.

    PubMed  Google Scholar 

  1428. Marshall BJ, Warren JR: Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet. 1984, 1: 1311-1315.

    CAS  PubMed  Google Scholar 

  1429. Davey HM, Kell DB: Flow cytometry and cell sorting of heterogeneous microbial populations: the importance of single-cell analysis. Microbiol Rev. 1996, 60: 641-696.

    CAS  PubMed  PubMed Central  Google Scholar 

  1430. Marshall B: Helicobacter connections. ChemMedChem. 2006, 1: 783-802.

    CAS  PubMed  Google Scholar 

  1431. Rakba N, Aouad F, Henry C, Caris C, Morel I, Baret P, Pierre JL, Brissot P, Ward RJ, Lescoat G, Crichton RR: Iron mobilisation and cellular protection by a new synthetic chelator O-Trensox. Biochem Pharmacol. 1998, 55: 1797-806.

    CAS  PubMed  Google Scholar 

  1432. Le NTV, Richardson DR: The role of iron in cell cycle progression and the proliferation of neoplastic cells. Biochimica Et Biophysica Acta-Reviews on Cancer. 2002, 1603: 31-46.

    CAS  Google Scholar 

  1433. Buss JL, Torti FM, Torti SV: The role of iron chelation in cancer therapy. Curr Med Chem. 2003, 10: 1021-34.

    CAS  PubMed  Google Scholar 

  1434. Lovejoy DB, Richardson DR: Iron chelators as anti-neoplastic agents: Current developments and promise of the PIH class of chelators. Current Medicinal Chemistry. 2003, 10: 1035-1049.

    CAS  PubMed  Google Scholar 

  1435. Buss JL, Greene BT, Turner J, Torti FM, Torti SV: Iron chelators in cancer chemotherapy. Curr Top Med Chem. 2004, 4: 1623-35.

    CAS  PubMed  Google Scholar 

  1436. Hanai J, Mammoto T, Seth P, Mori K, Karumanchi SA, Barasch J, Sukhatme VP: Lipocalin 2 diminishes invasiveness and metastasis of Ras-transformed cells. J Biol Chem. 2005, 280: 13641-7.

    CAS  PubMed  Google Scholar 

  1437. Kalinowski DS, Richardson DR: The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev. 2005, 57: 547-83.

    CAS  PubMed  Google Scholar 

  1438. Pahl PMB, Horwitz LD: Cell permeable iron chelators as potential cancer chemotherapeutic agents. Cancer Invest. 2005, 23: 683-691.

    CAS  PubMed  Google Scholar 

  1439. Nie GJ, Chen GH, Sheftel AD, Pantopoulos K, Ponka P: In vivo tumor growth is inhibited by cytosolic iron deprivation caused by the expression of mitochondrial ferritin. Blood. 2006, 108: 2428-2434.

    CAS  PubMed  Google Scholar 

  1440. Edgren G, Nyren O, Melbye M: Cancer as a ferrotoxic disease: are we getting hard stainless evidence?. J Natl Cancer Inst. 2008, 100: 976-7.

    PubMed  Google Scholar 

  1441. Huang X: Does iron have a role in breast cancer?. Lancet Oncol. 2008, 9: 803-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  1442. Zacharski LR, Chow BK, Howes PS, Shamayeva G, Baron JA, Dalman RL, Malenka DJ, Ozaki CK, Lavori PW: Decreased cancer risk after iron reduction in patients with peripheral arterial disease: results from a randomized trial. J Natl Cancer Inst. 2008, 100: 996-1002.

    CAS  PubMed  Google Scholar 

  1443. Lim R, Ahmed N, Borregaard N, Riley C, Wafai R, Thompson EW, Quinn MA, Rice GE: Neutrophil gelatinase-associated lipocalin (NGAL) an early-screening biomarker for ovarian cancer: NGAL is associated with epidermal growth factor-induced epithelio-mesenchymal transition. Int J Cancer. 2007, 120: 2426-2434.

    CAS  PubMed  Google Scholar 

  1444. Iannetti A, Pacifico F, Acquaviva R, Lavorgna A, Crescenzi E, Vascotto C, Tell G, Salzano AM, Scaloni A, Vuttariello E, Chiappetta G, Formisano S, Leonardi A: The neutrophil gelatinase-associated lipocalin (NGAL), a NF-{kappa}B-regulated gene, is a survival factor for thyroid neoplastic cells. Proc Natl Acad Sci USA. 2008, 105: 14058-63.

    CAS  PubMed  PubMed Central  Google Scholar 

  1445. Nelson RL, Davis FG, Sutter E, Sobin LH, Kikendall JW, Bowen P: Body iron stores and risk of colonic neoplasia. J Natl Cancer Inst. 1994, 86: 455-60.

    CAS  PubMed  Google Scholar 

  1446. Okada S: Iron-induced tissue damage and cancer: The role of reactive oxygen species-free radicals. Pathol Int. 1996, 46: 311-332.

    CAS  PubMed  Google Scholar 

  1447. Toyokuni S: Iron-induced carcinogenesis: The role of redox regulation. Free Radical Biology and Medicine. 1996, 20: 553-566.

    CAS  PubMed  Google Scholar 

  1448. Weinberg ED: The role of iron in cancer. Eur J Cancer Prev. 1996, 5: 19-36.

    CAS  PubMed  Google Scholar 

  1449. Weinberg ED: Iron loading and disease surveillance. Emerg Infect Dis. 1999, 5: 346-52.

    CAS  PubMed  PubMed Central  Google Scholar 

  1450. Núñez MT, Tapia V, Toyokuni S, Okada S: Iron-induced oxidative damage in colon carcinoma (Caco-2) cells. Free Radic Res. 2001, 34: 57-68.

    PubMed  Google Scholar 

  1451. Glei M, Latunde-Dada GO, Klinder A, Becker TW, Hermann U, Voigt K, Pool-Zobel BL: Iron-overload induces oxidative DNA damage in the human colon carcinoma cell line HT29 clone 19A. Mutat Res. 2002, 519: 151-61.

    CAS  PubMed  Google Scholar 

  1452. Toyokuni S: Iron and carcinogenesis: from Fenton reaction to target genes. Redox Rep. 2002, 7: 189-97.

    CAS  PubMed  Google Scholar 

  1453. Deugnier Y: Iron and liver cancer. Alcohol. 2003, 30: 145-150.

    CAS  PubMed  Google Scholar 

  1454. Klaunig JE, Kamendulis LM: The role of oxidative stress in carcinogenesis. Annu Rev Pharmacol Toxicol. 2004, 44: 239-67.

    CAS  PubMed  Google Scholar 

  1455. Storz P: Reactive oxygen species in tumor progression. Front Biosci. 2005, 10: 1881-96.

    CAS  PubMed  Google Scholar 

  1456. Lee SK, Lee JJ, Lee HJ, Lee J, Jeon BH, Jun CD, Lee SK, Kim EC: Iron chelator-induced growth arrest and cytochrome c-dependent apoptosis in immortalized and malignant oral keratinocytes. Journal of Oral Pathology & Medicine. 2006, 35: 218-226.

    CAS  Google Scholar 

  1457. Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M: Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chemico-Biological Interactions. 2006, 160: 1-40.

    CAS  PubMed  Google Scholar 

  1458. Wu J, Eckard J, Chen H, Costa M, Frenkel K, Huang X: Altered iron homeostasis involvement in arsenite-mediated cell transformation. Free Radic Biol Med. 2006, 40: 444-52.

    PubMed  Google Scholar 

  1459. Kallianpur AR, Lee SA, Gao YT, Lu W, Zheng Y, Ruan ZX, Dai Q, Gu K, Shu XO, Zheng W: Dietary animal-derived iron and fat intake and breast cancer risk in the Shanghai Breast Cancer Study. Breast Cancer Res Treat. 2008, 107: 123-32.

    CAS  PubMed  Google Scholar 

  1460. Prá D, Franke SI, Giulian R, Yoneama ML, Dias JF, Erdtmann B, Henriques JA: Genotoxicity and mutagenicity of iron and copper in mice. Biometals. 2008, 21: 289-97.

    PubMed  Google Scholar 

  1461. Yang WS, Stockwell BR: Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 2008, 15: 234-45.

    CAS  PubMed  PubMed Central  Google Scholar 

  1462. Richardson DR, Kalinowski DS, Lau S, Jansson PJ, Lovejoy DB: Cancer cell iron metabolism and the development of potent iron chelators as anti-tumour agents. Biochim Biophys Acta. 2008.

    Google Scholar 

  1463. Weitzman SA, Graceffa P: Asbestos catalyzes hydroxyl and superoxide radical generation from hydrogen peroxide. Arch Biochem Biophys. 1984, 228: 373-6.

    CAS  PubMed  Google Scholar 

  1464. Gulumian M, van Wyk JA: Hydroxyl radical production in the presence of fibres by a Fenton-type reaction. Chem Biol Interact. 1987, 62: 89-97.

    CAS  PubMed  Google Scholar 

  1465. Mossman BT, Marsh JP, Shatos MA, Doherty J, Gilbert R, Hill S: Implication of active oxygen species as second messengers of asbestos toxicity. Drug Chem Toxicol. 1987, 10: 157-80.

    CAS  PubMed  Google Scholar 

  1466. Shatos MA, Doherty JM, Marsh JP, Mossman BT: Prevention of asbestos-induced cell death in rat lung fibroblasts and alveolar macrophages by scavengers of active oxygen species. Environ Res. 1987, 44: 103-16.

    CAS  PubMed  Google Scholar 

  1467. Kamp DW, Graceffa P, Pryor WA, Weitzman SA: The role of free radicals in asbestos-induced diseases. Free Radic Biol Med. 1992, 12: 293-315.

    CAS  PubMed  Google Scholar 

  1468. Quinlan TR, Marsh JP, Janssen YM, Borm PA, Mossman BT: Oxygen radicals and asbestos-mediated disease. Environ Health Perspect. 1994, 102 (Suppl 10): 107-10.

    PubMed  PubMed Central  Google Scholar 

  1469. Hardy JA, Aust AE: Iron in asbestos chemistry and carcinogenicity. Chem Rev. 1995, 95: 97-118.

    CAS  Google Scholar 

  1470. Gilmour PS, Brown DM, Beswick PH, MacNee W, Rahman I, Donaldson K: Free radical activity of industrial fibers: role of iron in oxidative stress and activation of transcription factors. Environ Health Perspect. 1997, 105 (Suppl 5): 1313-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  1471. Governa M, Amati M, Fontana S, Visona I, Botta GC, Mollo F, Bellis D, Bo P: Role of iron in asbestos-body-induced oxidant radical generation. J Toxicol Environ Health A. 1999, 58: 279-87.

    CAS  PubMed  Google Scholar 

  1472. Kamp DW, Weitzman SA: The molecular basis of asbestos induced lung injury. Thorax. 1999, 54: 638-52.

    CAS  PubMed  PubMed Central  Google Scholar 

  1473. Xu A, Wu LJ, Santella RM, Hei TK: Role of oxyradicals in mutagenicity and DNA damage induced by crocidolite asbestos in mammalian cells. Cancer Res. 1999, 59: 5922-6.

    CAS  PubMed  Google Scholar 

  1474. Aljandali A, Pollack H, Yeldandi A, Li Y, Weitzman SA, Kamp DW: Asbestos causes apoptosis in alveolar epithelial cells: role of iron-induced free radicals. J Lab Clin Med. 2001, 137: 330-9.

    CAS  PubMed  Google Scholar 

  1475. Gazzano E, Turci F, Foresti E, Putzu MG, Aldieri E, Silvagno F, Lesci IG, Tomatis M, Riganti C, Romano C, Fubini B, Roveri N, Ghigo D: Iron-loaded synthetic chrysotile: a new model solid for studying the role of iron in asbestos toxicity. Chem Res Toxicol. 2007, 20: 380-7.

    CAS  PubMed  Google Scholar 

  1476. Pervaiz S: Pro-oxidant milieu blunts scissors: Insight into tumor progression, drug resistance, and novel druggable targets. Curr Pharmaceut Des. 2006, 12: 4469-4477.

    CAS  Google Scholar 

  1477. Perrone GG, Tan SX, Dawes IW: Reactive oxygen species and yeast apoptosis. Biochim Biophys Acta. 2008, 1783: 1354-68.

    CAS  PubMed  Google Scholar 

  1478. Scheibel LW, Adler A: Anti-malarial activity of selected aromatic chelators. Mol Pharmacol. 1980, 18: 320-325.

    CAS  PubMed  Google Scholar 

  1479. Scheibel LW, Rodriguez S: Antimalarial activity of selected aromatic chelators V. Localization of 59Fe in Plasmodium falciparum in the presence of oxines. Prog Clin Biol Res. 1989, 313: 119-49.

    CAS  PubMed  Google Scholar 

  1480. Hershko C, Gordeuk VR, Thuma PE, Theanacho EN, Spira DT, Hider RC, Peto TEA, Brittenham GM: The antimalaria effect of iron chelators – studies in animal models and in humans with mild Falciparum malaria. J Inorg Biochem. 1992, 47: 267-277.

    CAS  PubMed  Google Scholar 

  1481. Gordeuk V, Thuma P, Brittenham G, McLaren C, Parry D, Backenstose A, Biemba G, Msiska R, Holmes L, McKinley E, Vargas L, Gilkeson R, Poltera AA: Effect of iron chelation therapy on recovery from deep coma in children with cerebral malaria. New Engl J Med. 1992, 327: 1473-1477.

    CAS  PubMed  Google Scholar 

  1482. Gordeuk VR, Thuma PE, Brittenham GM, Biemba G, Zulu S, Simwanza G, Kalense P, Mhango A, Parry D, Poltera AA, Aikawa M: Iron chelation as a chemotherapeutic strategy for Falciparum malaria. Amer J Trop Med Hyg. 1993, 48: 193-197.

    CAS  Google Scholar 

  1483. Voest EE, Vreugdenhil G, Marx JJM: Iron-chelating agents in non-iron overload conditions. Ann Internal Med. 1994, 120: 490-499.

    CAS  Google Scholar 

  1484. Hider RC, Liu ZD: The treatment of malaria with iron chelators. J Pharm Pharmacol. 1997, 49: 59-64.

    CAS  Google Scholar 

  1485. Cabantchik ZI, Moody-Haupt S, Gordeuk VR: Iron chelators as anti-infectives; malaria as a paradigm. FEMS Immunol Med Microbiol. 1999, 26: 289-98.

    CAS  PubMed  Google Scholar 

  1486. Mabeza GF, Loyevsky M, Gordeuk VR, Weiss G: Iron chelation therapy for malaria: A review. Pharmacology & Therapeutics. 1999, 81: 53-75.

    CAS  Google Scholar 

  1487. Fritsche G, Larcher C, Schennach H, Weiss G: Regulatory interactions between iron and nitric oxide metabolism for immune defense against Plasmodium falciparum infection. J Infect Dis. 2001, 183: 1388-94.

    CAS  PubMed  Google Scholar 

  1488. Pradines B, Rolain JM, Ramiandrasoa F, Fusai T, Mosnier J, Rogier C, Daries W, Baret E, Kunesch G, Le Bras J, Parzy D: Iron chelators as antimalarial agents: in vitro activity of dicatecholate against Plasmodium falciparum. J Antimicrob Chemother. 2002, 50: 177-87.

    CAS  PubMed  Google Scholar 

  1489. Birch N, Wang X, Chong HS: Iron chelators as therapeutic iron depletion agents. Exp Opin Therapeut Pat. 2006, 16: 1533-1556.

    CAS  Google Scholar 

  1490. Golenser J, Domb A, Mordechai-Daniel T, Leshem B, Luty A, Kremsner P: Iron chelators: correlation between effects on Plasmodium spp. and immune functions. J Parasitol. 2006, 92: 170-7.

    CAS  PubMed  Google Scholar 

  1491. Melnyk P, Leroux V, Sergheraert C, Grellier P: Design, synthesis and in vitro antimalarial activity of an acylhydrazone library. Bioorg Med Chem Lett. 2006, 16: 31-35.

    CAS  PubMed  Google Scholar 

  1492. Pradines B, Tall A, Ramiandrasoa FR, Spiegel A, Sokhna C, Fusai T, Mosnier J, Daries W, Trape JF, Kunesch G, Parzy D, Rogier C: In vitro activity of iron-binding compounds against Senegalese isolates of Plasmodium falciparum. J Antimicrobial Chemotherapy. 2006, 57: 1093-1099.

    CAS  Google Scholar 

  1493. Doherty CP: Host-pathogen interactions: the role of iron. J Nutr. 2007, 137: 1341-4.

    CAS  PubMed  Google Scholar 

  1494. Vangapandu S, Jain M, Kaur K, Patil P, Patel SR, Jain R: Recent advances in antimalarial drug development. Medicinal Research Reviews. 2007, 27: 65-107.

    CAS  PubMed  Google Scholar 

  1495. Thuma PE, Mabeza GF, Biemba G, Bhat GJ, McLaren CE, Moyo VM, Zulu S, Khumalo H, Mabeza P, M'Hango A, Parry D, Poltera AA, Brittenham GM, Gordeuk VR: Effect of iron chelation therapy on mortality in Zambian children with cerebral malaria. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1998, 92: 214-218.

    CAS  PubMed  Google Scholar 

  1496. Meshnick SR, Yang YZ, Lima V, Kuypers F, Kamchonwongpaisan S, Yuthavong Y: Iron-dependent free radical generation from the antimalarial agent artemisinin (qinghaosu). Antimicrob Agents Chemother. 1993, 37: 1108-14.

    CAS  PubMed  PubMed Central  Google Scholar 

  1497. Kamchonwongpaisan S, Meshnick SR: The mode of action of the antimalarial artemisinin and its derivatives. Gen Pharmacol. 1996, 27: 587-92.

    CAS  PubMed  Google Scholar 

  1498. Meshnick SR, Taylor TE, Kamchonwongpaisan S: Artemisinin and the antimalarial endoperoxides: from herbal remedy to targeted chemotherapy. Microbiol Rev. 1996, 60: 301-15.

    CAS  PubMed  PubMed Central  Google Scholar 

  1499. Wu YK, Yue ZY, Wu YL: Interaction of Qinghaosu (artemisinin) with cysteine sulfhydryl mediated by traces of non-heme iron. Angewandte Chemie-International Edition. 1999, 38: 2580-2582.

    CAS  Google Scholar 

  1500. Li W, Mo W, Shen D, Sun L, Wang J, Lu S, Gitschier JM, Zhou B: Yeast model uncovers dual roles of mitochondria in action of artemisinin. PLoS Genet. 2005, 1: e36.

    PubMed  PubMed Central  Google Scholar 

  1501. Efferth T, Benakis A, Romero MR, Tomicic M, Rauh R, Steinbach D, Hafer R, Stamminger T, Oesch F, Kaina B, Marschall M: Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron. Free Radic Biol Med. 2004, 37: 998-1009.

    CAS  PubMed  Google Scholar 

  1502. Haynes RK, Chan WC, Lung CM, Uhlemann AC, Eckstein U, Taramelli D, Parapini S, Monti D, Krishna S: The Fe2+-mediated decomposition, PfATP6 Binding, and antimalarial activities of artemisone and other artemisinins: the unlikelihood of C-centered radicals as bioactive intermediates. Chem Med Chem. 2007, 2: 1480-1497.

    CAS  PubMed  Google Scholar 

  1503. Nakase I, Lai H, Singh NP, Sasaki T: Anticancer properties of artemisinin derivatives and their targeted delivery by transferrin conjugation. Int J Pharm. 2008, 354: 28-33.

    CAS  PubMed  Google Scholar 

  1504. White NJ: Qinghaosu (artemisinin): the price of success. Science. 2008, 320: 330-4.

    CAS  PubMed  Google Scholar 

  1505. Eckstein-Ludwig U, Webb RJ, Van Goethem ID, East JM, Lee AG, Kimura M, O'Neill PM, Bray PG, Ward SA, Krishna S: Artemisinins target the SERCA of Plasmodium falciparum. Nature. 2003, 424: 957-61.

    CAS  PubMed  Google Scholar 

  1506. Boyd PW, Jickells T, Law CS, Blain S, Boyle EA, Buesseler KO, Coale KH, Cullen JJ, de Baar HJ, Follows M, Harvey M, Lancelot C, Levasseur M, Owens NP, Pollard R, Rivkin RB, Sarmiento J, Schoemann V, Smetacek V, Takeda S, Tsuda A, Turner S, Watson AJ: Mesoscale iron enrichment experiments 1993–2005: synthesis and future directions. Science. 2007, 315: 612-7.

    CAS  PubMed  Google Scholar 

  1507. Blain S, Queguiner B, Armand L, Belviso S, Bombled B, Bopp L, Bowie A, Brunet C, Brussaard C, Carlotti F, Christaki U, Corbiere A, Durand I, Ebersbach F, Fuda JL, Garcia N, Gerringa L, Griffiths B, Guigue C, Guillerm C, Jacquet S, Jeandel C, Laan P, Lefevre D, Lo Monaco C, Malits A, Mosseri J, Obernosterer I, Park YH, Picheral M, Pondaven P, Remenyi T, Sandroni V, Sarthou G, Savoye N, Scouarnec L, Souhaut M, Thuiller D, Timmermans K, Trull T, Uitz J, van Beek P, Veldhuis M, Vincent D, Viollier E, Vong L, Wagener T: Effect of natural iron fertilization on carbon sequestration in the Southern Ocean. Nature. 2007, 446: 1070-4.

    CAS  PubMed  Google Scholar 

  1508. Boyd PW: Biogeochemistry: iron findings. Nature. 2007, 446: 989-91.

    CAS  PubMed  Google Scholar 

  1509. Watson AJ, Bakker DC, Ridgwell AJ, Boyd PW, Law CS: Effect of iron supply on Southern Ocean CO2 uptake and implications for glacial atmospheric CO2. Nature. 2000, 407: 730-3.

    CAS  PubMed  Google Scholar 

  1510. Collins HL: Withholding iron as a cellular defence mechanism–friend or foe?. Eur J Immunol. 2008, 38: 1803-6.

    CAS  PubMed  Google Scholar 

  1511. Weinberg ED: Iron and susceptibility to infectious disease. Science. 1974, 184: 952-6.

    CAS  PubMed  Google Scholar 

  1512. Weinberg ED: Iron and infection. Microbiol Rev. 1978, 42: 45-66.

    CAS  PubMed  PubMed Central  Google Scholar 

  1513. Hershko C, Peto TEA, Weatherall DJ: Iron and Infection. British Medical Journal. 1988, 296: 660-664.

    CAS  PubMed  PubMed Central  Google Scholar 

  1514. Oppenheimer SJ: Iron and infection: the clinical evidence. Acta Paediatr Scand Suppl. 1989, 361: 53-62.

    CAS  PubMed  Google Scholar 

  1515. Otto BR, Verweij vanVught A, Maclaren DM: Transferrins and heme compounds as iron sources for pathogenic bacteria. Critical Reviews in Microbiology. 1992, 18: 217-233.

    CAS  PubMed  Google Scholar 

  1516. Vasil ML, Ochsner UA: The response of Pseudomonas aeruginosa to iron: genetics, biochemistry and virulence. Mol Microbiol. 1999, 34: 399-413.

    CAS  PubMed  Google Scholar 

  1517. Bullen J, Griffiths E, Rogers H, Ward G: Sepsis: the critical role of iron. Microbes Infect. 2000, 2: 409-15.

    CAS  PubMed  Google Scholar 

  1518. Law D: Virulence factors of Escherichia coli O157 and other Shiga toxin-producing E. coli. J Appl Microbiol. 2000, 88: 729-45.

    CAS  PubMed  Google Scholar 

  1519. van Asbeck BS, Georgiou NA, Bruggen van der T, Oudshoorn M, Nottet HS, Marx JJ: Anti-HIV effect of iron chelators: different mechanisms involved. J Clin Virol. 2001, 20: 141-7.

    CAS  PubMed  Google Scholar 

  1520. Gordeuk VR, Delanghe JR, Langlois MR, Boelaert JR: Iron status and the outcome of HIV infection: an overview. J Clin Virol. 2001, 20: 111-5.

    CAS  PubMed  Google Scholar 

  1521. Rodriguez GM, Smith I: Mechanisms of iron regulation in mycobacteria: role in physiology and virulence. Mol Microbiol. 2003, 47: 1485-94.

    CAS  PubMed  Google Scholar 

  1522. Bullen JJ, Rogers HJ, Spalding PB, Ward CG: Iron and infection: the heart of the matter. FEMS Immunol Med Microbiol. 2005, 43: 325-30.

    CAS  PubMed  Google Scholar 

  1523. Bullen JJ, Rogers HJ, Spalding PB, Ward CG: Natural resistance, iron and infection: a challenge for clinical medicine. J Med Microbiol. 2006, 55: 251-8.

    CAS  PubMed  Google Scholar 

  1524. Maresso AW, Schneewind O: Iron acquisition and transport in Staphylococcus aureus. Biometals. 2006, 19: 193-203.

    CAS  PubMed  Google Scholar 

  1525. McDermid JM, Prentice AM: Iron and infection: effects of host iron status and the iron-regulatory genes haptoglobin and NRAMP1 (SLC11A1) on host-pathogen interactions in tuberculosis and HIV. Clin Sci (Lond). 2006, 110: 503-24.

    CAS  Google Scholar 

  1526. Meyer D: Iron chelation as therapy for HIV and Mycobacterium tuberculosis co-infection under conditions of iron overload. Curr Pharm Des. 2006, 12: 1943-7.

    CAS  PubMed  Google Scholar 

  1527. Sritharan M: Iron and bacterial virulence. Indian J Med Microbiol. 2006, 24: 163-4.

    CAS  PubMed  Google Scholar 

  1528. Boelaert JR, Vandecasteele SJ, Appelberg R, Gordeuk VR: The effect of the host's iron status on tuberculosis. J Infect Dis. 2007, 195: 1745-53.

    CAS  PubMed  Google Scholar 

  1529. Maynor L, Brophy DF: Risk of infection with intravenous iron therapy. Ann Pharmacother. 2007, 41: 1476-80.

    CAS  PubMed  Google Scholar 

  1530. Basaraba RJ, Bielefeldt-Ohmann H, Eschelbach EK, Reisenhauer C, Tolnay AE, Taraba LC, Shanley CA, Smith EA, Bedwell CL, Chlipala EA, Orme IM: Increased expression of host iron-binding proteins precedes iron accumulation and calcification of primary lung lesions in experimental tuberculosis in the guinea pig. Tuberculosis (Edinb). 2008, 88: 69-79.

    CAS  Google Scholar 

  1531. Weinberg ED: Iron availability and infection. Biochim Biophys Acta. 2008

    Google Scholar 

  1532. Ashrafian H: Hepcidin: the missing link between hemochromatosis and infections. Infect Immun. 2003, 71: 6693-700.

    CAS  PubMed  PubMed Central  Google Scholar 

  1533. Saiga H, Nishimura J, Kuwata H, Okuyama M, Matsumoto S, Sato S, Matsumoto M, Akira S, Yoshikai Y, Honda K, Yamamoto M, Takeda K: Lipocalin 2-dependent inhibition of mycobacterial growth in alveolar epithelium. J Immunol. 2008, 181: 8521-7.

    CAS  PubMed  Google Scholar 

  1534. Breidbach T, Scory S, Krauth-Siegel RL, Steverding D: Growth inhibition of bloodstream forms of Trypanosoma brucei by the iron chelator deferoxamine. Int J Parasitol. 2002, 32: 473-479.

    CAS  PubMed  Google Scholar 

  1535. Merschjohann K, Steverding D: In vitro growth inhibition of bloodstream forms of Trypanosoma brucei and Trypanosoma congolense by iron chelators. Kinetoplastid Biol Dis. 2006, 5: 3.

    PubMed  PubMed Central  Google Scholar 

  1536. Drakesmith H, Prentice A: Viral infection and iron metabolism. Nat Rev Microbiol. 2008, 6: 541-52.

    CAS  PubMed  Google Scholar 

  1537. Kaprelyants AS, Gottschal JC, Kell DB: Dormancy in non-sporulating bacteria. FEMS Microbiol Rev. 1993, 10: 271-286.

    CAS  PubMed  Google Scholar 

  1538. Kell DB, Kaprelyants AS, Weichart DH, Harwood CL, Barer MR: Viability and activity in readily culturable bacteria: a review and discussion of the practical issues. Antonie van Leeuwenhoek. 1998, 73: 169-187.

    CAS  PubMed  Google Scholar 

  1539. Goris RJ: Mediators of multiple organ failure. Intensive Care Med. 1990, 16 (Suppl 3): S192-6.

    PubMed  Google Scholar 

  1540. Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, Panoskaltsis N: Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006, 355: 1018-28.

    CAS  PubMed  Google Scholar 

  1541. Beal AL, Cerra FB: Multiple Organ Failure Syndrome in the 1990s – Systemic Inflammatory Response and Organ Dysfunction. Jama-Journal of the American Medical Association. 1994, 271: 226-233.

    CAS  Google Scholar 

  1542. Bolton CF: Sepsis and the systemic inflammatory response syndrome: Neuromuscular manifestations. Critical Care Medicine. 1996, 24: 1408-1416.

    CAS  PubMed  Google Scholar 

  1543. Bone RC: Toward a theory regarding the pathogenesis of the systemic inflammatory response syndrome: What we do and do not know about cytokine regulation. Critical Care Medicine. 1996, 24: 163-172.

    CAS  PubMed  Google Scholar 

  1544. Davies MG, Hagen PO: Systemic inflammatory response syndrome. British Journal of Surgery. 1997, 84: 920-935.

    CAS  PubMed  Google Scholar 

  1545. Baue AE, Durham R, Faist E: Systemic inflammatory response syndrome (SIRS), multiple organ dysfunction syndrome (MODS), multiple organ failure (MOF): Are we winning the battle?. Shock. 1998, 10: 79-89.

    CAS  PubMed  Google Scholar 

  1546. Gutteridge JMC, Mitchell J: Redox imbalance in the critically ill. British Medical Bulletin. 1999, 55: 49-75.

    CAS  PubMed  Google Scholar 

  1547. Johnson D, Mayers I: Multiple organ dysfunction syndrome: a narrative review. Can J Anaesth. 2001, 48: 502-9.

    CAS  PubMed  Google Scholar 

  1548. Martin GS, Mannino DM, Eaton S, Moss M: The epidemiology of sepsis in the United States from 1979 through 2000. New England Journal of Medicine. 2003, 348: 1546-1554.

    PubMed  Google Scholar 

  1549. Keel M, Trentz O: Pathophysiology of polytrauma. Injury. 2005, 36: 691-709.

    PubMed  Google Scholar 

  1550. Nguyen HB, Rivers EP, Abrahamian FM, Moran GJ, Abraham E, Trzeciak S, Huang DT, Osborn T, Stevens D, Talan DA: Severe sepsis and septic shock: Review of the literature and emergency department management guidelines. Ann Emergency Med. 2006, 48: 28-54.

    Google Scholar 

  1551. Protti A, Singer M: Bench-to-bedside review: Potential strategies to protect or reverse mitochondrial dysfunction in sepsis-induced organ failure. Critical Care. 2006, 10: 227-233.

    Google Scholar 

  1552. Papathanassoglou EDE, Moynihan JA, Ackerman MH: Does programmed cell death (apoptosis) play a role in the development of multiple organ dysfunction in critically ill patients? A review and a theoretical framework. Critical Care Medicine. 2000, 28: 537-549.

    CAS  PubMed  Google Scholar 

  1553. Festjens N, Berghe Vanden T, Vandenabeele P: Necrosis, a well-orchestrated form of cell demise: Signalling cascades, important mediators and concomitant immune response. Biochim Biophys Acta. 2006, 1757: 1371-1387.

    CAS  PubMed  Google Scholar 

  1554. Golstein P, Kroemer G: Cell death by necrosis: towards a molecular definition. Trends in Biochemical Sciences. 2007, 32: 37-43.

    CAS  PubMed  Google Scholar 

  1555. Goode HF, Webster NR: Free radicals and antioxidants in sepsis. Crit Care Med. 1993, 21: 1770-6.

    CAS  PubMed  Google Scholar 

  1556. Redl H, Gasser H, Schlag G, Marzi I: Involvement of oxygen radicals in shock related cell injury. Br Med Bull. 1993, 49: 556-65.

    CAS  PubMed  Google Scholar 

  1557. Horn KD: Evolving strategies in the treatment of sepsis and systemic inflammatory response syndrome (SIRS). QJM. 1998, 91: 265-77.

    CAS  PubMed  Google Scholar 

  1558. Bulger EM, Maier RV: Antioxidants in critical illness. Arch Surg. 2001, 136: 1201-7.

    CAS  PubMed  Google Scholar 

  1559. Cadenas S, Cadenas AM: Fighting the stranger-antioxidant protection against endotoxin toxicity. Toxicology. 2002, 180: 45-63.

    CAS  PubMed  Google Scholar 

  1560. Victor VM, Rocha M, De la Fuente M: Immune cells: free radicals and antioxidants in sepsis. Int Immunopharmacol. 2004, 4: 327-47.

    CAS  PubMed  Google Scholar 

  1561. Victor VM, Rocha M, Esplugues JV, De la Fuente M: Role of free radicals in sepsis: Antioxidant therapy. Current Pharmaceutical Design. 2005, 11: 3141-3158.

    CAS  PubMed  Google Scholar 

  1562. Vlessis AA, Goldman RK, Trunkey DD: New concepts in the pathophysiology of oxygen metabolism during sepsis. Br J Surg. 1995, 82: 870-6.

    CAS  PubMed  Google Scholar 

  1563. Quinlan GJ, Chen Y, Evans TW, Gutteridge JMC: Iron signalling regulated directly and through oxygen: implications for sepsis and the acute respiratory distress syndrome. Clinical Science. 2001, 100: 169-182.

    CAS  PubMed  Google Scholar 

  1564. Closa D, Folch-Puy E: Oxygen free radicals and the systemic inflammatory response. IUBMB Life. 2004, 56: 185-191.

    CAS  PubMed  Google Scholar 

  1565. Abdelrahman M, Mazzon E, Bauer M, Bauer I, Delbosc S, Cristol JP, Patel NS, Cuzzocrea S, Thiemermann C: Inhibitors of NADPH oxidase reduce the organ injury in hemorrhagic shock. Shock. 2005, 23: 107-14.

    CAS  PubMed  Google Scholar 

  1566. Javadi P, Buchman TG, Stromberg PE, Turnbull IR, Vyas D, Hotchkiss RS, Karl IE, Coopersmith CM: Iron dysregulation combined with aging prevents sepsis-induced apoptosis. J Surg Res. 2005, 128: 37-44.

    CAS  PubMed  PubMed Central  Google Scholar 

  1567. Crimi E, Sica V, Slutsky AS, Zhang HB, Williams-Ignarro S, Ignarro LJ, Napoli C: Role of oxidative stress in experimental sepsis and multisystem organ dysfunction. Free Radical Research. 2006, 40: 665-672.

    CAS  PubMed  Google Scholar 

  1568. Crimi E, Sica V, Williams-Ignarro S, Zhang H, Slutsky AS, Ignarro LJ, Napoli C: The role of oxidative stress in adult critical care. Free Radic Biol Med. 2006, 40: 398-406.

    CAS  PubMed  Google Scholar 

  1569. Lemineur T, Deby-Dupont G, Preiser JC: Biomarkers of oxidative stress in critically ill patients: what should be measured, when and how?. Curr Op Clin Nutr Metab Care. 2006, 9: 704-710.

    CAS  Google Scholar 

  1570. Lagan AL, Melley DD, Evans TW, Quinlan GJ: Pathogenesis of the systemic inflammatory syndrome and acute lung injury: role of iron mobilization and decompartmentalization. Am J Physiol Lung Cell Mol Physiol. 2008, 294: L161-74.

    CAS  PubMed  Google Scholar 

  1571. Galley HF, Webster NR: Elevated serum bleomycin-detectable iron concentrations in patients with sepsis syndrome. Intensive Care Med. 1996, 22: 226-9.

    CAS  PubMed  Google Scholar 

  1572. Galley HF, Davies MJ, Webster NR: Ascorbyl radical formation in patients with sepsis: effect of ascorbate loading. Free Radic Biol Med. 1996, 20: 139-43.

    CAS  PubMed  Google Scholar 

  1573. Galley HF, Howdle PD, Walker BE, Webster NR: The effects of intravenous antioxidants in patients with septic shock. Free Radic Biol Med. 1997, 23: 768-74.

    CAS  PubMed  Google Scholar 

  1574. Eaton JW, Qian MW: Molecular bases of cellular iron toxicity. Free Radical Biology and Medicine. 2002, 32: 833-840.

    CAS  PubMed  Google Scholar 

  1575. Yu Z, Persson HL, Eaton JW, Brunk UT: Intralysosomal iron: a major determinant of oxidant-induced cell death. Free Radic Biol Med. 2003, 34: 1243-52.

    CAS  PubMed  Google Scholar 

  1576. Guicciardi ME, Leist M, Gores GJ: Lysosomes in cell death. Oncogene. 2004, 23: 2881-90.

    CAS  PubMed  Google Scholar 

  1577. Terman A, Kurz T, Gustafsson B, Brunk UT: Lysosomal labilization. IUBMB Life. 2006, 58: 531-9.

    CAS  PubMed  Google Scholar 

  1578. Vulcano M, Meiss RP, Isturiz MA: Deferoxamine reduces tissue injury and lethality in LPS-treated mice. Int J Immunopharmacol. 2000, 22: 635-44.

    CAS  PubMed  Google Scholar 

  1579. Cuzzocrea S, Rossi A, Pisano B, Di Paola R, Genovese T, Patel NS, Cuzzocrea E, Ianaro A, Sautebin L, Fulia F, Chatterjee PK, Caputi AP, Thiemermann C: Pyrrolidine dithiocarbamate attenuates the development of organ failure induced by zymosan in mice. Intensive Care Med. 2003, 29: 2016-25.

    PubMed  Google Scholar 

  1580. Persson HL, Yu Z, Tirosh O, Eaton JW, Brunk UT: Prevention of oxidant-induced cell death by lysosomotropic iron chelators. Free Radic Biol Med. 2003, 34: 1295-305.

    CAS  PubMed  Google Scholar 

  1581. Messaris E, Antonakis PT, Memos N, Chatzigianni E, Leandros E, Konstadoulakis MM: Deferoxamine administration in septic animals: improved survival and altered apoptotic gene expression. Int Immunopharmacol. 2004, 4: 455-9.

    CAS  PubMed  Google Scholar 

  1582. Tracey KJ, Cerami A: Tumor Necrosis Factor: a pleiotropic cytokine and therapeutic target. Annu Rev Med. 1994, 45: 491-503.

    CAS  PubMed  Google Scholar 

  1583. Opal SM, Huber CE: Bench-to-bedside review: Toll-like receptors and their role in septic shock. Critical Care. 2002, 6: 125-136.

    PubMed  PubMed Central  Google Scholar 

  1584. Hopkins P, Cohen J: Toll-like receptors: the key to the stable door?. Crit Care. 2002, 6: 99-101.

    PubMed  PubMed Central  Google Scholar 

  1585. Reynolds FD, Dauchy R, Blask D, Dietz PA, Lynch D, Zuckerman R: The pineal gland hormone melatonin improves survival in a rat model of sepsis/shock induced by zymosan A. Surgery. 2003, 134: 474-479.

    PubMed  Google Scholar 

  1586. Carrillo-Vico A, Lardone PJ, Naji L, Fernández-Santos JM, Martín-Lacave I, Guerrero JM, Calvo JR: Beneficial pleiotropic actions of melatonin in an experimental model of septic shock in mice: regulation of pro-/anti-inflammatory cytokine network, protection against oxidative damage and anti-apoptotic effects. J Pineal Res. 2005, 39: 400-408.

    CAS  PubMed  Google Scholar 

  1587. Escames G, Acuna-Castroviejo D, López LC, Tan DX, Maldonado MD, Sanchez-Hidalgo M, León J, Reiter RJ: Pharmacological utility of melatonin in the treatment of septic shock: experimental and clinical evidence. J Pharm Pharmacol. 2006, 58: 1153-1165.

    CAS  PubMed  Google Scholar 

  1588. Cuzzocrea S, Riley DP, Caputi AP, Salvemini D: Antioxidant therapy: a new pharmacological approach in shock, inflammation, and ischemia/reperfusion injury. Pharmacol Rev. 2001, 53: 135-59.

    CAS  PubMed  Google Scholar 

  1589. Nathens AB, Neff MJ, Jurkovich GJ, Klotz P, Farver K, Ruzinski JT, Radella F, Garcia I, Maier RV: Randomized, prospective trial of antioxidant supplementation in critically ill surgical patients. Annals of Surgery. 2002, 236: 814-822.

    PubMed  PubMed Central  Google Scholar 

  1590. Cuzzocrea S, Thiemermann C, Salvemini D: Potential therapeutic effect of antioxidant therapy in shock and inflammation. Current Medicinal Chemistry. 2004, 11: 1147-1162.

    CAS  PubMed  Google Scholar 

  1591. Di Paola R, Mazzon E, Muia C, Crisafulli C, Genovese T, Di Bella P, Esposito E, Menegazzi M, Meli R, Suzuki H, Cuzzocrea S: Green tea polyphenol extract attenuates zymosan-induced non-septic shock in mice. Shock. 2006, 26: 402-9.

    CAS  PubMed  Google Scholar 

  1592. Ritter C, Andrades ME, Reinke A, Menna-Barreto S, Moreira JC, Dal-Pizzol F: Treatment with N-acetylcysteine plus deferoxamine protects rats against oxidative stress and improves survival in sepsis. Crit Care Med. 2004, 32: 342-9.

    CAS  PubMed  Google Scholar 

  1593. Ritter C, da Cunha AA, Echer IC, Andrades M, Reinke A, Lucchiari N, Rocha J, Streck EL, Menna-Barreto S, Moreira JC, Dal-Pizzol F: Effects of N-acetylcysteine plus deferoxamine in lipopolysaccharide-induced acute lung injury in the rat. Crit Care Med. 2006, 34: 471-7.

    CAS  PubMed  Google Scholar 

  1594. Pinho RA, Silveira PC, Silva LA, Luiz Streck E, Dal-Pizzol F, JC FM: N-acetylcysteine and deferoxamine reduce pulmonary oxidative stress and inflammation in rats after coal dust exposure. Environ Res. 2005, 99: 355-60.

    CAS  PubMed  Google Scholar 

  1595. Teixeira KC, Soares FS, Rocha LG, Silveira PC, Silva LA, Valenca SS, Pizzol FD, Streck EL, Pinho RA: Attenuation of bleomycin-induced lung injury and oxidative stress by N-acetylcysteine plus deferoxamine. Pulm Pharmacol Ther. 2008, 21: 309-16.

    CAS  PubMed  Google Scholar 

  1596. Abdelrahman M, Sharples EJ, McDonald MC, Collin M, Patel NS, Yaqoob MM, Thiemermann C: Erythropoietin attenuates the tissue injury associated with hemorrhagic shock and myocardial ischemia. Shock. 2004, 22: 63-9.

    CAS  PubMed  Google Scholar 

  1597. Cuzzocrea S, Di Paola R, Mazzon E, Patel NS, Genovese T, Muia C, Crisafulli C, Caputi AP, Thiemermann C: Erythropoietin reduces the development of nonseptic shock induced by zymosan in mice. Crit Care Med. 2006, 34: 1168-77.

    CAS  PubMed  Google Scholar 

  1598. Lovitt RW, Kell DB, Morris JG: Proline reduction by Clostridium sporogenes is coupled to vectorial proton ejection. FEMS Microbiology Letters. 1986, 36: 269-273.

    CAS  Google Scholar 

  1599. Kell DB, Mendes P: Snapshots of systems: metabolic control analysis and biotechnology in the post-genomic era. Technological and Medical Implications of Metabolic Control Analysis. Edited by: Cornish-Bowden A, Cárdenas ML. 2000, Kluwer Academic Publishers, Dordrecht, 3-25. [http://dbkgroup.org/WhitePapers/mcabio.htm]

    Google Scholar 

  1600. Wikström MKF: The different cytochrome b components in the respiratory chain of animal mitochondria and their role in electron transport and energy conservation. Biochim Biophys Acta. 1973, 301: 155-93.

    PubMed  Google Scholar 

  1601. John P, Papa S: Rapid oxygen-induced reduction of b-type cytochromes in Paracoccus denitrificans. FEBS Lett. 1978, 85: 179-82.

    CAS  PubMed  Google Scholar 

  1602. Reif DW: Ferritin as a source of iron for oxidative damage. Free Rad Biol Med. 1992, 12: 417-427.

    CAS  PubMed  Google Scholar 

  1603. Moison RM, Bloemhof FE, Geerdink JA, de Beaufort AJ, Berger HM: The capacity of different infusion fluids to lower the prooxidant activity of plasma iron: an important factor in resuscitation?. Transfusion. 2000, 40: 1346-51.

    CAS  PubMed  Google Scholar 

  1604. Fábián I, Csordás V: Metal ion catalyzed autoxidation reactions: Kinetics and mechanisms. Adv Inorg Chem. 2003, 54: 395-461.

    Google Scholar 

  1605. Duarte TL, Almeida GM, Jones GD: Investigation of the role of extracellular H2O2 and transition metal ions in the genotoxic action of ascorbic acid in cell culture models. Toxicol Lett. 2007, 170: 57-65.

    CAS  PubMed  Google Scholar 

  1606. Wang S, Geraci G, Kuhlmann MK, Levin NW, Handelman GJ: Chemical reactions of vitamin C with intravenous-iron formulations. Nephrol Dial Transplant. 2008, 23: 120-5.

    PubMed  Google Scholar 

  1607. Yagi K, Ishida N, Komura S, Ohishi N, Kusai M, Kohno M: Generation of hydroxyl radical from linoleic acid hydroperoxide in the presence of epinephrine and iron. Biochem Biophys Res Commun. 1992, 183: 945-51.

    CAS  PubMed  Google Scholar 

  1608. Everse J, Hsia N: The toxicities of native and modified hemoglobins. Free Radical Biology and Medicine. 1997, 22: 1075-1099.

    CAS  PubMed  Google Scholar 

  1609. Medina I, Tombo I, Satue-Gracia MT, German JB, Frankel EN: Effects of natural phenolic compounds on the antioxidant activity of lactoferrin in liposomes and oil-in-water emulsions. J Agric Food Chem. 2002, 50: 2392-9.

    CAS  PubMed  Google Scholar 

  1610. Hoppe M, Hulthen L, Hallberg L: The relative bioavailability in humans of elemental iron powders for use in food fortification. Eur J Nutrition. 2006, 45: 37-44.

    CAS  Google Scholar 

  1611. Goodell B, Jellison J, Liu J, Daniel G, Paszczynski A, Fekete F, Krishnamurthy S, Jun L, Xu G: Low molecular weight chelators and phenolic compounds isolated from wood decay fungi and their role in the fungal biodegradation of wood. J Biotechnol. 1997, 53: 133-162.

    CAS  Google Scholar 

  1612. Mályusz M, Kahler W, Gronow G: Hippurate metabolism as a hydroxyl radical trapping mechanism in the rat kidney. Kidney Blood Press Res. 2001, 24: 149-58.

    PubMed  Google Scholar 

  1613. Brzezinski A: Melatonin in humans. N Engl J Med. 1997, 336: 186-95.

    CAS  PubMed  Google Scholar 

  1614. Reiter R, Tang L, Garcia JJ, MunozHoyos A: Pharmacological actions of melatonin in oxygen radical pathophysiology. Life Sci. 1997, 60: 2255-2271.

    CAS  PubMed  Google Scholar 

  1615. Beyer CE, Steketee JD, Saphier D: Antioxidant properties of melatonin – An emerging mystery. Biochem Pharmacol. 1998, 56: 1265-1272.

    CAS  PubMed  Google Scholar 

  1616. Reiter RJ: Oxidative damage in the central nervous system: protection by melatonin. Progr Neurobiol. 1998, 56: 359-384.

    CAS  Google Scholar 

  1617. Cuzzocrea S, Costantino G, Gitto E, Mazzon E, Fulia F, Serraino I, Cordaro S, Barberi I, De Sarro A, Caputi AP: Protective effects of melatonin in ischemic brain injury. J Pineal Res. 2000, 29: 217-27.

    CAS  PubMed  Google Scholar 

  1618. Karbownik M, Lewinski A, Reiter RJ: Anticarcinogenic actions of melatonin which involve antioxidative processes: comparison with other antioxidants. International Journal of Biochemistry & Cell Biology. 2001, 33: 735-753.

    CAS  Google Scholar 

  1619. Letechipia-Vallejo G, Gonzalez-Burgos I, Cervantes M: Neuroprotective effect of melatonin on brain damage induced by acute global cerebral ischemia in cats. Arch Med Res. 2001, 32: 186-92.

    CAS  PubMed  Google Scholar 

  1620. Okatani Y, Wakatsuki A, Shinohara K, Taniguchi K, Fukaya T: Melatonin protects against oxidative mitochondrial damage induced in rat placenta by ischemia and reperfusion. J Pineal Res. 2001, 31: 173-8.

    CAS  PubMed  Google Scholar 

  1621. Reiter RJ, Tan DX, Manchester LC, Qi WB: Biochemical reactivity of melatonin with reactive oxygen and nitrogen species – A review of the evidence. Cell Biochemistry and Biophysics. 2001, 34: 237-256.

    CAS  PubMed  Google Scholar 

  1622. Allegra M, Reiter RJ, Tan DX, Gentile C, Tesoriere L, Livrea MA: The chemistry of melatonin's interaction with reactive species. Journal of Pineal Research. 2003, 34: 1-10.

    CAS  PubMed  Google Scholar 

  1623. Cheung RTF: The utility of melatonin in reducing cerebral damage resulting from ischemia and reperfusion. Journal of Pineal Research. 2003, 34: 153-160.

    CAS  PubMed  Google Scholar 

  1624. Okatani Y, Wakatsuki A, Reiter RJ, Enzan H, Miyahara Y: Protective effect of melatonin against mitochondrial injury induced by ischemia and reperfusion of rat liver. Eur J Pharmacol. 2003, 469: 145-152.

    CAS  PubMed  Google Scholar 

  1625. Pei Z, Pang SF, Cheung RTF: Administration of melatonin after onset of ischemia reduces the volume of cerebral infarction in a rat middle cerebral artery occlusion stroke model. Stroke. 2003, 34: 770-775.

    CAS  PubMed  Google Scholar 

  1626. Reiter RJ, Tan DX: Melatonin: a novel protective agent against oxidative injury of the ischemic/reperfused heart. Cardiovascular Research. 2003, 58: 10-19.

    CAS  PubMed  Google Scholar 

  1627. Reiter RJ: Melatonin: clinical relevance. Best Pract Res Clin Endocrinol Metab. 2003, 17: 273-85.

    CAS  PubMed  Google Scholar 

  1628. Weiss JF, Landauer MR: Protection against ionizing radiation by antioxidant nutrients and phytochemicals. Toxicology. 2003, 189: 1-20.

    CAS  PubMed  Google Scholar 

  1629. Macleod MR, O'Collins T, Horky LL, Howells DW, Donnan GA: Systematic review and meta-analysis of the efficacy of melatonin in experimental stroke. J Pineal Res. 2005, 38: 35-41.

    CAS  PubMed  Google Scholar 

  1630. Srinivasan V, Pandi-Perumal SR, Maestroni GJM, Esquifino AI, Hardeland R, Cardinali DP: Role of melatonin in neurodegenerative diseases. Neurotox Res. 2005, 7: 293-318.

    CAS  PubMed  Google Scholar 

  1631. Hardeland R, Pandi-Perumal SR, Cardinali DP: Melatonin. International Journal of Biochemistry & Cell Biology. 2006, 38: 313-316.

    CAS  Google Scholar 

  1632. Grootveld M, Halliwell B: Aromatic hydroxylation as a potential measure of hydroxyl-radical formation in vivo. Identification of hydroxylated derivatives of salicylate in human body fluids. Biochem J. 1986, 237: 499-504.

    CAS  PubMed  PubMed Central  Google Scholar 

  1633. Roob JM, Khoschsorur G, Tiran A, Horina JH, Holzer H, Winklhofer-Roob BM: Vitamin E attenuates oxidative stress induced by intravenous iron in patients on hemodialysis. J Am Soc Nephrol. 2000, 11: 539-49.

    CAS  PubMed  Google Scholar 

  1634. Oldham KM, Bowen PE: Oxidative stress in critical care: is antioxidant supplementation beneficial?. J Am Diet Assoc. 1998, 98: 1001-8.

    CAS  PubMed  Google Scholar 

  1635. Rehman A, Collis CS, Yang M, Kelly M, Diplock AT, Halliwell B, Rice-Evans C: The effects of iron and vitamin C co-supplementation on oxidative damage to DNA in healthy volunteers. Biochem Biophys Res Comm. 1998, 246: 293-298.

    CAS  PubMed  Google Scholar 

  1636. Gilgun-Sherki Y, Rosenbaum Z, Melamed E, Offen D: Antioxidant therapy in acute central nervous system injury: current state. Pharmacol Rev. 2002, 54: 271-84.

    CAS  PubMed  Google Scholar 

  1637. Miller ER, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E: Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med. 2005, 142: 37-46.

    CAS  PubMed  Google Scholar 

  1638. Rodrigo R, Guichard C, Charles R: Clinical pharmacology and therapeutic use of antioxidant vitamins. Fund Clin Pharmacol. 2007, 21: 111-127.

    CAS  Google Scholar 

  1639. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C: Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. JAMA. 2007, 297: 842-57.

    CAS  PubMed  Google Scholar 

  1640. Orrell RW, Lane RJ, Ross M: Antioxidant treatment for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2007, CD002829.

    Google Scholar 

  1641. Stephens NG, Parsons A, Schofield PM, Kelly F, Cheeseman K, Mitchinson MJ: Randomised controlled trial of vitamin E in patients with coronary disease: Cambridge Heart Antioxidant Study (CHAOS). Lancet. 1996, 347: 781-6.

    CAS  PubMed  Google Scholar 

  1642. Yusuf S, Dagenais G, Pogue J, Bosch J, Sleight P: Vitamin E supplementation and cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med. 2000, 342: 154-60.

    CAS  PubMed  Google Scholar 

  1643. Lee DH, Folsom AR, Harnack L, Halliwell B, Jacobs DR: Does supplemental vitamin C increase cardiovascular disease risk in women with diabetes?. Am J Clin Nutr. 2004, 80: 1194-200.

    CAS  PubMed  Google Scholar 

  1644. Williams KJ, Fisher EA: Oxidation, lipoproteins, and atherosclerosis: which is wrong, the antioxidants or the theory?. Curr Opin Clin Nutr Metab Care. 2005, 8: 139-46.

    CAS  PubMed  Google Scholar 

  1645. Papaharalambus CA, Griendling KK: Basic mechanisms of oxidative stress and reactive oxygen species in cardiovascular injury. Trends Cardiovasc Med. 2007, 17: 48-54.

    CAS  PubMed  PubMed Central  Google Scholar 

  1646. O'Collins VE, Macleod MR, Donnan GA, Horky LL, Worp van der BH, Howells DW: 1,026 experimental treatments in acute stroke. Ann Neurol. 2006, 59: 467-77.

    PubMed  Google Scholar 

  1647. Evans JR, Henshaw K: Antioxidant vitamin and mineral supplements for preventing age-related macular degeneration. Cochrane Database Syst Rev. 2008, CD000253.

    Google Scholar 

  1648. Jeyabalan A, Caritis SN: Antioxidants and the prevention of preeclampsia – Unresolved issues. N Engl J Med. 2006, 354: 1841-1843.

    CAS  PubMed  Google Scholar 

  1649. Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH: Vitamin C and vitamin E in pregnant women at risk for pre-eclampsia (VIP trial): randomised placebo-controlled trial. Lancet. 2006, 367: 1145-54.

    CAS  PubMed  Google Scholar 

  1650. Rumbold AR, Crowther CA, Haslam RR, Dekker GA, Robinson JS: Vitamins C and E and the risks of preeclampsia and perinatal complications. N Engl J Med. 2006, 354: 1796-806.

    CAS  PubMed  Google Scholar 

  1651. Spinnato JA: New therapies in the prevention of preeclampsia. Current Opinion in Obstetrics & Gynecology. 2006, 18: 601-604.

    Google Scholar 

  1652. Rumbold A, Duley L, Crowther CA, Haslam RR: Antioxidants for preventing pre-eclampsia. Cochrane Database Syst Rev. 2008, CD004227.

    Google Scholar 

  1653. Coleman NA: Antioxidants in critical care medicine. Env Toxicol Pharmacol. 2001, 10: 183-188.

    CAS  Google Scholar 

  1654. Bayne AC, Sohal RS: Effects of superoxide dismutase/catalase mimetics on life span and oxidative stress resistance in the housefly, Musca domestica. Free Radic Biol Med. 2002, 32: 1229-34.

    CAS  PubMed  Google Scholar 

  1655. Magwere T, West M, Riyahi K, Murphy MP, Smith RA, Partridge L: The effects of exogenous antioxidants on lifespan and oxidative stress resistance in Drosophila melanogaster. Mech Ageing Dev. 2006, 127: 356-70.

    CAS  PubMed  Google Scholar 

  1656. Omenn GS, Goodman GE, Thornquist MD, Balmes J, Cullen MR, Glass A, Keogh JP, Meyskens FL, Valanis B, Williams JH, Barnhart S, Hammar S: Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N Engl J Med. 1996, 334: 1150-5.

    CAS  PubMed  Google Scholar 

  1657. Bailey DM, Raman S, McEneny J, Young IS, Parham KL, Hullin DA, Davies B, McKeeman G, McCord JM, Lewis MH: Vitamin C prophylaxis promotes oxidative lipid damage during surgical ischemia-reperfusion. Free Radical Biology and Medicine. 2006, 40: 591-600.

    CAS  PubMed  Google Scholar 

  1658. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C: Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database of Systematic Reviews 2008. 2008, CD007176.

    Google Scholar 

  1659. Long LH, Clement MV, Halliwell B: Artifacts in cell culture: Rapid generation of hydrogen peroxide on addition of (-)-epigallocatechin, (-)-epigallocatechin gallate, (+)-catechin, and quercetin to commonly used cell culture media. Biochem Biophys Res Comm. 2000, 273: 50-53.

    CAS  PubMed  Google Scholar 

  1660. Hininger I, Waters R, Osman M, Garrel C, Fernholz K, Roussel AM, Anderson RA: Acute prooxidant effects of vitamin C in EDTA chelation therapy and long-term antioxidant benefits of therapy. Free Rad Biol Med. 2005, 38: 1565-1570.

    CAS  PubMed  Google Scholar 

  1661. Szewczyk A, Wojtczak L: Mitochondria as a pharmacological target. Pharmacol Rev. 2002, 54: 101-27.

    CAS  PubMed  Google Scholar 

  1662. Murphy MP, Smith RA: Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu Rev Pharmacol Toxicol. 2007, 47: 629-56.

    CAS  PubMed  Google Scholar 

  1663. Jou MJ: Pathophysiological and pharmacological implications of mitochondria-targeted reactive oxygen species generation in astrocytes. Adv Drug Deliv Rev. 2008, 60: 1512-26.

    CAS  PubMed  Google Scholar 

  1664. Balcerczyk A, Sowa K, Bartou G: Metal chelators react also with reactive oxygen and nitrogen species. Biochem Biophys Res Comm. 2007, 352: 522-525.

    CAS  PubMed  Google Scholar 

  1665. Hider RC, Zhou T: The design of orally active iron chelators. Ann N Y Acad Sci. 2005, 1054: 141-54.

    CAS  PubMed  Google Scholar 

  1666. Marcus RA: Theory of Oxidation-Reduction Reactions Involving Electron Transfer .1. J Chem Phys. 1956, 24: 966-978.

    CAS  Google Scholar 

  1667. Marcus RA: Chemical + Electrochemical Electron-Transfer Theory. Annu Rev Phys Chem. 1964, 15: 155.

    CAS  Google Scholar 

  1668. Chou M, Creutz C, Sutin N: Rate constants and activation parameters for outer-sphere electron transfer reactions and comparisons with predictions of Marcus theory. JACS. 1977, 99: 5615-5623.

    CAS  Google Scholar 

  1669. Marcus RA, Sutin N: Electron transfers in chemistry and biology. Biochim Biophys Acta. 1985, 811: 265-322.

    CAS  Google Scholar 

  1670. Liu ZD, Hider RC: Design of iron chelators with therapeutic application. Coord Chem Rev. 2002, 232: 151-171.

    CAS  Google Scholar 

  1671. Liu ZD, Hider RC: Design of clinically useful iron(III)-selective chelators. Medicinal Res Rev. 2002, 22: 26-64.

    CAS  Google Scholar 

  1672. Richardson DR, Ponka P: Development of iron chelators to treat iron overload disease and their use as experimental tools to probe intracellular iron metabolism. Am J Hematol. 1998, 58: 299-305.

    CAS  PubMed  Google Scholar 

  1673. Aouad F, Florence A, Zhang Y, Collins F, Henry C, Ward RJ, Crichton RR: Evaluation of new iron chelators and their therapeutic potential. Inorganica Chimica Acta. 2002, 339: 470-480.

    CAS  Google Scholar 

  1674. Bush AI: Metal complexing agents as therapies for Alzheimer's disease. Neurobiology of Aging. 2002, 23: 1031-1038.

    CAS  PubMed  Google Scholar 

  1675. Crichton RR, Florence A, Ward RJ: Aluminium and iron in the brain – prospects for chelation. Coordination Chemistry Reviews. 2002, 228: 365-371.

    CAS  Google Scholar 

  1676. Chaston TB, Richardson DR: Iron chelators for the treatment of iron overload disease: relationship between structure, redox activity, and toxicity. Amer J Hematol. 2003, 73: 200-210.

    CAS  Google Scholar 

  1677. Crichton RR, Ward RJ: Iron chelators and their therapeutic potential. Metal Ions in Biological Systems, Metal Ions and Their Complexes in Medication, Metal Ions in Biological Systems. 2004, 41: 185-219.

    CAS  Google Scholar 

  1678. Bernhardt PV: Coordination chemistry and biology of chelators for the treatment of iron overload disorders. Dalton Trans. 2007, 3214-3220.

    Google Scholar 

  1679. Leeson PD, Springthorpe B: The influence of drug-like concepts on decision-making in medicinal chemistry. Nat Rev Drug Discov. 2007, 6: 881-90.

    CAS  PubMed  Google Scholar 

  1680. Imbert M, Bechet M, Blondeau R: Comparison of the Main Siderophores Produced by Some Species of Streptomyces. Current Microbiology. 1995, 31: 129-133.

    CAS  Google Scholar 

  1681. Flores FJ, Rincón J, Martín JF: Characterization of the iron-regulated desA promoter of Streptomyces pilosus as a system for controlled gene expression in actinomycetes. Microb Cell Fact. 2003, 2: 5.

    PubMed  PubMed Central  Google Scholar 

  1682. Catnach SM, Fairclough PD, Hammond SM: Intestinal absorption of peptide drugs: advances in our understanding and clinical implications. Gut. 1994, 35: 441-4.

    CAS  PubMed  PubMed Central  Google Scholar 

  1683. Bailey PD, Boyd CA, Bronk JR, Collier ID, Meredith D, Morgan KM, Temple CS: How to make drugs orally active: a substrate template for peptide transporter PepT1. Angew Chem Int Ed Engl. 2000, 39: 505-508.

    PubMed  Google Scholar 

  1684. Terada T, Inui K: Peptide transporters: structure, function, regulation and application for drug delivery. Curr Drug Metab. 2004, 5: 85-94.

    CAS  PubMed  Google Scholar 

  1685. Bailey PD, Boyd CA, Collier ID, George JG, Kellett GL, Meredith D, Morgan KM, Pettecrew R, Price RA, Pritchard RG: Conformational and spacial preferences for substrates of PepT1. Chem Commun (Camb). 2005, 5352-4.

    Google Scholar 

  1686. Maggio A: Light and shadows in the iron chelation treatment of haematological diseases. Br J Haematol. 2007, 138: 407-21.

    CAS  PubMed  Google Scholar 

  1687. Kontoghiorghes GJ, Neocleous K, Kolnagou A: Benefits and risks of deferiprone in iron overload in Thalassaemia and other conditions: comparison of epidemiological and therapeutic aspects with deferoxamine. Drug Saf. 2003, 26: 553-84.

    CAS  PubMed  Google Scholar 

  1688. Anderson LJ, Wonke B, Prescott E, Holden S, Walker JM, Pennell DJ: Comparison of effects of oral deferiprone and subcutaneous desferrioxamine on myocardial iron concentrations and ventricular function in beta-thalassaemia. Lancet. 2002, 360: 516-20.

    CAS  PubMed  Google Scholar 

  1689. Heinz U, Hegetschweiler K, Acklin P, Faller B, Lattmann R, Schnebli HP: 4-[3,5-Bis(2-hydroxyphenyl)-1,2,4-triazol-1-yl]-benzoic acid: a novel efficient and selective iron(III) complexing agent. Angew Chem Int Ed Engl. 1999, 38: 2568-2570.

    CAS  PubMed  Google Scholar 

  1690. Nick H, Acklin P, Lattmann R, Buehlmayer P, Hauffe S, Schupp J, Alberti D: Development of tridentate iron chelators: From desferrithiocin to ICL670. Curr Med Chem. 2003, 10: 1065-1076.

    CAS  PubMed  Google Scholar 

  1691. Nisbet-Brown E, Olivieri NF, Giardina PJ, Grady RW, Neufeld EJ, Sechaud R, Krebs-Brown AJ, Anderson JR, Alberti D, Sizer KC, Nathan DG: Effectiveness and safety of ICL670 in iron-loaded patients with thalassaemia: a randomised, double-blind, placebo-controlled, dose-escalation trial. Lancet. 2003, 361: 1597-602.

    CAS  PubMed  Google Scholar 

  1692. Kontoghiorghes GJ: Effects of ICL670 (deferasirox) on cardiac iron concentrations. Lancet. 2005, 366: 804.

    PubMed  Google Scholar 

  1693. Cappellini MD, Cohen A, Piga A, Bejaoui M, Perrotta S, Agaoglu L, Aydinok Y, Kattamis A, Kilinc Y, Porter J, Capra M, Galanello R, Fattoum S, Drelichman G, Magnano C, Verissimo M, Athanassiou-Metaxa M, Giardina P, Kourakli-Symeonidis A, Janka-Schaub G, Coates T, Vermylen C, Olivieri N, Thuret I, Opitz H, Ressayre-Djaffer C, Marks P, Alberti D: A phase 3 study of deferasirox (ICL670), a once-daily oral iron chelator, in patients with beta-thalassemia. Blood. 2006, 107: 3455-62.

    CAS  PubMed  Google Scholar 

  1694. Chantrel-Groussard K, Gaboriau F, Pasdeloup N, Havouis R, Nick H, Pierre JL, Brissot P, Lescoat G: The new orally active iron chelator ICL670A exhibits a higher antiproliferative effect in human hepatocyte cultures than O-trensox. Eur J Pharmacol. 2006, 541: 129-137.

    CAS  PubMed  Google Scholar 

  1695. Glickstein H, El RB, Link G, Breuer W, Konijn AM, Hershko C, Nick H, Cabantchik ZI: Action of chelators in iron-loaded cardiac cells: Accessibility to intracellular labile iron and functional consequences. Blood. 2006, 108: 3195-203.

    CAS  PubMed  Google Scholar 

  1696. Kontoghiorghes GJ: Future chelation monotherapy and combination therapy strategies in thalassemia and other conditions. comparison of deferiprone deferoxamine, ICL670, GT56-252, L1NAll and starch deferoxamine polymers. Hemoglobin. 2006, 30: 329-47.

    CAS  PubMed  Google Scholar 

  1697. Piga A, Galanello R, Forni GL, Cappellini MD, Origa R, Zappu A, Donato G, Bordone E, Lavagetto A, Zanaboni L, Sechaud R, Hewson N, Ford JM, Opitz H, Alberti D: Randomized phase II trial of deferasirox (Exjade, ICL670), a once-daily, orally-administered iron chelator, in comparison to deferoxamine in thalassemia patients with transfusional iron overload. Haematologica. 2006, 91: 873-80.

    CAS  PubMed  Google Scholar 

  1698. Porter JB: Deferasirox: An effective once-daily orally active iron chelator. Drugs of Today. 2006, 42: 623-637.

    CAS  PubMed  Google Scholar 

  1699. VanOrden HE, Hagemann TM: Deferasirox–an oral agent for chronic iron overload. Ann Pharmacother. 2006, 40: 1110-7.

    CAS  PubMed  Google Scholar 

  1700. Nick H: Iron chelation, quo vadis?. Curr Opin Chem Biol. 2007, 11: 419-23.

    CAS  PubMed  Google Scholar 

  1701. Yang LP, Keam SJ, Keating GM: Deferasirox: a review of its use in the management of transfusional chronic iron overload. Drugs. 2007, 67: 2211-30.

    CAS  PubMed  Google Scholar 

  1702. Vermylen C: What is new in iron overload?. Eur J Pediatr. 2008, 167: 377-81.

    PubMed  Google Scholar 

  1703. Dobson PD, Patel Y, Kell DB: "Metabolite-likeness" as a criterion in the design and selection of pharmaceutical drug libraries. Drug Disc Today. 2008.

    Google Scholar 

  1704. Borisy AA, Elliott PJ, Hurst NW, Lee MS, Lehar J, Price ER, Serbedzija G, Zimmermann GR, Foley MA, Stockwell BR, Keith CT: Systematic discovery of multicomponent therapeutics. Proc Natl Acad Sci USA. 2003, 100: 7977-82.

    CAS  PubMed  PubMed Central  Google Scholar 

  1705. Keith CT, Borisy AA, Stockwell BR: Multicomponent therapeutics for networked systems. Nat Rev Drug Discov. 2005, 4: 71-8.

    CAS  PubMed  Google Scholar 

  1706. Costello F, Stuve O, Weber MS, Zamvil SS, Frohman E: Combination therapies for multiple sclerosis: scientific rationale, clinical trials, and clinical practice. Curr Opin Neurol. 2007, 20: 281-5.

    CAS  PubMed  Google Scholar 

  1707. Lehár J, Zimmermann GR, Krueger AS, Molnar RA, Ledell JT, Heilbut AM, Short GF, Giusti LC, Nolan GP, Magid OA, Lee MS, Borisy AA, Stockwell BR, Keith CT: Chemical combination effects predict connectivity in biological systems. Mol Syst Biol. 2007, 3: 80.

    PubMed  PubMed Central  Google Scholar 

  1708. Zimmermann GR, Lehár J, Keith CT: Multi-target therapeutics: when the whole is greater than the sum of the parts. Drug Discov Today. 2007, 12: 34-42.

    CAS  PubMed  Google Scholar 

  1709. Aruoma OI, Halliwell B: The iron-binding and hydroxyl radical scavenging action of anti-inflammatory drugs. Xenobiotica. 1988, 18: 459-70.

    CAS  PubMed  Google Scholar 

  1710. Cherny RA, Atwood CS, Xilinas ME, Gray DN, Jones WD, McLean CA, Barnham KJ, Volitakis I, Fraser FW, Kim YS, Huang XD, Goldstein LE, Moir RD, Lim JT, Beyreuther K, Zheng H, Tanzi RE, Masters CL, Bush AI: Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer's disease transgenic mice. Neuron. 2001, 30: 665-676.

    CAS  PubMed  Google Scholar 

  1711. Di Vaira M, Bazzicalupi C, Orioli P, Messori L, Bruni B, Zatta P: Clioquinol, a drug for Alzheimer's disease specifically interfering with brain metal metabolism: structural characterization of its zinc(II) and copper(II) complexes. Inorg Chem. 2004, 43: 3795-7.

    CAS  PubMed  Google Scholar 

  1712. Shiraki H: The neuropathology of subacute myelo-optico-neuropathy, "SMON", in the humans: –with special reference to the quinoform intoxication. Jpn J Med Sci Biol. 1975, 28 (Suppl): 101-64.

    PubMed  Google Scholar 

  1713. Arbiser JL, Kraeft SK, van Leeuwen R, Hurwitz SJ, Selig M, Dickersin GR, Flint A, Byers HR, Chen LB: Clioquinol-zinc chelate: a candidate causative agent of subacute myelo-optic neuropathy. Mol Med. 1998, 4: 665-70.

    CAS  PubMed  PubMed Central  Google Scholar 

  1714. Sai Y: Biochemical and molecular pharmacological aspects of transporters as determinants of drug disposition. Drug Metab Pharmacokinet. 2005, 20: 91-9.

    CAS  PubMed  Google Scholar 

  1715. Richardson DR, Ponka P: Orally effective iron chelators for the treatment of iron overload disease: the case for a further look at pyridoxal isonicotinoyl hydrazone and its analogs. J Lab Clin Med. 1998, 132: 351-2.

    CAS  PubMed  Google Scholar 

  1716. Richardson DR, Ponka P: Pyridoxal isonicotinoyl hydrazone and its analogs: potential orally effective iron-chelating agents for the treatment of iron overload disease. J Lab Clin Med. 1998, 131: 306-15.

    CAS  PubMed  Google Scholar 

  1717. Buss JL, Arduini E, Shephard KC, Ponka P: Lipophilicity of analogs of pyridoxal isonicotinoyl hydrazone (PTH) determines the efflux of iron complexes and toxicity in K562 cells. Biochem Pharmacol. 2003, 65: 349-360.

    CAS  PubMed  Google Scholar 

  1718. Buss JL, Neuzil J, Gellert N, Weber C, Ponka P: Pyridoxal isonicotinoyl hydrazone analogs induce apoptosis in hematopoietic cells due to their iron-chelating properties. Biochemical Pharmacology. 2003, 65: 161-172.

    CAS  PubMed  Google Scholar 

  1719. Lovejoy DB, Richardson DR: Iron chelators as anti-neoplastic agents: current developments and promise of the PIH class of chelators. Curr Med Chem. 2003, 10: 1035-49.

    CAS  PubMed  Google Scholar 

  1720. Simunek T, Sterba M, Popelova O, Kaiserova H, Potacova A, Adamcova M, Mazurova Y, Ponka P, Gersl V: Pyridoxal isonicotinoyl hydrazone (PIH) and its analogs as protectants against anthracycline-induced cardiotoxicity. Hemoglobin. 2008, 32: 207-15.

    CAS  PubMed  Google Scholar 

  1721. Buss JL, Ponka P: Hydrolysis of pyridoxal isonicotinoyl hydrazone and its analogs. Biochim Biophys Acta. 2003, 1619: 177-186.

    CAS  PubMed  Google Scholar 

  1722. Šimůnek T, Boer C, Bouwman RA, Vlasblom R, Versteilen AMG, Štĕrba M, Geršl V, Hrdina R, Poňka P, de Lange JJ, Paulus WJ, Musters RJP: SIH – a novel lipophilic iron chelator – protects H9c2 cardiomyoblasts from oxidative stress-induced mitochondrial injury and cell death. Journal of Molecular and Cellular Cardiology. 2005, 39: 345-354.

    PubMed  Google Scholar 

  1723. Kalinowski DS, Sharpe PC, Bernhardt PV, Richardson DR: Structure-Activity Relationships of Novel Iron Chelators for the Treatment of Iron Overload Disease: The Methyl Pyrazinylketone Isonicotinoyl Hydrazone Series. J Med Chem. 2007

    Google Scholar 

  1724. Mouralian C, Buss JL, Stranix B, Chin J, Ponka P: Mobilization of iron from cells by hydroxyquinoline-based chelators. Biochem Pharmacol. 2005, 71: 214-222.

    CAS  PubMed  Google Scholar 

  1725. Murakami K, Haneda M, Yoshino M: Prooxidant action of xanthurenic acid and quinoline compounds: role of transition metals in the generation of reactive oxygen species and enhanced formation of 8-hydroxy-2'-deoxyguanosine in DNA. Biometals. 2006, 19: 429-35.

    CAS  PubMed  Google Scholar 

  1726. Murakami K, Ishida K, Watakabe K, Tsubouchi R, Naruse M, Yoshino M: Maltol/iron-mediated apoptosis in HL60 cells: participation of reactive oxygen species. Toxicol Lett. 2006, 161: 102-7.

    CAS  PubMed  Google Scholar 

  1727. Youdim MBH, Stephenson G, Ben Shachar D: Ironing iron out in Parkinson's disease and other neurodegenerative diseases with iron chelators – A lesson from 6-hydroxydopamine and iron chelators, desferal and VK-28. Redox-Active Metals in Neurological Disorders, Annals of the New York Academy of Sciences. 2004, 1012: 306-325.

    CAS  Google Scholar 

  1728. Ben Shachar D, Kahana N, Kampel V, Warshawsky A, Youdim MBH: Neuroprotection by a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats. Neuropharmacology. 2004, 46: 254-263.

    CAS  Google Scholar 

  1729. Youdim MBH, Fridkin M, Zheng HL: Bifunctional drug derivatives of MAO-B inhibitor rasagiline and iron chelator VK-28 as a more effective approach to treatment of brain ageing and ageing neurodegenerative diseases. Mech Ageing Dev. 2005, 126: 317-326.

    CAS  PubMed  Google Scholar 

  1730. Bandyopadhyay S, Huang X, Cho H, Greig NH, Youdim MB, Rogers JT: Metal specificity of an iron-responsive element in Alzheimer's APP mRNA 5 ' untranslated region, tolerance of SH-SY5Y and H4 neural cells to desferrioxamine clioquinol, VK-28, and a piperazine chelator. Journal of Neural Transmission-Supplement. 2006, 237-247.

    Google Scholar 

  1731. Hopkins AL, Mason JS, Overington JP: Can we rationally design promiscuous drugs?. Curr Opin Struct Biol. 2006, 16: 127-36.

    CAS  PubMed  Google Scholar 

  1732. Overington JP, Al-Lazikani B, Hopkins AL: How many drug targets are there?. Nat Rev Drug Discov. 2006, 5: 993-6.

    CAS  PubMed  Google Scholar 

  1733. Paolini GV, Shapland RH, van Hoorn WP, Mason JS, Hopkins AL: Global mapping of pharmacological space. Nat Biotechnol. 2006, 24: 805-815.

    CAS  PubMed  Google Scholar 

  1734. Dey A, Tergaonkar V, Lane DP: Double-edged swords as cancer therapeutics: simultaneously targeting p53 and NF-kappaB pathways. Nat Rev Drug Discov. 2008, 7: 1031-40.

    CAS  PubMed  Google Scholar 

  1735. Baret P, Beguin CG, Boukhalfa H, Caris C, Laulhere JP, Pierre JL, Serratrice G: O-Trensox – a promising water-soluble iron chelator (both Fe-III and Fe-II) potentially suitable for plant nutrition and iron chelation therapy. JACS. 1995, 117: 9760-9761.

    CAS  Google Scholar 

  1736. Caris C, Baret P, Beguin C, Serratrice G, Pierre JL, Laulhere JP: Metabolization of iron by plant cells using O-Trensox, a high-affinity abiotic iron-chelating agent. Biochem J. 1995, 312 (Pt 3): 879-85.

    CAS  PubMed  PubMed Central  Google Scholar 

  1737. Henry C, Rakba N, Imbert D, Thomas F, Baret P, Serratrice G, Gaude D, Pierre JL, Ward RJ, Crichton RR, Lescoat G: New 8-hydroxyquinoline and catecholate iron chelators: Influence of their partition coefficient on their biological activity. Biochem Pharmacol. 2001, 62: 1355-1362.

    CAS  PubMed  Google Scholar 

  1738. Serratrice G, Boukhalfa H, Beguin C, Baret P, Caris C, Pierre JL: O-TRENSOX, a new tripodal iron chelator based on 8-hydroxyquinoline subunits: Thermodynamic and kinetic studies. Inorganic Chemistry. 1997, 36: 3898-3910.

    CAS  Google Scholar 

  1739. Imbert D, Baret P, Gaude D, Gautier-Luneau I, Gellon G, Thomas F, Serratrice G, Pierre JL: Hydrophilic and lipophilic iron chelators with the same complexing abilities. Chemistry-a European Journal. 2002, 8: 1091-1100.

    CAS  Google Scholar 

  1740. Pierre JL, Baret P, Serratrice G: Hydroxyquinolines as iron chelators. Curr Med Chem. 2003, 10: 1077-1084.

    CAS  PubMed  Google Scholar 

  1741. Apostol M, Baret P, Serratrice G, Desbrieres J, Putaux JL, Stebe MJ, Expert D, Pierre JL: Self-assembly of an amphiphilic iron(III) chelator: Mimicking iron acquisition in marine bacteria. Angewandte Chemie-International Edition. 2005, 44: 2580-2582.

    CAS  Google Scholar 

  1742. d'Hardemare AD, Torelli S, Serratrice G, Pierre JL: Design of iron chelators: Syntheses and iron (III) complexing abilities of tripodal tris-bidentate ligands. Biometals. 2006, 19: 349-366.

    Google Scholar 

  1743. Whitnall M, Howard J, Ponka P, Richardson DR: A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. Proc Natl Acad Sci USA. 2006, 103: 14901-6.

    CAS  PubMed  PubMed Central  Google Scholar 

  1744. Mello Filho AC, Hoffmann ME, Meneghini R: Cell killing and DNA damage by hydrogen peroxide are mediated by intracellular iron. Biochem J. 1984, 218: 273-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  1745. Tatsuta M, Iishi H, Baba M, Mikuni T, Narahara H, Uedo N, Yano H: Suppression by iron chelator phenanthroline of sodium chloride-enhanced gastric carcinogenesis induced by N-methyl-N'-nitro-N-nitrosoguanidine in Wistar rats. Cancer Lett. 2003, 191: 9-16.

    CAS  PubMed  Google Scholar 

  1746. Kalinowski DS, Yu Y, Sharpe PC, Islam M, Liao YT, Lovejoy DB, Kumar N, Bernhardt PV, Richardson DR: Design synthesis, and characterization of novel iron chelators: structure-activity relationships of the 2-benzoylpyridine thiosemicarbazone series and their 3-nitrobenzoyl analogues as potent antitumor agents. J Med Chem. 2007, 50: 3716-29.

    CAS  PubMed  Google Scholar 

  1747. Kalinowski DS, Sharpe PC, Bernhardt PV, Richardson DR: Design Synthesis, and Characterization of New Iron Chelators with Anti-Proliferative Activity: Structure-Activity Relationships of Novel Thiohydrazone Analogues. J Med Chem. 2007, 50: 6212-6225.

    CAS  PubMed  Google Scholar 

  1748. L'Eplattenier F, Murase I, Martell AE: New multidentate ligands .6. chelating tendencies of N,N'-di(2-hydroxybenzyl) ethylenediamine-N,N'-diacetic acid. JACS. 1967, 89: 837.

    Google Scholar 

  1749. Faller B, Spanka C, Sergejew T, Tschinke V: Improving the oral bioavailability of the iron chelator HBED by breaking the symmetry of the intramolecular H-bond network. J Med Chem. 2000, 43: 1467-1475.

    CAS  PubMed  Google Scholar 

  1750. Bergeron RJ, Wiegand J, Brittenham GM: HBED: The continuing development of a potential alternative to deferoxamine for iron-chelating therapy. Blood. 1999, 93: 370-375.

    CAS  PubMed  Google Scholar 

  1751. Samuni AM, Afeworki M, Stein W, Yordanov AT, DeGraff W, Krishna MC, Mitchell JB, Brechbiel MW: Multifunctional antioxidant activity of HBED iron chelator. Free Radical Biology and Medicine. 2001, 30: 170-177.

    CAS  PubMed  Google Scholar 

  1752. Bergeron RJ, Wiegand J, Brittenham GM: HBED ligand: preclinical studies of a potential alternative to deferoxamine for treatment of chronic iron overload and acute iron poisoning. Blood. 2002, 99: 3019-3026.

    CAS  PubMed  Google Scholar 

  1753. Lebedev AV, Ivanova MV, Levitsky DO: Iron chelators and free radical scavengers in naturally occurring polyhydroxylated 1,4-naphthoquinones. Hemoglobin. 2008, 32: 165-79.

    CAS  PubMed  Google Scholar 

  1754. Liu JK, Killilea DW, Ames BN: Age-associated mitochondrial oxidative decay: Improvement of carnitine acetyltransferase substrate-binding affinity and activity in brain by feeding old rats acetyl-L-carnitine and/or R-alpha-lipoic acid. Proc Natl Acad Sci. 2002, 99: 1876-1881.

    CAS  PubMed  PubMed Central  Google Scholar 

  1755. Suh JH, Moreau R, Heath SH, Hagen TM: Dietary supplementation with (R)-alpha-lipoic acid reverses the age-related accumulation of iron and depletion of antioxidants in the rat cerebral cortex. Redox Rep. 2005, 10: 52-60.

    CAS  PubMed  Google Scholar 

  1756. Skibska B, Jozelowicz-Okonkwo G, Goraca A: Protective effects of early administration of alpha-lipoic acid against lipopolysaccharide-induced plasma lipid peroxidation. Pharmacological Reports. 2006, 58: 399-404.

    CAS  PubMed  Google Scholar 

  1757. Holmquist L, Stuchbury G, Berbaum K, Muscat S, Young S, Hager K, Engel J, Munch G: Lipoic acid as a novel treatment for Alzheimer's disease and related dementias. Pharmacology & Therapeutics. 2007, 113: 154-164.

    CAS  Google Scholar 

  1758. Petersen Shay K, Moreau RF, Smith EJ, Hagen TM: Is alpha-lipoic acid a scavenger of reactive oxygen species in vivo? Evidence for its initiation of stress signaling pathways that promote endogenous antioxidant capacity. IUBMB Life. 2008, 60: 362-7.

    PubMed  Google Scholar 

  1759. Wei Y, Guo M: Hydrogen peroxide triggered prochelator activation, subsequent metal chelation, and attenuation of the fenton reaction. Angew Chem Int Ed Engl. 2007, 46: 4722-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  1760. Galey JB: Potential use of iron chelators against oxidative damage. Adv Pharmacol. 1997, 38: 167-203.

    CAS  PubMed  Google Scholar 

  1761. Faa G, Crisponi G: Iron chelating agents in clinical practice. Coordination Chemistry Reviews. 1999, 184: 291-310.

    CAS  Google Scholar 

  1762. Kontoghiorghes GJ, Pattichi K, Hadjigavriel M, Kolnagou A: Transfusional iron overload and chelation therapy with deferoxamine and deferiprone (L1). Transfusion Science. 2000, 23: 211-223.

    CAS  PubMed  Google Scholar 

  1763. Emerit J, Beaumont C, Trivin F: Iron metabolism, free radicals, and oxidative injury. Biomed Pharmacother. 2001, 55: 333-9.

    CAS  PubMed  Google Scholar 

  1764. Tam TF, Leung-Toung R, Li WR, Wang YS, Karimian K, Spino M: Iron chelator research: Past present, and future. Current Medicinal Chemistry. 2003, 10: 983-995.

    CAS  PubMed  Google Scholar 

  1765. Blanuša M, Varnai VM, Piasek M, Kostial K: Chelators as antidotes of metal toxicity: Therapeutic and experimental aspects. Curr Med Chem. 2005, 12: 2771-2794.

    PubMed  Google Scholar 

  1766. Kontoghiorghes GJ, Kolnagou A: Molecular factors and mechanisms affecting iron and other metal excretion or absorption in health and disease. The role of natural and synthetic chelators. Current Medicinal Chemistry. 2005, 12: 2695-2709.

    CAS  PubMed  Google Scholar 

  1767. Kontoghiorghes GJ: Iron mobilization from transferrin and non-transferrin-bound-iron by deferiprone. Implications in the treatment of thalassemia, anemia of chronic disease, cancer and other conditions. Hemoglobin. 2006, 30: 183-200.

    CAS  PubMed  Google Scholar 

  1768. Golenser J, Domb A, Leshem B, Kremsner P, Luty A: Iron chelators as drugs against malaria pose a potential risk. Redox Rep. 2003, 8: 268-71.

    CAS  PubMed  Google Scholar 

  1769. Cherny RA, Legg JT, McLean CA, Fairlie DP, Huang XD, Atwood CS, Beyreuther K, Tanzi RE, Masters CL, Bush AI: Aqueous dissolution of Alzheimer's disease A beta amyloid deposits by biometal depletion. J Biol Chem. 1999, 274: 23223-23228.

    CAS  PubMed  Google Scholar 

  1770. Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, MacGregor L, Kiers L, Cherny R, Li QX, Tammer A, Carrington D, Mavros C, Volitakis I, Xilinas M, Ames D, Davis S, Volitakis I, Xilinas M, Ames D, Davis S, Beyreuther K, Tanzi RE, Masters CL: Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting A beta amyloid deposition and toxicity in Alzheimer disease – A pilot phase 2 clinical trial. Archives of Neurology. 2003, 60: 1685-1691.

    PubMed  Google Scholar 

  1771. Regland B, Lehmann W, Abedini I, Blennow K, Jonsson M, Karlsson I, Sjogren M, Wallin A, Xilinas M, Gottfries CG: Treatment of Alzheimer's disease with clioquinol. Dementia and Geriatric Cognitive Disorders. 2001, 12: 408-414.

    CAS  PubMed  Google Scholar 

  1772. Kaur D, Yantiri F, Rajagopalan S, Kumar J, Mo JO, Boonplueang R, Viswanath V, Jacobs R, Yang L, Beal MF, DiMonte D, Volitaskis I, Ellerby L, Cherny RA, Bush AI, Andersen JK: Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: A novel therapy for Parkinson's disease. Neuron. 2003, 37: 899-909.

    CAS  PubMed  Google Scholar 

  1773. Ikeda Y, Ikeda K, Long DM: Comparative study of different iron-chelating agents in cold-induced brain edema. Neurosurgery. 1989, 24: 820-4.

    CAS  PubMed  Google Scholar 

  1774. Masuda T, Hida H, Kanda Y, Aihara N, Ohta K, Yamada K, Nishino H: Oral administration of metal chelator ameliorates motor dysfunction after a small hemorrhage near the internal capsule in rat. J Neurosci Res. 2007, 85: 213-222.

    CAS  PubMed  Google Scholar 

  1775. De Vries B, Walter SJ, Von Bonsdorff L, Wolfs TGAM, Van Heurn LWE, Parkkinen J, Buurman WA: Reduction of circulating redox-active iron by apotransferrin protects against renal ischemia-reperfusion injury. Transplantation. 2004, 77: 669-675.

    CAS  PubMed  Google Scholar 

  1776. Galey JB, Destree O, Dumats J, Pichaud P, Marche J, Genard S, Bracciolli G, Le Capitaine L, Plessix H, Brambilla L, Cantoni O: Protection of U937 cells against oxidative injury by a novel series of iron chelators. Free Radic Biol Med. 1998, 25: 881-90.

    CAS  PubMed  Google Scholar 

  1777. Galey JB, Destree O, Dumats J, Genard S, Tachon P: Protection against oxidative damage by iron chelators: effect of lipophilic analogues and prodrugs of N,N'-bis(3,4,5-trimethoxybenzyl)ethylenediamine- N,N'-diacetic acid (OR10141). J Med Chem. 2000, 43: 1418-21.

    CAS  PubMed  Google Scholar 

  1778. Galey JB: Recent advances in the design of iron chelators against oxidative damage. Mini Rev Med Chem. 2001, 1: 233-42.

    CAS  PubMed  Google Scholar 

  1779. Charkoudian LK, Pham DM, Franz KJ: A pro-chelator triggered by hydrogen peroxide inhibits iron-promoted hydroxyl radical formation. JACS. 2006, 128: 12424-12425.

    CAS  Google Scholar 

  1780. Gal S, Zheng H, Fridkin M, Youdim MBH: Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selective brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine depletion. Journal of Neurochemistry. 2005, 95: 79-88.

    CAS  PubMed  Google Scholar 

  1781. Zheng H, Weiner LM, Bar-Am O, Epsztejn S, Cabantchik ZI, Warshawsky A, Youdim MB, Fridkin M: Design synthesis, and evaluation of novel bifunctional iron-chelators as potential agents for neuroprotection in Alzheimer's, Parkinson's, and other neurodegenerative diseases. Bioorg Med Chem. 2005, 13: 773-83.

    CAS  PubMed  Google Scholar 

  1782. Zheng HL, Youdim MBH, Weiner LM, Fridkin M: Novel potential neuroprotective agents with both iron chelating and amino acid-based derivatives targeting central nervous system neurons. Biochemical Pharmacology. 2005, 70: 1642-1652.

    CAS  PubMed  Google Scholar 

  1783. Gal S, Fridkin M, Amit T, Zheng H, Youdim MBH: M30, a novel multifunctional neuroprotective drug with potent iron chelating and brain selective monoamine oxidase-ab inhibitory activity for Parkinson's disease. J Neural Transmission – Suppl. 2006, 447-456.

    Google Scholar 

  1784. Avramovich-Tirosh Y, Amit T, Bar-Am O, Zheng HL, Fridkin M, Youdim MBH: Therapeutic targets and potential of the novel brain- permeable multifunctional iron chelator-monoamine oxidase inhibitor drug, M-30, for the treatment of Alzheimer's disease. J Neurochem. 2007, 100: 490-502.

    CAS  PubMed  Google Scholar 

  1785. Kayyali R, Pannala AS, Khodr H, Hider RC: Comparative radical scavenging ability of bidentate iron (III) chelators. Biochem Pharmacol. 1998, 55: 1327-32.

    CAS  PubMed  Google Scholar 

  1786. Kontoghiorghes GJ, Weinberg ED: Iron – mammalian defense systems, mechanisms of disease, and chelation therapy approaches. Blood Rev. 1995, 9: 33-45.

    CAS  PubMed  Google Scholar 

  1787. Gabutti V, Piga A: Results of long-perm iron-chelating therapy. Acta Haematologica. 1996, 95: 26-36.

    CAS  PubMed  Google Scholar 

  1788. Song BB, Anderson DJ, Schacht J: Protection from gentamicin ototoxicity by iron chelators in guinea pig in vivo. Journal of Pharmacology and Experimental Therapeutics. 1997, 282: 369-377.

    CAS  PubMed  Google Scholar 

  1789. Song BB, Schacht J: Variable efficacy of radical scavengers and iron chelators to attenuate gentamicin ototoxicity in guinea pig in vivo. Hearing Research. 1996, 94: 87-93.

    CAS  PubMed  Google Scholar 

  1790. Wetli HA, Buckett PD, Wessling-Resnick M: Small-molecule screening identifies the selanazal drug ebselen as a potent inhibitor of DMT1-mediated iron uptake. Chemistry & Biology. 2006, 13: 965-972.

    CAS  Google Scholar 

  1791. Hovorka SW, Schoneich C: Oxidative degradation of pharmaceuticals: Theory, mechanisms and inhibition. J Pharm Sci. 2001, 90: 253-269.

    CAS  PubMed  Google Scholar 

  1792. England K, Driscoll CO, Cotter TG: ROS and protein oxidation in early stages of cytotoxic drug induced apoptosis. Free Radical Research. 2006, 40: 1124-1137.

    CAS  PubMed  Google Scholar 

  1793. Cha'on U, Valmas N, Collins PJ, Reilly PEB, Hammock BD, Ebert PR: Disruption of iron homeostasis increases phosphine toxicity in Caenorhabditis elegans. Toxicological Sciences. 2007, 96: 194-201.

    PubMed  Google Scholar 

  1794. Bus JS, Gibson JE, Aust SD: Superoxide-catalyzed and singlet oxygen-catalyzed lipid peroxidation as a possible mechanism for paraquat (methyl viologen) toxicity. Biochem Biophys Res Commun. 1974, 58: 749-755.

    CAS  PubMed  Google Scholar 

  1795. Doelman CJA, Bast A: Oxygen Radicals in Lung Pathology. Free Rad Biol Med. 1990, 9: 381-400.

    CAS  PubMed  Google Scholar 

  1796. Hippeli S, Elstner EF: Transition metal ion-catalyzed oxygen activation during pathogenic processes. Febs Letters. 1999, 443: 1-7.

    CAS  PubMed  Google Scholar 

  1797. Jones GM, Vale JA: Mechanisms of toxicity, clinical features, and management of diquat poisoning: A review. J Toxicol. 2000, 38: 123-128.

    CAS  Google Scholar 

  1798. Suntres ZE: Role of antioxidants in paraquat toxicity. Toxicology. 2002, 180: 65-77.

    CAS  PubMed  Google Scholar 

  1799. Abdollahi M, Ranjbar A, Shadnia S, Nikfar S, Rezaie A: Pesticides and oxidative stress: a review. Medical Science Monitor. 2004, 10: RA141-RA147.

    CAS  PubMed  Google Scholar 

  1800. Rogachev I, Kampel V, Gusis V, Cohen N, Gressel J, Warshawsky A: Synthesis properties, and use of copper-chelating amphiphilic dithiocarbamates as synergists of oxidant-generating herbicides. Pesticide Biochemistry and Physiology. 1998, 60: 133-145.

    CAS  Google Scholar 

  1801. Kotamraju S, Chitambar CR, Kalivendi SV, Joseph J, Kalyanaraman B: Transferrin receptor-dependent iron uptake is responsible for doxorubicin-mediated apoptosis in endothelial cells – Role of oxidant-induced iron signaling in apoptosis. Journal of Biological Chemistry. 2002, 277: 17179-17187.

    CAS  PubMed  Google Scholar 

  1802. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L: Anthracyclines: Molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacological Reviews. 2004, 56: 185-229.

    CAS  PubMed  Google Scholar 

  1803. Asare GA, Bronz M, Naidoo V, Kew MC: Interactions between aflatoxin B1 and dietary iron overload in hepatic mutagenesis. Toxicology. 2007, 234: 157-66.

    CAS  PubMed  Google Scholar 

  1804. Scalbert A, Williamson G: Dietary intake and bioavailability of polyphenols. J Nutr. 2000, 130: 2073S-85S.

    CAS  PubMed  Google Scholar 

  1805. Reed J: Cranberry flavonoids, atherosclerosis and cardiovascular health. Crit Rev Food Sci Nutr. 2002, 42: 301-16.

    CAS  PubMed  Google Scholar 

  1806. Benzie IF: Evolution of dietary antioxidants. Comp Biochem Physiol A Mol Integr Physiol. 2003, 136: 113-26.

    PubMed  Google Scholar 

  1807. Higdon JV, Frei B: Tea catechins and polyphenols: health effects metabolism, and antioxidant functions. Crit Rev Food Sci Nutr. 2003, 43: 89-143.

    CAS  PubMed  Google Scholar 

  1808. Surh YJ: Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer. 2003, 3: 768-80.

    CAS  PubMed  Google Scholar 

  1809. Manach C, Scalbert A, Morand C, Remesy C, Jimenez L: Polyphenols: food sources and bioavailability. Am J Clin Nutr. 2004, 79: 727-47.

    CAS  PubMed  Google Scholar 

  1810. Manach C, Mazur A, Scalbert A: Polyphenols and prevention of cardiovascular diseases. Curr Opin Lipidol. 2005, 16: 77-84.

    CAS  PubMed  Google Scholar 

  1811. Scalbert A, Manach C, Morand C, Remesy C, Jimenez L: Dietary polyphenols and the prevention of diseases. Crit Rev Food Sci Nutr. 2005, 45: 287-306.

    CAS  PubMed  Google Scholar 

  1812. Nichenametla SN, Taruscio TG, Barney DL, Exon JH: A review of the effects and mechanisms of polyphenolics in cancer. Crit Rev Food Sci Nutr. 2006, 46: 161-83.

    CAS  PubMed  Google Scholar 

  1813. Zhang HY, Yang DP, Tang GY: Multipotent antioxidants: from screening to design. Drug Discov Today. 2006, 11: 749-54.

    CAS  PubMed  Google Scholar 

  1814. Shahidi F, Wanasundara PK: Phenolic antioxidants. Crit Rev Food Sci Nutr. 1992, 32: 67-103.

    CAS  PubMed  Google Scholar 

  1815. Wright JS, Johnson ER, DiLabio GA: Predicting the activity of phenolic antioxidants: theoretical method, analysis of substituent effects, and application to major families of antioxidants. J Am Chem Soc. 2001, 123: 1173-83.

    CAS  PubMed  Google Scholar 

  1816. Block G, Patterson B, Subar A: Fruit vegetables, and cancer prevention: a review of the epidemiological evidence. Nutr Cancer. 1992, 18: 1-29.

    CAS  PubMed  Google Scholar 

  1817. Ness AR, Powles JW: Fruit and vegetables, and cardiovascular disease: a review. Int J Epidemiol. 1997, 26: 1-13.

    CAS  PubMed  Google Scholar 

  1818. Krauss RM, Eckel RH, Howard B, Appel LJ, Daniels SR, Deckelbaum RJ, Erdman JW, Kris-Etherton P, Goldberg IJ, Kotchen TA, Lichtenstein AH, Mitch WE, Mullis R, Robinson K, Wylie-Rosett J, St Jeor S, Suttie J, Tribble DL, Bazzarre TL: AHA Dietary Guidelines: revision 2000: A statement for healthcare professionals from the Nutrition Committee of the American Heart Association. Circulation. 2000, 102: 2284-99.

    CAS  PubMed  Google Scholar 

  1819. Yang CS, Landau JM, Huang MT, Newmark HL: Inhibition of carcinogenesis by dietary polyphenolic compounds. Annu Rev Nutr. 2001, 21: 381-406.

    CAS  PubMed  Google Scholar 

  1820. Lotito SB, Frei B: Consumption of flavonoid-rich foods and increased plasma antioxidant capacity in humans: Cause consequence, or epiphenomenon?. Free Rad Biol Med. 2006, 41: 1727-1746.

    CAS  PubMed  Google Scholar 

  1821. Cook NC, Samman S: Flavonoids – Chemistry metabolism, cardioprotective effects, and dietary sources. Journal of Nutritional Biochemistry. 1996, 7: 66-76.

    CAS  Google Scholar 

  1822. Bravo L: Polyphenols: chemistry, dietary sources metabolism, and nutritional significance. Nutrition Reviews. 1998, 56: 317-333.

    CAS  PubMed  Google Scholar 

  1823. Fang YZ, Yang S, Wu GY: Free radicals antioxidants, and nutrition. Nutrition. 2002, 18: 872-879.

    CAS  PubMed  Google Scholar 

  1824. Fraga CG, Oteiza PI: Iron toxicity and antioxidant nutrients. Toxicology. 2002, 180: 23-32.

    CAS  PubMed  Google Scholar 

  1825. Halvorsen BL, Holte K, Myhrstad MC, Barikmo I, Hvattum E, Remberg SF, Wold AB, Haffner K, Baugerod H, Andersen LF, Moskaug O, Jacobs DR, Blomhoff R: A systematic screening of total antioxidants in dietary plants. J Nutr. 2002, 132: 461-71.

    CAS  PubMed  Google Scholar 

  1826. Havsteen BH: The biochemistry and medical significance of the flavonoids. Pharmacol Ther. 2002, 96: 67-202.

    CAS  PubMed  Google Scholar 

  1827. Vertuani S, Angusti A, Manfredini S: The antioxidants and pro-antioxidants network: an overview. Curr Pharmaceut Design. 2004, 10: 1677-1694.

    CAS  Google Scholar 

  1828. Arts ICW, Hollman PCH: Polyphenols and disease risk in epidemiologic studies. American Journal of Clinical Nutrition. 2005, 81: 317S-325S.

    CAS  PubMed  Google Scholar 

  1829. Afanas'ev IB, Dorozhko AI, Brodskii AV, Kostyuk VA, Potapovitch AI: Chelating and free radical scavenging mechanisms of inhibitory action of rutin and quercetin in lipid peroxidation. Biochem Pharmacol. 1989, 38: 1763-9.

    PubMed  Google Scholar 

  1830. Morel I, Lescoat G, Cillard P, Cillard J: Role of flavonoids and iron chelation in antioxidant action. Methods Enzymol. 1994, 234: 437-43.

    CAS  PubMed  Google Scholar 

  1831. Moran JF, Klucas RV, Grayer RJ, Abian J, Becana M: Complexes of iron with phenolic compounds from soybean nodules and other legume tissues: prooxidant and antioxidant properties. Free Radic Biol Med. 1997, 22: 861-70.

    CAS  PubMed  Google Scholar 

  1832. Yoshino M, Murakami K: Interaction of iron with polyphenolic compounds: application to antioxidant characterization. Anal Biochem. 1998, 257: 40-4.

    CAS  PubMed  Google Scholar 

  1833. Grinberg LN, Newmark H, Kitrossky N, Rahamim E, Chevion M, Rachmilewitz EA: Protective effects of tea polyphenols against oxidative damage to red blood cells. Biochemical Pharmacology. 1997, 54: 973-978.

    CAS  PubMed  Google Scholar 

  1834. Lopes GKB, Schulman HM, Hermes-Lima M: Polyphenol tannic acid inhibits hydroxyl radical formation from Fenton reaction by complexing ferrous ions. Biochimica Et Biophysica Acta-General Subjects. 1999, 1472: 142-152.

    CAS  Google Scholar 

  1835. Packer L, Rimbach G, Virgili F: Antioxidant activity and biologic properties of a procyanidin-rich extract from pine (Pinus maritima) bark, pycnogenol. Free Rad Biol Med. 1999, 27: 704-724.

    CAS  PubMed  Google Scholar 

  1836. Robards K, Prenzler PD, Tucker G, Swatsitang P, Glover W: Phenolic compounds and their role in oxidative processes in fruits. Food Chem. 1999, 66: 401-436.

    CAS  Google Scholar 

  1837. Middleton E, Kandaswami C, Theoharides TC: The effects of plant flavonoids on mammalian cells: Implications for inflammation, heart disease, and cancer. Pharmacological Reviews. 2000, 52: 673-751.

    CAS  PubMed  Google Scholar 

  1838. Hider RC, Liu ZD, Khodr HH: Metal chelation of polyphenols. Flavonoids and Other Polyphenols, Methods in Enzymology. 2001, 335: 190-203.

    CAS  Google Scholar 

  1839. Levites Y, Youdim MBH, Maor G, Mandel S: Attenuation of 6-hydroxydopamine (6-OHDA)-induced nuclear factor-kappaB (NF-kappa B) activation and cell death by tea extracts in neuronal cultures. Biochemical Pharmacology. 2002, 63: 21-29.

    CAS  PubMed  Google Scholar 

  1840. Mandel S, Youdim MBH: Catechin polyphenols: Neurodegeneration and neuroprotection in neurodegenerative diseases. Free Radical Biology and Medicine. 2004, 37: 304-317.

    CAS  PubMed  Google Scholar 

  1841. Mandel S, Weinreb O, Amit T, Youdim MBH: Cell signaling pathways in the neuroprotective actions of the green tea polyphenol (-)-epigallocatechin-3-gallate: implications for neurodegenerative diseases. J Neurochem. 2004, 88: 1555-1569.

    CAS  PubMed  Google Scholar 

  1842. Hague T, Andrews PLR, Barker J, Naughton DP: Dietary chelators as antioxidant enzyme mimetics: implications for dietary intervention in neurodegenerative diseases. Behavioural Pharmacology. 2006, 17: 425-430.

    CAS  PubMed  Google Scholar 

  1843. Weinreb O, Amit T, Youdim MB: A novel approach of proteomics and transcriptomics to study the mechanism of action of the antioxidant-iron chelator green tea polyphenol (-)-epigallocatechin-3-gallate. Free Radic Biol Med. 2007, 43: 546-56.

    CAS  PubMed  Google Scholar 

  1844. Rice-Evans CA, Miller NJ, Paganga G: Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med. 1996, 20: 933-56.

    CAS  PubMed  Google Scholar 

  1845. Rice-Evans CA, Miller J, Paganga G: Antioxidant properties of phenolic compounds. Trends in Plant Science. 1997, 2: 152-159.

    Google Scholar 

  1846. Kähkönen MP, Hopia AI, Vuorela HJ, Rauha JP, Pihlaja K, Kujala TS, Heinonen M: Antioxidant activity of plant extracts containing phenolic compounds. J Agric Food Chem. 1999, 47: 3954-62.

    PubMed  Google Scholar 

  1847. Wang H, Cao GH, Prior RL: Oxygen radical absorbing capacity of anthocyanins. Journal of Agricultural and Food Chemistry. 1997, 45: 304-309.

    CAS  Google Scholar 

  1848. Harborne JB, Williams CA: Advances in flavonoid research since 1992. Phytochemistry. 2000, 55: 481-504.

    CAS  PubMed  Google Scholar 

  1849. Nijveldt RJ, van Nood E, van Hoorn DEC, Boelens PG, van Norren K, van Leeuwen PAM: Flavonoids: a review of probable mechanisms of action and potential applications. American Journal of Clinical Nutrition. 2001, 74: 418-425.

    CAS  PubMed  Google Scholar 

  1850. Heim KE, Tagliaferro AR, Bobilya DJ: Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. J Nutr Biochem. 2002, 13: 572-584.

    CAS  PubMed  Google Scholar 

  1851. Rice-Evans CA, Packer L: Flavonoids in health and disease. 2003, Marcel Dekker, New York, 2

    Google Scholar 

  1852. Robbins RJ: Phenolic acids in foods: an overview of analytical methodology. J Agric Food Chem. 2003, 51: 2866-87.

    CAS  PubMed  Google Scholar 

  1853. Sakakibara H, Honda Y, Nakagawa S, Ashida H, Kanazawa K: Simultaneous determination of all polyphenols in vegetables fruits, and teas. J Agric Food Chem. 2003, 51: 571-81.

    CAS  PubMed  Google Scholar 

  1854. Morel I, Lescoat G, Cogrel P, Sergent O, Pasdeloup N, Brissot P, Cillard P, Cillard J: Antioxidant and iron-chelating activities of the flavonoids catechin, quercetin and diosmetin on iron-loaded rat hepatocyte cultures. Biochem Pharmacol. 1993, 45: 13-19.

    CAS  PubMed  Google Scholar 

  1855. Ferrali M, Signorini C, Caciotti B, Sugherini L, Ciccoli L, Giachetti D, Comporti M: Protection against oxidative damage of erythrocyte membrane by the flavonoid quercetin and its relation to iron chelating activity. FEBS Lett. 1997, 416: 123-9.

    CAS  PubMed  Google Scholar 

  1856. Shoskes DA: Effect of bioflavonoids quercetin and curcumin on ischemic renal injury – A new class of renoprotective agents. Transplantation. 1998, 66: 147-152.

    CAS  PubMed  Google Scholar 

  1857. Kostyuk VA, Potapovich AI: Antiradical and chelating effects in flavonoid protection against silica-induced cell injury. Archives of Biochemistry and Biophysics. 1998, 355: 43-48.

    CAS  PubMed  Google Scholar 

  1858. Aherne SA, O'Brien NM: Mechanism of protection by the flavonoids, quercetin and rutin, against tert-butylhydroperoxide- and menadione induced DNA single strand breaks in Caco-2 cells. Free Radical Biology and Medicine. 2000, 29: 507-514.

    CAS  PubMed  Google Scholar 

  1859. Cheng IF, Breen K: On the ability of four flavonoids baicilein, luteolin naringenin, and quercetin, to suppress the Fenton reaction of the iron-ATP complex. Biometals. 2000, 13: 77-83.

    CAS  PubMed  Google Scholar 

  1860. Zhang Y, Li H, Zhao Y, Gao Z: Dietary supplementation of baicalin and quercetin attenuates iron overload induced mouse liver injury. Eur J Pharmacol. 2006, 535: 263-9.

    CAS  PubMed  Google Scholar 

  1861. Comporti M, Signorini C, Buonocore G, Ciccoli L: Iron release, oxidative stress and erythrocyte ageing. Free Radical Biology and Medicine. 2002, 32: 568-576.

    CAS  PubMed  Google Scholar 

  1862. Mira L, Fernandez MT, Santos M, Rocha R, Florencio MH, Jennings KR: Interactions of flavonoids with iron and copper ions: A mechanism for their antioxidant activity. Free Radical Research. 2002, 36: 1199-1208.

    CAS  PubMed  Google Scholar 

  1863. Melidou M, Riganakos K, Galaris D: Protection against nuclear DNA damage offered by flavonoids in cells exposed to hydrogen peroxide: The role of iron chelation. Free Rad Biol Med. 2005, 39: 1591-1600.

    CAS  PubMed  Google Scholar 

  1864. El Hajji H, Nkhili E, Tomao V, Dangles O: Interactions of quercetin with iron and copper ions: complexation and autoxidation. Free Radic Res. 2006, 40: 303-20.

    CAS  PubMed  Google Scholar 

  1865. Russo A, Acquaviva R, Campisi A, Sorrenti V, Di Giacomo C, Virgata G, Barcellona ML, Vanella A: Bioflavonoids as antiradicals, antioxidants and DNA cleavage protectors. Cell Biology and Toxicology. 2000, 16: 91-98.

    CAS  PubMed  Google Scholar 

  1866. Kaiserová H, Šimůnek T, Vijgh van der WJ, Bast A, Kvasničková E: Flavonoids as protectors against doxorubicin cardiotoxicity: role of iron chelation, antioxidant activity and inhibition of carbonyl reductase. Biochim Biophys Acta. 2007, 1772: 1065-74.

    PubMed  Google Scholar 

  1867. Zhao Y, Li H, Gao Z, Xu H: Effects of dietary baicalin supplementation on iron overload-induced mouse liver oxidative injury. Eur J Pharmacol. 2005, 509: 195-200.

    CAS  PubMed  Google Scholar 

  1868. Borsari M, Ferrari E, Grandi R, Saladini M: Curcuminoids as potential new iron-chelating agents: spectroscopic, polarographic and potentiometric study on their Fe(III) complexing ability. Inorganica Chimica Acta. 2002, 328: 61-68.

    CAS  Google Scholar 

  1869. Baum L, Ng A: Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer's disease animal models. J Alzheimers Dis. 2004, 6: 367-377.

    CAS  PubMed  Google Scholar 

  1870. Bernabé-Pineda M, Ramírez-Silva MT, Romero-Romo MA, González-Vergara E, Rojas-Hernández A: Spectrophotometric and electrochemical determination of the formation constants of the complexes Curcumin-Fe(Ill)-water and Curcumin-Fe(II)-water. Spectrochim Acta A. 2004, 60: 1105-1113.

    Google Scholar 

  1871. Jiao Y, Wilkinson J, Pletsch EC, Buss JL, Wang W, Planalp R, Torti FM, Torti SV: Iron chelation in the biological activity of curcumin. Free Radical Biology and Medicine. 2006, 40: 1152-1160.

    CAS  PubMed  Google Scholar 

  1872. Aggarwal BB, Sundaram C, Malani N, Ichikawa H: Curcumin: The Indian solid gold. Adv Exp Med Biol. 2007, 595: 1-75.

    PubMed  Google Scholar 

  1873. Farombi EO, Nwaokeafor IA: Anti-oxidant mechanisms of kolaviron: studies on serum lipoprotein oxidation, metal chelation and oxidative membrane damage in rats. Clin Exp Pharmacol Physiol. 2005, 32: 667-74.

    CAS  PubMed  Google Scholar 

  1874. Engelmann MD, Hutcheson R, Cheng IF: Stability of ferric complexes with 3-hydroxyflavone (flavonol), 5,7-dihydroxyflavone (chrysin), and 3', 4'-dihydroxyflavone. J Agric Food Chem. 2005, 53: 2953-2960.

    CAS  PubMed  Google Scholar 

  1875. Botelho FV, Alvarez-Leite JI, Lemos VS, Pimenta AM, Calado HD, Matencio T, Miranda CT, Pereira-Maia EC: Physicochemical study of floranol, its copper(II) and iron(III) complexes, and their inhibitory effect on LDL oxidation. J Inorg Biochem. 2007, 101: 935-43.

    CAS  PubMed  Google Scholar 

  1876. Gottlieb M, Leal-Campanario R, Campos-Esparza MR, Sanchez-Gomez MV, Alberdi E, Arranz A, Delgado-Garcia JM, Gruart A, Matute C: Neuroprotection by two polyphenols following excitotoxicity and experimental ischemia. Neurobiol Dis. 2006, 23: 374-86.

    CAS  PubMed  Google Scholar 

  1877. Pardo-Andreu GL, Sanchez-Baldoquin C, Avila-Gonzalez R, Delgado R, Naal Z, Curti C: Fe(III) improves antioxidant and cytoprotecting activities of mangiferin. European Journal of Pharmacology. 2006, 547: 31-36.

    CAS  PubMed  Google Scholar 

  1878. Pardo-Andreu GL, Cavalheiro RA, Dorta DJ, Naal Z, Delgado R, Vercesi AE, Curti C: Fe(III) shifts the mitochondria permeability transition-eliciting capacity of mangiferin to protection of organelle. J Pharmacol Exp Ther. 2007, 320: 646-53.

    CAS  PubMed  Google Scholar 

  1879. Pardo-Andreu GL, Barrios MF, Curti C, Hernandez I, Merino N, Lemus Y, Martinez I, Riano A, Delgado R: Protective effects of Mangifera indica L extract (Vimang), and its major component mangiferin, on iron-induced oxidative damage to rat serum and liver. Pharmacol Res. 2008, 57: 79-86.

    CAS  PubMed  Google Scholar 

  1880. Mandel S, Weinreb O, Reznichenko L, Kalfon L, Amit T: Green tea catechins as brain-permeable, non toxic iron chelators to "iron out iron" from the brain. Journal of Neural Transmission-Supplement. 2006, 249-257.

    Google Scholar 

  1881. Mandel S, Amit T, Reznichenko L, Weinreb O, Youdim MBH: Green tea catechins as brain-permeable, natural iron chelators-antioxidants for the treatment of neurodegenerative disorders. Molecular Nutrition & Food Research. 2006, 50: 229-234.

    CAS  Google Scholar 

  1882. Miller NJ, Castelluccio C, Tijburg L, Rice-Evans C: The antioxidant properties of theaflavins and their gallate esters–radical scavengers or metal chelators?. FEBS Lett. 1996, 392: 40-4.

    CAS  PubMed  Google Scholar 

  1883. Khokhar S, Apenten RKO: Iron binding characteristics of phenolic compounds: some tentative structure-activity relations. Food Chemistry. 2003, 81: 133-140.

    CAS  Google Scholar 

  1884. Limson J, Nyokong T, Daya S: The interaction of melatonin and its precursors with aluminium cadmium, copper iron, lead, and zinc: an adsorptive voltammetric study. J Pineal Res. 1998, 24: 15-21.

    CAS  PubMed  Google Scholar 

  1885. Lin AM, Ho LT: Melatonin suppresses iron-induced neurodegeneration in rat brain. Free Radic Biol Med. 2000, 28: 904-11.

    CAS  PubMed  Google Scholar 

  1886. Gulcin I, Buyukokuroglu ME, Kufrevioglu OI: Metal chelating and hydrogen peroxide scavenging effects of melatonin. J Pineal Res. 2003, 34: 278-281.

    CAS  PubMed  Google Scholar 

  1887. Chwelatiuk E, Wlostowski T, Krasowska A, Bonda E: The effect of orally administered melatonin on tissue accumulation and toxicity of cadmium in mice. Journal of Trace Elements in Medicine and Biology. 2006, 19: 259-265.

    CAS  PubMed  Google Scholar 

  1888. Belguendouz L, Fremont L, Linard A: Resveratrol inhibits metal ion-dependent and independent peroxidation of porcine low-density lipoproteins. Biochemical Pharmacology. 1997, 53: 1347-1355.

    CAS  PubMed  Google Scholar 

  1889. Cadenas S, Barja G: Resveratrol melatonin, vitamin E, and PBN protect against renal oxidative DNA damage induced by the kidney carcinogen KBrO3. Free Radic Biol Med. 1999, 26: 1531-7.

    CAS  PubMed  Google Scholar 

  1890. Hung LM, Chen JK, Huang SS, Lee RS, Su MJ: Cardioprotective effect of resveratrol, a natural antioxidant derived from grapes. Cardiovasc Res. 2000, 47: 549-55.

    CAS  PubMed  Google Scholar 

  1891. Giovannini L, Migliori M, Longoni BM, Das DK, Bertelli AAE, Panichi SV, Filippi C, Bertelli A: Resveratrol, a polyphenol found in wine, reduces ischemia reperfusion injury in rat kidneys. J Cardiovasc Pharmacol. 2001, 37: 262-270.

    CAS  PubMed  Google Scholar 

  1892. Huang SS, Tsai MC, Chih CL, Hung LM, Tsai SK: Resveratrol reduction of infarct size in Long-Evans rats subjected to focal cerebral ischemia. Life Sci. 2001, 69: 1057-65.

    CAS  PubMed  Google Scholar 

  1893. Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, Scherer B, Sinclair DA: Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003, 425: 191-6.

    CAS  PubMed  Google Scholar 

  1894. Leonard SS, Xia C, Jiang BH, Stinefelt B, Klandorf H, Harris GK, Shi X: Resveratrol scavenges reactive oxygen species and effects radical-induced cellular responses. Biochem Biophys Res Commun. 2003, 309: 1017-26.

    CAS  PubMed  Google Scholar 

  1895. Kundu JK, Surh YJ: Molecular basis of chemoprevention by resveratrol: NF-kappaB and AP-1 as potential targets. Mutat Res. 2004, 555: 65-80.

    CAS  PubMed  Google Scholar 

  1896. Signorelli P, Ghidoni R: Resveratrol as an anticancer nutrient: molecular basis, open questions and promises. J Nutr Biochem. 2005, 16: 449-66.

    CAS  PubMed  Google Scholar 

  1897. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA: Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006, 444: 337-42.

    CAS  PubMed  Google Scholar 

  1898. Baur JA, Sinclair DA: Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov. 2006, 5: 493-506.

    CAS  PubMed  Google Scholar 

  1899. Kotha A, Sekharam M, Cilenti L, Siddiquee K, Khaled A, Zervos AS, Carter B, Turkson J, Jove R: Resveratrol inhibits Src and Stat3 signaling and induces the apoptosis of malignant cells containing activated Stat3 protein. Mol Cancer Ther. 2006, 5: 621-9.

    CAS  PubMed  Google Scholar 

  1900. Russo GL: Ins and outs of dietary phytochemicals in cancer chemoprevention. Biochem Pharmacol. 2007, 74: 533-44.

    CAS  PubMed  Google Scholar 

  1901. Tsai SK, Hung LM, Fu YT, Cheng H, Nien MW, Liu HY, Zhang FB, Huang SS: Resveratrol neuroprotective effects during focal cerebral ischemia injury via nitric oxide mechanism in rats. J Vasc Surg. 2007, 46: 346-53.

    PubMed  Google Scholar 

  1902. Gatz SA, Wiesmuller L: Take a break–resveratrol in action on DNA. Carcinogenesis. 2008, 29: 321-32.

    CAS  PubMed  Google Scholar 

  1903. Chu YF, Sun J, Wu X, Liu RH: Antioxidant and antiproliferative activities of common vegetables. J Agric Food Chem. 2002, 50: 6910-6.

    CAS  PubMed  Google Scholar 

  1904. Afaq F, Saleem M, Krueger CG, Reed JD, Mukhtar H: Anthocyanin- and hydrolyzable tannin-rich pomegranate fruit extract modulates MAPK and NF-kappaB pathways and inhibits skin tumorigenesis in CD-1 mice. Int J Cancer. 2005, 113: 423-33.

    CAS  PubMed  Google Scholar 

  1905. Malik A, Afaq F, Sarfaraz S, Adhami VM, Syed DN, Mukhtar H: Pomegranate fruit juice for chemoprevention and chemotherapy of prostate cancer. Proc Natl Acad Sci USA. 2005, 102: 14813-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  1906. Butelli E, Titta L, Giorgio M, Mock HP, Matros A, Peterek S, Schijlen EG, Hall RD, Bovy AG, Luo J, Martin C: Enrichment of tomato fruit with health-promoting anthocyanins by expression of select transcription factors. Nat Biotechnol. 2008, 26: 1301-1308.

    CAS  PubMed  Google Scholar 

  1907. Joseph JA, Shukitt-Hale B, Denisova NA, Bielinski D, Martin A, McEwen JJ, Bickford PC: Reversals of age-related declines in neuronal signal transduction cognitive, and motor behavioral deficits with blueberry spinach, or strawberry dietary supplementation. Journal of Neuroscience. 1999, 19: 8114-8121.

    CAS  PubMed  Google Scholar 

  1908. Sweeney MI, Kalt W, MacKinnon SL, Ashby J, Gottschall-Pass KT: Feeding rats diets enriched in lowbush blueberries for six weeks decreases ischemia-induced brain damage. Nutritional Neuroscience. 2002, 5: 427-431.

    CAS  PubMed  Google Scholar 

  1909. Joseph JA, Fisher DR, Bielinski D: Blueberry extract alters oxidative stress-mediated signaling in COS-7 cells transfected with selectively vulnerable muscarinic receptor subtypes. J Alzheimers Dis. 2006, 9: 35-42.

    CAS  PubMed  Google Scholar 

  1910. Ramassamy C: Emerging role of polyphenolic compounds in the treatment of neurodegenerative diseases: A review of their intracellular targets. Eur J Pharmacol. 2006, 545: 51-64.

    CAS  PubMed  Google Scholar 

  1911. Kalt W, Joseph JA, Shukitt-Hale B: Blueberries and human health: a review of current reseach. J Amer Pomol Soc. 2007, 61: 151-160.

    Google Scholar 

  1912. Lau FC, Bielinski DF, Joseph JA: Inhibitory effects of blueberry extract on the production of inflammatory mediators in lipopolysaccharide-activated BV2 microglia. J Neurosci Res. 2007, 85: 1010-7.

    CAS  PubMed  Google Scholar 

  1913. Neto CC: Cranberry and blueberry: evidence for protective effects against cancer and vascular diseases. Mol Nutr Food Res. 2007, 51: 652-64.

    CAS  PubMed  Google Scholar 

  1914. McAnulty SR, McAnulty LS, Nieman DC, Dumke CL, Morrow JD, Utter AC, Henson DA, Proulx WR, George GL: Consumption of blueberry polyphenols reduces exercise-induced oxidative stress compared to vitamin C. Nutrition Research. 2004, 24: 209-221.

    CAS  Google Scholar 

  1915. Longpré F, Garneau P, Christen Y, Ramassamy C: Protection by EGb 761 against beta-amyloid-induced neurotoxicity: Involvement of NF-kappa B, SIRT1, and MAPKs pathways and inhibition of amyloid fibril formation. Free Rad Biol Med. 2006, 41: 1781-1794.

    PubMed  Google Scholar 

  1916. Ramassamy C, Longpré F, Christen Y: Ginkgo biloba extract (EGb 761) in Alzheimer's disease: Is there any evidence?. Current Alzheimer Research. 2007, 4: 253-262.

    CAS  PubMed  Google Scholar 

  1917. Wang Q, Simonyi A, Li WL, Sisk BA, Miller RL, MacDonald RS, Lubahn DE, Sun GY, Sun AY: Dietary grape supplement ameliorates cerebral ischemia-induced neuronal death in gerbils. Molecular Nutrition & Food Research. 2005, 49: 443-451.

    CAS  Google Scholar 

  1918. Curin Y, Andriantsitohaina R: Polyphenols as potential therapeutical agents against cardiovascular diseases. Pharmacological Reports. 2005, 57: 97-107.

    PubMed  Google Scholar 

  1919. Kuriyama S, Shimazu T, Ohmori K, Kikuchi N, Nakaya N, Nishino Y, Tsubono Y, Tsuji I: Green tea consumption and mortality due to cardiovascular disease cancer, and all causes in Japan – The Ohsaki Study. Jama-Journal of the American Medical Association. 2006, 296: 1255-1265.

    CAS  Google Scholar 

  1920. Fraser ML, Mok GS, Lee AH: Green tea and stroke prevention: emerging evidence. Complement Ther Med. 2007, 15: 46-53.

    PubMed  Google Scholar 

  1921. Ikeda M, Suzuki C, Umegaki K, Saito K, Tabuchi M, Tomita T: Preventive effects of green tea catechins on spontaneous stroke in rats. Medical Science Monitor. 2007, 13: BR40-BR45.

    CAS  PubMed  Google Scholar 

  1922. Nelson SK, Bose SK, Grunwald GK, Myhill P, McCord JM: The induction of human superoxide dismutase and catalase in vivo: A fundamentally new approach to antioxidant therapy. Free Radical Biology and Medicine. 2006, 40: 341-347.

    CAS  PubMed  Google Scholar 

  1923. Amidon GL, Lennernas H, Shah VP, Crison JR: A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995, 12: 413-20.

    CAS  PubMed  Google Scholar 

  1924. Lennernäs H, Abrahamsson B: The use of biopharmaceutic classification of drugs in drug discovery and development: current status and future extension. J Pharm Pharmacol. 2005, 57: 273-85.

    PubMed  Google Scholar 

  1925. Wu CY, Benet LZ: Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res. 2005, 22: 11-23.

    CAS  PubMed  Google Scholar 

  1926. Manach C, Williamson G, Morand C, Scalbert A, Remesy C: Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr. 2005, 81: 230S-242S.

    CAS  PubMed  Google Scholar 

  1927. Williamson G, Manach C: Bioavailability and bioefficacy of polyphenols in humans. II. Review of 93 intervention studies. Am J Clin Nutr. 2005, 81: 243S-255S.

    CAS  PubMed  Google Scholar 

  1928. Rechner AR, Kuhnle G, Bremner P, Hubbard GP, Moore KP, Rice-Evans CA: The metabolic fate of dietary polyphenols in humans. Free Radic Biol Med. 2002, 33: 220-35.

    CAS  PubMed  Google Scholar 

  1929. Ross JA, Kasum CM: Dietary flavonoids: bioavailability, metabolic effects, and safety. Annu Rev Nutr. 2002, 22: 19-34.

    CAS  PubMed  Google Scholar 

  1930. Scalbert A, Morand C, Manach C, Remesy C: Absorption and metabolism of polyphenols in the gut and impact on health. Biomedicine & Pharmacotherapy. 2002, 56: 276-282.

    CAS  Google Scholar 

  1931. Passamonti S, Vrhovsek U, Vanzo A, Mattivi F: Fast access of some grape pigments to the brain. J Agric Food Chem. 2005, 53: 7029-34.

    CAS  PubMed  Google Scholar 

  1932. Serra H, Mendes T, Bronze MR, Simplicio AL: Prediction of intestinal absorption and metabolism of pharmacologically active flavones and flavanones. Bioorg Med Chem. 2008, 16: 4009-18.

    CAS  PubMed  Google Scholar 

  1933. Heiss E, Herhaus C, Klimo K, Bartsch H, Gerhauser C: Nuclear factor kappa B is a molecular target for sulforaphane-mediated anti-inflammatory mechanisms. J Biol Chem. 2001, 276: 32008-15.

    CAS  PubMed  Google Scholar 

  1934. Gerhäuser C, Klimo K, Heiss E, Neumann I, Gamal-Eldeen A, Knauft J, Liu GY, Sitthimonchai S, Frank N: Mechanism-based in vitro screening of potential cancer chemopreventive agents. Mutat Res. 2003, 523–524: 163-72.

    PubMed  Google Scholar 

  1935. Manson MM: Cancer prevention – the potential for diet to modulate molecular signalling. Trends Mol Med. 2003, 9: 11-8.

    CAS  PubMed  Google Scholar 

  1936. Murakami A, Takahashi D, Hagihara K, Koshimizu K, Ohigashi H: Combinatorial effects of nonsteroidal anti-inflammatory drugs and food constituents on production of prostaglandin E2 and tumor necrosis factor-alpha in RAW264.7 murine macrophages. Biosci Biotechnol Biochem. 2003, 67: 1056-62.

    CAS  PubMed  Google Scholar 

  1937. Murakami A, Takahashi D, Koshimizu K, Ohigashi H: Synergistic suppression of superoxide and nitric oxide generation from inflammatory cells by combined food factors. Mutat Res. 2003, 523–524: 151-61.

    PubMed  Google Scholar 

  1938. Murakami A, Matsumoto K, Koshimizu K, Ohigashi H: Effects of selected food factors with chemopreventive properties on combined lipopolysaccharide- and interferon-gamma-induced IkappaB degradation in RAW264.7 macrophages. Cancer Lett. 2003, 195: 17-25.

    CAS  PubMed  Google Scholar 

  1939. Surh YJ, Ferguson LR: Dietary and medicinal antimutagens and anticarcinogens: molecular mechanisms and chemopreventive potential–highlights of a symposium. Mutat Res. 2003, 523–524: 1-8.

    PubMed  Google Scholar 

  1940. Alcaraz MJ, Vicente AM, Araico A, Dominguez JN, Terencio MC, Ferrandiz ML: Role of nuclear factor-kappaB and heme oxygenase-1 in the mechanism of action of an anti-inflammatory chalcone derivative in RAW 264.7 cells. Br J Pharmacol. 2004, 142: 1191-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  1941. Dorai T, Aggarwal BB: Role of chemopreventive agents in cancer therapy. Cancer Lett. 2004, 215: 129-40.

    CAS  PubMed  Google Scholar 

  1942. Feng R, Bowman LL, Lu Y, Leonard SS, Shi X, Jiang BH, Castranova V, Vallyathan V, Ding M: Blackberry extracts inhibit activating protein 1 activation and cell transformation by perturbing the mitogenic signaling pathway. Nutr Cancer. 2004, 50: 80-9.

    PubMed  Google Scholar 

  1943. Goyarzu P, Malin DH, Lau FC, Taglialatela G, Moon WD, Jennings R, Moy E, Moy D, Lippold S, Shukitt-Hale B, Joseph JA: Blueberry supplemented diet: effects on object recognition memory and nuclear factor-kappa B levels in aged rats. Nutr Neurosci. 2004, 7: 75-83.

    PubMed  Google Scholar 

  1944. Jeong WS, Kim IW, Hu R, Kong AN: Modulatory properties of various natural chemopreventive agents on the activation of NF-kappaB signaling pathway. Pharm Res. 2004, 21: 661-70.

    CAS  PubMed  Google Scholar 

  1945. Park OJ, Surh YJ: Chemopreventive potential of epigallocatechin gallate and genistein: evidence from epidemiological and laboratory studies. Toxicol Lett. 2004, 150: 43-56.

    CAS  PubMed  Google Scholar 

  1946. Pham NA, Jacobberger JW, Schimmer AD, Cao P, Gronda M, Hedley DW: The dietary isothiocyanate sulforaphane targets pathways of apoptosis, cell cycle arrest, and oxidative stress in human pancreatic cancer cells and inhibits tumor growth in severe combined immunodeficient mice. Mol Cancer Ther. 2004, 3: 1239-48.

    CAS  PubMed  Google Scholar 

  1947. Zhang Y: Cancer-preventive isothiocyanates: measurement of human exposure and mechanism of action. Mutat Res. 2004, 555: 173-90.

    CAS  PubMed  Google Scholar 

  1948. Bai SK, Lee SJ, Na HJ, Ha KS, Han JA, Lee H, Kwon YG, Chung CK, Kim YM: Beta-carotene inhibits inflammatory gene expression in lipopolysaccharide-stimulated macrophages by suppressing redox-based NF-kappaB activation. Exp Mol Med. 2005, 37: 323-34.

    CAS  PubMed  Google Scholar 

  1949. Chen C, Kong AN: Dietary cancer-chemopreventive compounds: from signaling and gene expression to pharmacological effects. Trends Pharmacol Sci. 2005, 26: 318-26.

    PubMed  Google Scholar 

  1950. Kundu JK, Surh YJ: Breaking the relay in deregulated cellular signal transduction as a rationale for chemoprevention with anti-inflammatory phytochemicals. Mutat Res. 2005, 591: 123-46.

    CAS  PubMed  Google Scholar 

  1951. Losso JN, Bawadi HA: Hypoxia inducible factor pathways as targets for functional foods. J Agric Food Chem. 2005, 53: 3751-68.

    CAS  PubMed  Google Scholar 

  1952. Ramos S, Alia M, Bravo L, Goya L: Comparative effects of food-derived polyphenols on the viability and apoptosis of a human hepatoma cell line (HepG2). J Agric Food Chem. 2005, 53: 1271-80.

    CAS  PubMed  Google Scholar 

  1953. Surh YJ, Kundu JK, Na HK, Lee JS: Redox-sensitive transcription factors as prime targets for chemoprevention with anti-inflammatory and antioxidative phytochemicals. J Nutr. 2005, 135: 2993S-3001S.

    CAS  PubMed  Google Scholar 

  1954. Xu C, Shen G, Chen C, Gelinas C, Kong AN: Suppression of NF-kappaB and NF-kappaB-regulated gene expression by sulforaphane and PEITC through IkappaBalpha, IKK pathway in human prostate cancer PC-3 cells. Oncogene. 2005, 24: 4486-95.

    CAS  PubMed  Google Scholar 

  1955. Zhang Y, Li J, Tang L: Cancer-preventive isothiocyanates: dichotomous modulators of oxidative stress. Free Radic Biol Med. 2005, 38: 70-7.

    PubMed  Google Scholar 

  1956. Wang SY, Feng R, Lu Y, Bowman L, Ding M: Inhibitory effect on activator protein-1, nuclear factor-kappaB, and cell transformation by extracts of strawberries (Fragaria × ananassa Duch.). J Agric Food Chem. 2005, 53: 4187-93.

    CAS  PubMed  Google Scholar 

  1957. Aggarwal BB, Shishodia S: Molecular targets of dietary agents for prevention and therapy of cancer. Biochem Pharmacol. 2006, 71: 1397-421.

    CAS  PubMed  Google Scholar 

  1958. Brigelius-Flohé R, Banning A: Sulforaphane and selenium, partners in adaptive response and prevention of cancer. Free Radic Res. 2006, 40: 775-87.

    PubMed  Google Scholar 

  1959. Jakubíkova J, Sedlák J, Bod'o J, Bao Y: Effect of isothiocyanates on nuclear accumulation of NF-kappaB, Nrf2, and thioredoxin in caco-2 cells. J Agric Food Chem. 2006, 54: 1656-62.

    PubMed  Google Scholar 

  1960. Jacob C: A scent of therapy: pharmacological implications of natural products containing redox-active sulfur atoms. Nat Prod Rep. 2006, 23: 851-63.

    CAS  PubMed  Google Scholar 

  1961. Keum YS, Yu S, Chang PP, Yuan X, Kim JH, Xu C, Han J, Agarwal A, Kong AN: Mechanism of action of sulforaphane: inhibition of p38 mitogen-activated protein kinase isoforms contributing to the induction of antioxidant response element-mediated heme oxygenase-1 in human hepatoma HepG2 cells. Cancer Res. 2006, 66: 8804-13.

    CAS  PubMed  Google Scholar 

  1962. Köhle C, Bock KW: Activation of coupled Ah receptor and Nrf2 gene batteries by dietary phytochemicals in relation to chemoprevention. Biochem Pharmacol. 2006, 72: 795-805.

    PubMed  Google Scholar 

  1963. Kresty LA, Frankel WL, Hammond CD, Baird ME, Mele JM, Stoner GD, Fromkes JJ: Transitioning from preclinical to clinical chemopreventive assessments of lyophilized black raspberries: interim results show berries modulate markers of oxidative stress in Barrett's esophagus patients. Nutr Cancer. 2006, 54: 148-56.

    CAS  PubMed  Google Scholar 

  1964. Lee KW, Kundu JK, Kim SO, Chun KS, Lee HJ, Surh YJ: Cocoa polyphenols inhibit phorbol ester-induced superoxide anion formation in cultured HL-60 cells and expression of cyclooxygenase-2 and activation of NF-kappaB and MAPKs in mouse skin in vivo. J Nutr. 2006, 136: 1150-5.

    CAS  PubMed  Google Scholar 

  1965. Lu H, Li J, Zhang D, Stoner GD, Huang C: Molecular mechanisms involved in chemoprevention of black raspberry extracts: from transcription factors to their target genes. Nutr Cancer. 2006, 54: 69-78.

    CAS  PubMed  Google Scholar 

  1966. Martin KR: Targeting apoptosis with dietary bioactive agents. Exp Biol Med. 2006, 231: 117-29.

    CAS  Google Scholar 

  1967. Na HK, Surh YJ: Transcriptional regulation via cysteine thiol modification: a novel molecular strategy for chemoprevention and cytoprotection. Mol Carcinog. 2006, 45: 368-80.

    CAS  PubMed  Google Scholar 

  1968. Pappa G, Lichtenberg M, Iori R, Barillari J, Bartsch H, Gerhauser C: Comparison of growth inhibition profiles and mechanisms of apoptosis induction in human colon cancer cell lines by isothiocyanates and indoles from Brassicaceae. Mutat Res. 2006, 599: 76-87.

    CAS  PubMed  Google Scholar 

  1969. Sandur SK, Ichikawa H, Sethi G, Ahn KS, Aggarwal BB: Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone) suppresses NF-kappaB activation and NF-kappaB-regulated gene products through modulation of p65 and IkappaBalpha kinase activation, leading to potentiation of apoptosis induced by cytokine and chemotherapeutic agents. J Biol Chem. 2006, 281: 17023-33.

    CAS  PubMed  Google Scholar 

  1970. Shen G, Xu C, Hu R, Jain MR, Gopalkrishnan A, Nair S, Huang MT, Chan JY, Kong AN: Modulation of nuclear factor E2-related factor 2-mediated gene expression in mice liver and small intestine by cancer chemopreventive agent curcumin. Mol Cancer Ther. 2006, 5: 39-51.

    CAS  PubMed  Google Scholar 

  1971. Thangapazham RL, Sharma A, Maheshwari RK: Multiple molecular targets in cancer chemoprevention by curcumin. Aaps J. 2006, 8: E443-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  1972. Vina J, Borras C, Gomez-Cabrera MC, Orr WC: Role of reactive oxygen species and (phyto)oestrogens in the modulation of adaptive response to stress. Free Radic Res. 2006, 40: 111-9.

    CAS  PubMed  Google Scholar 

  1973. Davis CD, Milner JA: Biomarkers for diet and cancer prevention research: potentials and challenges. Acta Pharmacol Sin. 2007, 28: 1262-73.

    CAS  PubMed  Google Scholar 

  1974. Higdon JV, Delage B, Williams DE, Dashwood RH: Cruciferous vegetables and human cancer risk: epidemiologic evidence and mechanistic basis. Pharmacol Res. 2007, 55: 224-36.

    CAS  PubMed  PubMed Central  Google Scholar 

  1975. Huber WW, Parzefall W: Thiols and the chemoprevention of cancer. Curr Opin Pharmacol. 2007, 7: 404-9.

    CAS  PubMed  Google Scholar 

  1976. Juge N, Mithen RF, Traka M: Molecular basis for chemoprevention by sulforaphane: a comprehensive review. Cell Mol Life Sci. 2007, 64: 1105-27.

    CAS  PubMed  Google Scholar 

  1977. Kampa M, Nifli AP, Notas G, Castanas E: Polyphenols and cancer cell growth. Rev Physiol Biochem Pharmacol. 2007, 159: 79-113.

    CAS  PubMed  Google Scholar 

  1978. Kwon KH, Barve A, Yu S, Huang MT, Kong AN: Cancer chemoprevention by phytochemicals: potential molecular targets, biomarkers and animal models. Acta Pharmacol Sin. 2007, 28: 1409-21.

    CAS  PubMed  Google Scholar 

  1979. Liby KT, Yore MM, Sporn MB: Triterpenoids and rexinoids as multifunctional agents for the prevention and treatment of cancer. Nat Rev Cancer. 2007, 7: 357-69.

    CAS  PubMed  Google Scholar 

  1980. Murakami A, Ohigashi H: Targeting NOX, INOS and COX-2 in inflammatory cells: chemoprevention using food phytochemicals. Int J Cancer. 2007, 121: 2357-63.

    CAS  PubMed  Google Scholar 

  1981. Nair S, Li W, Kong AN: Natural dietary anti-cancer chemopreventive compounds: redox-mediated differential signaling mechanisms in cytoprotection of normal cells versus cytotoxicity in tumor cells. Acta Pharmacol Sin. 2007, 28: 459-72.

    CAS  PubMed  Google Scholar 

  1982. Neto CC: Cranberry and its phytochemicals: a review of in vitro anticancer studies. J Nutr. 2007, 137: 186S-193S.

    CAS  PubMed  Google Scholar 

  1983. Syed DN, Afaq F, Mukhtar H: Pomegranate derived products for cancer chemoprevention. Semin Cancer Biol. 2007, 17: 377-85.

    CAS  PubMed  Google Scholar 

  1984. Zafra-Stone S, Yasmin T, Bagchi M, Chatterjee A, Vinson JA, Bagchi D: Berry anthocyanins as novel antioxidants in human health and disease prevention. Mol Nutr Food Res. 2007, 51: 675-83.

    CAS  PubMed  Google Scholar 

  1985. Gopalakrishnan A, Tony Kong AN: Anticarcinogenesis by dietary phytochemicals: cytoprotection by Nrf2 in normal cells and cytotoxicity by modulation of transcription factors NF-kappa B and AP-1 in abnormal cancer cells. Food Chem Toxicol. 2008, 46: 1257-70.

    CAS  PubMed  Google Scholar 

  1986. Nair S, Hebbar V, Shen G, Gopalakrishnan A, Khor TO, Yu S, Xu C, Kong AN: Synergistic effects of a combination of dietary factors sulforaphane and (-) epigallocatechin-3-gallate in HT-29 AP-1 human colon carcinoma cells. Pharm Res. 2008, 25: 387-99.

    CAS  PubMed  Google Scholar 

  1987. Neto CC, Amoroso JW, Liberty AM: Anticancer activities of cranberry phytochemicals: An update. Mol Nutr Food Res. 2008

    Google Scholar 

  1988. Pan MH, Ghai G, Ho CT: Food bioactives apoptosis, and cancer. Mol Nutr Food Res. 2008, 52: 43-52.

    CAS  PubMed  Google Scholar 

  1989. Ramos S: Cancer chemoprevention and chemotherapy: dietary polyphenols and signalling pathways. Mol Nutr Food Res. 2008, 52: 507-26.

    CAS  PubMed  Google Scholar 

  1990. Youn HS, Lim HJ, Lee HJ, Hwang D, Yang M, Jeon R, Ryu JH: Garlic (Allium sativum) extract inhibits lipopolysaccharide-induced Toll-like receptor 4 dimerization. Biosci Biotechnol Biochem. 2008, 72: 368-75.

    CAS  PubMed  Google Scholar 

  1991. Dower SK, Qwarnstrom EE: Signalling networks, inflammation and innate immunity. Biochem Soc Trans. 2003, 31: 1462-71.

    CAS  PubMed  Google Scholar 

  1992. Leonard SS, Harris GK, Shi XL: Metal-induced oxidative stress and signal transduction. Free Rad Biol Med. 2004, 37: 1921-1942.

    CAS  PubMed  Google Scholar 

  1993. Matsuzawa A, Saegusa K, Noguchi T, Sadamitsu C, Nishitoh H, Nagai S, Koyasu S, Matsumoto K, Takeda K, Ichijo H: ROS-dependent activation of the TRAF6-ASK1-p38 pathway is selectively required for TLR4-mediated innate immunity. Nat Immunol. 2005, 6: 587-92.

    CAS  PubMed  Google Scholar 

  1994. Schreck R, Rieber P, Baeuerle PA: Reactive oxygen intermediates as apparently widely used messengers in the activation of the NF-Kappa-B transcription Factor and HIV-1. EMBO J. 1991, 10: 2247-2258.

    CAS  PubMed  PubMed Central  Google Scholar 

  1995. Schreck R, Albermann K, Baeuerle PA: Nuclear factor kappa-B – an oxidative stress-responsive transcription factor of eukaryotic cells (a Review). Free Rad Res Comm. 1992, 17: 221-237.

    CAS  Google Scholar 

  1996. Schreck R, Meier B, Mannel DN, Droge W, Baeuerle PA: Dithiocarbamates as potent inhibitors of nuclear factor kappa B activation in intact cells. J Exp Med. 1992, 175: 1181-94.

    CAS  PubMed  Google Scholar 

  1997. Mohan N, Sadeghi K, Reiter RJ, Meltz ML: The neurohormone melatonin inhibits cytokine, mitogen and ionizing radiation induced NF-kappa B. Biochemistry and Molecular Biology International. 1995, 37: 1063-1070.

    CAS  PubMed  Google Scholar 

  1998. Baeuerle PA, Baltimore D: NF-kappa B: ten years after. Cell. 1996, 87: 13-20.

    CAS  PubMed  Google Scholar 

  1999. Baldwin AS: The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol. 1996, 14: 649-83.

    CAS  PubMed  Google Scholar 

  2000. Dalton TP, Shertzer HG, Puga A: Regulation of gene expression by reactive oxygen. Annu Rev Pharmacol Toxicol. 1999, 39: 67-101.

    CAS  PubMed  Google Scholar 

  2001. Li C, Browder W, Kao RL: Early activation of transcription factor NF-kappaB during ischemia in perfused rat heart. Am J Physiol. 1999, 276: H543-52.

    CAS  PubMed  Google Scholar 

  2002. Wang S, Leonard SS, Castranova V, Vallyathan V, Shi X: The role of superoxide radical in TNF-alpha induced NF-kappaB activation. Ann Clin Lab Sci. 1999, 29: 192-9.

    CAS  PubMed  Google Scholar 

  2003. Knight JA: Review: Free radicals, antioxidants, and the immune system. Ann Clin Lab Sci. 2000, 30: 145-158.

    CAS  PubMed  Google Scholar 

  2004. Schoonbroodt S, Piette J: Oxidative stress interference with the nuclear factor-kappa B activation pathways. Biochem Pharmacol. 2000, 60: 1075-83.

    CAS  PubMed  Google Scholar 

  2005. Forman HJ, Torres M: Reactive oxygen species and cell signaling: respiratory burst in macrophage signaling. Am J Respir Crit Care Med. 2002, 166: S4-8.

    PubMed  Google Scholar 

  2006. Wang SW, Kotamraju S, Konorev E, Kalivendi S, Joseph J, Kalyanaraman B: Activation of nuclear factor-kappa B during doxorubicin-induced apoptosis in endothelial cells and myocytes is pro-apoptotic: the role of hydrogen peroxide. Biochemical Journal. 2002, 367: 729-740.

    CAS  PubMed  PubMed Central  Google Scholar 

  2007. Haddad JJ: Science review: Redox and oxygen-sensitive transcription factors in the regulation of oxidant-mediated lung injury: role for nuclear factor-kappaB. Crit Care. 2002, 6: 481-90.

    PubMed  PubMed Central  Google Scholar 

  2008. Asehnoune K, Strassheim D, Mitra S, Kim JY, Abraham E: Involvement of reactive oxygen species in Toll-like receptor 4-dependent activation of NF-kappa B. J Immunol. 2004, 172: 2522-9.

    CAS  PubMed  Google Scholar 

  2009. Poli G, Leonarduzzi G, Biasi F, Chiarpotto E: Oxidative stress and cell signalling. Curr Med Chem. 2004, 11: 1163-1182.

    CAS  PubMed  Google Scholar 

  2010. Manna SK, Sarkar S, Barr J, Wise K, Barrera EV, Jejelowo O, Rice-Ficht AC, Ramesh GT: Single-walled carbon nanotube induces oxidative stress and activates nuclear transcription factor-kappaB in human keratinocytes. Nano Lett. 2005, 5: 1676-84.

    CAS  PubMed  PubMed Central  Google Scholar 

  2011. Bubici C, Papa S, Dean K, Franzoso G: Mutual cross-talk between reactive oxygen species and nuclear factor-kappa B: molecular basis and biological significance. Oncogene. 2006, 25: 6731-48.

    CAS  PubMed  Google Scholar 

  2012. Bubici C, Papa S, Pham CG, Zazzeroni F, Franzoso G: The NF-kappaB-mediated control of ROS and JNK signaling. Histol Histopathol. 2006, 21: 69-80.

    CAS  PubMed  Google Scholar 

  2013. Gloire G, Legrand-Poels S, Piette J: NF-kappaB activation by reactive oxygen species: fifteen years later. Biochem Pharmacol. 2006, 72: 1493-505.

    CAS  PubMed  Google Scholar 

  2014. Kim JY, Jung KJ, Choi JS, Chung HY: Modulation of the age-related nuclear factor-kappaB (NF-kappaB) pathway by hesperetin. Aging Cell. 2006, 5: 401-11.

    CAS  PubMed  Google Scholar 

  2015. Li LX, Frei B: Iron chelation inhibits NF-kappa B-mediated adhesion molecule expression by inhibiting p22(phox) protein expression and NADPH oxidase activity. Arteriosclerosis Thromb Vasc Biol. 2006, 26: 2638-2643.

    CAS  Google Scholar 

  2016. Kramer HF, Goodyear LJ: Exercise MAPK, and NF-kappaB signaling in skeletal muscle. J Appl Physiol. 2007, 103: 388-95.

    CAS  PubMed  Google Scholar 

  2017. Glauert HP: Vitamin E and NF-kappaB activation: a review. Vitam Horm. 2007, 76: 135-53.

    CAS  PubMed  Google Scholar 

  2018. Perkins ND: Integrating cell-signalling pathways with NF-kappaB and IKK function. Nat Rev Mol Cell Biol. 2007, 8: 49-62.

    CAS  PubMed  Google Scholar 

  2019. Uwe S: Anti-inflammatory interventions of NF-kappaB signaling: potential applications and risks. Biochem Pharmacol. 2008, 75: 1567-79.

    CAS  PubMed  Google Scholar 

  2020. Ghosh S, Karin M: Missing pieces in the NF-kappaB puzzle. Cell. 2002, 109 (Suppl): S81-96.

    CAS  PubMed  Google Scholar 

  2021. Hayden MS, Ghosh S: Signaling to NF-kappaB. Genes Dev. 2004, 18: 2195-224.

    CAS  PubMed  Google Scholar 

  2022. Nelson DE, Ihekwaba AEC, Elliott M, Gibney CA, Foreman BE, Nelson G, See V, Horton CA, Spiller DG, Edwards SW, McDowell HP, Unitt JF, Sullivan E, Grimley R, Benson N, Broomhead DS, Kell DB, White MRH: Oscillations in NF-κB signalling control the dynamics of target gene expression. Science. 2004, 306: 704-708.

    CAS  PubMed  Google Scholar 

  2023. Hoffmann A, Baltimore D: Circuitry of nuclear factor kappaB signaling. Immunol Rev. 2006, 210: 171-86.

    PubMed  Google Scholar 

  2024. Perkins ND, Gilmore TD: Good cop, bad cop: the different faces of NF-kappaB. Cell Death Differ. 2006, 13: 759-72.

    CAS  PubMed  Google Scholar 

  2025. Chen CC, Wang JK, Lin SB: Antisense oligonucleotides targeting protein kinase C-alpha, -beta I, or -delta but not -eta inhibit lipopolysaccharide-induced nitric oxide synthase expression in RAW 264.7 macrophages: involvement of a nuclear factor kappa B-dependent mechanism. J Immunol. 1998, 161: 6206-14.

    CAS  PubMed  Google Scholar 

  2026. Sanlioglu S, Williams CM, Samavati L, Butler NS, Wang G, McCray PB, Ritchie TC, Hunninghake GW, Zandi E, Engelhardt JF: Lipopolysaccharide induces Rac1-dependent reactive oxygen species formation and coordinates tumor necrosis factor-alpha secretion through IKK regulation of NF-kappa B. J Biol Chem. 2001, 276: 30188-98.

    CAS  PubMed  Google Scholar 

  2027. Hsu HY, Wen MH: Lipopolysaccharide-mediated reactive oxygen species and signal transduction in the regulation of interleukin-1 gene expression. J Biol Chem. 2002, 277: 22131-9.

    CAS  PubMed  Google Scholar 

  2028. Macdonald J, Galley HF, Webster NR: Oxidative stress and gene expression in sepsis. Br J Anaesth. 2003, 90: 221-32.

    CAS  PubMed  Google Scholar 

  2029. Park HS, Jung HY, Park EY, Kim J, Lee WJ, Bae YS: Cutting edge: direct interaction of TLR4 with NAD(P)H oxidase 4 isozyme is essential for lipopolysaccharide-induced production of reactive oxygen species and activation of NF-kappa B. J Immunol. 2004, 173: 3589-93.

    CAS  PubMed  Google Scholar 

  2030. Pham CG, Bubici C, Zazzeroni F, Papa S, Jones J, Alvarez K, Jayawardena S, De Smaele E, Cong R, Beaumont C, Torti FM, Torti SV, Franzoso G: Ferritin heavy chain upregulation by NF-kappaB inhibits TNFalpha-induced apoptosis by suppressing reactive oxygen species. Cell. 2004, 119: 529-42.

    CAS  PubMed  Google Scholar 

  2031. Allan SM, Tyrrell PJ, Rothwell NJ: Interleukin-1 and neuronal injury. Nat Rev Immunol. 2005, 5: 629-40.

    CAS  PubMed  Google Scholar 

  2032. Ihekwaba AEC, Broomhead DS, Grimley R, Benson N, Kell DB: Sensitivity analysis of parameters controlling oscillatory signalling in the NF-κB pathway: the roles of IKK and IκBα. Systems Biology. 2004, 1: 93-103.

    CAS  PubMed  Google Scholar 

  2033. Yue H, Brown M, Knowles J, Wang H, Broomhead DS, Kell DB: Insights into the behaviour of systems biology models from dynamic sensitivity and identifiability analysis: a case study of an NF-kappaB signalling pathway. Mol Biosyst. 2006, 2: 640-649.

    CAS  PubMed  Google Scholar 

  2034. Ihekwaba AEC, Broomhead DS, Grimley R, Benson N, White MRH, Kell DB: Synergistic control of oscillations in the NF-κB signalling pathway. IEE Systems Biology. 2005, 152: 153-160.

    CAS  PubMed  Google Scholar 

  2035. Mignotte B, Vayssiere JL: Mitochondria and apoptosis. Eur J Biochem. 1998, 252: 1-15.

    CAS  PubMed  Google Scholar 

  2036. Fleury C, Mignotte B, Vayssiere JL: Mitochondrial reactive oxygen species in cell death signaling. Biochimie. 2002, 84: 131-41.

    CAS  PubMed  Google Scholar 

  2037. Bharti AC, Aggarwal BB: Nuclear factor-kappa B and cancer: its role in prevention and therapy. Biochem Pharmacol. 2002, 64: 883-8.

    CAS  PubMed  Google Scholar 

  2038. Richmond A: NF-κB, chemokine gene transcription and tumour growth. Nat Rev Immunol. 2002, 2: 664-74.

    CAS  PubMed  PubMed Central  Google Scholar 

  2039. Zhang Z, Rigas B: NF-kappaB, inflammation and pancreatic carcinogenesis: NF-kappaB as a chemoprevention target (review). Int J Oncol. 2006, 29: 185-92.

    CAS  PubMed  Google Scholar 

  2040. Calzado MA, Bacher S, Schmitz ML: NF-kappaB inhibitors for the treatment of inflammatory diseases and cancer. Curr Med Chem. 2007, 14: 367-76.

    CAS  PubMed  Google Scholar 

  2041. Roman-Blas JA, Jimenez SA: NF-kappaB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis. Osteoarthritis Cartilage. 2006, 14: 839-48.

    CAS  PubMed  Google Scholar 

  2042. Saklatvala J: Inflammatory signaling in cartilage: MAPK and NF-kappaB pathways in chondrocytes and the use of inhibitors for research into pathogenesis and therapy of osteoarthritis. Curr Drug Targets. 2007, 8: 305-13.

    CAS  PubMed  Google Scholar 

  2043. Simmonds RE, Foxwell BM: NF-{kappa}B and its relevance to arthritis and inflammation. Rheumatology (Oxford). 2008

    Google Scholar 

  2044. Smahi A, Courtois G, Rabia SH, Doffinger R, Bodemer C, Munnich A, Casanova JL, Israel A: The NF-kappaB signalling pathway in human diseases: from incontinentia pigmenti to ectodermal dysplasias and immune-deficiency syndromes. Hum Mol Genet. 2002, 11: 2371-5.

    CAS  PubMed  Google Scholar 

  2045. Lezoualc'h F, Sparapani M, Behl C: N-acetyl-serotonin (normelatonin) and melatonin protect neurons against oxidative challenges and suppress the activity of the transcription factor NF-kappaB. J Pineal Res. 1998, 24: 168-78.

    PubMed  Google Scholar 

  2046. Post A, Holsboer F, Behl C: Induction of NF-kappaB activity during haloperidol-induced oxidative toxicity in clonal hippocampal cells: suppression of NF-kappaB and neuroprotection by antioxidants. J Neurosci. 1998, 18: 8236-46.

    CAS  PubMed  Google Scholar 

  2047. Herrera J, Nava M, Romero F, Rodriguez-Iturbe B: Melatonin prevents oxidative stress resulting from iron and erythropoietin administration. Am J Kidney Dis. 2001, 37: 750-7.

    CAS  PubMed  Google Scholar 

  2048. Beni SM, Kohen R, Reiter RJ, Tan DX, Shohami E: Melatonin-induced neuroprotection after closed head injury is associated with increased brain antioxidants and attenuated late-phase activation of NF-kappaB and AP-1. FASEB J. 2004, 18: 149-51.

    CAS  PubMed  Google Scholar 

  2049. Rodriguez MI, Escames G, Lopez LC, Lopez A, Garcia JA, Ortiz F, Acuna-Castroviejo D: Chronic melatonin treatment reduces the age-dependent inflammatory process in senescence-accelerated mice. J Pineal Res. 2007, 42: 272-9.

    CAS  PubMed  Google Scholar 

  2050. Takada Y, Mukhopadhyay A, Kundu GC, Mahabeleshwar GH, Singh S, Aggarwal BB: Hydrogen peroxide activates NF-kappa B through tyrosine phosphorylation of I kappa B alpha and serine phosphorylation of p65: evidence for the involvement of I kappa B alpha kinase and Syk protein-tyrosine kinase. J Biol Chem. 2003, 278: 24233-41.

    CAS  PubMed  Google Scholar 

  2051. Hsieh CC, Papaconstantinou J: Thioredoxin-ASK1 complex levels regulate ROS-mediated p38 MAPK pathway activity in livers of aged and long-lived Snell dwarf mice. FASEB J. 2006, 20: 259-68.

    CAS  PubMed  PubMed Central  Google Scholar 

  2052. Walter DH, Dimmeler S, Zeiher AM: Effects of statins on endothelium and endothelial progenitor cell recruitment. Semin Vasc Med. 2004, 4: 385-93.

    PubMed  Google Scholar 

  2053. Madonna R, Di Napoli P, Massaro M, Grilli A, Felaco M, De Caterina A, Tang D, De Caterina R, Geng YJ: Simvastatin attenuates expression of cytokine-inducible nitric-oxide synthase in embryonic cardiac myoblasts. J Biol Chem. 2005, 280: 13503-11.

    CAS  PubMed  Google Scholar 

  2054. Urbich C, Dimmeler S: Risk factors for coronary artery disease, circulating endothelial progenitor cells, and the role of HMG-CoA reductase inhibitors. Kidney Int. 2005, 67: 1672-6.

    CAS  PubMed  Google Scholar 

  2055. Gilmore TD, Herscovitch M: Inhibitors of NF-kappaB signaling: 785 and counting. Oncogene. 2006, 25: 6887-99.

    CAS  PubMed  Google Scholar 

  2056. Lazzerini PE, Lorenzini S, Selvi E, Capecchi PL, Chindamo D, Bisogno S, Ghittoni R, Natale MR, Caporali F, Giuntini S, Marcolongo R, Galeazzi M, Laghi-Pasini F: Simvastatin inhibits cytokine production and nuclear factor-kB activation in interleukin 1beta-stimulated synoviocytes from rheumatoid arthritis patients. Clin Exp Rheumatol. 2007, 25: 696-700.

    CAS  PubMed  Google Scholar 

  2057. Lee JY, Kim JS, Kim JM, Kim N, Jung HC, Song IS: Simvastatin inhibits NF-kappaB signaling in intestinal epithelial cells and ameliorates acute murine colitis. Int Immunopharmacol. 2007, 7: 241-8.

    CAS  PubMed  Google Scholar 

  2058. Lin SJ, Chen YH, Lin FY, Hsieh LY, Wang SH, Lin CY, Wang YC, Ku HH, Chen JW, Chen YL: Pravastatin induces thrombomodulin expression in TNFalpha-treated human aortic endothelial cells by inhibiting Rac1 and Cdc42 translocation and activity. J Cell Biochem. 2007, 101: 642-53.

    CAS  PubMed  Google Scholar 

  2059. Dolga AM, Nijholt IM, Ostroveanu A, Ten Bosch Q, Luiten PG, Eisel UL: Lovastatin induces neuroprotection through tumor necrosis factor receptor 2 signaling pathways. J Alzheimers Dis. 2008, 13: 111-22.

    CAS  PubMed  Google Scholar 

  2060. Gao F, Linhartova L, Johnston AM, Thickett DR: Statins and sepsis. British Journal of Anaesthesia. 2008, 100: 288-298.

    CAS  PubMed  Google Scholar 

  2061. Galter D, Mihm S, Dröge W: Distinct effects of glutathione disulphide on the nuclear transcription factor kappa B and the activator protein-1. Eur J Biochem. 1994, 221: 639-48.

    CAS  PubMed  Google Scholar 

  2062. Kretz-Remy C, Mehlen P, Mirault ME, Arrigo AP: Inhibition of I kappa B-alpha phosphorylation and degradation and subsequent NF-kappa B activation by glutathione peroxidase overexpression. J Cell Biol. 1996, 133: 1083-93.

    CAS  PubMed  Google Scholar 

  2063. García-Ruiz C, Colell A, Morales A, Kaplowitz N, Fernández-Checa JC: Role of oxidative stress generated from the mitochondrial electron transport chain and mitochondrial glutathione status in loss of mitochondrial function and activation of transcription factor nuclear factor-kappa B: studies with isolated mitochondria and rat hepatocytes. Mol Pharmacol. 1995, 48: 825-34.

    PubMed  Google Scholar 

  2064. Mihm S, Galter D, Dröge W: Modulation of transcription factor NF kappa B activity by intracellular glutathione levels and by variations of the extracellular cysteine supply. Faseb J. 1995, 9: 246-52.

    PubMed  Google Scholar 

  2065. Ginn-Pease ME, Whisler RL: Optimal NF kappa B mediated transcriptional responses in Jurkat T cells exposed to oxidative stress are dependent on intracellular glutathione and costimulatory signals. Biochem Biophys Res Commun. 1996, 226: 695-702.

    CAS  PubMed  Google Scholar 

  2066. Pinkus R, Weiner LM, Daniel V: Role of oxidants and antioxidants in the induction of AP-1, NF-kappaB, and glutathione S-transferase gene expression. J Biol Chem. 1996, 271: 13422-9.

    CAS  PubMed  Google Scholar 

  2067. Brigelius-Flohé R, Friedrichs B, Maurer S, Schultz M, Streicher R: Interleukin-1-induced nuclear factor kappa B activation is inhibited by overexpression of phospholipid hydroperoxide glutathione peroxidase in a human endothelial cell line. Biochem J. 1997, 328 (Pt 1): 199-203.

    PubMed  PubMed Central  Google Scholar 

  2068. Sen CK, Khanna S, Reznick AZ, Roy S, Packer L: Glutathione regulation of tumor necrosis factor-alpha-induced NF-kappa B activation in skeletal muscle-derived L6 cells. Biochem Biophys Res Commun. 1997, 237: 645-9.

    CAS  PubMed  Google Scholar 

  2069. Cho S, Urata Y, Iida T, Goto S, Yamaguchi M, Sumikawa K, Kondo T: Glutathione downregulates the phosphorylation of I kappa B: autoloop regulation of the NF-kappa B-mediated expression of NF-kappa B subunits by TNF-alpha in mouse vascular endothelial cells. Biochem Biophys Res Commun. 1998, 253: 104-8.

    CAS  PubMed  Google Scholar 

  2070. Rokutan K, Teshima S, Miyoshi M, Kawai T, Nikawa T, Kishi K: Glutathione depletion inhibits oxidant-induced activation of nuclear factor-kappa B, AP-1, and c-Jun/ATF-2 in cultured guinea-pig gastric epithelial cells. J Gastroenterol. 1998, 33: 646-55.

    CAS  PubMed  Google Scholar 

  2071. Fernandez PC, Machado J, Heussler VT, Botteron C, Palmer GH, Dobbelaere DA: The inhibition of NF-kappaB activation pathways and the induction of apoptosis by dithiocarbamates in T cells are blocked by the glutathione precursor N-acetyl-L-cysteine. Biol Chem. 1999, 380: 1383-94.

    CAS  PubMed  Google Scholar 

  2072. Haddad JJ, Olver RE, Land SC: Antioxidant/pro-oxidant equilibrium regulates HIF-1alpha and NF-kappa B redox sensitivity. Evidence for inhibition by glutathione oxidation in alveolar epithelial cells. J Biol Chem. 2000, 275: 21130-9.

    CAS  PubMed  Google Scholar 

  2073. Haddad JJ, Land SC: O2-evoked regulation of HIF-1alpha and NF-kappaB in perinatal lung epithelium requires glutathione biosynthesis. Am J Physiol Lung Cell Mol Physiol. 2000, 278: L492-503.

    CAS  PubMed  Google Scholar 

  2074. Rahman I, MacNee W: Oxidative stress and regulation of glutathione in lung inflammation. Eur Respir J. 2000, 16: 534-54.

    CAS  PubMed  Google Scholar 

  2075. Rahman I, MacNee W: Regulation of redox glutathione levels and gene transcription in lung inflammation: therapeutic approaches. Free Radic Biol Med. 2000, 28: 1405-20.

    CAS  PubMed  Google Scholar 

  2076. Davis W, Ronai Z, Tew KD: Cellular thiols and reactive oxygen species in drug-induced apoptosis. J Pharmacol Exp Ther. 2001, 296: 1-6.

    CAS  PubMed  Google Scholar 

  2077. Nishi T, Shimizu N, Hiramoto M, Sato I, Yamaguchi Y, Hasegawa M, Aizawa S, Tanaka H, Kataoka K, Watanabe H, Handa H: Spatial redox regulation of a critical cysteine residue of NF-kappa B in vivo. J Biol Chem. 2002, 277: 44548-56.

    CAS  PubMed  Google Scholar 

  2078. Biswas SK, McClure D, Jimenez LA, Megson IL, Rahman I: Curcumin induces glutathione biosynthesis and inhibits NF-kappaB activation and interleukin-8 release in alveolar epithelial cells: mechanism of free radical scavenging activity. Antioxid Redox Signal. 2005, 7: 32-41.

    CAS  PubMed  Google Scholar 

  2079. Biswas SK, Newby DE, Rahman I, Megson IL: Depressed glutathione synthesis precedes oxidative stress and atherogenesis in Apo-E(-/-) mice. Biochem Biophys Res Commun. 2005, 338: 1368-73.

    CAS  PubMed  Google Scholar 

  2080. Rahman I, Biswas SK, Jimenez LA, Torres M, Forman HJ: Glutathione, stress responses, and redox signaling in lung inflammation. Antioxid Redox Signal. 2005, 7: 42-59.

    CAS  PubMed  Google Scholar 

  2081. Cuzzocrea S, Chatterjee PK, Mazzon E, Dugo L, Serraino I, Britti D, Mazzullo G, Caputi AP, Thiemermann C: Pyrrolidine dithiocarbamate attenuates the development of acute and chronic inflammation. Br J Pharmacol. 2002, 135: 496-510.

    CAS  PubMed  PubMed Central  Google Scholar 

  2082. Desoize B: Metals and metal compounds in carcinogenesis. In Vivo. 2003, 17: 529-539.

    CAS  PubMed  Google Scholar 

  2083. Xiong SG, She HY, Takeuchi H, Han B, Engelhardt JF, Barton CH, Zandi E, Giulivi C, Tsukamoto H: Signaling role of intracellular iron in NF-kappa B activation. Journal of Biological Chemistry. 2003, 278: 17646-17654.

    CAS  PubMed  Google Scholar 

  2084. Parodi FE, Mao D, Ennis TL, Bartoli MA, Thompson RW: Suppression of experimental abdominal aortic aneurysms in mice by treatment with pyrrolidine dithiocarbamate, an antioxidant inhibitor of nuclear factor-kappaB. J Vasc Surg. 2005, 41: 479-89.

    PubMed  Google Scholar 

  2085. Antosiewicz J, Ziolkowski W, Kaczor JJ, Herman-Antosiewicz A: Tumor necrosis factor-alpha-induced reactive oxygen species formation is mediated by JNK1-dependent ferritin degradation and elevation of labile iron pool. Free Radic Biol Med. 2007, 43: 265-70.

    CAS  PubMed  Google Scholar 

  2086. Vélez-Pardo C, Ospina GG, Del Río MJ: Abeta[25–35] peptide and iron promote apoptosis in lymphocytes by an oxidative stress mechanism: involvement of H2O2, caspase-3, NF-kappaB, p53 and c-Jun. Neurotoxicology. 2002, 23: 351-65.

    PubMed  Google Scholar 

  2087. Bisti S, Soteriadou K: Is the reactive oxygen species-dependent-NF-kappaB activation observed in iron-loaded BALB/c mice a key process preventing growth of Leishmania major progeny and tissue-damage?. Microbes Infect. 2006, 8: 1473-82.

    CAS  PubMed  Google Scholar 

  2088. Bu DX, Hemdahl AL, Gabrielsen A, Fuxe J, Zhu C, Eriksson P, Yan ZQ: Induction of neutrophil gelatinase-associated lipocalin in vascular injury via activation of nuclear factor-kappaB. Am J Pathol. 2006, 169: 2245-53.

    CAS  PubMed  PubMed Central  Google Scholar 

  2089. Sommer G, Weise S, Kralisch S, Lossner U, Bluher M, Stumvoll M, Fasshauer M: Lipocalin-2 is induced by interleukin-1beta in murine adipocytes in vitro. J Cell Biochem. 2008

    Google Scholar 

  2090. Lee P, Peng H, Gelbart T, Wang L, Beutler E: Regulation of hepcidin transcription by interleukin-1 and interleukin-6. Proc Natl Acad Sci USA. 2005, 102: 1906-10.

    CAS  PubMed  PubMed Central  Google Scholar 

  2091. Duvigneau JC, Piskernik C, Haindl S, Kloesch B, Hartl RT, Huttemann M, Lee I, Ebel T, Moldzio R, Gemeiner M, Redl H, Kozlov AV: A novel endotoxin-induced pathway: upregulation of heme oxygenase 1, accumulation of free iron, and free iron-mediated mitochondrial dysfunction. Lab Invest. 2008, 88: 70-7.

    CAS  PubMed  Google Scholar 

  2092. Whitcomb DC, Aoun E, Vodovotz Y, Clermont G, Barmada MM: Evaluating disorders with a complex genetics basis. the future roles of meta-analysis and systems biology. Dig Dis Sci. 2005, 50: 2195-202.

    PubMed  Google Scholar 

  2093. Xiong M, Feghali-Bostwick CA, Arnett FC, Zhou X: A systems biology approach to genetic studies of complex diseases. FEBS Lett. 2005, 579: 5325-32.

    CAS  PubMed  Google Scholar 

  2094. Ideker T, Galitski T, Hood L: A new approach to decoding life: systems biology. Annu Rev Genomics Hum Genet. 2001, 2: 343-72.

    CAS  PubMed  Google Scholar 

  2095. Barabási A-L, Oltvai ZN: Network biology: understanding the cell's functional organization. Nat Rev Genet. 2004, 5: 101-113.

    PubMed  Google Scholar 

  2096. Morel NM, Holland JM, Greef van der J, Marple EW, Clish C, Loscalzo J, Naylor S: Primer on medical genomics. Part XIV: Introduction to systems biology–a new approach to understanding disease and treatment. Mayo Clin Proc. 2004, 79: 651-8.

    CAS  PubMed  Google Scholar 

  2097. Khalil IG, Hill C: Systems biology for cancer. Curr Opin Oncol. 2005, 17: 44-8.

    CAS  PubMed  Google Scholar 

  2098. Moore JH, Boczko EM, Summar ML: Connecting the dots between genes biochemistry, and disease susceptibility: systems biology modeling in human genetics. Mol Genet Metab. 2005, 84: 104-11.

    CAS  PubMed  Google Scholar 

  2099. Hornberg JJ, Bruggeman FJ, Westerhoff HV, Lankelma J: Cancer: a Systems Biology disease. Biosystems. 2006, 83: 81-90.

    CAS  PubMed  Google Scholar 

  2100. Lemberger T: Systems biology in human health and disease. Mol Syst Biol. 2007, 3: 136.

    PubMed  PubMed Central  Google Scholar 

  2101. Loscalzo J, Kohane I, Barabasi AL: Human disease classification in the postgenomic era: a complex systems approach to human pathobiology. Mol Syst Biol. 2007, 3: 124.

    PubMed  PubMed Central  Google Scholar 

  2102. Schnackenberg LK, Beger RD: Monitoring the health to disease continuum with global metabolic profiling and systems biology. Pharmacogenomics. 2006, 7: 1077-86.

    CAS  PubMed  Google Scholar 

  2103. Featherstone DE, Broadie K: Wrestling with pleiotropy: genomic and topological analysis of the yeast gene expression network. Bioessays. 2002, 24: 267-74.

    CAS  PubMed  Google Scholar 

  2104. Castrillo JI, Zeef LA, Hoyle DC, Zhang N, Hayes A, Gardner DCJ, Cornell MJ, Petty J, Hakes L, Wardleworth L, Rash B, Brown M, Dunn WB, Broadhurst D, O'Donoghue K, Hester SS, Dunkley TPJ, Hart SR, Swainston N, Li P, Gaskell SJ, Paton NW, Lilley KS, Kell DB, Oliver SG: Growth control of the eukaryote cell: a systems biology study in yeast. J. Biol. 2007, 6: 4.

    PubMed  PubMed Central  Google Scholar 

  2105. Raamsdonk LM, Teusink B, Broadhurst D, Zhang N, Hayes A, Walsh M, Berden JA, Brindle KM, Kell DB, Rowland JJ, Westerhoff HV, van Dam K, Oliver SG: A functional genomics strategy that uses metabolome data to reveal the phenotype of silent mutations. Nature Biotechnol. 2001, 19: 45-50.

    CAS  Google Scholar 

  2106. Mo ML, Jamshidi N, Palsson BO: A genome-scale, constraint-based approach to systems biology of human metabolism. Mol Biosyst. 2007, 3: 598-603.

    CAS  PubMed  Google Scholar 

  2107. Kell DB: Metabolomics and systems biology: making sense of the soup. Curr Op Microbiol. 2004, 7: 296-307.

    CAS  Google Scholar 

  2108. Kell DB, Brown M, Davey HM, Dunn WB, Spasic I, Oliver SG: Metabolic footprinting and Systems Biology: the medium is the message. Nat Rev Microbiol. 2005, 3: 557-565.

    CAS  PubMed  Google Scholar 

  2109. Kell DB: Metabolomics, modelling and machine learning in systems biology: towards an understanding of the languages of cells. The 2005 Theodor Bücher lecture. FEBS J. 2006, 273: 873-894.

    CAS  PubMed  Google Scholar 

  2110. Kell DB: Systems biology, metabolic modelling and metabolomics in drug discovery and development. Drug Disc Today. 2006, 11: 1085-1092.

    CAS  Google Scholar 

  2111. Kell DB: Metabolomic biomarkers: search, discovery and validation. Exp Rev Mol Diagnost. 2007, 7: 329-333.

    CAS  Google Scholar 

  2112. Greef van der J, Martin S, Juhasz P, Adourian A, Plasterer T, Verheij ER, McBurney RN: The art and practice of systems biology in medicine: mapping patterns of relationships. J Proteome Res. 2007, 6: 1540-59.

    PubMed  Google Scholar 

  2113. Kacser H, Burns JA: The control of flux. Rate Control of Biological Processes. Symposium of the Society for Experimental Biology. Edited by: Davies DD. 1973, Cambridge University Press, Cambridge, 27: 65-104.

    Google Scholar 

  2114. Heinrich R, Rapoport TA: A linear steady-state treatment of enzymatic chains. General properties, control and effector strength. Eur J Biochem. 1974, 42: 89-95.

    CAS  PubMed  Google Scholar 

  2115. Kell DB, Westerhoff HV: Metabolic control theory: its role in microbiology and biotechnology. FEMS Microbiol Rev. 1986, 39: 305-320.

    CAS  Google Scholar 

  2116. Westerhoff HV, van Dam K: Thermodynamics and control of biological free energy transduction. 1987, Elsevier, Amsterdam

    Google Scholar 

  2117. Westerhoff HV, Kell DB: Matrix method for determining the steps most rate-limiting to metabolic fluxes in biotechnological processes. Biotechnol Bioeng. 1987, 30: 101-107.

    CAS  PubMed  Google Scholar 

  2118. Fell DA: Understanding the control of metabolism. 1996, Portland Press, London

    Google Scholar 

  2119. Heinrich R, Schuster S: The regulation of cellular systems. 1996, Chapman & Hall, New York

    Google Scholar 

  2120. Murphy MP, Partridge L: Toward a control theory analysis of aging. Annu Rev Biochem. 2008, 77: 777-98.

    CAS  PubMed  PubMed Central  Google Scholar 

  2121. Morris JG: The physiology of obligate anaerobiosis. Adv Microb Physiol. 1975, 12: 169-246.

    CAS  Google Scholar 

  2122. Kirkwood TB, Boys RJ, Gillespie CS, Proctor CJ, Shanley DP, Wilkinson DJ: Towards an e-biology of ageing: integrating theory and data. Nat Rev Mol Cell Biol. 2003, 4: 243-9.

    CAS  PubMed  Google Scholar 

  2123. Hofmeyr JH, Westerhoff HV: Building the cellular puzzle: control in multi-level reaction networks. J Theor Biol. 2001, 208: 261-85.

    CAS  PubMed  Google Scholar 

  2124. Saltelli A, Tarantola S, Campolongo F, Ratto M: Sensitivity analysis in practice: a guide to assessing scientific models. 2004, Wiley, New York

    Google Scholar 

  2125. Sauro HM, Kholodenko BN: Quantitative analysis of signaling networks. Prog Biophys Mol Biol. 2004, 86: 5-43.

    CAS  PubMed  Google Scholar 

  2126. Lüdtke N, Panzeri S, Brown M, Broomhead DS, Knowles J, Montemurro MA, Kell DB: Information-theoretic Sensitivity Analysis: a general method for credit assignment in complex networks. J Roy Soc Interface. 2008, 5: 223-235.

    Google Scholar 

  2127. Saltelli A, Ratto M, Andres T, Campolongo F, Cariboni J, Gatelli D, Saisana M, Tarantola S: Global sensitivity analysis: the primer. 2008, WileyBlackwell, New York

    Google Scholar 

  2128. Smallbone K, Simeonidis E, Broomhead DS, Kell DB: Something from nothing: bridging the gap between constraint-based and kinetic modelling. FEBS J. 2007, 274: 5576-5585.

    CAS  PubMed  Google Scholar 

  2129. Mendes P, Kell DB: Non-linear optimization of biochemical pathways: applications to metabolic engineering and parameter estimation. Bioinformatics. 1998, 14: 869-883.

    CAS  PubMed  Google Scholar 

  2130. Moles CG, Mendes P, Banga JR: Parameter estimation in biochemical pathways: a comparison of global optimization methods. Genome Res. 2003, 13: 2467-74.

    CAS  PubMed  PubMed Central  Google Scholar 

  2131. Rodriguez-Fernandez M, Mendes P, Banga JR: A hybrid approach for efficient and robust parameter estimation in biochemical pathways. Biosystems. 2006, 83: 248-65.

    CAS  PubMed  Google Scholar 

  2132. Wilkinson SJ, Benson N, Kell DB: Proximate parameter tuning for biochemical networks with uncertain kinetic parameters. Mol Biosyst. 2008, 4: 74-97.

    CAS  PubMed  Google Scholar 

  2133. Klipp E, Herwig R, Kowald A, Wierling C, Lehrach H: Systems biology in practice: concepts, implementation and clinical application. 2005, Wiley/VCH, Berlin

    Google Scholar 

  2134. Alon U: An introduction to systems biology: design principles of biological circuits. 2006, Chapman and Hall/CRC, London

    Google Scholar 

  2135. Palsson BØ: Systems biology: properties of reconstructed networks. 2006, Cambridge University Press, Cambridge

    Google Scholar 

  2136. Doyle FJ, Stelling J: Systems interface biology. J R Soc Interface. 2006, 3: 603-16.

    PubMed  PubMed Central  Google Scholar 

  2137. Noble D: The music of life: biology beyond genes. 2006, Oxford University Press, Oxford

    Google Scholar 

  2138. Hopkins AL: Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol. 2008, 4: 682-690.

    CAS  PubMed  Google Scholar 

  2139. Rand DA: Mapping global sensitivity of cellular network dynamics: sensitivity heat maps and a global summation law. J R Soc Interface. 2008, 5 (Suppl 1): S59-69.

    CAS  PubMed  PubMed Central  Google Scholar 

  2140. Tagkopoulos I, Liu YC, Tavazoie S: Predictive behavior within microbial genetic networks. Science. 2008, 320: 1313-7.

    CAS  PubMed  PubMed Central  Google Scholar 

  2141. Muscoli C, Cuzzocrea S, Riley DP, Zweier JL, Thiemermann C, Wang ZQ, Salvemini D: On the selectivity of superoxide dismutase mimetics and its importance in pharmacological studies. British Journal of Pharmacology. 2003, 140: 445-460.

    CAS  PubMed  PubMed Central  Google Scholar 

  2142. Fell DA: Increasing the flux in metabolic pathways: A metabolic control analysis perspective. Biotechnol Bioeng. 1998, 58: 121-124.

    CAS  PubMed  Google Scholar 

  2143. Sai Y, Tsuji A: Transporter-mediated drug delivery: recent progress and experimental approaches. Drug Discov Today. 2004, 9: 712-20.

    CAS  PubMed  Google Scholar 

  2144. Schulze MB, Manson JE, Willett WC, Hu FB: Processed meat intake and incidence of Type 2 diabetes in younger and middle-aged women. Diabetologia. 2003, 46: 1465-73.

    CAS  PubMed  Google Scholar 

  2145. Miyake T, Kung CK, Goldwasser E: Purification of human erythropoietin. J Biol Chem. 1977, 252: 5558-64.

    CAS  PubMed  Google Scholar 

  2146. Jacobs K, Shoemaker C, Rudersdorf R, Neill SD, Kaufman RJ, Mufson A, Seehra J, Jones SS, Hewick R, Fritsch EF, et al: Isolation and characterization of genomic and cDNA clones of human erythropoietin. Nature. 1985, 313: 806-10.

    CAS  PubMed  Google Scholar 

  2147. Lin FK, Suggs S, Lin CH, Browne JK, Smalling R, Egrie JC, Chen KK, Fox GM, Martin F, Stabinsky Z, et al: Cloning and expression of the human erythropoietin gene. Proc Natl Acad Sci USA. 1985, 82: 7580-4.

    CAS  PubMed  PubMed Central  Google Scholar 

  2148. Fenjves ES, Ochoa MS, Cabrera O, Mendez AJ, Kenyon NS, Inverardi L, Ricordi C: Human, nonhuman primate, and rat pancreatic islets express erythropoietin receptors. Transplantation. 2003, 75: 1356-60.

    CAS  PubMed  Google Scholar 

  2149. Ghezzi P, Brines M: Erythropoietin as an antiapoptotic, tissue-protective cytokine. Cell Death Differ. 2004, 11 (Suppl 1): S37-44.

    CAS  PubMed  Google Scholar 

  2150. Brines M, Cerami A: Emerging biological roles for erythropoietin in the nervous system. Nat Rev Neurosci. 2005, 6: 484-94.

    CAS  PubMed  Google Scholar 

  2151. Chong ZZ, Li F, Maiese K: Oxidative stress in the brain: novel cellular targets that govern survival during neurodegenerative disease. Prog Neurobiol. 2005, 75: 207-46.

    CAS  PubMed  Google Scholar 

  2152. Brines M, Cerami A: Discovering erythropoietin's extra-hematopoietic functions: biology and clinical promise. Kidney Int. 2006, 70: 246-50.

    CAS  PubMed  Google Scholar 

  2153. Coleman TR, Westenfelder C, Togel FE, Yang Y, Hu Z, Swenson L, Leuvenink HG, Ploeg RJ, d'Uscio LV, Katusic ZS, Ghezzi P, Zanetti A, Kaushansky K, Fox NE, Cerami A, Brines M: Cytoprotective doses of erythropoietin or carbamylated erythropoietin have markedly different procoagulant and vasoactive activities. Proc Natl Acad Sci USA. 2006, 103: 5965-70.

    CAS  PubMed  PubMed Central  Google Scholar 

  2154. Ghezzi P, Mengozzi M: Activities of erythropoietin on tumors: an immunological perspective. Eur J Immunol. 2007, 37: 1427-30.

    CAS  PubMed  Google Scholar 

  2155. Tönges L, Schlachetzki JCM, Weishaupt JH, Bähr M: Hematopoietic cytokines – on the verge of conquering neurology. Current Molecular Medicine. 2007, 7: 157-170.

    PubMed  Google Scholar 

  2156. Cai Z, Manalo DJ, Wei G, Rodriguez ER, Fox-Talbot K, Lu H, Zweier JL, Semenza GL: Hearts from rodents exposed to intermittent hypoxia or erythropoietin are protected against ischemia-reperfusion injury. Circulation. 2003, 108: 79-85.

    CAS  PubMed  Google Scholar 

  2157. Tramontano AF, Muniyappa R, Black AD, Blendea MC, Cohen I, Deng L, Sowers JR, Cutaia MV, El-Sherif N: Erythropoietin protects cardiac myocytes from hypoxia-induced apoptosis through an Akt-dependent pathway. Biochem Biophys Res Commun. 2003, 308: 990-4.

    CAS  PubMed  Google Scholar 

  2158. Bogoyevitch MA: An update on the cardiac effects of erythropoietin cardioprotection by erythropoietin and the lessons learnt from studies in neuroprotection. Cardiovasc Res. 2004, 63: 208-16.

    CAS  PubMed  Google Scholar 

  2159. Cai Z, Semenza GL: Phosphatidylinositol-3-kinase signaling is required for erythropoietin-mediated acute protection against myocardial ischemia/reperfusion injury. Circulation. 2004, 109: 2050-3.

    CAS  PubMed  Google Scholar 

  2160. Wright GL, Hanlon P, Amin K, Steenbergen C, Murphy E, Arcasoy MO: Erythropoietin receptor expression in adult rat cardiomyocytes is associated with an acute cardioprotective effect for recombinant erythropoietin during ischemia-reperfusion injury. FASEB J. 2004, 18: 1031-3.

    CAS  PubMed  Google Scholar 

  2161. Fiordaliso F, Chimenti S, Staszewsky L, Bai A, Carlo E, Cuccovillo I, Doni M, Mengozzi M, Tonelli R, Ghezzi P, Coleman T, Brines M, Cerami A, Latini R: A nonerythropoietic derivative of erythropoietin protects the myocardium from ischemia-reperfusion injury. Proc Natl Acad Sci USA. 2005, 102: 2046-51.

    CAS  PubMed  PubMed Central  Google Scholar 

  2162. Moon C, Krawczyk M, Paik D, Lakatta EG, Talan MI: Cardioprotection by recombinant human erythropoietin following acute experimental myocardial infarction: dose response and therapeutic window. Cardiovasc Drugs Ther. 2005, 19: 243-50.

    CAS  PubMed  Google Scholar 

  2163. Rui T, Feng Q, Lei M, Peng T, Zhang J, Xu M, Abel ED, Xenocostas A, Kvietys PR: Erythropoietin prevents the acute myocardial inflammatory response induced by ischemia/reperfusion via induction of AP-1. Cardiovasc Res. 2005, 65: 719-27.

    CAS  PubMed  Google Scholar 

  2164. Lee ST, Chu K, Sinn DI, Jung KH, Kim EH, Kim SJ, Kim JM, Ko SY, Kim M, Roh JK: Erythropoietin reduces perihematomal inflammation and cell death with eNOS and STAT3 activations in experimental intracerebral hemorrhage. J Neurochem. 2006, 96: 1728-39.

    CAS  PubMed  Google Scholar 

  2165. Li Y, Takemura G, Okada H, Miyata S, Maruyama R, Li L, Higuchi M, Minatoguchi S, Fujiwara T, Fujiwara H: Reduction of inflammatory cytokine expression and oxidative damage by erythropoietin in chronic heart failure. Cardiovasc Res. 2006, 71: 684-94.

    CAS  PubMed  Google Scholar 

  2166. Li L, Takemura G, Li Y, Miyata S, Esaki M, Okada H, Kanamori H, Khai NC, Maruyama R, Ogino A, Minatoguchi S, Fujiwara T, Fujiwara H: Preventive effect of erythropoietin on cardiac dysfunction in doxorubicin-induced cardiomyopathy. Circulation. 2006, 113: 535-43.

    CAS  PubMed  Google Scholar 

  2167. Miki T, Miura T, Yano T, Takahashi A, Sakamoto J, Tanno M, Kobayashi H, Ikeda Y, Nishihara M, Naitoh K, Ohori K, Shimamoto K: Alteration in erythropoietin-induced cardioprotective signaling by postinfarct ventricular remodeling. J Pharmacol Exp Ther. 2006, 317: 68-75.

    CAS  PubMed  Google Scholar 

  2168. Moon C, Krawczyk M, Paik D, Coleman T, Brines M, Juhaszova M, Sollott SJ, Lakatta EG, Talan MI: Erythropoietin, modified to not stimulate red blood cell production, retains its cardioprotective properties. J Pharmacol Exp Ther. 2006, 316: 999-1005.

    CAS  PubMed  Google Scholar 

  2169. Nishihara M, Miura T, Miki T, Sakamoto J, Tanno M, Kobayashi H, Ikeda Y, Ohori K, Takahashi A, Shimamoto K: Erythropoietin affords additional cardioprotection to preconditioned hearts by enhanced phosphorylation of glycogen synthase kinase-3 beta. Am J Physiol Heart Circ Physiol. 2006, 291: H748-55.

    CAS  PubMed  Google Scholar 

  2170. Binbrek AS, Mittal B, Rao KN, Sobel BE: The potential of erythropoietin for conferring cardioprotection complementing reperfusion. Coron Artery Dis. 2007, 18: 583-5.

    PubMed  Google Scholar 

  2171. Chen X, Chen Y, Bi Y, Fu N, Shan C, Wang S, Aslam S, Wang PW, Xu J: Preventive cardioprotection of erythropoietin against doxorubicin-induced cardiomyopathy. Cardiovasc Drugs Ther. 2007, 21: 367-74.

    CAS  PubMed  Google Scholar 

  2172. Gao E, Boucher M, Chuprun JK, Zhou RH, Eckhart AD, Koch WJ: Darbepoetin alfa, a long-acting erythropoietin analog, offers novel and delayed cardioprotection for the ischemic heart. Am J Physiol Heart Circ Physiol. 2007, 293: H60-8.

    CAS  PubMed  Google Scholar 

  2173. Putten van der K, Braam B, Jie KE, Gaillard CA: Mechanisms of Disease: erythropoietin resistance in patients with both heart and kidney failure. Nat Clin Pract Nephrol. 2008, 4: 47-57.

    PubMed  Google Scholar 

  2174. Furlani D, Klopsch C, Gabel R, Ugurlucan M, Pittermann E, Klee D, Wagner K, Li W, Wang W, Ong LL, Nizze H, Titze U, Lutzow K, Lendlein A, Steinhoff G, Ma N: Intracardiac erythropoietin injection reveals antiinflammatory potential and improved cardiac functions detected by Forced Swim Test. Transplant Proc. 2008, 40: 962-6.

    CAS  PubMed  Google Scholar 

  2175. Robey TE, Saiget MK, Reinecke H, Murry CE: Systems approaches to preventing transplanted cell death in cardiac repair. J Mol Cell Cardiol. 2008, 45: 567-81.

    CAS  PubMed  PubMed Central  Google Scholar 

  2176. Sirén AL, Ehrenreich H: Erythropoietin–a novel concept for neuroprotection. Eur Arch Psychiatry Clin Neurosci. 2001, 251: 179-84.

    PubMed  Google Scholar 

  2177. Buemi M, Cavallaro E, Floccari F, Sturiale A, Aloisi C, Trimarchi M, Grasso G, Corica F, Frisina N: Erythropoietin and the brain: from neurodevelopment to neuroprotection. Clin Sci (Lond). 2002, 103: 275-82.

    CAS  Google Scholar 

  2178. Junk AK, Mammis A, Savitz SI, Singh M, Roth S, Malhotra S, Rosenbaum PS, Cerami A, Brines M, Rosenbaum DM: Erythropoietin administration protects retinal neurons from acute ischemia-reperfusion injury. Proc Natl Acad Sci USA. 2002, 99: 10659-64.

    CAS  PubMed  PubMed Central  Google Scholar 

  2179. Juul S: Erythropoietin in the central nervous system, and its use to prevent hypoxic-ischemic brain damage. Acta Paediatr Suppl. 2002, 91: 36-42.

    CAS  PubMed  Google Scholar 

  2180. Ruscher K, Freyer D, Karsch M, Isaev N, Megow D, Sawitzki B, Priller J, Dirnagl U, Meisel A: Erythropoietin is a paracrine mediator of ischemic tolerance in the brain: evidence from an in vitro model. J Neurosci. 2002, 22: 10291-301.

    CAS  PubMed  Google Scholar 

  2181. Campana WM, Myers RR: Exogenous erythropoietin protects against dorsal root ganglion apoptosis and pain following peripheral nerve injury. Eur J Neurosci. 2003, 18: 1497-506.

    PubMed  Google Scholar 

  2182. Prass K, Scharff A, Ruscher K, Lowl D, Muselmann C, Victorov I, Kapinya K, Dirnagl U, Meisel A: Hypoxia-induced stroke tolerance in the mouse is mediated by erythropoietin. Stroke. 2003, 34: 1981-6.

    CAS  PubMed  Google Scholar 

  2183. Villa P, Bigini P, Mennini T, Agnello D, Laragione T, Cagnotto A, Viviani B, Marinovich M, Cerami A, Coleman TR, Brines M, Ghezzi P: Erythropoietin selectively attenuates cytokine production and inflammation in cerebral ischemia by targeting neuronal apoptosis. J Exp Med. 2003, 198: 971-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  2184. Digicaylioglu M, Garden G, Timberlake S, Fletcher L, Lipton SA: Acute neuroprotective synergy of erythropoietin and insulin-like growth factor I. Proc Natl Acad Sci USA. 2004, 101: 9855-60.

    CAS  PubMed  PubMed Central  Google Scholar 

  2185. Ehrenreich H, Aust C, Krampe H, Jahn H, Jacob S, Herrmann M, Siren AL: Erythropoietin: novel approaches to neuroprotection in human brain disease. Metab Brain Dis. 2004, 19: 195-206.

    CAS  PubMed  Google Scholar 

  2186. Genc S, Koroglu TF, Genc K: Erythropoietin and the nervous system. Brain Res. 2004, 1000: 19-31.

    CAS  PubMed  Google Scholar 

  2187. Keswani SC, Buldanlioglu U, Fischer A, Reed N, Polley M, Liang H, Zhou C, Jack C, Leitz GJ, Hoke A: A novel endogenous erythropoietin mediated pathway prevents axonal degeneration. Ann Neurol. 2004, 56: 815-26.

    CAS  PubMed  Google Scholar 

  2188. Lipton SA: Erythropoietin for neurologic protection and diabetic neuropathy. N Engl J Med. 2004, 350: 2516-7.

    CAS  PubMed  Google Scholar 

  2189. Marti HH: Erythropoietin and the hypoxic brain. J Exp Biol. 2004, 207: 3233-42.

    CAS  PubMed  Google Scholar 

  2190. Prass K, Ruscher K, Karsch M, Isaev N, Megow D, Priller J, Scharff A, Dirnagl U, Meisel A: Desferrioxamine induces delayed tolerance against cerebral ischemia in vivo and in vitro. J Cereb Blood Flow Metab. 2002, 22: 520-5.

    CAS  PubMed  Google Scholar 

  2191. Diem R, Sättler MB, Merkler D, Demmer I, Maier K, Stadelmann C, Ehrenreich H, Bähr M: Combined therapy with methylprednisolone and erythropoietin in a model of multiple sclerosis. Brain. 2005, 128: 375-85.

    PubMed  Google Scholar 

  2192. Yatsiv I, Grigoriadis N, Simeonidou C, Stahel PF, Schmidt OI, Alexandrovitch AG, Tsenter J, Shohami E: Erythropoietin is neuroprotective, improves functional recovery, and reduces neuronal apoptosis and inflammation in a rodent model of experimental closed head injury. FASEB J. 2005, 19: 1701-3.

    CAS  PubMed  Google Scholar 

  2193. Savino C, Pedotti R, Baggi F, Ubiali F, Gallo B, Nava S, Bigini P, Barbera S, Fumagalli E, Mennini T, Vezzani A, Rizzi M, Coleman T, Cerami A, Brines M, Ghezzi P, Bianchi R: Delayed administration of erythropoietin and its non-erythropoietic derivatives ameliorates chronic murine autoimmune encephalomyelitis. J Neuroimmunol. 2006, 172: 27-37.

    CAS  PubMed  Google Scholar 

  2194. Sirén AL, Radyushkin K, Boretius S, Kammer D, Riechers CC, Natt O, Sargin D, Watanabe T, Sperling S, Michaelis T, Price J, Meyer B, Frahm J, Ehrenreich H: Global brain atrophy after unilateral parietal lesion and its prevention by erythropoietin. Brain. 2006, 129: 480-9.

    PubMed  Google Scholar 

  2195. Chen G, Shi JX, Hang CH, Xie W, Liu J, Liu X: Inhibitory effect on cerebral inflammatory agents that accompany traumatic brain injury in a rat model: a potential neuroprotective mechanism of recombinant human erythropoietin (rhEPO). Neurosci Lett. 2007, 425: 177-82.

    CAS  PubMed  Google Scholar 

  2196. Cherian L, Goodman JC, Robertson C: Neuroprotection with erythropoietin administration following controlled cortical impact injury in rats. J Pharmacol Exp Ther. 2007, 322: 789-94.

    CAS  PubMed  Google Scholar 

  2197. Ehrenreich H, Fischer B, Norra C, Schellenberger F, Stender N, Stiefel M, Siren AL, Paulus W, Nave KA, Gold R, Bartels C: Exploring recombinant human erythropoietin in chronic progressive multiple sclerosis. Brain. 2007, 130: 2577-88.

    PubMed  Google Scholar 

  2198. Esiri MM: The interplay between inflammation and neurodegeneration in CNS disease. J Neuroimmunol. 2007, 184: 4-16.

    CAS  PubMed  Google Scholar 

  2199. Juul S, Felderhoff-Mueser U: Epo and other hematopoietic factors. Semin Fetal Neonatal Med. 2007, 12: 250-8.

    PubMed  PubMed Central  Google Scholar 

  2200. Lykissas MG, Korompilias AV, Vekris MD, Mitsionis GI, Sakellariou E, Beris AE: The role of erythropoietin in central and peripheral nerve injury. Clin Neurol Neurosurg. 2007, 109: 639-44.

    PubMed  Google Scholar 

  2201. Noguchi CT, Asavaritikrai P, Teng R, Jia Y: Role of erythropoietin in the brain. Crit Rev Oncol Hematol. 2007, 64: 159-71.

    PubMed  PubMed Central  Google Scholar 

  2202. Wang Y, Zhang ZG, Rhodes K, Renzi M, Zhang RL, Kapke A, Lu M, Pool C, Heavner G, Chopp M: Post-ischemic treatment with erythropoietin or carbamylated erythropoietin reduces infarction and improves neurological outcome in a rat model of focal cerebral ischemia. Br J Pharmacol. 2007, 151: 1377-84.

    CAS  PubMed  PubMed Central  Google Scholar 

  2203. Wu Y, Shang Y, Sun SG, Liu RG, Yang WQ: Protective effect of erythropoietin against 1-methyl-4-phenylpyridinium-induced neurodegenaration in PC12 cells. Neurosci Bull. 2007, 23: 156-64.

    CAS  PubMed  Google Scholar 

  2204. Wiese L, Hempel C, Penkowa M, Kirkby N, Kurtzhals JA: Recombinant human erythropoietin increases survival and reduces neuronal apoptosis in a murine model of cerebral malaria. Malar J. 2008, 7: 3.

    PubMed  PubMed Central  Google Scholar 

  2205. Bianchi R, Buyukakilli B, Brines M, Savino C, Cavaletti G, Oggioni N, Lauria G, Borgna M, Lombardi R, Cimen B, Comelekoglu U, Kanik A, Tataroglu C, Cerami A, Ghezzi P: Erythropoietin both protects from and reverses experimental diabetic neuropathy. Proc Natl Acad Sci USA. 2004, 101: 823-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  2206. İnal M, Kanbak G, Şen S, Akyüz F, Sunal E: Antioxidant status and lipid peroxidation in hemodialysis patients undergoing erythropoietin and erythropoietin-vitamin E combined therapy. Free Radic Res. 1999, 31: 211-6.

    PubMed  Google Scholar 

  2207. Yang CW, Li C, Jung JY, Shin SJ, Choi BS, Lim SW, Sun BK, Kim YS, Kim J, Chang YS, Bang BK: Preconditioning with erythropoietin protects against subsequent ischemia-reperfusion injury in rat kidney. FASEB J. 2003, 17: 1754-5.

    CAS  PubMed  Google Scholar 

  2208. Patel NS, Sharples EJ, Cuzzocrea S, Chatterjee PK, Britti D, Yaqoob MM, Thiemermann C: Pretreatment with EPO reduces the injury and dysfunction caused by ischemia/reperfusion in the mouse kidney in vivo. Kidney Int. 2004, 66: 983-9.

    CAS  PubMed  Google Scholar 

  2209. Sharples EJ, Patel N, Brown P, Stewart K, Mota-Philipe H, Sheaff M, Kieswich J, Allen D, Harwood S, Raftery M, Thiemermann C, Yaqoob MM: Erythropoietin protects the kidney against the injury and dysfunction caused by ischemia-reperfusion. J Am Soc Nephrol. 2004, 15: 2115-24.

    CAS  PubMed  Google Scholar 

  2210. Ates E, Yalcin AU, Yilmaz S, Koken T, Tokyol C: Protective effect of erythropoietin on renal ischemia and reperfusion injury. ANZ J Surg. 2005, 75: 1100-5.

    PubMed  Google Scholar 

  2211. Fliser D, Bahlmann FH, Haller H: EPO: renoprotection beyond anemia correction. Pediatr Nephrol. 2006, 21: 1785-9.

    PubMed  Google Scholar 

  2212. Jie KE, Verhaar MC, Cramer MJ, Putten van der K, Gaillard CA, Doevendans PA, Koomans HA, Joles JA, Braam B: Erythropoietin and the cardiorenal syndrome: cellular mechanisms on the cardiorenal connectors. Am J Physiol Renal Physiol. 2006, 291: F932-44.

    CAS  PubMed  Google Scholar 

  2213. Guneli E, Cavdar Z, Islekel H, Sarioglu S, Erbayraktar S, Kiray M, Sokmen S, Yilmaz O, Gokmen N: Erythropoietin protects the intestine against ischemia/reperfusion injury in rats. Mol Med. 2007, 13: 509-17.

    CAS  PubMed  PubMed Central  Google Scholar 

  2214. Mitra A, Bansal S, Wang W, Falk S, Zolty E, Schrier RW: Erythropoietin ameliorates renal dysfunction during endotoxaemia. Nephrol Dial Transplant. 2007, 22: 2349-53.

    CAS  PubMed  Google Scholar 

  2215. Ashikawa K, Majumdar S, Banerjee S, Bharti AC, Shishodia S, Aggarwal BB: Piceatannol inhibits TNF-induced NF-kappaB activation and NF-kappaB-mediated gene expression through suppression of IkappaBalpha kinase and p65 phosphorylation. J Immunol. 2002, 169: 6490-7.

    CAS  PubMed  Google Scholar 

  2216. Paur I, Austenaa LM, Blomhoff R: Extracts of dietary plants are efficient modulators of nuclear factor kappa B. Food Chem Toxicol. 2008, 46: 1288-97.

    CAS  PubMed  Google Scholar 

  2217. Wojchowski DM, Gregory RC, Miller CP, Pandit AK, Pircher TJ: Signal transduction in the erythropoietin receptor system. Exp Cell Res. 1999, 253: 143-56.

    CAS  PubMed  Google Scholar 

  2218. Coleman T, Brines M: Science review: recombinant human erythropoietin in critical illness: a role beyond anemia?. Crit Care. 2004, 8: 337-41.

    PubMed  PubMed Central  Google Scholar 

  2219. Hanlon PR, Fu P, Wright GL, Steenbergen C, Arcasoy MO, Murphy E: Mechanisms of erythropoietin-mediated cardioprotection during ischemia-reperfusion injury: role of protein kinase C and phosphatidylinositol 3-kinase signaling. FASEB J. 2005, 19: 1323-5.

    CAS  PubMed  Google Scholar 

  2220. Maiese K, Li F, Chong ZZ: Erythropoietin in the brain: can the promise to protect be fulfilled?. Trends Pharmacol Sci. 2004, 25: 577-83.

    CAS  PubMed  Google Scholar 

  2221. Chong ZZ, Li F, Maiese K: Erythropoietin requires NF-kappaB and its nuclear translocation to prevent early and late apoptotic neuronal injury during beta-amyloid toxicity. Curr Neurovasc Res. 2005, 2: 387-99.

    CAS  PubMed  PubMed Central  Google Scholar 

  2222. Maiese K, Li F, Chong ZZ: New avenues of exploration for erythropoietin. JAMA. 2005, 293: 90-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  2223. Xu B, Dong GH, Liu H, Wang YQ, Wu HW, Jing H: Recombinant human erythropoietin pretreatment attenuates myocardial infarct size: a possible mechanism involves heat shock Protein 70 and attenuation of nuclear factor-kappaB. Ann Clin Lab Sci. 2005, 35: 161-8.

    CAS  PubMed  Google Scholar 

  2224. Aerbajinai W, Zhu J, Gao Z, Chin K, Rodgers GP: Thalidomide induces gamma-globin gene expression through increased reactive oxygen species-mediated p38 MAPK signaling and histone H4 acetylation in adult erythropoiesis. Blood. 2007, 110: 2864-71.

    CAS  PubMed  PubMed Central  Google Scholar 

  2225. Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X, Choi AM: Mechanisms of cell death in oxidative stress. Antioxid Redox Signal. 2007, 9: 49-89.

    CAS  PubMed  Google Scholar 

  2226. Wu Y, Shang Y, Sun S, Liang H, Liu R: Erythropoietin prevents PC12 cells from 1-methyl-4-phenylpyridinium ion-induced apoptosis via the Akt/GSK-3beta/caspase-3 mediated signaling pathway. Apoptosis. 2007, 12: 1365-75.

    CAS  PubMed  Google Scholar 

  2227. Cavdar C, Camsari T, Semin I, Gonenc S, Acikgoz O: Lipid peroxidation and antioxidant activity in chronic haemodialysis patients treated with recombinant human erythropoietin. Scand J Urol Nephrol. 1997, 31: 371-5.

    CAS  PubMed  Google Scholar 

  2228. Katavetin P, Inagi R, Miyata T, Shao J, Sassa R, Adler S, Eto N, Kato H, Fujita T, Nangaku M: Erythropoietin induces heme oxygenase-1 expression and attenuates oxidative stress. Biochem Biophys Res Commun. 2007, 359: 928-34.

    CAS  PubMed  Google Scholar 

  2229. Katavetin P, Tungsanga K, Eiam-Ong S, Nangaku M: Antioxidative effects of erythropoietin. Kidney Int Suppl. 2007, S10-5.

    Google Scholar 

  2230. Chattopadhyay A, Das Choudhury TD, Basu MK, Datta AG: Effect of Cu2+-ascorbic acid on lipid peroxidation, Mg2+-ATPase activity and spectrin of RBC membrane and reversal by erythropoietin. Mol Cell Biochem. 1992, 118: 23-30.

    CAS  PubMed  Google Scholar 

  2231. Chattopadhyay A, Choudhury TD, Bandyopadhyay D, Datta AG: Protective effect of erythropoietin on the oxidative damage of erythrocyte membrane by hydroxyl radical. Biochem Pharmacol. 2000, 59: 419-25.

    CAS  PubMed  Google Scholar 

  2232. Akisu M, Tuzun S, Arslanoglu S, Yalaz M, Kultursay N: Effect of recombinant human erythropoietin administration on lipid peroxidation and antioxidant enzyme(s) activities in preterm infants. Acta Med Okayama. 2001, 55: 357-62.

    CAS  PubMed  Google Scholar 

  2233. Hirayama A, Nagase S, Gotoh M, Ueda A, Ishizu T, Yoh K, Aoyagi K, Terao J, Koyama A: Reduced serum hydroxyl radical scavenging activity in erythropoietin therapy resistant renal anemia. Free Radic Res. 2002, 36: 1155-61.

    CAS  PubMed  Google Scholar 

  2234. Yankowitz J, Piraino B, Laifer SA, Frassetto L, Gavin L, Kitzmiller JL, Crombleholme W: Erythropoietin in pregnancies complicated by severe anemia of renal failure. Obstet Gynecol. 1992, 80: 485-8.

    CAS  PubMed  Google Scholar 

  2235. Braga J, Marques R, Branco A, Goncalves J, Lobato L, Pimentel JP, Flores MM, Goncalves E, Jorge CS: Maternal and perinatal implications of the use of human recombinant erythropoietin. Acta Obstet Gynecol Scand. 1996, 75: 449-3.

    CAS  PubMed  Google Scholar 

  2236. Jungers P, Chauveau D: Pregnancy in renal disease. Kidney Int. 1997, 52: 871-85.

    CAS  PubMed  Google Scholar 

  2237. Vora M, Gruslin A: Erythropoietin in obstetrics. Obstet Gynecol Surv. 1998, 53: 500-8.

    CAS  PubMed  Google Scholar 

  2238. Breymann C, Visca E, Huch R, Huch A: Efficacy and safety of intravenously administered iron sucrose with and without adjuvant recombinant human erythropoietin for the treatment of resistant iron-deficiency anemia during pregnancy. Am J Obstet Gynecol. 2001, 184: 662-7.

    CAS  PubMed  Google Scholar 

  2239. Sifakis S, Angelakis E, Vardaki E, Koumantaki Y, Matalliotakis I, Koumantakis E: Erythropoietin in the treatment of iron deficiency anemia during pregnancy. Gynecol Obstet Invest. 2001, 51: 150-6.

    CAS  PubMed  Google Scholar 

  2240. Sesti F, Ticconi C, Bonifacio S, Piccione E: Preoperative administration of recombinant human erythropoietin in patients undergoing gynecologic surgery. Gynecol Obstet Invest. 2002, 54: 1-5.

    CAS  PubMed  Google Scholar 

  2241. Donato E, Guinot M, Vilar C, Garcia R, Canigral G: rHuEPO in the management of pregnancy complicated by anti-Dib. Transfusion. 2003, 43: 681-2.

    PubMed  Google Scholar 

  2242. Kaupke CJ, Vaziri ND, Powers DR, Gonzales E: Erythropoietin in preeclampsia. Obstet Gynecol. 1991, 78: 795-9.

    CAS  PubMed  Google Scholar 

  2243. Buescher U, Hertwig K, Wolf C, Dudenhausen JW: Erythropoietin in amniotic fluid as a marker of chronic fetal hypoxia. Int J Gynaecol Obstet. 1998, 60: 257-63.

    CAS  PubMed  Google Scholar 

  2244. Goldstein JD, Garry DJ, Maulik D: Obstetric conditions and erythropoietin levels. Am J Obstet Gynecol. 2000, 182: 1055-7.

    CAS  PubMed  Google Scholar 

  2245. Troeger C, Holzgreve W, Ladewig A, Zhong XY, Hahn S: Examination of maternal plasma erythropoietin and activin A concentrations with regard to circulatory erythroblast levels in normal and preeclamptic pregnancies. Fetal Diagn Ther. 2006, 21: 156-60.

    PubMed  Google Scholar 

  2246. Semenza GL: HIF-1 and human disease: one highly involved factor. Genes Dev. 2000, 14: 1983-91.

    CAS  PubMed  Google Scholar 

  2247. Semenza GL: Hypoxia-inducible factor 1: oxygen homeostasis and disease pathophysiology. Trends Mol Med. 2001, 7: 345-50.

    CAS  PubMed  Google Scholar 

  2248. Wenger RH: Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression. FASEB J. 2002, 16: 1151-62.

    CAS  PubMed  Google Scholar 

  2249. Pugh CW, Ratcliffe PJ: Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med. 2003, 9: 677-84.

    CAS  PubMed  Google Scholar 

  2250. Haase VH: Hypoxia-inducible factors in the kidney. Amer J Physiol. 2006, 291: F271-F281.

    CAS  Google Scholar 

  2251. Yoon D, Pastore YD, Divoky V, Liu E, Mlodnicka AE, Rainey K, Ponka P, Semenza GL, Schumacher A, Prchal JT: Hypoxia-inducible factor-1 deficiency results in dysregulated erythropoiesis signaling and iron homeostasis in mouse development. J Biol Chem. 2006, 281: 25703-11.

    CAS  PubMed  Google Scholar 

  2252. Zhang B, Tanaka J, Yang L, Yang L, Sakanaka M, Hata R, Maeda N, Mitsuda N: Protective effect of vitamin E against focal brain ischemia and neuronal death through induction of target genes of hypoxia-inducible factor-1. Neuroscience. 2004, 126: 433-440.

    CAS  PubMed  Google Scholar 

  2253. Rius J, Guma M, Schachtrup C, Akassoglou K, Zinkernagel AS, Nizet V, Johnson RS, Haddad GG, Karin M: NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature. 2008

    Google Scholar 

  2254. Peyssonnaux C, Zinkernagel AS, Schuepbach RA, Rankin E, Vaulont S, Haase VH, Nizet V, Johnson RS: Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs). J Clin Invest. 2007, 117: 1926-32.

    CAS  PubMed  PubMed Central  Google Scholar 

  2255. Peyssonnaux C, Cejudo-Martin P, Doedens A, Zinkernagel AS, Johnson RS, Nizet V: Cutting edge: Essential role of hypoxia inducible factor-1alpha in development of lipopolysaccharide-induced sepsis. J Immunol. 2007, 178: 7516-9.

    CAS  PubMed  Google Scholar 

  2256. Peyssonnaux C, Nizet V, Johnson RS: Role of the hypoxia inducible factors HIF in iron metabolism. Cell Cycle. 2008, 7: 28-32.

    CAS  PubMed  Google Scholar 

  2257. Hamrick SE, McQuillen PS, Jiang X, Mu D, Madan A, Ferriero DM: A role for hypoxia-inducible factor-1alpha in desferoxamine neuroprotection. Neurosci Lett. 2005, 379: 96-100.

    CAS  PubMed  Google Scholar 

  2258. Caniggia I, Winter J, Lye SJ, Post M: Oxygen and placental development during the first trimester: implications for the pathophysiology of pre-eclampsia. Placenta. 2000, 21 (Suppl A): S25-30.

    PubMed  Google Scholar 

  2259. Qutub AA, Popel AS: A computational model of intracellular oxygen sensing by hypoxia-inducible factor HIF1 alpha. J Cell Sci. 2006, 119: 3467-80.

    CAS  PubMed  PubMed Central  Google Scholar 

  2260. Qutub AA, Popel AS: Three autocrine feedback loops determine HIF1 alpha expression in chronic hypoxia. Biochim Biophys Acta. 2007, 1773: 1511-25.

    CAS  PubMed  PubMed Central  Google Scholar 

  2261. von Dassow G, Meir E, Munro EM, Odell GM: The segment polarity network is a robust development module. Nature. 2000, 406: 188-192.

    CAS  PubMed  Google Scholar 

  2262. Brandman O, Ferrell JE, Li R, Meyer T: Interlinked fast and slow positive feedback loops drive reliable cell decisions. Science. 2005, 310: 496-8.

    CAS  PubMed  PubMed Central  Google Scholar 

  2263. Kell DB, Knowles JD: The role of modeling in systems biology. System modeling in cellular biology: from concepts to nuts and bolts. Edited by: Szallasi Z, Stelling J, Periwal V. 2006, MIT Press, Cambridge, 3-18.

    Google Scholar 

  2264. Wei YH, Lee HC: Oxidative stress, mitochondrial DNA mutation, and impairment of antioxidant enzymes in aging. Exp Biol Med (Maywood). 2002, 227: 671-82.

    CAS  Google Scholar 

  2265. McCord JM: Iron, free radicals, and oxidative injury. Semin Hematol. 1998, 35: 5-12.

    CAS  PubMed  Google Scholar 

  2266. Barkai N, Leibler S: Robustness in simple biochemical networks. Nature. 1997, 387: 913-7.

    CAS  PubMed  Google Scholar 

  2267. Bornholdt S, Sneppen K: Robustness as an evolutionary principle. Proc. R. Soc. B-Biological Sciences. 2000, 267: 2281-2286.

    CAS  Google Scholar 

  2268. Kohn MC, Tohmaz AS, Giroux KJ, Blumenthal GM, Feezor MD, Millington DS: Robustness of MetaNet graph models: Predicting control of urea production in humans. Biosystems. 2002, 65: 61-78.

    CAS  PubMed  Google Scholar 

  2269. Ma L, Iglesias PA: Quantifying robustness of biochemical network models. BMC Bioinformatics. 2002, 3: [http://www.biomedcentral.com/1471-2105/3/38]

    Google Scholar 

  2270. Morohashi M, Winn AE, Borisuk MT, Bolouri H, Doyle J, Kitano H: Robustness as a measure of plausibility in models of biochemical networks. J Theor Biol. 2002, 216: 19-30.

    CAS  PubMed  Google Scholar 

  2271. Aldana M, Cluzel P: A natural class of robust networks. Proc Natl Acad Sci USA. 2003, 100: 8710-4.

    CAS  PubMed  PubMed Central  Google Scholar 

  2272. Ebenhoh O, Heinrich R: Stoichiometric design of metabolic networks: multifunctionality clusters, optimization, weak and strong robustness. Bull Math Biol. 2003, 65: 323-57.

    PubMed  Google Scholar 

  2273. Kitano H, Oda K, Kimura T, Matsuoka Y, Csete M, Doyle J, Muramatsu M: Metabolic syndrome and robustness tradeoffs. Diabetes. 2004, 53 (Suppl 3): S6-S15.

    CAS  PubMed  Google Scholar 

  2274. Kitano H: Biological robustness. Nat Rev Genet. 2004, 5: 826-37.

    CAS  PubMed  Google Scholar 

  2275. Stelling J, Sauer U, Szallasi Z, Doyle FJ, Doyle J: Robustness of cellular functions. Cell. 2004, 118: 675-85.

    CAS  PubMed  Google Scholar 

  2276. Stelling J, Gilles ED, Doyle FJ: Robustness properties of circadian clock architectures. Proc Natl Acad Sci USA. 2004, 101: 13210-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  2277. Chen BS, Wang YC, Wu WS, Li WH: A new measure of the robustness of biochemical networks. Bioinformatics. 2005, 21: 2698-705.

    CAS  PubMed  Google Scholar 

  2278. Wagner A: Robustness evolvability, and neutrality. FEBS Lett. 2005, 579: 1772-8.

    CAS  PubMed  Google Scholar 

  2279. Wagner A: Circuit topology and the evolution of robustness in two-gene circadian oscillators. Proc Natl Acad Sci USA. 2005, 102: 11775-80.

    CAS  PubMed  PubMed Central  Google Scholar 

  2280. Kurata H, El-Samad H, Iwasaki R, Ohtake H, Doyle JC, Grigorova I, Gross CA, Khammash M: Module-based analysis of robustness tradeoffs in the heat shock response system. PLoS Comput Biol. 2006, 2: e59.

    PubMed  PubMed Central  Google Scholar 

  2281. Wagner A: Robustness and evolvability in living systems. 2005, Princeton University Press Princeton, NJ

    Google Scholar 

  2282. Moriya H, Shimizu-Yoshida Y, Kitano H: In vivo robustness analysis of cell division cycle genes in Saccharomyces cerevisiae. PLoS Genet. 2006, 2: e111.

    PubMed  PubMed Central  Google Scholar 

  2283. Ma W, Lai L, Ouyang Q, Tang C: Robustness and modular design of the Drosophila segment polarity network. Mol Syst Biol. 2006, 2: 70.

    PubMed  PubMed Central  Google Scholar 

  2284. Kitano H: A robustness-based approach to systems-oriented drug design. Nat Rev Drug Discov. 2007, 6: 202-210.

    CAS  PubMed  Google Scholar 

  2285. Vizan P, Mazurek S, Cascante M: Robust metabolic adaptation underlying tumor progression. Metabolomics. 2008, 4: 1-12.

    CAS  Google Scholar 

  2286. Jacobsen EW, Cedersund G: Structural robustness of biochemical network models-with application to the oscillatory metabolism of activated neutrophils. IET Syst Biol. 2008, 2: 39-47.

    CAS  PubMed  Google Scholar 

  2287. Tsai TY, Choi YS, Ma W, Pomerening JR, Tang C, Ferrell JE: Robust, tunable biological oscillations from interlinked positive and negative feedback loops. Science. 2008, 321: 126-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  2288. Daniels BC, Chen YJ, Sethna JP, Gutenkunst RN, Myers CR: Sloppiness robustness, and evolvability in systems biology. Curr Opin Biotechnol. 2008, 19: 389-95.

    CAS  PubMed  Google Scholar 

  2289. Martin OC, Wagner A: Multifunctionality and robustness trade-offs in model genetic circuits. Biophys J. 2008, 94: 2927-37.

    CAS  PubMed  PubMed Central  Google Scholar 

  2290. Wagner A: Robustness and evolvability: a paradox resolved. Proc Biol Sci. 2008, 275: 91-100.

    PubMed  Google Scholar 

  2291. Lehár J, Krueger A, Zimmermann G, Borisy A: High-order combination effects and biological robustness. Molecular Systems Biology. 2008, 4.

    Google Scholar 

  2292. Medawar P: Pluto's republic. 1982, Oxford University Press, Oxford

    Google Scholar 

  2293. Gilbert GN, Mulkay M: Opening Pandora's box: a sociological analysis of scientists' discourse. 1984, Cambridge University Press, Cambridge

    Google Scholar 

  2294. Breen AP, Murphy JA: Reactions of oxyl radicals with DNA. Free Radic Biol Med. 1995, 18: 1033-77.

    CAS  PubMed  Google Scholar 

  2295. Burrows CJ, Muller JG: Oxidative nucleobase modifications leading to strand scission. Chem Rev. 1998, 98: 1109-1152.

    CAS  PubMed  Google Scholar 

  2296. Bolton JL, Trush MA, Penning TM, Dryhurst G, Monks TJ: Role of quinones in toxicology. Chem Res Toxicol. 2000, 13: 135-60.

    CAS  PubMed  Google Scholar 

  2297. Cooke MS, Evans MD, Dizdaroglu M, Lunec J: Oxidative DNA damage: mechanisms mutation, and disease. Faseb J. 2003, 17: 1195-214.

    CAS  PubMed  Google Scholar 

  2298. Haggarty SJ, Clemons PA, Schreiber SL: Chemical genomic profiling of biological networks using graph theory and combinations of small molecule perturbations. J Am Chem Soc. 2003, 125: 10543-5.

    CAS  PubMed  Google Scholar 

  2299. Csermely P, Agoston V, Pongor S: The efficiency of multi-target drugs: the network approach might help drug design. Trends Pharmacol Sci. 2005, 26: 178-82.

    CAS  PubMed  Google Scholar 

  2300. Ambesi-Impiombato A, di Bernardo D: Computational biology and drug discovery: From single-target to network drugs. Current Bioinformatics. 2006, 1: 3-13.

    CAS  Google Scholar 

  2301. Yeh P, Tschumi AI, Kishony R: Functional classification of drugs by properties of their pairwise interactions. Nat Genet. 2006, 38: 489-94.

    CAS  PubMed  Google Scholar 

  2302. Lee MS, Johansen L, Zhang Y, Wilson A, Keegan M, Avery W, Elliott P, Borisy AA, Keith CT: The novel combination of chlorpromazine and pentamidine exerts synergistic antiproliferative effects through dual mitotic action. Cancer Res. 2007, 67: 11359-67.

    CAS  PubMed  Google Scholar 

  2303. Abbott A: Pharmaceutical futures: a fiendish puzzle. Nature. 2008, 455: 1164-7.

    CAS  PubMed  Google Scholar 

  2304. Henney A, Superti-Furga G: A network solution. Nature. 2008, 455: 730-1.

    CAS  PubMed  Google Scholar 

  2305. Hoon S, Smith AM, Wallace IM, Suresh S, Miranda M, Fung E, Proctor M, Shokat KM, Zhang C, Davis RW, Giaever G, St Onge RP, Nislow C: An integrated platform of genomic assays reveals small-molecule bioactivities. Nat Chem Biol. 2008, 4: 498-506.

    CAS  PubMed  Google Scholar 

  2306. Kvien TK, Fjeld E, Slatkowsky-Christensen B, Nichols M, Zhang Y, Proven A, Mikkelsen K, Palm O, Borisy AA, Lessem J: Efficacy and safety of a novel synergistic drug candidate, CRx-102, in hand osteoarthritis. Ann Rheum Dis. 2008, 67: 942-8.

    CAS  PubMed  Google Scholar 

  2307. Lehár J, Stockwell BR, Giaever G, Nislow C: Combination chemical genetics. Nat Chem Biol. 2008, 4: 674-81.

    PubMed  PubMed Central  Google Scholar 

  2308. Nelander S, Wang W, Nilsson B, She QB, Pratilas C, Rosen N, Gennemark P, Sander C: Models from experiments: combinatorial drug perturbations of cancer cells. Mol Syst Biol. 2008, 4: 216.

    PubMed  PubMed Central  Google Scholar 

  2309. Westerhoff HV: Systems biology: the grand challenge for European biology. Screening. 2008, 9: 34-37.

    Google Scholar 

  2310. Westerhoff HV, Mosekilde E, Noe CR, Clemensen AM: Integrating systems approaches into pharmaceutical sciences. Eur J Pharm Sci. 2008, 35: 1-4.

    CAS  PubMed  Google Scholar 

  2311. Tong AH, Lesage G, Bader GD, Ding H, Xu H, Xin X, Young J, Berriz GF, Brost RL, Chang M, Chen Y, Cheng X, Chua G, Friesen H, Goldberg DS, Haynes J, Humphries C, He G, Hussein S, Ke L, Krogan N, Li Z, Levinson JN, Lu H, Menard P, Munyana C, Parsons AB, Ryan O, Tonikian R, Roberts T, Sdicu AM, Shapiro J, Sheikh B, Suter B, Wong SL, Zhang LV, Zhu H, Burd CG, Munro S, Sander C, Rine J, Greenblatt J, Peter M, Bretscher A, Bell G, Roth FP, Brown GW, Andrews B, Bussey H, Boone C: Global mapping of the yeast genetic interaction network. Science. 2004, 303: 808-13.

    CAS  PubMed  Google Scholar 

  2312. Parsons AB, Brost RL, Ding H, Li Z, Zhang C, Sheikh B, Brown GW, Kane PM, Hughes TR, Boone C: Integration of chemical-genetic and genetic interaction data links bioactive compounds to cellular target pathways. Nat Biotechnol. 2004, 22: 62-9.

    CAS  PubMed  Google Scholar 

  2313. Parsons AB, Lopez A, Givoni IE, Williams DE, Gray CA, Porter J, Chua G, Sopko R, Brost RL, Ho CH, Wang J, Ketela T, Brenner C, Brill JA, Fernandez GE, Lorenz TC, Payne GS, Ishihara S, Ohya Y, Andrews B, Hughes TR, Frey BJ, Graham TR, Andersen RJ, Boone C: Exploring the mode-of-action of bioactive compounds by chemical-genetic profiling in yeast. Cell. 2006, 126: 611-25.

    CAS  PubMed  Google Scholar 

  2314. Pearl J: Probabilistic reasoning in intelligent systems: networks of plausible inference. 1988, Morgan Kaufmann, San Francisco

    Google Scholar 

  2315. Pearl J: Causality: models, reasoning and inference. 2000, Cambridge University Press, Cambridge

    Google Scholar 

  2316. Hastie T, Tibshirani R, Friedman J: The elements of statistical learning: data mining, inference and prediction. 2001, Springer-Verlag, Berlin

    Google Scholar 

  2317. Shipley B: Cause and correlation in biology: a user's guide to path analysis, structural equations and causal inference. 2001, Cambridge University Press, Cambridge

    Google Scholar 

  2318. Mackay DJC: Information theory, inference and learning algorithms. 2003, Cambridge University Press, Cambridge

    Google Scholar 

  2319. Needham CJ, Bradford JR, Bulpitt AJ, Westhead DR: Inference in Bayesian networks. Nat Biotechnol. 2006, 24: 51-53.

    CAS  PubMed  Google Scholar 

  2320. Jayawardhana B, Kell DB, Rattray M: Bayesian inference of the sites of perturbations in metabolic pathways via Markov Chain Monte Carlo. Bioinformatics. 2008, 24: 1191-1197.

    CAS  PubMed  Google Scholar 

  2321. Rohr JR, Raffel TR, Sessions SK, Hudson PJ: Understanding the net effects of pesticides on amphibian trematode infections. Ecol Appl. 2008, 18: 1743-53.

    PubMed  Google Scholar 

  2322. Calabrese EJ: Converging concepts: adaptive response preconditioning, and the Yerkes-Dodson Law are manifestations of hormesis. Ageing Res Rev. 2008, 7: 8-20.

    CAS  PubMed  Google Scholar 

  2323. Calabrese EJ: Hormesis and medicine. Br J Clin Pharmacol. 2008

    Google Scholar 

  2324. Calabrese E: Hormesis: why it is important to toxicology and toxicologists. Environ Toxicol Chem. 2008, 1.

    Google Scholar 

  2325. Mattson MP: Hormesis and disease resistance: activation of cellular stress response pathways. Hum Exp Toxicol. 2008, 27: 155-62.

    PubMed  Google Scholar 

  2326. Mattson MP: Hormesis defined. Ageing Res Rev. 2008, 7: 1-7.

    CAS  PubMed  Google Scholar 

  2327. The Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group: Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. N Engl J Med. 1998, 339: 1349-57.

    Google Scholar 

  2328. Liappis AP, Kan VL, Rochester CG, Simon GL: The effect of statins on mortality in patients with bacteremia. Clin Infect Dis. 2001, 33: 1352-7.

    CAS  PubMed  Google Scholar 

  2329. Almog Y, Shefer A, Novack V, Maimon N, Barski L, Eizinger M, Friger M, Zeller L, Danon A: Prior statin therapy is associated with a decreased rate of severe sepsis. Circulation. 2004, 110: 880-5.

    CAS  PubMed  Google Scholar 

  2330. Merx MW, Liehn EA, Janssens U, Lutticken R, Schrader J, Hanrath P, Weber C: HMG-CoA reductase inhibitor simvastatin profoundly improves survival in a murine model of sepsis. Circulation. 2004, 109: 2560-5.

    CAS  PubMed  Google Scholar 

  2331. Merx MW, Liehn EA, Graf J, Sandt van de A, Schaltenbrand M, Schrader J, Hanrath P, Weber C: Statin treatment after onset of sepsis in a murine model improves survival. Circulation. 2005, 112: 117-24.

    CAS  PubMed  Google Scholar 

  2332. Zhang L, Zhang ZG, Ding GL, Jiang Q, Liu X, Meng H, Hozeska A, Zhang C, Li L, Morris D, Zhang RL, Lu M, Chopp M: Multitargeted effects of statin-enhanced thrombolytic therapy for stroke with recombinant human tissue-type plasminogen activator in the rat. Circulation. 2005, 112: 3486-94.

    CAS  PubMed  Google Scholar 

  2333. Hackam DG, Mamdani M, Li P, Redelmeier DA: Statins and sepsis in patients with cardiovascular disease: a population-based cohort analysis. Lancet. 2006, 367: 413-8.

    CAS  PubMed  Google Scholar 

  2334. Terblanche M, Almog Y, Rosenson RS, Smith TS, Hackam DG: Statins: panacea for sepsis?. Lancet Infect Dis. 2006, 6: 242-8.

    CAS  PubMed  Google Scholar 

  2335. Thomsen RW, Hundborg HH, Johnsen SP, Pedersen L, Sorensen HT, Schonheyder HC, Lervang HH: Statin use and mortality within 180 days after bacteremia: a population-based cohort study. Crit Care Med. 2006, 34: 1080-6.

    CAS  PubMed  Google Scholar 

  2336. Aneja R, Fink MP: Promising therapeutic agents for sepsis. Trends Microbiol. 2007, 15: 31-7.

    CAS  PubMed  Google Scholar 

  2337. Martin CP, Talbert RL, Burgess DS, Peters JI: Effectiveness of statins in reducing the rate of severe sepsis: a retrospective evaluation. Pharmacotherapy. 2007, 27: 20-6.

    CAS  PubMed  Google Scholar 

  2338. Merx MW, Weber C: Sepsis and the heart. Circulation. 2007, 116: 793-802.

    CAS  PubMed  Google Scholar 

  2339. Schlienger RG, Fedson DS, Jick SS, Jick H, Meier CR: Statins and the risk of pneumonia: a population-based, nested case-control study. Pharmacotherapy. 2007, 27: 325-32.

    CAS  PubMed  Google Scholar 

  2340. Falagas ME, Makris GC, Matthaiou DK, Rafailidis PI: Statins for infection and sepsis: a systematic review of the clinical evidence. J Antimicrob Chemother. 2008, 61: 774-85.

    CAS  PubMed  Google Scholar 

  2341. Lipinski MJ, Abbate A, Fuster V, Vetrovec GW: Drug insight: statins for nonischemic heart failure–evidence and potential mechanisms. Nat Clin Pract Cardiovasc Med. 2007, 4: 196-205.

    CAS  PubMed  Google Scholar 

  2342. Santodomingo-Garzón T, Cunha TM, Verri WA, Valerio DA, Parada CA, Poole S, Ferreira SH, Cunha FQ: Atorvastatin inhibits inflammatory hypernociception. Br J Pharmacol. 2006, 149: 14-22.

    PubMed  PubMed Central  Google Scholar 

  2343. Ferreira GA, Navarro TP, Telles RW, Andrade LE, Sato EI: Atorvastatin therapy improves endothelial-dependent vasodilation in patients with systemic lupus erythematosus: an 8 weeks controlled trial. Rheumatology (Oxford). 2007, 46: 1560-5.

    CAS  Google Scholar 

  2344. Rydgren T, Vaarala O, Sandler S: Simvastatin protects against multiple low-dose streptozotocin-induced type 1 diabetes in CD-1 mice and recurrence of disease in nonobese diabetic mice. J Pharmacol Exp Ther. 2007, 323: 180-5.

    CAS  PubMed  Google Scholar 

  2345. Leung BP, Sattar N, Crilly A, Prach M, McCarey DW, Payne H, Madhok R, Campbell C, Gracie JA, Liew FY, McInnes IB: A novel anti-inflammatory role for simvastatin in inflammatory arthritis. J Immunol. 2003, 170: 1524-30.

    CAS  PubMed  Google Scholar 

  2346. Sattar N, McCarey DW, Capell H, McInnes IB: Explaining how "high-grade" systemic inflammation accelerates vascular risk in rheumatoid arthritis. Circulation. 2003, 108: 2957-63.

    PubMed  Google Scholar 

  2347. Barsante MM, Roffe E, Yokoro CM, Tafuri WL, Souza DG, Pinho V, Castro MS, Teixeira MM: Anti-inflammatory and analgesic effects of atorvastatin in a rat model of adjuvant-induced arthritis. Eur J Pharmacol. 2005, 516: 282-9.

    CAS  PubMed  Google Scholar 

  2348. Dombrecht EJ, Van Offel JF, Bridts CH, Ebo DG, Seynhaeve V, Schuerwegh AJ, Stevens WJ, De Clerck LS: Influence of simvastatin on the production of pro-inflammatory cytokines and nitric oxide by activated human chondrocytes. Clin Exp Rheumatol. 2007, 25: 534-9.

    CAS  PubMed  Google Scholar 

  2349. Haruna Y, Morita Y, Yada T, Satoh M, Fox DA, Kashihara N: Fluvastatin reverses endothelial dysfunction and increased vascular oxidative stress in rat adjuvant-induced arthritis. Arthritis Rheum. 2007, 56: 1827-35.

    CAS  PubMed  Google Scholar 

  2350. Yamagata T, Kinoshita K, Nozaki Y, Sugiyama M, Ikoma S, Funauchi M: Effects of pravastatin in murine collagen-induced arthritis. Rheumatol Int. 2007, 27: 631-9.

    CAS  PubMed  Google Scholar 

  2351. Epstein M, Campese VM: Pleiotropic effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors on renal function. Am J Kidney Dis. 2005, 45: 2-14.

    CAS  PubMed  Google Scholar 

  2352. Yasuda H, Yuen PS, Hu X, Zhou H, Star RA: Simvastatin improves sepsis-induced mortality and acute kidney injury via renal vascular effects. Kidney Int. 2006, 69: 1535-42.

    CAS  PubMed  PubMed Central  Google Scholar 

  2353. Campese VM, Park J: HMG-CoA reductase inhibitors and the kidney. Kidney Int. 2007, 71: 1215-22.

    CAS  PubMed  Google Scholar 

  2354. Otuki MF, Pietrovski EF, Cabrini DA: Topical simvastatin: preclinical evidence for a treatment of skin inflammatory conditions. J Dermatol Sci. 2006, 44: 45-7.

    CAS  PubMed  Google Scholar 

  2355. Lee JH, Lee DS, Kim EK, Choe KH, Oh YM, Shim TS, Kim SE, Lee YS, Lee SD: Simvastatin inhibits cigarette smoking-induced emphysema and pulmonary hypertension in rat lungs. Am J Respir Crit Care Med. 2005, 172: 987-93.

    PubMed  Google Scholar 

  2356. Pirat A, Zeyneloglu P, Aldemir D, Yücel M, Özen O, Candan S, Arslan G: Pretreatment with simvastatin reduces lung injury related to intestinal ischemia-reperfusion in rats. Anesth Analg. 2006, 102: 225-32.

    CAS  PubMed  Google Scholar 

  2357. Endres M, Laufs U, Huang Z, Nakamura T, Huang P, Moskowitz MA, Liao JK: Stroke protection by 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors mediated by endothelial nitric oxide synthase. Proc Natl Acad Sci USA. 1998, 95: 8880-5.

    CAS  PubMed  PubMed Central  Google Scholar 

  2358. Vaughan CJ, Delanty N: Neuroprotective properties of statins in cerebral ischemia and stroke. Stroke. 1999, 30: 1969-73.

    CAS  PubMed  Google Scholar 

  2359. Amin-Hanjani S, Stagliano NE, Yamada M, Huang PL, Liao JK, Moskowitz MA: Mevastatin, an HMG-CoA reductase inhibitor, reduces stroke damage and upregulates endothelial nitric oxide synthase in mice. Stroke. 2001, 32: 980-6.

    CAS  PubMed  Google Scholar 

  2360. Sironi L, Cimino M, Guerrini U, Calvio AM, Lodetti B, Asdente M, Balduini W, Paoletti R, Tremoli E: Treatment with statins after induction of focal ischemia in rats reduces the extent of brain damage. Arterioscler Thromb Vasc Biol. 2003, 23: 322-7.

    CAS  PubMed  Google Scholar 

  2361. Endres M, Laufs U, Liao JK, Moskowitz MA: Targeting eNOS for stroke protection. Trends Neurosci. 2004, 27: 283-9.

    CAS  PubMed  Google Scholar 

  2362. Amarenco P, Moskowitz MA: The dynamics of statins: from event prevention to neuroprotection. Stroke. 2006, 37: 294-6.

    PubMed  Google Scholar 

  2363. Amarenco P, Lavallee PC, Mazighi M, Labreuche J: The role of statins in the prevention of stroke. Archives of Medical Science. 2007, 3: S109-S114.

    CAS  Google Scholar 

  2364. DeFaria Yeh D, Waters DD: Preventing and treating stroke and transient ischemic attack. Am J Cardiol. 2008, 101: 270-3.

    PubMed  Google Scholar 

  2365. Chen SF, Hung TH, Chen CC, Lin KH, Huang YN, Tsai HC, Wang JY: Lovastatin improves histological and functional outcomes and reduces inflammation after experimental traumatic brain injury. Life Sci. 2007, 81: 288-98.

    CAS  PubMed  Google Scholar 

  2366. Faden AI, Stoica B: Neuroprotection: challenges and opportunities. Arch Neurol. 2007, 64: 794-800.

    PubMed  Google Scholar 

  2367. Wu H, Lu D, Jiang H, Xiong Y, Qu C, Li B, Mahmood A, Zhou D, Chopp M: Simvastatin-mediated upregulation of VEGF and BDNF, activation of the PI3K/Akt pathway, and increase of neurogenesis are associated with therapeutic improvement after traumatic brain injury. J Neurotrauma. 2008, 25: 130-9.

    PubMed  Google Scholar 

  2368. Jick H, Zornberg GL, Jick SS, Seshadri S, Drachman DA: Statins and the risk of dementia. Lancet. 2000, 356: 1627-31.

    CAS  PubMed  Google Scholar 

  2369. Wolozin B, Kellman W, Ruosseau P, Celesia GG, Siegel G: Decreased prevalence of Alzheimer disease associated with 3-hydroxy-3-methyglutaryl coenzyme A reductase inhibitors. Arch Neurol. 2000, 57: 1439-43.

    CAS  PubMed  Google Scholar 

  2370. Fassbender K, Simons M, Bergmann C, Stroick M, Lutjohann D, Keller P, Runz H, Kuhl S, Bertsch T, von Bergmann K, Hennerici M, Beyreuther K, Hartmann T: Simvastatin strongly reduces levels of Alzheimer's disease beta -amyloid peptides Abeta 42 and Abeta 40 in vitro and in vivo. Proc Natl Acad Sci USA. 2001, 98: 5856-61.

    CAS  PubMed  PubMed Central  Google Scholar 

  2371. Refolo LM, Pappolla MA, LaFrancois J, Malester B, Schmidt SD, Thomas-Bryant T, Tint GS, Wang R, Mercken M, Petanceska SS, Duff KE: A cholesterol-lowering drug reduces beta-amyloid pathology in a transgenic mouse model of Alzheimer's disease. Neurobiol Dis. 2001, 8: 890-9.

    CAS  PubMed  Google Scholar 

  2372. Hajjar I, Schumpert J, Hirth V, Wieland D, Eleazer GP: The impact of the use of statins on the prevalence of dementia and the progression of cognitive impairment. J Gerontol A Biol Sci Med Sci. 2002, 57: M414-8.

    PubMed  Google Scholar 

  2373. Zamvil SS, Steinman L: Cholesterol-lowering statins possess anti-inflammatory activity that might be useful for treatment of MS. Neurology. 2002, 59: 970-1.

    PubMed  Google Scholar 

  2374. Aktas O, Waiczies S, Smorodchenko A, Dorr J, Seeger B, Prozorovski T, Sallach S, Endres M, Brocke S, Nitsch R, Zipp F: Treatment of relapsing paralysis in experimental encephalomyelitis by targeting Th1 cells through atorvastatin. J Exp Med. 2003, 197: 725-33.

    CAS  PubMed  PubMed Central  Google Scholar 

  2375. Casserly I, Topol E: Convergence of atherosclerosis and Alzheimer's disease: inflammation cholesterol, and misfolded proteins. Lancet. 2004, 363: 1139-46.

    CAS  PubMed  Google Scholar 

  2376. Nath N, Giri S, Prasad R, Singh AK, Singh I: Potential targets of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor for multiple sclerosis therapy. J Immunol. 2004, 172: 1273-86.

    CAS  PubMed  Google Scholar 

  2377. Neuhaus O, Stuve O, Zamvil SS, Hartung HP: Are statins a treatment option for multiple sclerosis?. Lancet Neurol. 2004, 3: 369-71.

    CAS  PubMed  Google Scholar 

  2378. Vollmer T, Key L, Durkalski V, Tyor W, Corboy J, Markovic-Plese S, Preiningerova J, Rizzo M, Singh I: Oral simvastatin treatment in relapsing-remitting multiple sclerosis. Lancet. 2004, 363: 1607-8.

    CAS  PubMed  Google Scholar 

  2379. Neuhaus O, Stuve O, Zamvil SS, Hartung HP: Evaluation of HMG-CoA reductase inhibitors for multiple sclerosis: opportunities and obstacles. CNS Drugs. 2005, 19: 833-41.

    CAS  PubMed  Google Scholar 

  2380. Mok SW, Thelen KM, Riemer C, Bamme T, Gultner S, Lutjohann D, Baier M: Simvastatin prolongs survival times in prion infections of the central nervous system. Biochem Biophys Res Commun. 2006, 348: 697-702.

    CAS  PubMed  Google Scholar 

  2381. Schmeer C, Kretz A, Isenmann S: Statin-mediated protective effects in the central nervous system: general mechanisms and putative role of stress proteins. Restor Neurol Neurosci. 2006, 24: 79-95.

    CAS  PubMed  Google Scholar 

  2382. Wolozin B, Wang SW, Li NC, Lee A, Lee TA, Kazis LE: Simvastatin is associated with a reduced incidence of dementia and Parkinson's disease. BMC Med. 2007, 5: 20.

    PubMed  PubMed Central  Google Scholar 

  2383. Cavalli A, Bolognesi ML, Minarini A, Rosini M, Tumiatti V, Recanatini M, Melchiorre C: Multi-target-directed ligands to combat neurodegenerative diseases. J Med Chem. 2008, 51: 347-72.

    CAS  PubMed  Google Scholar 

  2384. Paul F, Waiczies S, Wuerfel J, Bellmann-Strobl J, Dorr J, Waiczies H, Haertle M, Wernecke KD, Volk HD, Aktas O, Zipp F: Oral high-dose atorvastatin treatment in relapsing-remitting multiple sclerosis. PLoS ONE. 2008, 3: e1928.

    PubMed  PubMed Central  Google Scholar 

  2385. Bösel J, Gandor F, Harms C, Synowitz M, Harms U, Djoufack PC, Megow D, Dirnagl U, Hortnagl H, Fink KB, Endres M: Neuroprotective effects of atorvastatin against glutamate-induced excitotoxicity in primary cortical neurones. J Neurochem. 2005, 92: 1386-98.

    PubMed  Google Scholar 

  2386. Wong WW, Dimitroulakos J, Minden MD, Penn LZ: HMG-CoA reductase inhibitors and the malignant cell: the statin family of drugs as triggers of tumor-specific apoptosis. Leukemia. 2002, 16: 508-19.

    CAS  PubMed  Google Scholar 

  2387. Kaushal V, Kohli M, Mehta P, Mehta JL: Potential anticancer effects of statins: fact or fiction?. Endothelium. 2003, 10: 49-58.

    CAS  PubMed  Google Scholar 

  2388. Wu J, Wong WW, Khosravi F, Minden MD, Penn LZ: Blocking the Raf/MEK/ERK pathway sensitizes acute myelogenous leukemia cells to lovastatin-induced apoptosis. Cancer Res. 2004, 64: 6461-8.

    CAS  PubMed  Google Scholar 

  2389. Demierre MF, Higgins PD, Gruber SB, Hawk E, Lippman SM: Statins and cancer prevention. Nat Rev Cancer. 2005, 5: 930-42.

    CAS  PubMed  Google Scholar 

  2390. Fritz G: HMG-CoA reductase inhibitors (statins) as anticancer drugs (review). Int J Oncol. 2005, 27: 1401-9.

    CAS  PubMed  Google Scholar 

  2391. Katz MS: Therapy insight: Potential of statins for cancer chemoprevention and therapy. Nat Clin Pract Oncol. 2005, 2: 82-9.

    CAS  PubMed  Google Scholar 

  2392. Shannon J, Tewoderos S, Garzotto M, Beer TM, Derenick R, Palma A, Farris PE: Statins and prostate cancer risk: a case-control study. Am J Epidemiol. 2005, 162: 318-25.

    PubMed  Google Scholar 

  2393. Campbell MJ, Esserman LJ, Zhou Y, Shoemaker M, Lobo M, Borman E, Baehner F, Kumar AS, Adduci K, Marx C, Petricoin EF, Liotta LA, Winters M, Benz S, Benz CC: Breast cancer growth prevention by statins. Cancer Res. 2006, 66: 8707-14.

    CAS  PubMed  Google Scholar 

  2394. Cauley JA, McTiernan A, Rodabough RJ, LaCroix A, Bauer DC, Margolis KL, Paskett ED, Vitolins MZ, Furberg CD, Chlebowski RT: Statin use and breast cancer: prospective results from the Women's Health Initiative. J Natl Cancer Inst. 2006, 98: 700-7.

    CAS  PubMed  Google Scholar 

  2395. Platz EA, Leitzmann MF, Visvanathan K, Rimm EB, Stampfer MJ, Willett WC, Giovannucci E: Statin drugs and risk of advanced prostate cancer. J Natl Cancer Inst. 2006, 98: 1819-25.

    CAS  PubMed  Google Scholar 

  2396. Jacobs EJ, Rodriguez C, Bain EB, Wang Y, Thun MJ, Calle EE: Cholesterol-lowering drugs and advanced prostate cancer incidence in a large U.S. cohort. Cancer Epidemiol Biomarkers Prev. 2007, 16: 2213-7.

    CAS  PubMed  Google Scholar 

  2397. Kodach LL, Bleuming SA, Peppelenbosch MP, Hommes DW, Brink van den GR, Hardwick JC: The effect of statins in colorectal cancer is mediated through the bone morphogenetic protein pathway. Gastroenterology. 2007, 133: 1272-81.

    CAS  PubMed  Google Scholar 

  2398. Farwell WR, Scranton RE, Lawler EV, Lew RA, Brophy MT, Fiore LD, Gaziano JM: The association between statins and cancer incidence in a veterans population. J Natl Cancer Inst. 2008, 100: 134-9.

    CAS  PubMed  Google Scholar 

  2399. Karp I, Behlouli H, Lelorier J, Pilote L: Statins and cancer risk. Am J Med. 2008, 121: 302-9.

    CAS  PubMed  Google Scholar 

  2400. Sassano A, Platanias LC: Statins in tumor suppression. Cancer Lett. 2008, 260: 11-9.

    CAS  PubMed  Google Scholar 

  2401. Brenner S: Loose ends. 1997, Current Biology, London

    Google Scholar 

  2402. Kacser H: On parts and wholes in metabolism. The organization of cell metabolism. Edited by: Welch GR, Clegg JS. 1986, Plenum Press, New York, 327-337.

    Google Scholar 

  2403. Pennisi E: A new window on how genomes work. Science. 2007, 316: 1120-1.

    CAS  PubMed  Google Scholar 

  2404. Kell DB, Oliver SG: Here is the evidence, now what is the hypothesis? The complementary roles of inductive and hypothesis-driven science in the post-genomic era. Bioessays. 2004, 26: 99-105.

    PubMed  Google Scholar 

  2405. Russell B: Am I an atheist or an agnostic?. 1947

    Google Scholar 

  2406. Kostoff RN: Overcoming specialization. Bioscience. 2002, 52: 937-941.

    Google Scholar 

  2407. Noble D: Modeling the heart – from genes to cells to the whole organ. Science. 2002, 295: 1678-1682.

    CAS  PubMed  Google Scholar 

  2408. Hunter PJ: The IUPS Physiome Project: a framework for computational physiology. Prog Biophys Mol Biol. 2004, 85: 551-69.

    CAS  PubMed  Google Scholar 

  2409. Duarte NC, Becker SA, Jamshidi N, Thiele I, Mo ML, Vo TD, Srvivas R, Palsson BØ: Global reconstruction of the human metabolic network based on genomic and bibliomic data. Proc Natl Acad Sci. 2007, 104: 1777-1782.

    CAS  PubMed  PubMed Central  Google Scholar 

  2410. Kell DB: The virtual human: towards a global systems biology of multiscale, distributed biochemical network models. IUBMB Life. 2007, 59: 689-95.

    CAS  PubMed  Google Scholar 

  2411. Ma H, Sorokin A, Mazein A, Selkov A, Selkov E, Demin O, Goryanin I: The Edinburgh human metabolic network reconstruction and its functional analysis. Mol Syst Biol. 2007, 3: 135.

    PubMed  PubMed Central  Google Scholar 

  2412. Feist AM, Palsson BØ: The growing scope of applications of genome-scale metabolic reconstructions using Escherichia coli. Nat Biotechnol. 2008, 26 (6): 659-67.

    CAS  PubMed  PubMed Central  Google Scholar 

  2413. Ma H, Goryanin I: Human metabolic network reconstruction and its impact on drug discovery and development. Drug Discov Today. 2008, 13: 402-8.

    CAS  PubMed  Google Scholar 

  2414. Jamshidi N, Palsson BØ: Formulating genome-scale kinetic models in the post-genome era. Mol Syst Biol. 2008, 4: 171.

    PubMed  PubMed Central  Google Scholar 

  2415. Clapworthy G, Viceconti M, Coveney PV, Kohl P: The virtual physiological human: building a framework for computational biomedicine I. Editorial. Philos Transact A Math Phys Eng Sci. 2008, 366: 2975-8.

    Google Scholar 

  2416. Fenner JW, Brook B, Clapworthy G, Coveney PV, Feipel V, Gregersen H, Hose DR, Kohl P, Lawford P, McCormack KM, Pinney D, Thomas SR, Van Sint Jan S, Waters S, Viceconti M: The EuroPhysiome, STEP and a roadmap for the virtual physiological human. Philos Transact A Math Phys Eng Sci. 2008, 366: 2979-99.

    CAS  Google Scholar 

  2417. Snoep JL, Bruggeman F, Olivier BG, Westerhoff HV: Towards building the silicon cell: a modular approach. Biosystems. 2006, 83: 207-16.

    CAS  PubMed  Google Scholar 

  2418. Jongeneel CV, Delorenzi M, Iseli C, Zhou D, Haudenschild CD, Khrebtukova I, Kuznetsov D, Stevenson BJ, Strausberg RL, Simpson AJ, Vasicek TJ: An atlas of human gene expression from massively parallel signature sequencing (MPSS). Genome Res. 2005, 15: 1007-14.

    CAS  PubMed  PubMed Central  Google Scholar 

  2419. Persson A, Hober S, Uhlen M: A human protein atlas based on antibody proteomics. Curr Opin Mol Ther. 2006, 8: 185-90.

    CAS  PubMed  Google Scholar 

  2420. Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, Boe AF, Boguski MS, Brockway KS, Byrnes EJ, Chen L, Chen L, Chen TM, Chin MC, Chong J, Crook BE, Czaplinska A, Dang CN, Datta S, Dee NR, Desaki AL, Desta T, Diep E, Dolbeare TA, Donelan MJ, Dong HW, Dougherty JG, Duncan BJ, Ebbert AJ, Eichele G, Estin LK, Faber C, Facer BA, Fields R, Fischer SR, Fliss TP, Frensley C, Gates SN, Glattfelder KJ, Halverson KR, Hart MR, Hohmann JG, Howell MP, Jeung DP, Johnson RA, Karr PT, Kawal R, Kidney JM, Knapik RH, Kuan CL, Lake JH, Laramee AR, Larsen KD, Lau C, Lemon TA, Liang AJ, Liu Y, Luong LT, Michaels J, Morgan JJ, Morgan RJ, Mortrud MT, Mosqueda NF, Ng LL, Ng R, Orta GJ, Overly CC, Pak TH, Parry SE, Pathak SD, Pearson OC, Puchalski RB, Riley ZL, Rockett HR, Rowland SA, Royall JJ, Ruiz MJ, Sarno NR, Schaffnit K, Shapovalova NV, Sivisay T, Slaughterbeck CR, Smith SC, Smith KA, Smith BI, Sodt AJ, Stewart NN, Stumpf KR, Sunkin SM, Sutram M, Tam A, Teemer CD, Thaller C, Thompson CL, Varnam LR, Visel A, Whitlock RM, Wohnoutka PE, Wolkey CK, Wong VY, et al: Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2007, 445: 168-76.

    CAS  PubMed  Google Scholar 

  2421. Hober S, Uhlén M: Human protein atlas and the use of microarray technologies. Curr Opin Biotechnol. 2008, 19: 30-5.

    CAS  PubMed  Google Scholar 

  2422. Barbe L, Lundberg E, Oksvold P, Stenius A, Lewin E, Björling E, Asplund A, Ponten F, Brismar H, Uhlén M, Andersson-Svahn H: Toward a confocal subcellular atlas of the human proteome. Mol Cell Proteomics. 2008, 7: 499-508.

    CAS  PubMed  Google Scholar 

  2423. Sultan M, Schulz MH, Richard H, Magen A, Klingenhoff A, Scherf M, Seifert M, Borodina T, Soldatov A, Parkhomchuk D, Schmidt D, O'Keeffe S, Haas S, Vingron M, Lehrach H, Yaspo ML: A global view of gene activity and alternative splicing by deep sequencing of the human transcriptome. Science. 2008, 321: 956-60.

    CAS  PubMed  Google Scholar 

  2424. Shlomi T, Cabili MN, Herrgard MJ, Palsson BØ, Ruppin E: Network-based prediction of human tissue-specific metabolism. Nat Biotechnol. 2008, 26: 1003-10.

    CAS  PubMed  Google Scholar 

  2425. Hucka M, Finney A, Sauro HM, Bolouri H, Doyle JC, Kitano H, Arkin AP, Bornstein BJ, Bray D, Cornish-Bowden A, Cuellar AA, Dronov S, Gilles ED, Ginkel M, Gor V, Goryanin II, Hedley WJ, Hodgman TC, Hofmeyr JH, Hunter PJ, Juty NS, Kasberger JL, Kremling A, Kummer U, Le Novere N, Loew LM, Lucio D, Mendes P, Minch E, Mjolsness ED, Nakayama Y, Nelson MR, Nielsen PF, Sakurada T, Schaff JC, Shapiro BE, Shimizu TS, Spence HD, Stelling J, Takahashi K, Tomita M, Wagner J, Wang J: The systems biology markup language (SBML): a medium for representation and exchange of biochemical network models. Bioinformatics. 2003, 19: 524-31.

    CAS  PubMed  Google Scholar 

  2426. Le Novère N, Finney A, Hucka M, Bhalla US, Campagne F, Collado-Vides J, Crampin EJ, Halstead M, Klipp E, Mendes P, Nielsen P, Sauro H, Shapiro B, Snoep JL, Spence HD, Wanner BL: Minimum information requested in the annotation of biochemical models (MIRIAM). Nat Biotechnol. 2005, 23: 1509-15.

    PubMed  Google Scholar 

  2427. Taylor CF, Field D, Sansone SA, Aerts J, Apweiler R, Ashburner M, Ball CA, Binz PA, Bogue M, Booth T, Brazma A, Brinkman RR, Michael Clark A, Deutsch EW, Fiehn O, Fostel J, Ghazal P, Gibson F, Gray T, Grimes G, Hancock JM, Hardy NW, Hermjakob H, Julian RK, Kane M, Kettner C, Kinsinger C, Kolker E, Kuiper M, Novere NL, Leebens-Mack J, Lewis SE, Lord P, Mallon AM, Marthandan N, Masuya H, McNally R, Mehrle A, Morrison N, Orchard S, Quackenbush J, Reecy JM, Robertson DG, Rocca-Serra P, Rodriguez H, Rosenfelder H, Santoyo-Lopez J, Scheuermann RH, Schober D, Smith B, Snape J, Stoeckert CJ, Tipton K, Sterk P, Untergasser A, Vandesompele J, Wiemann S: Promoting coherent minimum reporting guidelines for biological and biomedical investigations: the MIBBI project. Nat Biotechnol. 2008, 26: 889-96.

    CAS  PubMed  PubMed Central  Google Scholar 

  2428. Kell DB: Progress being made on standards for use in data sharing. Nature. 2008, 456: 29.

    CAS  PubMed  Google Scholar 

  2429. Kell DB, Mendes P: The markup is the model: reasoning about systems biology models in the Semantic Web era. J Theoret Biol. 2008, 252: 538-543.

    CAS  Google Scholar 

  2430. Surowiecki J: The wisdom of crowds: why the many are smarter than the few. 2004, Abacus, London

    Google Scholar 

  2431. Anderson CM: The long tail: how endless choice is creating unlimited demand. 2006, Random House, London

    Google Scholar 

  2432. Sunstein CR: Infotopia: how many minds produce knowledge. 2006, Oxfor University Press, Oxford

    Google Scholar 

  2433. Tapscott D, Williams A: Wikinomics: how masscollaboration changes everything. 2007, New Paradigm

    Google Scholar 

  2434. Leadbeater C: We-think. 2008, Profile Books, London

    Google Scholar 

  2435. Shirky C: Here comes everybody. 2008, Allen Lane, London

    Google Scholar 

  2436. Li P, Oinn T, Soiland S, Kell DB: Automated manipulation of systems biology models using libSBML within Taverna workflows. Bioinformatics. 2008, 24: 287-289.

    PubMed  Google Scholar 

  2437. Li P, Castrillo JI, Velarde G, Wassink I, Soiland-Reyes S, Owen S, Withers D, Oinn T, Pocock MR, Goble CA, Oliver SG, Kell DB: Performing statistical analyses on quantitative data in Taverna workflows: an example using R and maxdBrowse to identify differentially expressed genes from microarray data. BMC Bioinformatics. 2008, 9: 334.

    PubMed  PubMed Central  Google Scholar 

  2438. De Roure D, Goble C: myExperiment – A Web 2.0 Virtual Research Environment. Proc International Workshop on Virtual Research Environments and Collaborative Work Environments, May 2007, Edinburgh, UK. 2007, [http://eprints.ecs.soton.ac.uk/13961/]

    Google Scholar 

  2439. Suber P: Open access to the scientific journal literature. J Biol. 2002, 1: 3.

    PubMed  PubMed Central  Google Scholar 

  2440. Bourne PE, Fink JL, Gerstein M: Open access: taking full advantage of the content. PLoS Comput Biol. 2008, 4: e1000037.

    PubMed  PubMed Central  Google Scholar 

  2441. Johnson RK: Institutional Repositories: partnering with faculty to enhance scholarly communication. D-Lib Magazine. 2002, 8: [http://www.dlib.org/dlib/november02/johnson/11johnson.html]

    Google Scholar 

  2442. Lynch CA: Institutional repositories: Essential infrastructure for scholarship in the digital age. Libr Acad. 2003, 3: 327-336.

    Google Scholar 

  2443. Lee E, Chuang HY, Kim JW, Ideker T, Lee D: Inferring pathway activity toward precise disease classification. PLoS Comput Biol. 2008, 4: e1000217.

    PubMed  PubMed Central  Google Scholar 

  2444. Stein LD: Integrating biological databases. Nat Rev Genet. 2003, 4: 337-45.

    CAS  PubMed  Google Scholar 

  2445. Lord P, Bechhofer S, Wilkinson MD, Schiltz G, Gessler D, Hull D, Goble CA, Stein L: Applying Semantic Web services to bioinformatics: Experiences gained, lessons learnt. LNCS. 2004, 3298: 350-364.

    Google Scholar 

  2446. Curcin V, Ghanem M, Guo Y: Web services in the life sciences. Drug Discov Today. 2005, 10: 865-71.

    CAS  PubMed  Google Scholar 

  2447. Neerincx PB, Leunissen JA: Evolution of web services in bioinformatics. Brief Bioinform. 2005, 6: 178-88.

    CAS  PubMed  Google Scholar 

  2448. Le Novère N, Bornstein B, Broicher A, Courtot M, Donizelli M, Dharuri H, Li L, Sauro H, Schilstra M, Shapiro B, Snoep JL, Hucka M: BioModels Database: a free, centralized database of curated published, quantitative kinetic models of biochemical and cellular systems. Nucleic Acids Res. 2006, 34: D689-91.

    PubMed  Google Scholar 

  2449. Richardson L, Riuby S: RESTful web services. 2007, O'Reilly Sebastopol, CA

    Google Scholar 

  2450. Fernández JM, Hoffmann R, Valencia A: iHOP web services. Nucleic Acids Res. 2007, 35: W21-6.

    PubMed  PubMed Central  Google Scholar 

  2451. Stein LD: Towards a cyberinfrastructure for the biological sciences: progress, visions and challenges. Nat Rev Genet. 2008, 9: 678-88.

    CAS  PubMed  Google Scholar 

  2452. Williams AJ: A perspective of publicly accessible/open-access chemistry databases. Drug Discov Today. 2008, 13: 495-501.

    CAS  PubMed  Google Scholar 

  2453. Margulies M, Egholm M, Altman WE, Attiya S, Bader JS, Bemben LA, Berka J, Braverman MS, Chen YJ, Chen Z, Dewell SB, Du L, Fierro JM, Gomes XV, Godwin BC, He W, Helgesen S, Ho CH, Irzyk GP, Jando SC, Alenquer ML, Jarvie TP, Jirage KB, Kim JB, Knight JR, Lanza JR, Leamon JH, Lefkowitz SM, Lei M, Li J, Lohman KL, Lu H, Makhijani VB, McDade KE, McKenna MP, Myers EW, Nickerson E, Nobile JR, Plant R, Puc BP, Ronan MT, Roth GT, Sarkis GJ, Simons JF, Simpson JW, Srinivasan M, Tartaro KR, Tomasz A, Vogt KA, Volkmer GA, Wang SH, Wang Y, Weiner MP, Yu P, Begley RF, Rothberg JM: Genome sequencing in microfabricated high-density picolitre reactors. Nature. 2005, 437: 376-380.

    CAS  PubMed  PubMed Central  Google Scholar 

  2454. Shendure J, Porreca GJ, Reppas NB, Lin X, McCutcheon JP, Rosenbaum AM, Wang MD, Zhang K, Mitra RD, Church GM: Accurate multiplex polony sequencing of an evolved bacterial genome. Science. 2005, 309: 1728-32.

    CAS  PubMed  Google Scholar 

  2455. Bentley DR, Balasubramanian S, Swerdlow HP, Smith GP, Milton J, Brown CG, Hall KP, Evers DJ, Barnes CL, Bignell HR, Boutell JM, Bryant J, Carter RJ, Keira Cheetham R, Cox AJ, Ellis DJ, Flatbush MR, Gormley NA, Humphray SJ, Irving LJ, Karbelashvili MS, Kirk SM, Li H, Liu X, Maisinger KS, Murray LJ, Obradovic B, Ost T, Parkinson ML, Pratt MR, Rasolonjatovo IM, Reed MT, Rigatti R, Rodighiero C, Ross MT, Sabot A, Sankar SV, Scally A, Schroth GP, Smith ME, Smith VP, Spiridou A, Torrance PE, Tzonev SS, Vermaas EH, Walter K, Wu X, Zhang L, Alam MD, Anastasi C, Aniebo IC, Bailey DM, Bancarz IR, Banerjee S, Barbour SG, Baybayan PA, Benoit VA, Benson KF, Bevis C, Black PJ, Boodhun A, Brennan JS, Bridgham JA, Brown RC, Brown AA, Buermann DH, Bundu AA, Burrows JC, Carter NP, Castillo N, Chiara ECM, Chang S, Neil Cooley R, Crake NR, Dada OO, Diakoumakos KD, Dominguez-Fernandez B, Earnshaw DJ, Egbujor UC, Elmore DW, Etchin SS, Ewan MR, Fedurco M, Fraser LJ, Fuentes Fajardo KV, Scott Furey W, George D, Gietzen KJ, Goddard CP, Golda GS, Granieri PA, Green DE, Gustafson DL, Hansen NF, Harnish K, Haudenschild CD, Heyer NI, Hims MM, Ho JT, Horgan AM, et al: Accurate whole human genome sequencing using reversible terminator chemistry. Nature. 2008, 456: 53-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  2456. Harris TD, Buzby PR, Babcock H, Beer E, Bowers J, Braslavsky I, Causey M, Colonell J, Dimeo J, Efcavitch JW, Giladi E, Gill J, Healy J, Jarosz M, Lapen D, Moulton K, Quake SR, Steinmann K, Thayer E, Tyurina A, Ward R, Weiss H, Xie Z: Single-molecule DNA sequencing of a viral genome. Science. 2008, 320: 106-9.

    CAS  PubMed  Google Scholar 

  2457. Gupta PK: Single-molecule DNA sequencing technologies for future genomics research. Trends Biotechnol. 2008, 26: 602-11.

    CAS  PubMed  Google Scholar 

  2458. Kahvejian A, Quackenbush J, Thompson JF: What would you do if you could sequence everything?. Nat Biotechnol. 2008, 26: 1125-33.

    CAS  PubMed  PubMed Central  Google Scholar 

  2459. Zerbino DR, Birney E: Velvet: algorithms for de novo short read assembly using de Bruijn graphs. Genome Res. 2008, 18: 821-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  2460. Pavlopoulos GA, Wegener A-L, Schneider R: A survey of visualization tools for biological network analysis. Bio-data mining. 2008, 1: 12.

    PubMed  PubMed Central  Google Scholar 

  2461. Garny A, Nickerson DP, Cooper J, Weber dos Santos R, Miller AK, McKeever S, Nielsen PM, Hunter PJ: CellML and associated tools and techniques. Philos Transact A Math Phys Eng Sci. 2008, 366: 3017-43.

    Google Scholar 

  2462. Li XJ, Brazhnik O, Kamal A, Guo D, Lee C, Hoops S, Mendes P: Databases and visualization for metabolomics. Metabolic profiling: its role in biomarker discovery and gene function analysis. Edited by: Harrigan GG, Goodacre R. 2003, Kluwer Academic Publishers, Boston, 293-309.

    Google Scholar 

  2463. Jenkinson AM, Albrecht M, Birney E, Blankenburg H, Down T, Finn RD, Hermjakob H, Hubbard TJ, Jimenez RC, Jones P, Kähäri A, Kulesha E, Macias JR, Reeves GA, Prlić A: Integrating biological data – the Distributed Annotation System. BMC Bioinformatics. 2008, 9 (Suppl 8): S3.

    PubMed  PubMed Central  Google Scholar 

  2464. Köhler J, Baumbach J, Taubert J, Specht M, Skusa A, Ruegg A, Rawlings C, Verrier P, Philippi S: Graph-based analysis and visualization of experimental results with ONDEX. Bioinformatics. 2006, 22: 1383-90.

    PubMed  Google Scholar 

  2465. Pettifer SR, Sinnott JR, Attwood TK: UTOPIA – user-friendly tools for operating informatics applications. Comp Func Genom. 2004, 5: 56-60.

    CAS  Google Scholar 

  2466. McDermott P, Sinnott J, Thorne D, Pettifer S, Attwood T: An architecture for visualisation and interactive analysis of proteins. Proc 4th Int Conf Coordinated and Multiple Views in Exploratory Visualization (CMV06). 2006, 55-68.

    Google Scholar 

  2467. Thorne D, Pettifer S, Attwood T: Integrating Abstract and Physical Molecular Model Interaction. EG UK Theory and Practice of Computer Graphics (2005). Edited by: Lever L, McDerby M. 2005, Eurographics Association, 75-82.

    Google Scholar 

  2468. Pettifer S, Wolstencroft K, Alper P, Attwood T, Coletta A, Goble C, Li P, McDermott J, Marsh J, Oinn T, Sinnott J, Thorne D: myGrid and UTOPIA: an integrated approach to enacting and visualising in silico experiments in the life sciences. Proc Conf Data Integration in the Life Sciences. 2007, 59-70.

    Google Scholar 

  2469. Neufeld EJ: Oral chelators deferasirox and deferiprone for transfusional iron overload in thalassemia major: new data, new questions. Blood. 2006, 107: 3436-41.

    CAS  PubMed  PubMed Central  Google Scholar 

Pre-publication history

Download references

Acknowledgements

I thank Jon Barasch (Columbia University) for drawing my attention to the role of NGAL in human physiology, and Phil Baker, David Brough and Louise Kenny for many further useful and enjoyable discussions. I thank Katya Tarasova for considerable assistance with literature gathering and Julie Cowley for assistance with the proofs. Much of my work when this was written has been supported by the UK BBSRC and EPSRC, for which I am most grateful, and I am also a recipient of funding from the British Heart Foundation. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. This is a contribution from the Manchester Centre for Integrative Systems Biology http://www.mcisb.org.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Douglas B Kell.

Additional information

Competing interests

The author declares that he has no competing interests.

Authors’ original submitted files for images

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Kell, D.B. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics 2, 2 (2009). https://doi.org/10.1186/1755-8794-2-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1755-8794-2-2

Keywords