ArticlePDF Available

Iron Behaving Badly: Inappropriate Iron Chelation as a Major Contributor to the Aetiology of Vascular and Other Progressive Inflammatory and Degenerative Diseases

Authors:

Abstract and Figures

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.
Content may be subject to copyright.
BMC Medical Genomics
Review
Iron behaving badly: inappropriate iron chelation as a major
contributor to the aetiology of vascular and other progressive
inflammatory and degenerative diseases
Douglas B Kell*
Address: School of Chemistry and Manchester Interdisciplinary Biocentre, The University of Manchester, 131 Princess St, Manchester, M1 7DN, UK
E-mail: Douglas B Kell* - dbk@manchester.ac.uk
*Corresponding author
Published: 08 January 2009 Received: 2 September 2008
BMC Medical Genomics 2009, 2:2 doi: 10.1186/1755- 8794-2-2 Accepted: 8 January 2009
This article is available from: http://www.biomedcentral.com/1755-8794/2/2
© 2009 Kell; licensee BioMed Central Ltd.
This is an Open Access articl e distributed under t he te rms of the Creative Commons Attribution License (
http://creativecommons.org/license s/by/2.0),
which permits unre stricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Background: The production of peroxide and superox ide 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 t he 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 o f 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 si tes of plaques an d lesions, as well as studies
showing the significance of iron to aging and l ongevity. 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 isolati on leads to inconsistentpatternsofapparentcausalitywhenitisthe
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 antio xidants can actually act as pro-oxid ants. The reduction of redox stress thus re quires
suitable levels of both antioxidants and effective iron chelators. Some polyphenolic antioxidants
may serve both roles.
Understanding the exact speciation and ligand ing 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 involvi ng 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 p ossible to identify mechanisms by which ROSs and poorly
Page 1 of 79
(page number n ot f or citation purposes)
BioMed Central
Open Access
liganded iron act synergi stical ly and autocatalytically, leadi ng 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 li ganded 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 physiolo gical
processes that degrade over time. Understanding these requires an integrative, systems-level
approach that may lead to novel therapeutic targets.
Background a nd 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 increas ing by
something approaching 2 per minute at PubMed/Med-
line 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 mi ning and natural language
processing (e.g. [3-5]), for joining disparate concepts [6],
for literature-based discovery (e.g. [7-11], 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 h ard way', i.e. with a f ew bibl iographic
and bibliometric tools but without the kind of automa-
tion implied above. For the record, the main tools used
(see a review in [2]) were Web of Knowledge a nd Scopus
for literature and citation searching, supplemented by
Google Scholar. Some use was also made of ARROW-
SMITH [6, 19, 20] and GOPubMed [21], as well as
various workflows in t he Taverna environment [22-26],
includ ing the BioAID_DiseaseDiscovery workflow http://
www.myexperiment.org/workflows/72 writt en 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 t o water is also accompanied by the
production, at considerable rates (ca 14% of O
2
reduced), of partially reduced forms of dioxygen such
as hydrogen peroxide and superoxide (e.g. [27-45]).
These 1- and 2-electron reductions of O
2
are necessarily
exacerbated when the redox poise of t he b-type
cytochromes is low, for instance when substrate supplies
are in ex cess o r w hen the terminal electron acceptor O
2
is
abnormally low due to hypoxia or ischaemia. Various
other oxygenases, oxidases and peroxidases can also lead
direct ly to the production of such 'reduc ed' forms of
dioxygen in vivo (e.g.[46-48]),withH
2
O
2
from xanthine
oxidase being especially implicated in ischaemia/reper-
fusion injury (e.g. [47, 49-54]). These molecules (per-
oxide and superoxide) can cause or contribute to various
kinds of oxidative stress. However, this is mainly not in
fact because they can react d irectly with tissue compo-
nents th em selves, since they a re
compar atively non-
toxic, cells have well-known means of dealing with them
[55], and they are even used in cellular signalling (e.g.
[56-60]). 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 [61-63].
The themes of this r eview are thus (i) that it i s this
combination of poorly ligan ded iron spec ies, 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 chang es leading to a variety of
disease manifestations, and in particular those where
the development of the disease is manifestly progres-
sive and degenerative.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 2 of 79
(page number n ot f or citation purposes)
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 30
th
, 2008, with
some updates being added following the refereeing process.
Some relevant chemistry of iron and
reduced forms of oxygen
While superoxide and peroxide are the proximate forms
of incomplete O
2
reduction in biology, a reaction
catalysed by the enzyme superoxide dismutase [65]
serves to equilibrate superoxide and peroxide:
2O
2
-
+2H
+
Æ H
2
O
2
+O
2
(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
)
Fe(II) + H
2
O
2
Æ Fe(III) + OH
-
+OH
(2)
Superoxide can also react with ferric iron in the Haber-
Weiss reaction [67] to produce Fe(II) again, thereby
effecting redox cycling:
O
2
-
+ Fe(III) Æ O
2
+ Fe(II) (3)
Figure 1
An overview of this article, set out in the f orm of a 'mind map' [64].
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 3 of 79
(page number n ot f or citation purposes)
Ascorbate can replace O
2
-
within the cell for reducing
the Fe(III) to Fe(II) [68]. F urther 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
[69-73], Fe(II) and certain Fe(II) chelates react w ith lipid
hydroperoxides (ROOH), as they do with hydrogen
peroxide, splitting the OO 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 [74-76] and to the
polymerisation and denaturation of proteins [77-79]
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 r adical and
other 'reactive oxygen species' (ROSs) are known [ 43,
82-89], and include 8-oxo-guanine [90-94], 8-hydroxy
guanine [95], 8-hydroxy-2'-deox y-guanosine [96, 97],
8-oxo-GTP [98 ], 4-hydroxy-2-hexenal [99], 4-hydroxy-
nonenal [100], 4-hydroperoxy-2-nonenal, various iso-
prostanes [101-107], 7-keto- cholesterol [108], many
other cholesterol derivatives [109], malondialdehyde
[110], neopterin [111], nitrotyrosine [112-115] 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 th em either to molecules
identified in persistent curated datbases [119] such as
ChEBI [120] or KEGG [121], or better to describe t hem
via database-independent encodings such as SMILES
[122] or InChI [123-128] strings. (There are other
oxidative markers that may be less direct, such as the
ratio of 6-keto-prostaglandin F1a to thromboxane B2
[129], but these are not our focus here.)
Overall, it i s in fact well e stablis hed 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, 1 30-139]), 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,
140-143]. While the role of ROSs generally in these
processes has been widely discussed, the general
recognition of the impor tance of inadequately liganded
iron in each of them has perhaps been less than fully
appreciated. One of our tas ks 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 ligan ds, though we absolutely
stress that it is the
exact speciation and liganding that
determines the reactivity of 'iron' in catalysing r eactions
such as that of hydroxyl radical formation, and indeed its
bioavailability generally inadequate liganding of iron
in the r equired 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
2O
2
-
+2H
+
Æ O
2
+H
2
O
2
(4)
and by catalase
H
2
O
2
Æ H
2
O+1/2O
2
(5)
These to gether, were th eir activi ty in the relevant
locations sufficiently gr eat, 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, 145-147]. 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, 149-154] leading to
nitrotyrosine [112], or nitro-fatty acid [155, 156] or
protein cystein nitrosylation [157, 158 ] being a common
means of t heir detection downstream. Other toxic
products of the reactions of NO include NO
2
,N
2
O
3
,
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 irre versible [161], that they are consequently
age-related, and (see [162-165] 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. Consequen tly, and because these reflect
fundamental physicochemical properties of such ions,
the problems of both solubility and toxicity were faced
by bacteria (and indeed fungi [166-169]) long ago in
evolution, and were solved by their creation and
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 4 of 79
(page number n ot f or citation purposes)
excretion of (mainly ferric-)iron chelators known as
siderophores [170-189] (and for haemophores see
[190]). These typically have extremely tight binding
constants (K
f
>10
30
[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 o ctahedral complexes with
ferric iron and typical ly employ hydroxamates, a-
hydroxycarboxylates and catechols as extremely effective
Fe
3+
ligands [182]. Since bacterial growth requires iron,
it is unsurprising that siderophores are effectively
virulence factors ( e.g. [174, 193-196]). 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, desfer riox amine 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 dete cted
nearly 30 years ago [198, 199 ] (see also [200- 205]). As
noted by Kaplan [206], "a discovery that m ammals
produce siderophores would lead to an epochal change
in the parad igm of mammalian iron ho meos tasis." 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 2mgofironperday
[210, 211]. The care ful 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 s ince 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,
212-241]), including a series on 'iron imports'
[242-248], and for our present purposes ( Fig 2) mainly
involves the intestinal (mainly duodenal) uptake of Fe
(II) (produced from Fe(III) using a luminal ferrireduc-
tase) 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 th e heme carrier protein-1 (HCP1) [251] and
it is thereby internalized, while the iron in heme is
liberated by heme oxygenase-1 ( HO1) [25 2-254]. Haem
is synthesised in many tissues, e specially liver and
erythroid cells [255]. Vesicular routes of intestinal
transfer may also occur [25 6, 257]. Low MW cytoplasmic
chelators such as citrate can bind iron fairl y weakly and
thereby contribute to a labile iron pool (LIP) in the
cytoplasm and especially the lysosomes and mitochon-
dria (see [258-262]), while ferritin [263] too can bind
cytoplasmic iron (via a chaperone [264]) and is seen as a
good overall marker of iron status [265-267]. Iron(II) is
subsequently exported through the basolateral mem-
brane of the enterocyte by ferroportin-1 (FPN1)
[268-270]. 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 t he peripheral tissues, with significant
transfer of iron from transferrin occurring via t he
transferrin receptor [273].
'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 ce llular 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, 278-282], even
though one of the products of its reaction is Fe that must
eventually be liganded (or e.g. incorporated into
Figure 2
Schematic overview of the main elements considered
to participate in mammalian iron metabolism.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 5 of 79
(page number n ot f or citation purposes)
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://ww w.proteinatlas.org/ and see later). It
also ignores any discussion on the genetic regulation of
iron metabolism (e.g. [285-288]), 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, 289-327] that acts in part as a negative regulator of
iron efflux [328] by causing the internalisation of
ferroportin [329-333]. 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 6 0 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 int o the circulation. In view of the inter-
reactivity of superoxide and iron this could be antici-
pated 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. How-
ever, hepcidin is overexpressed in inflammatory disease
and is an early inflammatory marker [245, 341-3 45]. 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 metabo-
lism, it is going to be especially important to understand
where it is expressed; f ortunately, such studies are
beginning to emerge [350].
Overall there is a c omplex interplay b etween 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 [35 2, 353]. Specifically, lowered hepcidin
causes hyperferraemia. Hepcidin is also activated by p53
[354], and may play a rol e in the degradation of
atherosclerotic plaques [355]. Another recently discov-
ered 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 b
barrel that defines a calyx, or cup-shaped structure,
enclosing the ligand binding site.
NGAL neutrophil gelatinase-associated l ipocalin is a
21 kDal glycoprotein first isolated by Kjeldsen and
colleagues in 1993 [358]. Synonyms i nclude lipocalin 2,
siderocalin, Lcn2, a2-microglobulin-related protein or
neu-related lipocalin (in rats) [359, 360] and (in m ice)
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 t he hypoxia-mediated regulati on of hepcidin
can have especially d amaging effects.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 6 of 79
(page number n ot f or citation purposes)
24p3 or uterocalin [361]. Although lipocalins are well
knowntobeinvolvedinthesequestrationandtransport
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 purifie d recombinant NGAL from a parti-
cular strain of E. coli and found that its structure
contained a negativel y charged f erric 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 [3 63-367]). 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 met al ion and in the complex
decrease its degradation [371 ].
The findi ng that NGAL was one of the most highly
expressed proteins following ischaemia-reperfusion
injury in kidney cells [ 372-374], and prognostic of
kidney damage long before the more traditional marker
creatinine was raised significantly, has led to consider-
able i nteres t in this protein, especially as a marker of
renal injury [375-389], 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 side-
rophore payload) and mediate its intracellular uptake
[392]. Oxidative stress can also induce its expression
[393], and it is protective against i t [394].
Exogenously administered NGAL also markedly upregu-
lates 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 produc-
tion 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 hydroxama te- 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 [362-367] and that bacteria can 'evade' this by
synthesising siderophores that siderocalin cannot bind (e.g.
[186, 187, 399-401]), 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 sequestrat-
ing 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, concentra-
tions of NGAL can increase to levels, with concentrations
approaching 2030 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 [406-409 ] and in
cancer [410-412].
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
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.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 7 of 79
(page number n ot f or citation purposes)
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
pertinentliteraturethatweherehighlightasthefocusof
this review.
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 m ortality in pregnant women [414]. The chief
clinical manifestations at time of diagnosis are a raised
blood pressure (hypertension) [415] and proteinuria,
together with raised creatinin e, co nsi stent with t he
reversible existence (since it is rel ieved upon delivery of
the baby) of rena l impairm ent. How ever, prognosti c
markers that might manifest early in pregnancy are
lacking, and would be highly desirable. There is wide-
spread 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 [417-423].
Since preeclampsia-like syndromes can be induced in
pregnant animals by surgical restriction of the uteropla-
cental 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 [425-489], while some have noted t he importance o f
iron status [70, 133, 450, 490-511], and so far as is
known the transporters of iron in the placenta are similar
tothoseinothercells[512].Oxidativestressofthistypeis
of course inflammatory in nature and inflammation is
observed in PE [472, 476, 484, 486, 513-519]. W e 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
approp riat e removal by liganding/chel ation or otherwise
of these ions would prove of therapeutic benefit. (Iron
status has also been implic ated in other pregnancy and
neonatal disorders [520-524].) 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 t hat this does of course lead
to oxidative stress [ 528, 529].
Oxidative stress is caused both by the init ial rate of
production of superoxide and the rate of their conver-
sion 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 h igh altitude.
Erythropoietin may be a marker for oxidative stress in
pre-eclampsia [531].
Regarding the second stage, predictions include that PE
should be m ore common in those suffering from
diseases of iron metabolism. Although such mothers
are of course less well apriori, this prediction is borne out
for a-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, 53 5], with consequent
and inevitable iron dysregulation.
Another prediction is then that hepcidin should be
changed in pre-eclampsia. Although no serum measure-
ments 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 u terus [361, 537-539]. 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. [541-54 5]), and also
predispose towards PE. In a similar vein, various types of
pregnancy-related intrauterine growth restriction predis-
pose towards diabetes in later l ife [546, 547], pointing
up the progre ssive nature of these syndromes. Metabolic
biomarkers for the one can thus be predictive of the
other [548], consistent with a common cause. Certainly
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 8 of 79
(page number n ot f or citation purposes)
ROSs are known to play a substantive role in both
insulin resistance [549- 556] and in a variety of diabetic
sequelae [95, 55 7-559], 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)g,
may also act by decreasing ROS production (e.g.
[561-565]), and are even active aganst cerebral isc haemia
and stroke [566-569]. As with most if not all of the other
diseases we review here, studies of pro-inflammatory
markers (such as TNF-a, IL-1 and C-reactive protein
[570]) during the development of diabetes show its
aetiology to be inflammatory in nature [553, 571-590].
Iron 'excess' is also a known feature of gestational
diabetes [591-593], and is a clear risk factor for the
disease even in 'normal' populations [594-603], and
indeed diabetes is a classical cons equence of iron
overloading as seen in hereditary haemochromatosis
[604]. Serum ferritin and body iron stores are strongly
associated with diabetes [603, 605-609], 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 d iabetes [612, 613]. Lowering
iron improves insulin sensitivity [598, 614], and
metallothionein is also protective [615-619]. 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 metabo-
lism [598 ]. Iron is also strongly i mplicated in non-
alcoholic steatohepatitis, considered an early marker of
insulin r esistance [622-624]. Well-known diabetic com-
plications 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 [627 -631]. Thus, serum
ferritin is also relate d 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 t oo are closely intertwined with inflammation
[575, 581, 587, 638], that is of course stimulated by
ROSs whose generation i s 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 i ncrease 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 chan ging environ-
ment interacting w ith 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. [641-643] 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, 647-664],
not least because adipocytes produce and release various
adipokines including pro-inflammatory cytokines such as
IL-6 and TNF-a [575, 649, 650, 665-671]. It is likely that it is
the
combination of overfe eding-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
2a
, 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.
Hypertension
As well its significance in pre-eclampsia (see above),
hypertension is a well known risk f actor for many
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 9 of 79
(page number n ot f or citation purposes)
cardiovascular and related disease (e.g. [677]), and there
is considerable evidence that its underlying cause is
inflammatory in nature [678-686], is related to the
metabolic syndrome and obesity (e.g. [648, 687-691]),
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 [695-698]
(and see also [699, 700]). (In this s ense, 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
[701-703], and that reducing homocysteine (e.g. by
folate supplementation) in the absence of lowering iron
has shown no cli nical b enefi t 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 cardio-
vascular, coronary a rtery disease and related diseases.
Indeed kidney disease is well established as a risk factor
forcardiovasculardisease[706-708](andindeedstroke
[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 o f inappropriately chelated iron in
cardiovascular diseases too. There is no doubt that the
iron-mediated causal chain of ischaemia Æ (su)peroxide
Æ OH
radical formation occurs during the develop-
ment of heart disease, especially during reperfusion
injury [710-713], and suitable iron chelators inhibit this
[714, 715] (see also [716, 717], and f or 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 [727-729],
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
[731-762] characterized by the accumulation of both
oxidised lipids and various fibrous elements in arteries,
often as plaques [763, 764]. Iron and oxidised lipids a re
both found in atherosclerotic lesions [141 , 76 5-777],
and iron depletion by dietary or other means delays this
[778-782]. There is a correlation between iron status and
atherosclerosis [766, 776, 783-794], evidently c aused in
part by the known ability of poorly liganded i ron to
effect lipid [76 5, 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 ir on chelati on
improves it [797-800]. 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 ti ssue; this is not a small effect . (Iron,
Figure 5
Role of inflammation caused by hydroxyl radical
formation in the interactive development of obesity,
the metabolic syndrome and diabetes.Interventionat
multiple steps is likely to be most beneficial i n alleviating this
kind of progression.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 10 of 79
(page number n ot f or citation purposes)
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 m any populations (e .g. [805-807]) 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 b
2
-integrin
leukocyte function antigen-1 (LFA-1) [80 8, 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-lowe ring ability via the inhibition of HMG-
CoA reductase (see e.g. [163, 281, 650, 807, 808,
810-840]), and dif ferent statins can cause a variet y 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 arg uably wel l explained,
especially within the context of the present review, via
their additional anti-inflammatory e ffects [817, 819,
843-863, 163, 807, 823-825, 830, 836, 837, 864-892],
acting upst ream of the nuclear trans cripti on factor NF-B
(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 i nvolved in oxidative stress. Serum
ferritin is considered by some to be the most reliable
marker of iron status in general [894], although it i s not
well correlated with iron dist ribution 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 [896-899].
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, 905-914]. 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 v arie ty of ot her studies have sho wn the beneficial
treatments in stroke models of anti-inflamm atory and
antioxidant treatment, i .e. treatments that lower the
amountofROSs(e.g.[915-922]),aswellasof
preconditioning [923]. Given its r ole in iron metabo-
lism, it is of considerable interest that erythropoietin
also seems to be very effective in protecting against brain
ischaemia/reperfusion injury and stroke [924-939], by a
mechanism independent of erythropoiesis [940-942],
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, 944-957] such as
Alzheimer's dis ease (e.g. [958-973]), 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, 982-985, 962, 986-989, 950, 990-1021, 917,
1022-1034, 278, 1035-1055, 43, 1056-1059, 285,
1060, 972, 1061-1065, 141, 975, 1066-1087 ].
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]. Kostof f [ 1090] has
used a very interesting literature-based discovery
approach to highlight the role of oxidative stress i n the
development of Parkinson's disease.
Other papers highlight the role of iron in multiple
sclerosis [899, 991, 1091-1098] and in prion diseases
[967, 1099, 1100]. However, a particularly cle ar 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).
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 11 of 79
(page number n ot f or citation purposes)
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 [1102 -1109], 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 [1114-1123]. 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,
1124-1127]. As well as t he 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 o f iron dysregulation, and in
which suit able chelation may be benef icial [1130, 1131]
(but cf. [1132]).
ROSs are undoubtedly involved in FA [1133, 1134],
specifically via Fenton chemistry [1135, 1136], since the
attenuation of H
2
O
2
production (but not of superoxide)
[1110] ameliorates the disease [1137]. The deficit in
frataxin causes both an increase in ROS (H
2
O
2
)
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 i t is the combination
of free iron with H
2
O
2
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 BCS1 L 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 p redisposition [1143 ],
save for a very clear association with defects in a Cu/Zn
superoxide dismutase [33, 1144-1146] that can
obviously lead to an increase in the steady-state levels
of superoxide (and hence hydroxyl radical formation).
There i s 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 defi cie ncies than in humans , th ough i ron
chelation therapy does not seem to have figured heavily,
and it is recognised t hat combination therapies might
offer better prognoses [114 9].
Aging
Aging or senescence is defined as a decline in perfor-
mance and fitness with advancing age [1150]. Iron stores
tend to increase with age [1151-1155], 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, 1162-1180] recognise t he relationship between
progressive inflammation, cellular damage and repair
and the higher-level manifestations of the aging process,
and ('the fre e 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, 1 182-1206]). Needless to say, not least because of
the low stea dy-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 metabo-
lism, 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 S ystems Biology
section, below). The iron content of cells also increases
as cells age normally [1209]. As many diseases increase
with age, probably via mechanisms h ighlighte d herein,
treating aging can thereby t reat 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 c hanges, referred to as frailty [1210], in
which ROSs have also been strongly implic ated. Indeed,
there are many parts o f physiol ogy and me tabolism 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 i ncrease 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
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 12 of 79
(page number n ot f or citation purposes)
metabolism as a major part of ROS-induced injuries that
may accelerate the aging process.
Longevity
Although aging and longe vity 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 cert ainly long-
evity 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 s pecies [1218-1 223] although possibly
not H. sapiens [1180]), and appears to act at the root of
the processes involved [1224]. Caloric r estriction appears
to be associated with a considerably lowered rate of
production of ROSs and accrual of ROS-induced damage
[1207, 1225-1228], and this is to be expecte d 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 O
2
to form peroxide and super-
oxide. It is therefore also of great interest that caloric
restriction also b enefits iron status [1230] and that it is
this improved iron status that in part promotes longevity
[1231]. Caloric restriction has also been shown t o effect
a differential stress res ponse 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 Drosop hila [1191, 1233-1235] w hile
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.Mutantswitha
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 inappropri-
ately chelated iron, that causes a 'double whammy'.
Indeed, iron, copper and H
2
O
2
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 art hritis (RA) [36,
1241-1246]. What is known of the r ole of iron
metabolism? G enerally an overall low iron status
anaemia is a char acteris tic of r heumatoid arthriti s
[1247-1251], whereas by contrast iron is elevated in the
synovial fluid of arthritic joints [12 52-1255]. This
suggests a significant derangement of iron m etabolism
in RA as well, and a mechanism [1256-1259] in which
elevated superoxide liberates free iron from ferritin in
synovial fluid (and elsewhere [1260]), thereby catalysing
further t he 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].
One interesting feature of RA is that in 75% of women it
is strongly ameliorated during pregnancy [1265-1267];
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' [1268-1270], that is often associated with
Figure 6
CatalysisandautocatalysisintheHaber-Weissand
Fenton reactions leading to the production of the
hydroxyl radical, including the liberation by
superoxide of free iron from ferritin.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 13 of 79
(page number n ot f or citation purposes)
iron deficiency and pregnancy. Serum transferrin recep-
tor seems to be a rather sensitive measure of this iron
deficiency [273, 1271, 1272]. There are relationships
with other syndromes that we discuss h ere, 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 E ryt hematosus )
Lupus, or Systemic Lupus Erythematosus (SLE) [1275,
1276] describes a syndrome, somewhat related to
arthritis and rheumatism, of broadly auto-immunologi-
cal or inflammatory origin, and with a large variety of
manifestations (e.g. [1 277-1285]), 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 ca uses), and there is
certain some evidence for t his [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 m etabolism [1290], something that has also
come up in relation to the s tatins 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 gene ration [1297] catalysed by
iron [276, 1298-1300].
Inflammatory bowel diseases (IBD)
By definition, IBD such as Crohn's disease and ulcerative
colitis are inflammatory diseases, and while the inflam-
mation and ROS production are well established here,
their origins are somewhat uncertain [1301-1304]. They
are frequently accompanied by anaemia, implying a
derangement in iron absorption and/or metabolism
[1305, 1306], and very probably absorption [ 1307,
1308]. T he anaemia may be monitored by ferritin and
transferrin receptor levels, and its correction is possible
by iron supplementation plus erythropoietin
[1309-1315]. 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 [1317-1320]. 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 pathogen-
esis between AMD and atherosclerosis. Retinal iron levels
increase with age [1322], iron is significantly implicated in
AMD [1323-1329], 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 [1332-1334] against VEGF
(pegaptanib) or a monoclonal antibody (ranibizumab)
have shown significant promise in the treatment of macular
degeneration [1335-1340]. Plausibly a combination ther-
apy with one of these plus a suitable iron chelator might be
even more effective.
Psoriasis
Psoriasis i s an anflammatory disease in which the
production of free radicals and ROSs are strongly
implicated [1341-1343]. Here too there is clear evidence
for the involvement of a deranged iron metabolism
[1343-1345]. 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, char-
acterised 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 [1352-1358]. Iron overload is well known to
lead to hepatotoxicity [1359-1362] 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 l ung diseases
Chronic obstructive pulmonary disease (COPD) is a
progressive and chronic disease which is characterised by
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 14 of 79
(page number n ot f or citation purposes)
an apparently inexorable decline in respiratory function,
exercise capacity, and health status. It is also charac-
terised by periods in which the symptoms are consider-
ably exacerbated [1368-1371]. Such an acute
exacerbation of COPD (AECOPD) is d efined [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 i nflammation and ROSs
in both the 'stable' and 'exacerbated' stages [1374-1381].
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 develop-
ment 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 l ung diseases in which ROS and iron have been
implicated include Adult Respiratory Distress Syndrome
[1387-1390] and cystic fibrosis [139 1].
Smoking
Tobacco smoke contains many unpleasant and carcino-
genic 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. [1392-1395]). 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 sub-
stantial contributor as well (see e.g. [1373, 1384,
1396-1400]).
Cancer and oncology
In addition to the issues of smoking, the development of
cancer can certainly contain an inflammatory compo-
nent (e .g. [1401-1425]), 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 [1428-1430] and e.g.
bowel cancer is probably initiated by chronic inflamma-
tion.) Given that cells require iron, restricting its supply
can also limit the growth of cells, including tumour cells
[1431-1442]. Conversely the iron carrier NGAL is
overexpressed in tumours [410, 411, 1443], a process
mediated via NF-B [1444 ] (and see later). Furthe r, the
roles of iron, not least in t he mutagenic effects of metal-
catalysed Fenton chemistry, are also of significance in
promoting oncogenesis [1445-1461 ]. 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 [1463-1475], while there seem to be com-
plex relations between the likelihood of apoptosis and
the differential concentrations of superoxide and H
2
O
2
[1476, 1477]. Overall, it is becoming increasingly widely
recognised that anti-inflammatory agents have a role to
play in the treatment of cancers; we woul d s uggest that
iron chelat ion may be a usefu l component of such
treatments.
Malaria
Just as do tumour cells, the malarial parasite Plasmodium
falciparum re quires considerable iron for growth, and
there is evidence that lowering the amount of available
iron provides a promising route to antimalarials (e.g.
[1478-1494], but cf. [1495]). Note in this context that
iron-catalysed radi cal forma tion is also significantly
involved in the antimalarial (i.e. cytotoxic) mode of
action of artemisinin [1494, 1496-15 04], 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 CO
2
fixation [1506-1509 ], its
excess can in some c ircumstances [ 1510] correlate with
infectivity or virulence (see above and e.g. [180, 193,
1493, 1511-1531]) , 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, 15 35], 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 t hat one consequence of bacterial
infection (sepsis) can be septic shock , that this can be
mimicked by the Gram-negative bacterial outer mem-
brane component LPS (lipopolysaccharide), and that in
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 15 of 79
(page number n ot f or citation purposes)
the worst cases this leads via multiple organ failure to
death. However, whatever LPS does it is quite indepen-
dent of the present of viable (i.e. growing or culturable
see [1537, 1538]) bacteria as the same phe nomena
leading to multiple organ failure (MOF) are seen
without infection [1539, 1540]. Consequently, the
recognition of a series of symptoms contingent on this
initial i nflammatory response has led to the development
of the idea of a Systemic Inflammatory Response Syndrome
(SIRS) [1541-1551] 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 hyperproducti on of ROSs [1546, 1551,
1555-1570]. Circulating free iron is raised in sepsis and
related conditions [1400, 1571-1573]. Direct assays of
oxidant induced cell death indicate that most 'free' iron is
concentrated in lysosomes [1574-1577], that its decom-
partmentation is substantially involved [1570], and that its
chelation can thus prevent cell death [1578-1581].
Many circulating inflammatory factors have been identi-
fied 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 pre-
venting septic shock [1585-1587], and a variety of
suitable antioxidants have shown potential here [1573,
1588-1591], notably in combination with i ron chelators
[1592, 1593] (and see a lso [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 acces-
sible t o 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
reduc-
tion 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 O
2
,
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 phenom-
enon that explains the otherwise possibly puzzling
observation of the oxidant-induced reduction of respira-
tory chain components (see e.g. [1600, 1601]). Conse-
quently, 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 O
2
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 pre-
ferred 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, 1602-1606]. In a similar vein, 'free' or inade-
quately 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-b 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, 1613-1631] 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 describe d above, many investi-
gators have considered the use of known antioxidants
such as vitamins C (ascorbate) and E (a-tocopherol) in
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 16 of 79
(page number n ot f or citation purposes)
preventative therapy. Although there have been some
successes (e.g. [1633]), the results have generally b een
decidedly mixed, with little clinical benefit (or even
actual disbenefit) following from their administration
[692, 1588, 1634-1639], e .g. f or ALS [1640], athero-
sclerosis [106, 764], cardiovascular disease [1641 -1645],
neuroprotection [1646], macular degeneration [1647],
pre-eclampsia [1648-1652], 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 t his r eview, 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 inappro-
priately 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
[1661-1663].) Thus any predictions about the utility or
otherwise of
antioxidants need t o take into account the
amount of 'free' iron present. In partic ular , we would
suggest that future trials of this type might beneficially
include appropriate iron chelators, whether alone or
with
antioxidants.
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 o ne
would wish [43]. This is because the OH
radical is so
reactive that its p roduction 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
gen erated. The abili ty 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 O H
itself,aswellasbyothermechanisms
[1664]. This said, there is little doubt that iron chelators
can be highly pro tective, and it is man y 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]"Oxygenligands prefer Fe(III); thus, the
reduction potential of the iron is decreased. Conversely,
nitrogen and sulfur liga nds 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
coordinationnumber 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
[1666-1669] 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 vi a 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 F e
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 i s a
very effective antioxidant through i ts ability to bind Fe."
By cont rast, 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 i n 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
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 17 of 79
(page number n ot f or citation purposes)
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-catecho-
lates, 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 thoseapprovedandusedclinically
A number of reviews (e.g. [1437, 1438, 1672-1678])
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 a ctivity, 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 com-
pounds will beneficially bind both FeIII and FeII u nder
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 sidero-
phores compose d of alternating dicarboxylic acid and
diamine u nits. linked by amide bonds. They are
produced by many Streptomyces species [1680]. Desfer-
rioxamine B is a linear (acyclic) substance produced
(industrially) by the actinobacterium Streptomy ces 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 poten-
tial. 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 trans-
porters [1682-1685]) 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 app roval 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
Table 1: Comparison of the main av ailable iron c helat ors to an ide al chelation drug (modified from [2469])
"Ideal chelator" Deferoxamine Deferiprone Deferasirox
Route of administration Oral Parenteral, usually
subcutaneous or
intravenous
Oral Oral
Plasma half-life Long enough to give
constant protection from
labile plasma iron
Short (minutes); requires
constant delivery
Moderate (< 2 hours).
Requires at least 3-times-
per-day dosing
Long, 8 16 hours; remains
in plasma at 24 h
Therapeutic index High High at moderate doses in
iron-overloaded subjects
Idiosyncratic side effects are
most important
Probably high in iron
overloaded subjects*
Molar iron chelating
efficiency; charge of i ron (III)
complex
High, uncharged High (hexadentate); charged Low (bidentate); uncharged Moderate (tridentate);
uncharged
Important side effects None or only in
iron-depleted s ubjects
Auditory and retinal
toxicity; effect s on bones
and growth; potential lung
toxicity, all at high doses;
local skin reactions at
infusion sites
Rare but severe
agranulocytosis; mild
neutropenia; common
abdominal discomfort;
erosive arthritis
Abdominal discomfort;
rash or m ild diarrhoea
upon initiation of therapy;
mild increased creatinine
level
Ability to chelate
intracellular cardiac and
other tissue iron in humans
High Probably lower than
deferiprone and deferasirox
(it is not clear why)
High in clinical and in in vitro
studies
Insufficient clinical data
available; promising in
laboratory studies
*Nephrotoxicity that has been observed i n non-iron-loaded animals has been minimal in iron-overloaded hum ans, but effectiveness is demonstrated
only at higher end of tested doses, a s discussed in [1693].
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 18 of 79
(page number n ot f or citation purposes)
common, viz. desferal (deferoxamine), ferriprox (L1 or
deferiprone) and exjade (ICL670 or deferasirox). (Dex-
razoxane, a hexadentate chelator, is also marketed
[1686].)
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 bioavail able 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 50120 mg/kg/day appear to b e effective
in bringing patients to negative iron balance" [1687]. It
can have s omewhat better properties than desferal
[1688]. Finally, Exjade (ICL670) (deferasirox)
[1689-1702] 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?disclaim er=Y. The recommen ded 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 defe riprone to deferasirox there
has been a noti ceable improvement in the general
properties of iron chelators, although there are few
published data on the quantitative s tructure-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 forclinicaluseforother
purposes, but that also happen to be iron chelators
The high investment of time, money and int ellectual
activity ne cessary 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 c an yet serve for novel indications (e.g.
[1704-1708]). 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 h as indeed
enjoyed some success in this role. However, clioquinol
toxicity has been reported if it is used over a n 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 assump-
tion that lipophilicity or log P is s ufficient 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, th e iron- chelating 8-hydroxy
quinoline motif contained in molecules such as clioqui-
nol is also pr esent in the tryptophan catabolite
xanthurenic acid (Fig 7), and it is likely that transmem-
brane 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
investigat ors have sough t to develop their own iron
chelators th at might exhibit the desirable properties
listed above. One class of molecule includes
Figure 7
Some iron chelators that are in clinical use (left hand
side) or that have been proposed.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 19 of 79
(page number n ot f or citation purposes)
isonicotinylhydrazones. Thus, pyridoxal isonicotinyl
hydrazone (PIH) [1672, 1715-1720] is a promising
molecule (also proposed in anti-cancer therapy),
although it is hydrolysed both in v ivo and in vitro
[1721]. Other analogues include salicylaldehyde (SIH)
[1722] and 2-hydroxy-1-napthylaldehyde (NIH) isoni-
cotinyl hydrazones. PIH was disclosed before being
patented, and is thus seen as having no pharmaceutical
(company) interest. Various other derivatives are there-
fore being considered [1035, 1462], including pyrazi-
nylketone isonicotinoyl hydrazones [1723].
A variety of 8-hydroxyquinolines (8HQs) [1724] have
been considered, although as with other bidentate and
tridentate ligands that cannot necessarily ef fect complete
liganding of iron there is always a danger that
inadequate concentrations might be pro-oxidant (e.g.
[1725, 1726]). One mole cule, VK-28, combines various
pertinent moieties and has shown some promise in the
treatment of neurological disorders [1727-1730]. This
strategy of combining drug elements that can hit
multiple targets ('polypharmacology' [807,
1731-1734]) has m uch to commend i t, including on
theoretical grounds, and we discuss these in the section
on systems biology below. Another 8HQ that has el icited
interest is O-trensox [1431, 1735-1742].
Other ligands or motifs that might be considered include
di-2-pyridylketone-4,4,-dimethyl-3-thiosemicarbazone
(Dp44mT), t hat 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 m ore effective than is DFO
[1750-1752]. Poly-hydroxylated 1,4-naphthoquinones
occur as sea urchin pigments and have shown protective
effects [1753].
Continuing the theme of polypharmacol ogy, R-(a)-
lipoic acid [1754-1757] 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, 1760-1767]).
Many problems remain, such as bioavailability, mis-
dosing [1768] (t oo 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 thera-
peutic benef its in Alzheimer's [1674, 1710, 1769-1771],
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 i nteresting strategy is to devise
chelators that are only activated by oxidative stress
[1760, 1776-1779]. 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-hydroxyquino-
line [1740] derivatives that are effective bidentate iron
chelators and that seem to sh ow con siderable promise in
the treatment of a variety of neurodegenerative diseases
[1037, 1727-1729, 1780-1784] (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 direc tly [1785],
a fact that a lso 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 eff ects if used carefully [1 483,
1762, 1786, 1787]
Interaction of xenobiotics with iron metabolism
As Cherny and colleague s 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 metabo-
lism. 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 symp-
toms such as hearing loss; this is reversed by iron
chelators [1788, 1 789]. E xisting d rugs 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 t hat 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 [1794-179 9] (whose
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 20 of 79
(page number n ot f or citation purposes)
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 F e(n) that it contains to catalyse
hydroxyl radical production [1469, 1475], while carci-
nogenic 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 t heir chelation of iron. Indeed, polyphenolic
compounds, many of which have known health benefits
[1804-1813], are widely used as food antioxidants [1814,
1815]. There is of course considerable epidemiological
evidence for the benefits of consuming f ruit and
vegetables that are likely to contain such antioxidants
(e.g. [1816-1819]), 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'dietthatareconsideredtobebeneficial
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 ant hocyanidins) [1821 -1828] so implicated
may also a ct to chelate iron as well [1073, 1829-1843].
This is reasonable given that many o f these polyphenols
and flavonoid compounds [1821, 1844-1853] have
groups such as the catechol moiety that are part of the
known iron-binding elements of microbial siderophores.
Examples include flavones s uch as q uercetin [914, 1813,
1829, 1854-1864], rutin [1829, 1857, 1858, 1865, 1866],
baicalin [1860, 1867], curcumin [1813, 1868-1872],
kolaviron [1873], flavonol [1874], floranol [1875],
xanthones such as mangiferin [1876-1879], morin
[1876], catechins [107 3, 1807 , 1838 , 1 854, 1 880, 18 81]
and the aflavins [1882], a s well as procyanidins [1835,
1883] and melatonin [1628, 1 884-1887 ]. However, the
celebr ated (trans- )-resveratrol molecule [1888-1902] 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 stre ss is implicated [1825,
1903-1906]. For instance in stroke and related neuronal
aging and s tress conditions, preventative activity can be
found in blueberries [1907-1913] (and see [1914]),
Ginkgo biloba extract (EGb 761) [1910 , 1915, 1916],
grapes [1917], green tea [1807 , 1918- 1921], Mangifera
indica extract [187 9], 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,
1923-1925], there are significant problems with bioa-
vailability [1926, 1927], although the necessary mea-
surements are starting to come forward [1 804, 1809,
1926-1932]. There is now increasing evidence for the
mechanisms with which these dietary c omponent s and
related natural products and derivatives (often with anti-
inflammatory, anti-mutagenic or anti-carcinogenic prop-
erties) interact with well r ecognised cellular signalling
pathways (e.g. [1402, 1935-1944, 1895 , 1945-1952,
1896, 1 410, 1953-1960, 1413, 1961-1981 , 1913, 1982,
1900, 1933, 1983-1990]).
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 me diated via the inflamma-
tory signalling pathways involved in 'innate immunity'
[900, 1991-1993]. NF-B is an important transcription
factor, and the NF-B system is intimately i nvolved in this
signalling [588, 672, 719, 1408, 1409, 1454, 1994 -2019 ].
In the NF-B system (e.g. [2020-2024]) (Fig 8), NF-Bis
normally held inactive in the cytoplasm by binding to an
inhibitor IB(oftenIBa). Pro-inflammatory cytokines
such as TNF-a, LPS [2025-2030] 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, IB kinase or IKK. This kinase phosphor-
ylates the IB causing it to be released (and ubiquitinated
and degraded by the proteasome), allowing the NF-Bto
be translocated to the nucleus where it can activate as many
as 300 genes. Simple models of the NF-B system show the
main control elements [2032, 2033] and their synergistic
interaction [2034]. The NF-B system is implicated in
apoptosis [2035, 2036], aging [1199], and in diseases such
as cancer [1405, 1444, 1454, 1808, 2037-2040], arthritis
[2040-2043] and a variety of other diseases [2044].
Antioxidants such as vitamin E [552, 2017] and melatonin
[2045-2049] are at least partially protective. Oxidative
stress seems to act upstream of IKK [2014], on IBa
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-Bpathwaystoo
[819, 883, 2052-2060]. A considerable number of inhibi-
tors of the NF-B system exist [2055], many exhibiting
cross-reactivity [1734].
The induction of NF-B by ROSs appears to involve a
coupling via the glutathione system [2007, 2035, 2036,
2061-2078] (and see also [2079, 2080]).
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 21 of 79
(page number n ot f or citation purposes)
A variety of studies have shown that iron is involved in
these signalling processes [1839, 1996, 2015,
2081-2085], probably again acting upstream of the IKK
[557, 2083, 2086, 2087].
Interestingly, there is interplay between the NF-B
pathway and the regulation of NGAL [404, 405, 2 088,
2089], ferritin [2030] and hepcidin [2090], p resumably
acting as a negative feedback as the cell tries to control
and ligand its free Fe(n) i n 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 conse-
quence, that we ne ed 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, 2094-2102]. Changes in individual reac-
tion 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 [2105-2112].
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 h uge n umber of reacti ons, whether dir ectly or
Figure 8
An overview of cellular signaling using the NF-Bandp38systems. Note that some of the extracellular effectors that
mediate NF-B activation are themselves produced and secreted as a result of the activation, potentially creating an
autocatalytic system.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 22 of 79
(page number n ot f or citation purposes)
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. [2113-2120]), 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. [2123-2127]). 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 representa-
tions 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. [2129-2132].
Indeed, the analysis of the properties and behaviour of
networks is at the core of modern systems biology (e.g.
[2095, 2133-2140]).
A corollary of such considerations is that to decrease the
amount of damage caused by OH
(or any ot her)
radicals we need both to decrease their production and
increase their removal to harmless substances [2141],
and that on general grounds [1706-1708, 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, 170 3, 1714, 2143]. At all events, i t
is undoubtedly the case that the steady-state rate of
production of a molecule such as t he 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 p resent review a large
number of reactions invol ved 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 i s o ne [165] as damage accumu-
lates. 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-
B and p38, there are also anti-inflammatory cytokines.
A particularly interesting exampleisthatoferythropoie-
tin (also discussed above as being protective in a number
of iron-mediated diseases).
Erythropoietin was originally recognized via its role in
erythropoiesis [2145-2147] (hence its name, of course),
but it has become evident that it has many other roles,
and in partic ular it is observed phenomenologically that
erythropoietin (and non-erthyropoetic derivatives) is
protective in a n umber of inflammatory conditions
that accompany many diseases such as those listed
above [940, 942, 2148-2155]. These included
Figure 9
General view of the role of iron, antioxidants and
ROSs in aging and degenerative processes.Someofthe
decay may be ameliorated by lifestyle and dietary means.
Based in part on [1175].
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 23 of 79
(page number n ot f or citation purposes)
cardiovascular disease [721, 722, 2156-2175], stroke and
other related neurological diseases [924-926, 928, 929,
933, 937, 938, 2155, 2176-2204], diabetic neuropathy
[2205], kidney injury [21 73, 2206- 2212], intest inal
injury [2213] and shock (both septic and non-septic)
[1596, 1597, 2214].
The question t hen arises as to how it is doing this
mechanistically, and the proximate answe r is that it (and
other anti-inflammatory agents, e.g. [180 8, 2215, 2216])
seem to act via many of the same signalling pathways as
do inflammatory agent s [943, 2150, 2217-2226]. 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 l iganded iron [2229]
and interact with hydroxyl radical directly [2230-2233].
It is notable that appropriate levels of erythropoietin
appear not o nly to be efficacious but to be safe, even in
pregnancy [2234-2241]. Erythropoietin may it self be a
marker of hypoxia and oxidative stress in pregnancy
[531, 2242-2245], 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 syn-
dromes described above are accompanied by hypoxia,
and this causes levels of the HIF to increase. HIF is a
transcription factor that can activate a considerabl e
number of genes, including VEGF [1951, 2246-2250 ].
In contrast to the constitutive expression of HIF-1a,HIF-
1b protein levels are regulated in response to the cellular
oxygen concentration [2251]. The active HIF is the HIF-
1ab heterodimer [2252]. HIF couples anoxia to innate
immunity via the NF-B system [2253]. In particular,
HIF effects (via hepcidin) the mobilisation o r iron and
can cause the expression of inflammatory cytokines such
as IL-1, IL-6 and TNF- a [2254-2256] 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, 2 257]. H IF also appears to
have a significant role in placental development, and
defective HIF expression may be involved in pre-
eclampsia and intra-uterin e growth retardation [435,
2246, 2258]. Qutub and colleagues provide useful
models [2259, 2260] of HIF activation under a variety
of conditions of iron, O
2
, 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 indivi-
dual 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-a systems in which IL-1 or TNF-a
(oxidative stress) acting on one cell can effect the secretion
of further IL-1/TNF-a 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. [2266-2291]).
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
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 24 of 79
(page number n ot f or citation purposes)
is the prediction that t hey 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 biomolec ules [43, 1 055], at l east
the following appear to be a result of reactions involving
OH
[2294, 2295]: 8-oxo-2'-deoxyguanosine (oxo
8
dG)
[98, 2296, 2297], 8-oxo-7,8- dihydro-2'-deoxyguanosine
[90, 92] and thymine glycol [92, 229 4].
Another set of predictions from the systems biology
perspective [1704-1708, 1731, 2138, 2291, 22 98-23 10]
is that combinations of chemical agents (or manipula-
tions such as those of transcription f actors 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. [2314-2320]) for a recent exam ple see [2321]. The
nonlinear behaviour of biochemical networks also serves
to explain the bell-shaped dose-response curves under-
pinning the hormesis [2322-2326] often observed.
Iron-mediated oxidative stress is arguably the cause of
much of the inf lammation typically observed in biologi-
cal systems, often further mediated via pro-inflammatory
cytokines. Another major prediction that come s 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
thisregardaretobefoundamongthestatins,sinceitis
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 [83 9, 2060,
2328-2340], heart failure [2341 ], pain perception [2342],
lupus and related diseases [1293, 2343], diabetes [877,
2344], rheumatoid art hrit is [866, 869, 890, 2345-2350],
kidney disease [2351- 2353], inflammatory skin disease
[2354], emphysema [2355], ischaemia-reperfusion injury
[2356], stroke [864, 872, 2357-2364], traumatic brain
injury [2365-2367], neurodegenerative diseases
[860-862, 920, 1294, 2059, 2368-2384], neurotoxicity
[2385] and cancer [2386-24 00].
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 th inking about your experime nts 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 m ust, I t hink, als o
admit t hat some things are much mor e nearly cer tain
than others." However, as mentioned above, the huge
volume of scientific activity has in many ways led to a
'balkanisation' of the literatu re [2406] in which scient ists
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,
2407-2416]. The 'digital human' is thus an important
research goal [2410, 2415-2417]. Expression profiling
atlases are becoming increasingly widespread (e.g.
[2418-2423]), and one can anticipate using these
straightforwardly to extend these 'generalised' (sub)
cellular network models in a tissue-specific manner
[2424]. With the ability t o exchange models of bio-
chemical networks in a princ ipled way [2425-2428],
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 m essage of the
present overview. Thus, while iron and me tabolism
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.
[2430-2435]), in which dispersed agents contribute their
different skills to the solution of a complex problem, may
well help this happen effectively. Developments in dis-
tributed workflow technology, such as the Taverna [22-26,
2436, 2437]http://taverna.sourceforge.net/ and the
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 25 of 79
(page number n ot f or citation purposes)
myExperiment [2438]http://w ww.myexperiment.org/
environments, r epresent 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 network s 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 inte rventions. In o rder to
develop and exploit this distributed approach, it will
also be necessary for those generating them t o m ake
their data and metadata available (preferably in a
semantically marked up way), probably as Web Services
(e.g. [24, 2444 -2452]) , and to g ive 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. [2453-2457]) are generating quality data at truly
enormous rates [2458], and innovative but computa-
tionally demanding algorithms are required t o deal with
them (e.g. [2459]). In particular, however, we need tools
for manipulating and visual ising b ioche mical m odels
[2110, 2410, 2429, 2460]. As well a s storing these
models (e.g. as SBML or C ellML [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 architec-
tures such as ONDEX [2464] and Utopia [2465-2468]
are similarly showing considerable promise for integra-
tive 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 t hat 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 Macu-
lar Degeneration; BBB: Blood-brain barrier; ChEBI:
Chemical Entities of Biological Interest; CQ: Clioquinol;
COPD: C hronic Obstructive Pulmonary Disorder;
DFO: Desferrioxamine; Dp44mT: di-2-pyridylketone-
4,4,-dimethyl-3-thiosemicarbazone; EDTA:
Ethylenediamine-tetraacetic Acid; F A: Friedrich's ataxia;
FPN1: Ferr oportin- 1; HBED: N,N'- bis-(2-hydroxybenzyl)
ethylenediamine-N,N'-diacetic acid; HCP1: Heme Car-
rier Protein-1; HIF: Hyp oxia-Inducible Factor; H O1:
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: IBkinase;
KEGG: Kyoto Encyclopedia of Genes and Genomes; L1:
Deferriprone;LFA-1:LeukocyteFunctionAntigen-1;LIP:
Labile Iron Pool; LIPID: Long-Term Intervention with
Pravastatin in Ischaemic Disease; LPS: Lipopolysacchar-
ide; MOF: Mult iple organ failure; NF- B: Nuclear Factor
B; NGAL: Neutrophil Gelatinase-Associated Lipocalin
(Also k nown as lipocalin-2 or siderocalin); NIH:
2-hydroxy-1-napthylaldehyde isonicotinyl hydrazo ne;
PE: Pre-eclampsia; PIH: Pyridoxal isonicotinyl hydra-
zone; PPAR: Peroxisome Proliferator Activated Receptor;
RA: Rheumatoid Arthritis; ROS: Reactive Oxygen Species;
SBML: Systems Biology Markup Language; SIH: Salicy-
laldehyde isonicotinyl hyd razone; SIRS: Systemic Inflam-
matory Response Syndrome; SLE: Systemic Lupus
Erythematosus; SMILES: Simplified Molecular Input
Line Entry Specif ication; SOD: Superoxide Dismut ase;
Tf:Transferrin;TNF:TissueNecrosisFactor;VEGF:
Vascular Endothelial Growth Factor.
Competing interests
The author declares t hat he has no competing interests.
Acknowledgements
I thank Jon Barasch (Columb ia 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 ha d no role in study design, d ata collection
and analysis, decision to publish, or preparation of the manuscript. This is a
contribution from the Manchest er Centre for Integrat ive Systems Biology
http://www.mcisb.org.
References
1. Harnad S, Brody T, Vallieres F, Carr L, Hitchcock S, Gingras Y,
Oppenheim C, Hajjem C a nd Hilf ER: The a ccess/ impact
problem and the green and gold roads to open access: An
update. Ser ials Review 2008, 34:3640.
2. Hull D, Petti fer SR and Kell DB: Def rosting the digital library:
bibliographic t ools for the next generation web. PLoS C omput
Biol 2008, 4:e1000204, doi:10.1371/journal .pcbi.1000204.
3. Ananiadou S and McNaught J: Text mining in biology and
biomedicine.Artech House, London; 20 06.
4. A nania dou S, Kell DB and Tsujii Ji: Text Mining and its potential
applications in Systems Biolog y. Tr ends Biot echnol 2006,
24:571579.
5. Jensen LJ, Saric J and Bork P: Literature mining for the biologist:
from information retrieval to biological discovery. Nat Rev
Genet 2006, 7:11929.
6. Smalheiser NR: The Arrowsmith project: 2005 status report.
LNCS 2005, 3735:2643.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 26 of 79
(page number n ot f or citation purposes)
7. Hristovski D, Peterlin B, Mitchell JA and Humphre y SM: Using
literature-based discovery to identify disease candidate
genes. Int J Med Inform 2005, 74:28998.
8. Kostoff RN: Systematic acceleratio n of radical discovery and
innovation in science and technology. Technol Forecasting Soc
Change 2006, 73:923936.
9. Yetisgen-Yildiz M and Pratt W: Using statistical and knowledge-
based approaches for literature-based discovery. J Biomed
Informatics 2006, 39:600611.
10. Kostoff RN: Validating discovery in literature-based discov-
ery. JBiomedInform2007, 40:44850, author reply 4502..
11. Kostoff RN, Briggs MB, Solka JL and Rushenberg RL: Literature-
related discovery (LRD): Methodology. Technol Forecast Soc
Change 2008.
12. Cronin B: Bibliometrics and beyond: Some thoughts on web-
based citation analysis. Journal o f Information Science 2001, 27:17.
13. Kasztler A and Leitner KH: Bibliometric analysis and visualisa-
tion of intellectual capital. J Unive rsal Comp Sci 2002, 8:516 525.
14. Tabah AN: Literature dynamics: Studies on growth, diffusion,
and epidemic s. Annu Rev Inf Sci Technol 1999, 34:24 9286.
15. Börner K, Chen CM and Boyack KW: Visualizing knowledge
domains. Annual Review of Information Science and Technology 2003,
37:179255.
16. Errami M, Hicks JM, Fisher W, Trusty D, Wren JD, Long TC and
Garner HR: Déjà vu: a study of duplicate citations in Medline.
Bioinformatics 2008, 24:2439.
17. Kell DB: Iron behaving badly: inappropr iate iron chelation as
a major contributor to the aetiology of vascular and other
progressive inflammator y and degenerative diseases.2008
http://arxiv.org/ftp/arxiv/papers/0808/0808. 1371.pdf.
18. Dobson PD and Kell DB: Carrie r-mediated cellular uptake of
pharmaceutical drugs: an exception or the rule? Nat Rev Drug
Discov 2008, 7:205220.
19. Smalheiser NR, Torvik VI, Bischoff-Grethe A, Burhans LB, Gabriel M,
Homayouni R, Kashef A, Martone ME, Perkins GA, Price DL,
Talk AC and West R: Collaborative development of the
Arrowsmith two node search interface designed for labora-
tory investigators. J Biomed Discov Collab 2006, 1:8.
20. Smalheiser NR and Swanson DR: Using ARROWSMITH: a
computer-assisted approach to formulating and assessing
scientific hypotheses. Comput Methods Progra ms Biomed 1998,
57:14953.
21. Doms A and Schroeder M: GoPubMed: exploring PubMed with
theGeneOntology.Nucleic Acids Res 2005, 33:W7836.
22. Oinn T, Addis M, Ferris J, Marvin D, Senger M, Greenwood M,
Carver T, Glover K, Pocock MR, Wipat A and Li P: Taverna: a tool
for the composition and enactment of bioinformatics
workflows. Bioinformatics 2004, 20:304554.
23. Oinn T, Li P, Kell DB, Goble C, Goderis A , Greenwood M, Hu ll D,
Stevens R, Turi D and Zhao J: Tav erna/myGrid: aligning a
wor kflow system with the life sciences community. Workfl ows
for e-Science: scientific workflows for Grids Springer, Guil dford: Taylo r
IJ, Dee lman E, Gannon DB, Shields M 2007, 300319.
24. Hull D, Wolstencroft K, St evens R, Goble C, Pocock MR, Li P and
Oinn T: Taverna: a tool for building and runnin g workflows of
services. Nucleic Acids Res 2006, 34:W72932.
25. Oinn T, Greenwo od 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 and Wroe C: Taverna: les sons in
creating a workf low envir onment fo r the life sciences.
Concurrency and Computation: Practice & Experience 2006,
18:10671100.
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 and De
Roure D: Knowledge discovery for biology with Taverna:
producing and consuming semantics in the Web of Science.
Semantic Web: revolutionising knowledge d iscovery in the life sciences
Springer, New Yor k: Baker CJO, Cheung K-H 2007.
27. Chance B, Sies H and Boveris A: Hydroperoxide metabolism in
mammalian organs. Physiol Rev 1979, 59:527605.
28. Lenaz G: Role of mitochondri a in oxidative stress and ageing.
Biochim Biophys Acta 1998, 1366:53
67.
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,
31:34766.
30. Kowaltowski AJ and Vercesi AE: Mitochondrial damage induced
by conditions of oxidative stress. Free Radic Biol Med 1999,
26:46371.
31. Caden as E and Davies KJ: Mitocho ndrial free radical genera-
tion, oxidative stress, and aging. Free Radic Biol Med 2000,
29:22230.
32. Lenaz G, D'Aurelio M, Merlo Pich M, Genova ML, Ventura B,
Bov ina C, Formiggini G and Parenti Castelli G: Mitochondrial
bioenergetics in aging. Bi ochim Biophys Acta 2000, 1459:397404.
33. Raha S and Robinson BH: Mitochondria, oxygen free radicals,
disease and ageing. Trends Biochem Sci 2000, 25:5028.
34. Lenaz G: The mitochondrial production of reactive oxygen
species: mechanisms and im plications in human pathology.
IUBMB Life 2001, 52:15964.
35. Raha S and Robinson BH: Mitochondria, oxygen free radicals,
and apoptosis. Am J Med Genet 2001, 106:6270.
36. Dröge W: Free radicals in the physi ological contr ol of cell
function. Physiol Rev 2002, 82:4795.
37. TaylorER,HurrellF,ShannonRJ,LinTK,HirstJandMurphyMP:
Reversible glutathionylation of complex I increases mito-
chondrial superoxide formation. JBiolChem2003,
278:19603 10.
38. Turrens JF: Mitochondrial f ormation of reactive oxygen
species. JPhysiol2003, 552:335 44.
39. Adam-Vizi V: P roduction of reactive oxygen species in brain
mitochondria: Contribution by electron transport chain and
non-elec tron transport chain sources. Antioxidants & Redox
Signaling 2005, 7:
11401149.
40. Andreyev AY, Kushnareva YE and Starkov AA: Mitochondrial
metabolism of reactive oxygen species. Biochemistry (Mosc)
2005, 70:20014.
41. Balab an RS, Nemoto S and Finkel T: Mitochondria, oxidants, and
aging. Cell 2005, 120:483495.
42. Adam-Vizi V and Chinopoulos C: Bioenergetics and the
formation of mitochondrial reactive oxygen species. Trends
Pharmacol Sci 2006, 27:63945.
43. Halliwell B and Gutteridge JMC: Free Radicals in Biology and Medicine
Oxford University Press, Oxford; 42006.
44. O rreni us S, Gogvadze A and Zhivotov sky B: Mitochondrial
oxidative stress: Implications for cell death. Annual Review of
Pharmacology and Toxicology 2007, 47:143 183.
45. Fato R, Bergamini C, Leoni S, Strocchi P and Lenaz G: Generatio n
of reactive oxygen species by mitochondrial complex I:
implications in neurodegeneration. Neurochem Res 2008,
33:2487501.
46. Babior BM: Phagocytes and oxidative stress. Am J Med 2000,
109:3344.
47. Cave AC, Brewer AC, Narayanapanicker A, R ay R, Grieve DJ,
Walker S and Shah AM: NADPH oxidases in cardiovascular
hea lth and disease. Antioxidants & Redox Signaling 2006, 8:691728.
48. Bedard K and Krause KH: The N OX fam ily of ROS-generating
NADPH oxidases: physiology and pathophysiology. Physiol Rev
2007, 87:245313.
49. Brown JM, Terada LS, Grosso MA, Whitmann GJ, Velasco SE, Patt A,
Harken AH and Repine JE: Xanthine oxidase produces hyd ro-
gen peroxide which contributes to reperfu sion injury of
ischemic, isolated, perfused rat hearts. JClinInvest1988,
81:12971301.
50. Granger DN: Role of xanthine oxidas e and granulocytes in
ischemia repe rfusion injur y. Amer J Physi ol 1988, 255:
H1269H1275.
51. Linas SL, Whittenburg D and Repine JE: Ro le of xanthine oxidase
in ischemia reperfusion injury. Amer J Physiol 1990, 258:
F711F716.
52. Thompson-Gorman SL and Zweier JL: Evaluation of the role of
xan thine oxidase in myocardial reperfusion injury. J Biol Chem
1990, 265:66566663.
53. Adkins WK and Taylor AE: Role of xanthine oxidase and
neutrophils in ischemia-reperfusion injury in rabbit lung.
J Appl Physi ol 1990, 69:20122018.
54. Müller MJ, Vol lmar B, Friedl HP and Menger MD: Xanthine oxidase
and superoxide radicals in portal triad crossclamping-
induced microvascular reperfusi on injury of the liver. Free
Radic Biol Med 1996, 21:18997.
55. Imlay JA: Cellular defenses against superoxide and hydrogen
peroxide. Annu Rev Biochem 2008, 77:755 76.
56. Sundaresan M, Yu ZX, Ferrans VJ, Irani K and Finkel T: Require-
ment for gener ation of H
2
O
2
for platelet-derived growth
factor signal transduction. Science 1995, 270:2969.
57. Finkel T: Oxidant signals and o xidat ive stress. Curr Opin Cell Biol
2003, 15:24754.
58. Rhee SG: Cell signaling. H2O2, a necessary evil for cell
signaling. Science 2006, 312:18823.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 27 of 79
(page number n ot f or citation purposes)
59. Miller EW, Tulyathan O, Isacoff EY and Chang CJ: Molecular
imaging of hydrogen peroxide produced for cell signaling.
Nat Chem Biol 2007, 3:2637.
60. Forman HJ, Fukuto JM, Miller T, Zhang H, Rinna A and Levy S: The
chemistry of cell si gnaling by reactive oxygen and nitrogen
species and 4-hydroxynonenal. Arch Biochem Biophys 2008,
477:18395.
61. Halliwell B and Gutteridge JM: Oxygen toxicity, oxygen radicals ,
tra nsition metals and d isease. Biochem J 1984, 219:114.
62. Halliwell B and Gutteridge JMC: Role of free radicals and
catalyti c metal ions in human disease an overview. Meth
Enzymol 19 90, 186:185.
63. G alaris D and Pantopoulos K: Oxidative stress and iron
homeostasis: mechanisti c and he alth aspects. Crit Rev Clin
Lab Sci 2008, 45:123.
64. Buzan T: How to mind map Thorsons, London; 2002.
65. Fridovich I: Superoxide radical and sup eroxide dismutases.
Annu Rev Biochem 1995, 64:97112.
66. Wardman P and Candeias LP: Fenton chemistry: An introduc-
tion. Rad Res 1996, 145:523531.
67. Kehrer JP: Th e Haber-Weiss reaction and mechanisms of
toxicity. Toxicology 2000, 149:4350.
68. Hershko C and Weatherall DJ: Iron-chelating therapy. Crit Rev
Clin Lab Sci 1988, 26:30345.
69. Minotti G: Sources and role of iron in lipid peroxidation. Chem
Res Toxicol 1993, 6:134146.
70. Rayman MP, Barlis J, Evans RW, Redman CW and King LJ:
Abnormal iron parameters in t he pregnancy syndrome
preeclam psia. Am J Obstet Gynec ol 2002,
187:4128.
71. Welch KD , Davis TZ, Van Ed en ME and Aust SD: Deleterious iron-
mediated oxidation of biomolecules. Free Radic Biol Med 2002,
32:57783.
72. Cheng Z and Li Y: What is responsible for the initiating
chemistry of iron-mediated lipid peroxidation: an update.
Chem Rev 2007, 107:74866 .
73. Ts u kahar a H: Biomarkers for oxidative stress: Clinical
application in pediatric medicine. Curr Med Chem 2007,
14:339351.
74. Evans MD, Dizdaroglu M and Cooke MS: Oxidative DNA damage
and disease: induction, repair and significance. Mut Res 2004,
567:161.
75. Valko M, Izakovic M, Mazur M, Rhodes CJ and Tels er J: Role of
oxygen radicals in DNA d amage and cancer incidence. Mol
Cell Biochem 2004, 266:3756.
76. David SS, O'Shea VL and Kundu S: Base-excision repair of
oxidative DNA damage. Nature 2007, 447:94150.
77. Stadtman ER and Oliver CN: Metal-catalyzed oxidation of
proteins physiological consequences. JBiolChem1991,
266:20052008.
78. Davies MJ, Fu S, W ang H and Dean RT: S table markers o f oxidant
damage to proteins and their application in the study of
human disease. Free Radic Biol Med 1999, 27:115163.
79. Davies MJ: The oxidative envir onment and pr otein dama ge.
Biochim Biophys Acta 2005, 1703:93 109.
80. Terman A and Brunk UT: Lipofuscin. Int J Biochem Cell Bi ol 2004,
36:14001404.
81. Jung T, Bader N and Grune T: Lipofuscin: formation, distribu-
tion, and metabolic consequences. Ann N Y Acad Sci 2007,
1119:97111.
82. Halliwell B: Free radicals and antio xidants: a personal view.
Nutr Rev 1994, 52:25365.
83. De Zwart LL, Meerman JHN, Commandeur JNM and
Vermeulen NPE: Biomarker s of free radical damage applica-
tions in experimental animals and in humans. Free Rad Biol
Med 1999, 26:202226.
84. K ohen R and N yska A: Oxidation of biological systems:
oxidative stress phenomena, antioxidant s, red ox reactions,
and methods for their quantification. Toxicol Pat hol 2002,
30:62050.
85. Tarpey MM, Wink DA and 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:R43144.
86. Halliwell B and 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,
142:23155.
87. Cherubini A, Ruggiero C, Polidori MC and Mecocci P: Potential
markers of oxidative stress in str oke. Free Radical Biology and
Medicine 2005 , 39:841 852.
88. Shulaev V and Oliver DJ: Metabolic and proteo mic markers f or
oxidative stress. New tools fo r reactive oxygen sp ecies
research. Plant Physiol 2006, 141:36772.
89. Se rkov a NJ, Reisdo rph NA and van Patot MCT: Metabolic
markers of hypoxia: Systems biology application in biome-
dicine. Toxicology Mechanisms and Methods 2008, 18:8195.
90. Aust AE and Eveleigh JF: Mechanisms of DNA oxidation. Proc Soc
Exp Biol Med 1999, 222:24652.
91. Cooke MS, Lunec J and Evans MD: Progress in t he analysis of
urinary oxidative DNA damage. Free Radic Biol Med 2002,
33:16011614.
92. Dizdaroglu M, Jaruga P, Birincioglu M and Rodriguez H: Free
radical-induced damage to DNA: Mechanism s and measure-
ment. Free Radical Biology and Medicine 2002, 32:11021115.
93. Neeley WL and Essigmann JM: Mechanisms of formation,
gen otoxicity, and mutation of guanine oxidation products.
Chem Res Toxicol 2006, 19:491505.
94. Hwang ES and Bowen PE: DNA damage, a biomarker of
carcinogenesis: Its measurement and modulation by diet
and environment. Crit Rev Food Sci Nutrition 2007, 47:2750.
95. Shin CS, Moon BS, Park KS, Ki m S Y, Park S J, Chung MH and Le e HK:
Serum 8-hydroxy-guanine levels are increased in diabetic
patients. Diabetes Care 2001, 24:7337.
96. Loft S, Fischer-N ielsen A, Jeding IB, Vistisen K and Poulsen HE:
8-Hydrox ydeoxyguanosine as a urinary biomar ker of oxida-
tive DNA damage. J Tox icol Environ Health 1993, 40:39140 4.
97. Migliore L, Fontana I, Colognato R, Coppede F, Siciliano G and
Murri L: Searchingfortheroleandthemostsuitable
biomarkers of oxidative stress in Alzheimer's disease and
in other neurodegenerative diseases. Neurobiol Aging 2005,
26:58795.
98. Bolin C and Cardozo-Pelaez F: Assessing biomarkers of
oxidative stress: Analysis of guanosine and oxidized guano-
sine nucleotide triphosphates by high performance liquid
chr omatography with electrochemical detection. JChroma-
togr B Analyt Technol Biomed Life Sci 2007.
99. Yamad a S, Funada T, Shibata N, Kobayashi M, Kawai Y, Tats uda E,
Furuhata A and Uchida K: Protein-bound 4-hydroxy-2-hexenal
as a marker of oxidized n-3 polyunsaturated fa tty acids.
J L ipid Res 2004, 45:626634.
100. Petersen DR and Doorn JA: Reactions of 4-hydroxynonenal
with proteins and cellular targets. Free Radic Biol Med 2004,
37:93745.
101. Montuschi P, Ciabattoni G, Paredi P, Pantelidis P, du Bois RM,
Kharitonov SA and Barnes PJ: 8-Isop rostane as a bio marker of
oxidative stress in interstitial lung diseases. Amer J R esp Crit
Care Med 1998, 158:15241527.
102. Montuschi P, Collins JV, Ciabattoni G, Lazzeri N, Corradi M,
Kharitonov SA and Barnes PJ: Exhaled 8-isoprostane as an in
viv o biomarker of lung oxidative st ress in patie nts with
COPD and healthy smokers . Amer J Resp Crit Care Med 2000,
162:11751177.
103. Schwedhelm E and Boger RH: Application of gas chromato-
graphy-mass spectrometry for analysis of isoprostanes : their
role in cardiovascular disease. Clin Chem Lab Med 2003,
41:155261.
104. Davì G, Falco A and Patrono C: Determinants of F2-isoprostane
biosynth esis and inhibition in man. Chem Phys Lipids 2004,
128:14963.
105. Montuschi P, Barnes PJ and Rober ts LJ 2nd : Isoprostanes: markers
and mediators of oxidative stress. FASEB J 2004, 18:1791800.
106. Morrow JD: Quantification of isoprostanes as indices of
oxidant stress and the risk of atherosclerosis in humans.
Arterioscler Thromb Vasc Biol 2005, 25:27986.
107. Montuschi P, Barnes P a nd Roberts LJ 2nd: Insights into oxi dative
stress: the isoprostane s. Curr Med Chem 2007, 14:703 17.
108. Mori TA, Croft KD, Puddey IB and Beilin LJ: Analysis of native and
oxidized low-density lipop rotein oxysterols using gas chro-
matography mass spectrometry with selective ion monitor-
ing. Redox Report 1996, 2:2534.
109. Murphy RC and Johnson KM: Cholesterol, reactive oxygen
species, and the formation of biologically active mediators.
J Biol Chem 2008, 283:155215.
110. Janero DR: Malondialdehyde and thiobarbituric acid reactiv-
ity as diagnostic indexes of lipid peroxidation and perox-
ida tive tissue injury. Free Radic Biol Med 1990, 9:515540.
111. Gieseg SP, Crone EM, Flavall EA and Amit Z: Potential to inhibit
growth of atherosclerotic p laque development through
modulati on of macrophage neopterin/7,8-dihydroneopterin
synthesis. Br J Pharmacol 2008, 153:627 35.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 28 of 79
(page number n ot f or citation purposes)
112. Herce- Pagliai C, Kotecha S and Shuker DEG: Analytical methods
for 3-nitrotyrosine as a marker of exp osure to rea ctive
nitrogen species: A review. Ni tric Oxide-Biology and Chemistry
1998, 2:324336.
113. Bian K, Gao Z, Weisbrodt N a nd Murad F: The nature of heme/
iron-induced protein tyrosine nitration. Proc Natl Acad Sci USA
2003, 100:57127.
114. D uncan MW: A review of approaches to the analysis of
3-nitrotyrosine. Amino Acids 2003, 25:351361.
115. R yberg H and Caidahl K : Chromatographic and mass spectro-
metric methods for quantitative determination of 3-nitro-
tyrosine in biological samples and their application to
human samples. JChromatogrB2007, 851:160171.
116. Cao EH and Wang JJ: Oxidative Damage to DNA Levels of
Thymine Glycol and Thymidine Glycol in Neoplastic Human
Urines. Carcinogenesis 1993, 14:1359 1362.
117. Makropoulos W, Kocher K, Heintz B, Schwarz ER, Mertens PR and
Stefanidis I: Urinary thymidine glycol as a biomarker for
oxidative stress after kidney transplantation. Renal Failure
2000, 22:499510.
118. Herrgå rd MJ, Swainston N, Dobson P, Dunn WB, A rga 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 and Kell DB: A
consensus yeast metabolic network obtained from a com-
munity approach to systems biology. Nature Biotechnol 2008,
26:11551160.
119. Howe D, Costanzo M, F ey P, Gojobori T, Han nick L, Hide W,
Hill DP, Kania R, Schaeffer M, St Pierre S, Twigger S, White O and
Yon Rhee S : Big data: The future of biocuration. Nature 2008,
455:4750.
120. Brooksbank C, Cameron G and Thornton J: The European
Bioinfor matics Inst itute's data resources: towards systems
biology. Nucleic Acids Res 2005, 33:D4653.
121. Kanehisa M, Goto S, Hattori M, Aoki-Kinoshita KF, Itoh M,
Kawashima S, Katayama T, Araki M and Hi rakawa M: From
genomics to chemical genomics: new developments in
KEGG. Nucleic Acids Res 2006, 34:D3547.
122. Weininger D: SMILES, a chemical language and information
system .1. Introduction to methodology and encoding rules.
J Chem Inf Comput Sci 1988, 28:3136.
123. Stein SE, Heller SR and Tchekhovski D: An open standard for
chemical structure rep resentation the IUPAC chemical
identifier. Proc 2003 N imes Int Chem Info Con f 2003, 131143.
124. Coles SJ, Day NE, Murray-Rust P, Rzepa HS and Zhang Y:
Enhanceme nt of the chemical semantic web through the
use of InChI identifi ers. Organic & Biomolecular Chemistry 2005,
3:18321834.
125. Cash er O an d Rzepa HS: SemanticEye: a semantic web
applicat ion to rationalize and enhance chemical electronic
publishing. J Chem Inf Model 2006, 46:2396411.
126. Monge A, Arrault A, Marot C and Morin-Allory L: Managing,
profilin g and analyzin g a library of 2.6 million compounds
gathered from 32 chemic al pr oviders. Mol Divers 2006,
10:389403.
127. Knox C, Shrivastava S, Sothard P, Eisner R and Wishart DS:
Biospider: a web server for automating metabolome
annotations. Pac Symp Biocomputing 2007, 12:145 156.
128. Klinger R, Kolářik C, Fluck J, Hofmann-Apitius M and Friedrich CM:
Detection of IUPAC and IUPAC-like chemical names.
Bioinformatics 2008, 24:i26876.
129. Chavarria ME, Lara-Gonzalez L, Gonzalez-Gleason A, Garci a-
Paleta Y, Vital-Re yes VS and Reyes A: Prostacyclin/thromboxane
early changes in pregnancies that are complicated by
preeclam psia. Am J Obstet Gynec ol 2003, 188:98692.
130. Miller DM, Buettner GR and Aust SD: Transition metals as
catalysts of "autoxidation" reactions. Free Radic Biol Med 1990,
8:95108.
131. Stohs SJ and Bagchi D: Oxidative mechanisms in the toxicity of
metal ions. Free Rad Biol Med 1995, 18:321
336.
132. Meneghini R: Iron homeostasis, oxidative stress, and DNA
damage. Free Radical Biology and Medicine 1997, 23:783792.
133. H ube l CA : Dyslipidemia, iron, and oxidative stress in
preeclam psia: assessment of maternal and feto-placental
interact ions. SeminReprodEndocrinol1998, 16:7592.
134. Aisen P, Enn s C and Wessling-Resnick M: Chemistry and biology
of eukaryotic iron metabolism. Int J Biochem Cell Biol 2001,
33:940959.
135. Reddy MB and Clark L: Iron, oxidative stress, and disease risk.
Nutr Rev 2004, 62:1204.
136. T hompson KJ, Shoham S and Connor JR: Iron and neurodegen-
erative disorders. Brain Res Bull 2001, 55:15564.
137. V alko M, Morri s H and Cronin MTD: Metals, toxicity and
oxidative str ess. Curr Med Chem 2005, 12:11611208.
138. Carbon ell T and Rama R: Ir on, oxidative stress and early
neurological deterioration in ischemic stroke. Current Medic-
inal Chemistry 20 07, 14:857 874.
139. Dunn LL, Rahmanto YS and Richardson DR: Iron uptake and
metabolism in the new mi llennium. Tre nds Cell Biol 2007,
17:93100.
140. Weinberg ED: Exposing the hidden dangers of iron: what every medical
professional should kno w about t he i mpact of iron on the disease pro cess
Cum berland House; 2004.
141. Brewer GJ: Iron and copper toxicity in diseases of aging,
particularly atherosclerosis and Alzheimer's disease. Exp Biol
Med 2007, 232:323335.
142. V alko M, Leibfri tz D, Moncol J, Cronin MTD, Mazur M and Telser J:
Free radicals and antioxidants in normal physiological
functions and human disease. Int J Biochem Cell Biol 2007,
39:4484.
143. Winterbourn CC: Reconciling the chemistry and biology of
reactive oxygen species. Nat Chem Biol
2008, 4:27886.
144. Qian SY and Buettner GR: Iron and dioxygen chemist ry is an
important route to initiation o f biolog ical free rad ical
oxidatio ns: an electron paramagnetic resonance spin trap-
ping study. Free Radic Biol Med 1999, 26:1447 56.
145. Brown GC and Borutaite V: Nitric oxide, mi tochondria, and
cell death. IUBMB Life 2001, 52:18995.
146. Gomes A, Fernandes E an d Lima J: Use of fluorescence probes
for dete ction of reactive nitrogen species: A review.
JFluorescence2006, 16:119 139.
147. Y okoyama H, Kuroiwa H, Yano R and A raki T: Targeting reactive
oxygen species, reactive nitrogen speci es and inflammation
in MPTP neurotoxicity and Parkins on's disea se. Neurol Sci
2008, 29:293301.
148. Moncada S, Palmer RMJ and Higgs EA: Nitric-Oxide Physiology,
Pathophysiology, and Pharmacology. Pharmacological Reviews
1991, 43:109142.
149. Beckma n JS, Beckman TW, Chen J, Marshall PA and Freeman BA:
Apparent hydroxyl radical production by peroxynitrite:
imp lications for endothelial injury from nit ric oxide and
superoxide. Proc Natl Acad Sc i USA 1990, 87:1620 4.
150. R adi R, Cas sina A, Hodara R, Quijano C and Castro L: Peroxyni-
trite reactions and formation in mitochondria. Free Radic Biol
Med 2002, 33:145164.
151. Rubbo H and O'Donnell V: Nitric oxide, peroxynitrite and
lipoxygenase in atherogen esis: mechanistic insights. Toxicol-
ogy 2005, 208:30 517.
152. Zimmet JM and Hare JM: Nitroso-redox interactions in the
cardiovascular system. Circulation 2006, 114:153144.
153. Pacher P, Beckman JS and Liaudet L: Nitric oxide and peroxyni-
trite in health and disease. Physiol Rev 2007, 87:315
424.
154. Pavlovic R and Santaniello E: Peroxynitrite and nitrosoperox-
ycarbonate, a tightly connected oxidizing-nitrating couple in
the reactive nitrogen-oxygen species family: new perspec-
tives for protection from radical-promoted injury by
flavonoids. J Pharm Pharmacol 2007, 59:168795.
155. Aslan M, Ry an TM, Adler B, Townes TM, Parks DA, T hompson JA,
Tousson A, Gladwin MT, Patel RP, Tarpey MM, Batini c-Haberle I,
White CR and Freeman BA: Oxygen radical in hibition of nitric
oxide-dependent vascular function in sickle cell disease. Proc
Natl Acad Sci 2001, 98:1521515220.
156. O'Donnell VB and Freeman BA: Interactions between nitric
oxide and lipid oxidation pathways Implications for
vascular disease. Circulation Research 2001, 88:1221.
157. Lancaster JR Jr: Protein cysteine thio l nitrosati on: maker or
marker of reactive nitrogen species-induced nonerythroid
cellular signaling? Nitric Oxide 2008, 19:6872.
158. V a z S M and Au g ust o O : Inhib ition of myel operoxidas e-
mediated protein nitration by tempol: Kin etics, mechanism,
and implications. Proc Natl Acad Sci USA 2008, 105: 81916.
159. Brown GC and Bal-Price A: Inflammatory neurodegeneration
mediated by nitric oxide, glutamate, and mitochondria. Mol
Neurobiol 2003, 27:325 55.
160. Kim YM, Chung HT, Simmons RL and Billiar TR: Cellular non-
heme iron content is a determinant of nitric oxide-media ted
apoptosis, necrosis, and caspase inhibition. JBiolChem2000,
275:10954 61.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 29 of 79
(page number n ot f or citation purposes)
161. Galli SJ, Tsai M and Piliponsky AM: The development of allergic
inflammation. Nature 20 08, 454:445 54.
162. Fi nkel T: Radical medicine: tr eating ageing to cure disease.
Nat Rev Mol Cell Biol 2005, 6:9716.
163. Finch CE: The biology of human longevity: inflammati on, nutrition and
aging in the evolution of lifespans Academic Press, Amsterdam; 2007.
164. SerranoMandBlascoMA:Cancer and ageing: convergent and
divergent mechanisms. Nat Rev Mol Cell Biol 2007, 8:71522.
165. Brown GM: The living end: the future of death, aging and immortality
Macmillan, London; 2008.
166. R enshaw JC, Robson GD, Trinci APJ, Wiebe MG, Livens F R,
Collison D and Taylor RJ: Fungal siderophores: structures,
functions and applications. Mycol Re s 2002, 106:11231142.
167. Haas H: Molecular genetics of fungal siderophore biosynth-
esis and uptake: the role of siderophore s in iron uptake and
storage. Applied Microbiology and Biotechnology 2003, 62:316330.
168. Haas H, Eisendle M and Turgeon BG: Siderophores in fungal
physiology and virule nce. Annu Rev Phytopathol 2008, 46:14987.
169. Johnson L: Iron an d siderophores in fungal-host interactions.
Mycol Res 20 08, 112:17083.
170. R aymond KN, Muller G and Mat zanke BF: Complexation of iron
by sider ophores a review of their solution and structural
chemistry and biological function. Top Curr Chem 1984,
123:49102.
171. Guerinot ML: Microbial iron transport. An nu Rev Microbiol 1994,
48:74372.
172. Jurado RL: Iron, infections, and anemia of inflammation. Clin
Infect Dis 1997, 25:888 95.
173. Drechsel H and Jung G: Peptide siderophores.
Journal of Peptide
Science 1998, 4:147181.
174. Ratledge C and Dover LG: Iron metabolism in pathogenic
bacteria. Annu Rev Microbiol 2000, 54:88194 1.
175. Boukhalfa H and Crumbliss AL: Chemical aspect s of siderophore
mediated iron transport. Biometals 2002, 15:325 339.
176. Braun V and Braun M: A ctive transport of iron and siderophore
antibiotics. Curr Opin Microbiol 2002, 5:194201.
177. Brown JS and Holde n DW: Iron acquisition by Gram-positive
bacterial pathogens. Microb es I nfect 2002, 4:114956.
178. Cornelis P and Matthijs S: Diversity of siderophore-mediated
iron uptake systems in fluorescent pseudomonads: not only
pyoverdines. Environ Microbiol 2002, 4:787798.
179. Crosa JH and Walsh CT: Genetics and assembly line enzymol-
ogy of siderophore biosynthesis in bacteria. Micro biolo gy and
Molecular Biology Reviews 2002 , 66:223.
180. Marx JJ: Iron and infection: competition between host and
microbes fo r a precious element. Best Pract Res Clin Haematol
2002, 15:41126.
181. Winkelmann G: Microbial siderophore-mediated transport.
Biochem Soc Trans 2002, 30:6916.
182. Andrews SC, Robinson AK and Rodriguez -Quinones F: Bacterial
iron homeostasis. FEMS Microbiol Rev 2003, 27:21537.
183. Faraldo-Gómez JD and Sansom MSP: Acquisition of siderophores
in Gram-negative bacteria. Nat Rev Mol Cell Biol 2003,
4:105116.
184. Raymond KN, Dertz EA and Kim SS: Enterobactin: an archetype
for microbial iron trans port. Proc Natl Acad Sci USA 2003,
100:35848.
185. Wandersman C and Delepelaire P: Bacterial iron sources: From
siderophores to hemophores. Annual Review of Microbiology 2004,
58:611647.
186. Abergel RJ, Wilson MK, Arceneaux JE, Hoette TM, Strong RK ,
Byers BR and R aymond KN: Anthrax pathogen evades the
mammalian immune system through stealth siderophore
production. Proc Natl Acad Sci USA 2006, 103:18499503.
187.FischbachMA,LinHN,LiuDRandWalshCT:How pathogenic
bacteria evade mammalian sabotage in the battle for iron.
Nature Chemical Biology 2006, 2:132138.
188. Grass G: Iron transport in Escherichia coli: All has not been
said and done. Bi ometals 20 06, 19:159 172.
189. Miethke M and Marahiel MA: Siderophore-based iron acquisi-
tion and pathogen control. Microbiol Mol Biol Rev 2007,
71:41351.
190. Genco CA and Dixon DW: Emerging strategies in microbial
haem capture. Molecular Microbiology 2001, 39:1 11.
191. Loomis LD and Raymond KN: Solution equilibria of enterobac-
tin and metal enterobactin c omplexes. Inorganic Chemistry
1991, 30:906911.
192. Stintzi A, Barnes C, Xu J and Raymond KN: Micr obial iron
transport via a siderophore shuttle: a membrane ion
tra nsport pa radigm. Proc Natl Acad Sci USA 2000, 97:106916.
193. Braun V: Iron uptake mechanisms and their regulation in
pathogenic bacteria. Int J Med Microbiol 2001, 291:67 79.
194. Cendrowski S, MacAr thur W and Hanna P: Bacillus anthracis
requires siderophore biosynt hesis for growth in macr o-
phages and mouse virulence. Mol Microbiol 2004, 51: 407417.
195. Schaible ME and Kaufmann SHE:
Iron and microbial infection.
Nat Rev Microbiol 2004, 2:946953.
196. Ong ST, Ho JZS, Ho B and Ding JL: Iron -withholding strategy in
innate immunity. Immunobiology 2006, 211:295314.
197. Challis GL and Ravel J: Coelichelin, a new peptide siderophore
encoded by the Streptomyces coelicolor genome: structure
prediction from the sequence of its non-ribosomal peptide
syntheta se. FEMS Microbiol Lett 2000, 187:1114.
198. Fernandez-Pol JA: Isolation and characterization of a side-
rophore-like growth factor from mutants of SV40-t rans-
formed cells adapted to picolinic acid. Cell 1978, 14:48999.
199. Jones RL, Peterson CM, Grady RW and Cerami A: Low molecular
weight iron-bindin g factor from mammalian tissue that
potentiates bacterial g rowth. J Exp Med 1980, 151:41828.
200. Primosigh JV and Thomas ED: Studies on the partition of iron in
bone marrow cells. JClinInvest1968, 47:147382.
201. Jacobs A: Low molecular weight intracellular iron transport
compounds. Blood 1977, 50:433 9.
202. Hershko C, Graham G, Bates GW and Rachmilewitz EA: Non-
specific serum iron in thalassaemia: an abnormal serum iron
fraction of potential toxicity. Br J Haematol 1978, 40:25563.
203. Graham G, Bates GW, Rachmilewitz EA and Hershko C: Non-
specific serum iron in thalassem ia: quantitation and chemi-
cal reactivity. Am J Hema tol 1979, 6:20717.
204. Craven CM, Alexander J, Eldridge M, Kushner JP, Bernstein S and
Kaplan J: Tissue distribution and clearance ki netic s of non-
tra nsferrin-bound iron in the hypotransfer rinemic mouse: a
rodent model for hemochromatosis. Proc Natl Acad Sci 1987,
84:345761.
205. Fontecave M and Pierre JL: Iron metabolism the low
mol ecular mass iron p ool. Biology of Metals
1991, 4:133135.
206. Kaplan J: Mechanisms of cellular iron acquisition: another
iron in the fire. Cell 2002, 111:6036.
207. Ganz T: Hepcidin, a key regulator of iron metaboli sm and
mediator of anemia of inflammation. Blood 2003, 102:783 788.
208. Finch C: Regulators of iron balance in humans. Blood 1994,
84:1697702.
209. Kohgo Y, Ikuta K, Ohtake T, Torimoto Y and Kato J: Body iron
metabolism and pathophysiology of iron o verload. Int J
Hematol 2008, 88:715.
210. Crichton RR and Ward RJ: Iron metabolismnew persp ectives
in view. Biochemistry 1992, 31:1125564.
211. Andrews NC: Medical progress: Dis orders of iron metabo-
lism. New England Journal of Medicine 1999, 341:19861995.
212. Beard JL, Dawson H and Piñero DJ: Iron metabolism: a
comprehensive review. Nutr Rev 1996, 54:295317.
213. Lieu PT, Heiskala M, Peterson PA and Yang Y: The roles of iron in
hea lth and disease. Mol Aspects Med 2001, 22:187.
214. Chung JY and Wessling-Resnick M: Molecular mechanisms and
regulation of iron transport. Crit Rev Clin Lab Sci 2003,
40:151182.
215. Frazer DM a nd Anderson GJ: The orchestration of body iron
intake: how and where do enterocytes receive their cues?
Blood Cells Molecules a nd Diseases 20 03, 30:288 297.
216. Knutson M and Wessling-Resnick M: Iron metabolism in the
reticuloendothelial system. Crit Rev Bioch em Mol Biol
2003,
38:6188.
217. McArdle HJ, Danzeisen R, Fosset C and Gamb ling L: The role of
the placenta in iron transfer from mot her to fetus and the
relationship between iron status and fetal outcome. Biometals
2003, 16:1617.
218. Hentze MW, Muckenthaler MU and Andrews NC: Balancing acts:
mol ecular control of m ammalian iron metabolism. Cel l 2004,
117:28597.
219. Andrews NC: Molecular control of iron metabolism. Best Pract
Res Clin Haematol 2005, 18:15969 .
220. Papanikolaou G and Pantopoulos K: Iron metabolism and
toxicity. ToxicolApplPharmacol2005, 202:199211.
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, 205:297318.
222. D onovan A, Roy CN and Andrews NC: The ins and outs of iron
homeostasis. P hysiology (Bethesda) 2006, 21:11523.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 30 of 79
(page number n ot f or citation purposes)
223. Ganz T and Nemeth E: Regulation of iron acquisition and iro n
distribution in mammals. Biochim Biophys Acta 2006,
1763:690699.
224. Huang XP, O'Brien PJ and Templeton DM: Mitoc hondrial
involvement in genetically determined transition metal
toxicity I. Iron toxicity. Chemico-Biological Interactions 2006,
163:6876.
225. Nairz M and Weiss G: Molecular and clinical aspects of iron
homeostasi s: from anemia to hemochromatosis. Wiener
Klinische Wochenschrift 2006, 118:442462.
226. Anderson GJ: Mechanisms of iron loading and toxicity. Am J
Hematol 2007, 82:1128 31.
227. Andrews NC and Schmidt PJ: Iron homeostasis. Annu Rev Physiol
2007, 69:6985.
228. Harrison-Findik DD: Role of alcohol in the regulation of iron
metabolism. World J Gastroenterol 2007, 13:492530.
229. Jones BC, Beard JL, Gibson JN, Ung er EL, Allen RP, McCarthy KA
and Earley CJ: Systems genetic analysis of peripheral iron
parameters in the mouse. Am J Physiol Regul Integr Comp Physiol
2007, 293:R11624.
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.
231. Vallerio LG: Mammalian iron me tabolism. Toxicology Mechanisms
and Methods 2007, 17:497517.
232. Andrews NC: Forging a field: the golden age of iron biology.
Blood 2008, 112:21930.
233. Beard J and Han O: Systemic iron status. Bio chim Bio phys Acta
2008 in press.
234. D eugnier Y, Brissot P and Loréal O: Iron and the liver: update
2008. JHepatol
2008, 48(Suppl 1):S11323.
235. Ganz T: Iron homeostasis: fitting the puzzle pieces together .
Cell Metab 2008, 7:28890.
236. Handelman GJ and Levin NW: Iron an d anemia in human
biology: a review of mechanisms. Heart Fail Rev 2008,
13:393404.
237. HowerV,MendesP,TortiFM,LaubenbacherR,AkmanS,ShulaevV
and Torti SV: A gener al map of iron metabolism and tissue-
specific subnetworks. Mol B iosyst 2008.
238. Kemna EH, Tjalsma H, Willems HL and Swinkels DW: Hepcidin:
from discovery to differential diagno sis. Haematologica 2008,
93:907.
239. Kong W, Dua n X, Shi Z and Chang Y: Ir on metabolism in the
mononuclear phagocyte system. Progr Nat Sci 2008,
18:11971202.
240. Levi S and Rovida E: The role of iron in mit ochondrial function.
Biochim Biophys Acta 2008 in press.
241. Wrighting DM and Andre ws NC: Iron homeostasis and
erythrop oiesis. Red Cell Development 2008, 82:141167.
242. Frazer DM and Anderson GJ: Iron Imports. I. Intestinal iron
absorption and its regulation. Amer J Physiol 2005, 289:
G631G635.
243. Mackenzie B and Garrick MD: Iron Imports. II. Iron uptake at
the apical membrane in the intestine. American Journal of
Physiology-Gastrointestinal and Liver Physiology 2005, 289:G981G986.
244. Wessling-Resnick M: Iron Imports. III. Transfer of iron fr om
the mucosa into circulation. American Journal of Physiology-
Gastrointestinal and Liver Physiology 2006, 290:G1G6.
245. Ganz T and Nemeth E: Iron imports. IV. Hepcidin and
regulation of body iron metabolism. Amer J Physiol 2006, 290:
G199G203.
246. Fleming RE a nd Britton RS: Iron Imports. VI. HFE and
regulation of intestinal iron absorption. American Journal of
Physiology-Gastrointestinal and Liver Physiology 2006, 290:G590G594.
247. Ma YX, Yeh M, Yeh KY and Glass J: Iron imports. V. Transport of
iron through the intestinal epithelium. American Journa l of
Physiology-Gastrointestinal and Liver Physiology 2006, 290:G417G422.
248. D e Domenico I, Ward DM an d Kaplan J: Regulation of iron
acquisition and storage: consequences for iron-linked dis-
orders. Nature Reviews Molecular Cell Biology 2008, 9:7281.
249. Gunshin H, Mackenzie B, Berger UV, Gunshin Y, Romero MF,
Boron WF, Nussberger S, Gollan JL and Hediger MA: Cloning and
characterization of a mammalian prot on-coupled metal-ion
transporter. Nature 1997, 388:4828.
250. Forbes JR and Gros P: Divalent-metal transport b y NRAMP
proteins at the interface of host-pathogen interactions.
Trends Microbiol 2001, 9:397403.
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 and McKie AT: Identification
of an intestinal heme transporter. Cell 2005, 122:789801.
252. R yter SW, Alam J and Choi AM: Heme oxyge nase-1/carbon
monoxide: from basic science to therap eutic applica tions.
Physiol Rev 2006, 86:583650.
253. V ítek L and Schwertner HA: The heme catabolic pathway and
its protective effects on oxidative stress-mediated diseases.
Adv Clin Chem 2007, 43:157.
254. Abraham NG and Kappas A: Pharmacological and clinical
aspects of heme o xygenase. Pharmaco l Rev 2008, 60:79 127.
255. Ponka P: Tissue-specific regulation of iron metabolism and
heme synthesis: Distinc t control mechanisms in erythroid
cells. Blood 1997, 89:125.
256. Linder MC, Mortya M, Whon A, Kassa A and Gilley C: Vesicular
tra nsport of Fe and interaction with other metal ions in
polarize d Caco2 Cell monolayers. Biological Research 2006,
39:143
156.
257. Moriya M and Linder MC: Vesicular transport and apotrans -
ferrin in intestinal iron absorption, as shown in the Caco-2
cell model. Amer J Physiol 2006, 290:G301G309.
258. Petrat F, de Groot H, Sust mann R and R auen U: The chelatable
iron pool in living cells: A methodically defined quantity.
Biological Chemistry 2002, 383:489502.
259. T enopoulou M, Kurz T, Doulias PT, Galaris D and Brunk UT: Does
the calcein-AM method assay the total ce llular 'labile iron
pool' or only a fraction of it? Biochem J 2007, 403:2616.
260. Shvartsman M, Kikkeri R, Shanzer A and Cabantchik ZI: Non-
tra nsferrin-bound iron reaches mitochondria by a chelator-
ina ccessible mechanism: biological and cli nical implications.
Am J Physiol Cell Phy siol 2007, 293:C138394.
261. Breuer W, Shvartsman M and Cabantchik ZI: Intracellular labile
iron. Int J Biochem Cell Biol 2008, 40: 3504.
262. Fakih S, Podinovskaia M, Kong X, Collins HL, Schaible UE and
Hider RC: Targeting t he lysosome: fluorescent iron(III)
chelators to selectively monitor endosomal/lysosomal labi le
iron pools. JMedChem2008, 51:453952.
263. Arosio P, Ingrassia R and Cavadini P: Ferritins: A family of
mol ecules for iron stora ge, antioxid ation a nd more. Biochim
Biophys Acta 2008.
264. Shi H, Bencze KZ, Stemmler TL and Philpott CC: A cytosolic iron
chaperone that delivers iron t o ferritin. Science 2008,
320:120710.
265. Hallberg L and Hulthen L: Perspectives on iron absorption. Bl ood
Cells Molecules and Diseases 2002, 29:562573.
266. Arosio P and Levi S: Ferritin, iron homeostasis, and oxidat ive
damage. Free Radic Biol Med 2002, 33:45763.
267. Lee DH, Zacharski LR and Jacobs DR Jr: Compar ison of the
serum ferritin and percentage of transferrin saturation as
exp osure markers of iron-driven oxidative stress-related
disease outcomes. Am Heart J 2006,
151:1247 e1 7.
268. M cKie AT a nd Barl ow DJ: The SLC40 basolateral iron
tra nsporter family (IREG1/ferroportin/MTP1). Pflugers Arch
2004, 447:8016.
269. D onovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, Robine S and
Andrews NC: The iron exporter ferroportin/Slc40a1 is
essential for iron homeostasis. Cell Metabolism 2005, 1:191200.
270. Ganz T: Cel lular iron: ferroportin i s the only way ou t. Cell
Metab 2005, 1:1557.
271. Oates PS: Th e role of hepcid in and ferroportin in iron
absorption. Histol Histopathol 2007, 22:791804.
272. V ulpe CD, Kuo YM, Murphy TL, Cowley L, Askwith C, Libina N,
Gitschier J and Anderson GJ: Hephaestin, a ceruloplasmin
homologue implicated in intestinal iron transport, is
defective in the sla mouse. Nature Genetics 1999, 21:195199.
273. Worwood M: Serum transferrin receptor assays an d their
applicat ion. Annals of Clinical Biochemistry 2002, 39:221230.
274. Mense SM and Zhang L: Herne: a versatile signaling molecule
controll ing the activities of diverse regulators ranging from
transcription factors to MAP kinases. Cell Res 2 006,
16:681692.
275. Balla J, Vercellotti GM, Nath K, Yachie A, Nagy E, Eaton JW and
Balla G: Haem, haem oxygenase and ferrit in in vascular
endothelial cell injury. Nephrology Dialysis Transplantation 2003,
18:812.
276. Choi AM and Alam J: Heme oxyg enase-1: function, regulation,
and implication of a novel stress-inducible protein i n
oxidant-induced lung injury. Am J Respir Cell Mol Biol 1996,
15:919.
277.ForestiR,GreenCJandMotterliniR:Generation of bile
pigments by haem oxygenase: a refined cellular strategy i n
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 31 of 79
(page number n ot f or citation purposes)
response to stressful insults. Biochem Soc Symp 200 4,
71:177192.
278. Poon HF, Calabrese V, Scapagnini G and Butterfield DA: Free
radicals: key to brain aging and heme oxygena se as a cellular
response to oxidative stress. J Gerontol A Biol Sci Med Sci 2004,
59:47893.
279. Akagi R, Takahashi T and Sassa S: Cytoprotective effects of heme
oxygenase in acute renal failure. Cellular Stress Responses in Renal
Diseases, Contrib Nephrol 2005, 148:7085.
280. Fredenburgh LE, Perrella MA and Mitsialis SA: The role of heme
oxygenase-1 in pulmonary disease. Amer J Resp Cell Mol Biol
2007, 36:158165.
281. Gueler F, Park JK, Rong S, Kirsch T, Lindschau C, Zheng W, Elger M,
Fiebeler A, Fliser D, Luft FC and Haller H: Statins attenuate
ischemia-reperfusion injury b y inducing heme oxygenase-1
in infiltrating macrophages. Am J Pat hol 2007, 170:11921199.
282. Koizumi S: Human h eme oxygenase-1 deficie ncy: a lesson on
serendipity in the discovery of t he novel disease. P ediatr Int
2007, 49:12532.
283.HorváthI,DonnellyLE,KissA,ParediP,KharitonovSAand
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 str ess. Thorax 1998, 53:668 72.
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, Ce rjan D , E kstrom 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, Larss on K, Lindskog M,
Lodewyckx W, Lund J, Lundeberg J, Magnusson K, Malm E, Nilsson P,
Odl ing J, Oksvold P, Olsson I, Oster E, Ottos son J, Paavilainen L,
PerssonA,RiminiR,RockbergJ,RunesonM,SivertssonA,
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, Wre thagen U, Xu LL, Hob er S
andPontenF:A human protein atlas for normal and cancer
tissues based on an tibody proteomics. Mol Ce ll Proteomics 2005,
4:192032.
285. Lee DW and Andersen JK: Role of HIF-1 in iron regulation:
potential therapeutic strategy for neurodegenera tive dis-
orders. Curr Mol Med 2006, 6:883893.
286. Rouault TA: The role of ir on regulatory prot eins in mamma-
lian iron homeostasis and disease. Nature Chem Biol
2006,
2:406414.
287. Wallander ML, Leibold EA and Eisenstein RS: Molecular control of
vertebrate iron homeostasis by iron regulatory proteins.
Biochim Biophys Acta 2006, 1763:668689.
288. Lee K, Roth R A and LaPres JJ: Hypoxia, d rug therapy and
toxicity. Pharmacology & Therapeutics 2007, 113:229246.
289. Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P an d
Loreal O: A new mouse liver-specific gene, encoding a
protein ho mologous to human antimicrobial peptide hepci-
din, is overexpressed during iron overload. JBiolChem2001,
276:78117819.
290. Fl eming RE and Sly WS: Hepcidin: A putative iron-regulatory
hormone relevant to hereditary hemochromatosis and the
anemia of chronic disease. Proc Nal Acad Sci 2001, 98:81608162.
291. Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B,
Kah n A and 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, 98:87808785.
292. Weinstein DA, Roy CN, Fleming MD, Loda MF, Wolfsdorf JI and
Andrews NC: Ina ppropriate expression of hepcidin is asso-
ciated with iron refractory anemia: implications for the
anemia of ch ronic disease. Blood 2002 , 100:377681.
293. Br idle KR , Frazer DM, Wilkins SJ, Dixon JL, Purdie DM,
Crawford DHG, Subramaniam VN, Powell LW, Anderson GJ and
Ramm GA: Disrupted hepcidin regulation in HFE-associated
haemochromatosis and the liver as a regulator of body iron
homoeostasis. Lancet 2003, 361:669673.
294. Nicolas G, Viatte L, Lou DQ, Bennoun M, Beaumont C, Kahn A,
Andrews NC and Vaulont S: Constitutive hepcidin expression
prevents iron overload in a mouse model of hemochroma-
tosis. Nature Genetics 2003, 34:97101.
295. Roetto A, Papanikolaou G, Politou M, Alberti F, Girelli D,
Christakis J, Loukopoulos D a nd Camaschella C: Mutant anti-
microbial peptide hepcidin is associated with severe juvenile
hemochromatosis. Nature Genetics 2003, 33:2122.
296. D eicher R and Horl WH: Hepcidin: a molecular link betwee n
inflammation and anaemia. NephrolDialTransplant2004,
19:
5214.
297. Leong WI and Lönn erdal B: Hepcidin, the recently identifi ed
peptide that appears to regulate i ron absorption. JNutr2004,
134:14.
298. R obson KJ: Hepcidin and its role in iron absorption. Gut 2004,
53:6179.
299. Camaschella C: Understanding i ron homeostasis through
genetic analysis of hemochromatosis and related disorders.
Blood 2005, 106:37107.
300. Loréal O, Haziza-Pigeon C, Troadec MB, Detivaud L , Turlin B,
Courselaud B, Ilyin G a nd Brissot P: Hepcidin in iron metabo-
lism. Curr Pr otein Pept Sci 2005, 6:27991.
301. R oetto A and Camaschella C: New insights into iron home-
ostasis through the study of non-HFE hereditary haemo-
chromatosis. Best Pract Res Clin Haematol 2005, 18:23550.
302. Roy CN and Andrews NC: Anemia of inflammati on: the
hepcidin lin k. Curr Opin Hematol 2005, 12:10711.
303. Beutler E: Hemochromatosis: Genetics and pathophysiology.
Annu Rev Med 2006, 57:331347.
304. D eicher R and Hörl WH: New insights into the regulation of
iron homeostasis. Eur J Clin Invest 2006, 36:301309.
305. Farnaud S, Patel A and Evans RW: Modelling of a metal-
containing hepcidin. Biometals 2006, 19:527533.
306. Ganz T: Hepcidin A peptide hormone at the interface of
innate immunity and iron metabolism. Antimicrobial Peptides
and Human Disease, Current T opics in Microbiology and Immunology
2006, 306:183198.
307. Harrison-Findik DD, Schafer D, Klein E, Timchenko NA, Kulaksiz H,
Clemens D, Fein E, Andriopoulos B, Pantopoulos K and Gollan J:
Alc ohol metabolism-mediated oxidative stress down-regu-
lates hep cidin transcription and leads to increased duodenal
iron transporter expression. J Biol Chem 2006,
281:22974 22982.
308. Hugman A: Hepcidin: an important new regulator of iron
homeostasis. Clin Lab Haematol 2006, 28:7583.
309. Mena NP, Esparza AL and Núñez MT: Regulation of transepithe-
lial transport of iron by hepcidin. Biol Res 2006, 39:191 193.
310. Mok H, Mlodnicka AE, Hentze MW, Muckenthaler M and
Schumacher A: The molecular circuitry regulating the switch
between iron deficiency and overload in mice. J Biol Chem
2006, 281:794651.
311. Nemeth E and Ganz T: Regulation of iron metabo lism by
hepcidin. Annual Review of Nutrition 2006, 26:323342.
312. Atanasiu V, Manolescu B and Stoian I: Hepcidin central
regulator of iron metabolism. European Journal of Haemat ology
2007, 78:110.
313. Malyszko J and Mysliwiec M: He pcidin in an emia and inflamma-
tion in chronic kidney disease. Kidney & Blood Pressure Research
2007, 30:1530.
314. Oates PS and Ahmed U: Molecular regulation of hepatic
exp ression of iron regulatory hormone hepcidin. JGastro-
enterol Hepatol 2007, 22:137887 .
315. Barisani D, Pelucchi S, Mariani R, Galimberti S, Trombini P,
Fumagalli D, Meneve ri R, Nemeth E, Ganz T and Pip erno A:
Hepcidin and iron-related gene expression i n subjects with
Dysmetabolic Hepatic Iron Overload. JHepatol2008,
49:12333.
316. Camaschella C and Silvestri L: New and old players in the
hepcidin pathway. Haematologica 2008, 93:14414.
317. De Domenico I, Nemeth E, Nelson JM, Phillips JD, Ajioka RS,
Kay MS, Kushner J P, Ganz T, Ward D M and Kaplan J: The hepcidin-
binding site on ferroportin is evolutiona rily conserved. Cell
Metab 2008, 8:14656.
318. Farnaud S, Rapisarda C, Bui T, Drake A, Cammack R and Evans RW:
Identification of an iron-hepcidin complex. Biochem J 2008,
413:5537.
319. Kartikasari AE, Roelofs R, Schaeps RM, Kemna EH, Peters WH,
Swinkels DW and Tjalsma H: Secretion of bioactive hepcidin-25
by liver cells correlates with its gene transcription and
points towards synergism between iron and inflammation
signaling pathway s. Biochim Biophys Acta 2008, 1784:202937.
320. Muckenthaler MU, Gal y B and Hentze MW: Systemic i ron
homeostasis a nd the iron-respon sive element/iron-regula-
tory protein (IRE/IRP) regulatory network. Annu Rev Nutr
2008, 28:197213.
321. Nemeth E: Iron regulation and erythropo iesis. Curr Opin
Hematol 2008, 15:16975.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 32 of 79
(page number n ot f or citation purposes)
322. Nishina S, Hino K, Kor enaga M, Vec chi C, Pietrangelo A, Mizukami Y,
Furutani T, Sakai A , Okuda M, Hidaka I, Okita K and Sakaida I:
Hepatitis C virus-induced reactive oxygen species raise
hepatic iron level in mice by reducing hepcidin transcrip-
tion. Ga stroenterology 2008, 134:22638.
323. Peeling P, Dawson B, Goodman C, Landers G and Trinder D:
Athletic induced iron deficiency: new insights int o the role
of in flammation, cytokines and hormones. Eur J Appl Physiol
2008, 103:38191.
324. Swinkels DW, Girelli D, Laarakkers C, Kroot J, Campostri ni N,
Kemna EH and Tjalsma H: Advances in quantitative hepcidin
measurements by t ime-of-flight mass spectrometry. PLoS
ONE 2008, 3:e270 6.
325. Swinkels DW and Wetzels JF: Hepcidin: a new tool in the
management of anaemia in patients with chronic kidney
disease? Nephrol Dial Transplant 2008, 23 :2450 3.
326. T oblli JE, Cao G, Rivas C and Kulaksiz H: Heart and iron
deficiency anaemia in rats with renal insufficiency: The role
of hepci din. Nephrology (Carlton) 2008, 13:636 645.
327. Trinder D, Ayonrinde OT and Olynyk JK: HCV, iron, and
oxidative stress: the new choreography of hepcidin. Ga stro-
enterology 2008, 134:34851.
328. Knutson MD, Oukka M, Koss LM, Aydemir F and Wessling-
Resnick M: Iron releas e from macrophages after erythropha-
gocytosis is up-regulated by ferroportin 1 overexpression
and down-regulated by hepcidin. Proc Natl Acad Sci USA 2005,
102:13248.
329. Nemet h E, Tuttle MS, Powelson J, V aughn MB, Donovan A,
Ward DM, Ganz T and Kaplan J: Hepcidin regulates cellular
iron efflux by binding to ferroportin and inducing its
internalization. Science 2004, 306:20902093.
330. D elaby C, Pilard N, Goncalves AS, Beaumont C and Canonne-
Hergaux F: Presence of the iron exporter ferroportin at the
pla sma membrane of macrophages is enhanced by iron
loading and down-regulated by hepcidin. Blood 2005,
106:397984.
331. Ganz T: Hepcidin a regulator of intestinal iron absorption
and iron recycling b y macrophages. Best Practice & Research
Clinical Haematology 2005, 18:171
182.
332. Papanikolaou G, Tzilianos M, Christakis JI, Bogdanos D, Tsimirika K,
MacFarlane J, Goldberg YP, Sakellaropoulos N, Ganz T and
Nemeth E: Hepcidin in iron overload disorders. Blood 2005,
105:41034105.
333. Lesbordes-Brion J C, Viatte L, Bennoun M, Lou DQ, Ramey G,
Houbron C, Hamard G, Kahn A and Va ulont S: Targeted
disruption of the hepcidin 1 gene results in severe hemo-
chromatosis. Bl ood 2006, 108:14021405.
334. Anderson GJ and Frazer DM: Iron metabolism meets signal
tra nsduction. Nature Genetics 2006, 38:503504.
335.KulaksizH,GehrkeSG,JanetzkoA,RostD,BrucknerT,
Kallinowski B and Stremmel W: Pro-hepcidin: expression and
cell specific localis ation in the liver and its regulat ion in
hereditary haemochromatosis, chronic renal insufficiency,
and renal anaemia. Gut 2004, 53:735743.
336. Kulaksiz H, Theilig F, Bachmann S, Gehrke SG, Rost D, Janetzko A,
CetinYandStremmelW:The iron-regulatory peptide
hormone hepcidin: expression and cellular localization in
the mammalian kidney. Journal of Endocrin ology 2005,
184:361370.
337. Tiker F, Celik B, Tarcan A, Kilicdag H, Ozbek N and Gurakan B:
Serum pro-hepcidin levels and relationships with iron
parameters in healthy preterm and term newborns. Pediatr
Hematol Oncol 2006, 23:293297.
338. R oe MA, Spinks C, Heath ALM, Harvey LJ, Foxall R, Wimper is J,
Wolf C and F airweather-Tait SJ: Serum prohepcidin concentra-
tion: no association with iron absorption in healthy men; and
no relationship wi th iron status in men carrying. HFE
mutations, hereditary haemochromatosis patients under-
going phlebotomy treatment, or pregnant women. Br J Nutr
2007, 97:544549.
339. Nicolas G, Chauvet C, Viatte L, Danan JL, Bigard X, Devaux I,
Beaumont C, Kahn A and Vaulont S: The gene encoding the iron
regulatory peptide hepcidin is regulated by anemia, hypoxia,
and inflammation. J Clin Invest 2002, 110:10371044.
340. Pak M, Lop ez MA, Gabayan V, Ganz T and Rivera S: Su ppression of
hepcidin during anemia requires erythropoietic activity.
Blood 2006, 108:37305.
341. Nemeth E, Valore EV, Territo M, Schiller G, Lichtenstein A and
Ganz T: Hepcidin, a putative mediator of anemia of
inflammation, is a type II acute-phase protein. Blood 2003,
101:24612463.
342. Nemeth E, Riv era S, Gabayan V, Keller C, Taudorf S, Pedersen BK
and Ganz T: IL-6 mediates hypoferremia of inflammation by
inducing the synthesis of the iron regulatory hormone
hepcidin. J Clin Invest 2004, 113:12711276.
343. Kemna E, Pickkers P, Nemeth E, Hoeven van der H and Swinkels D:
Time-course analysis of hepcidin, se rum iron, and plasma
cytokine levels in humans injected with LPS. Blood 2005,
106:18641866.
344. Fl eming RE: Iron and inflammation: cross-t alk between path-
ways regulating hepcidin. JMolMed2008, 86:4914.
345. Malyszko J, Malyszko JS, Pawlak K, Drozdowska-Rams L, Brzosko S
and Mysliwie c M: Hepcidin is linked to anemia and inflamma-
tion in peritoneal dialysis patients. Perit Dial Int 2008,
28:41821.
346. Wang RH, Li CL, Xu XL, Zheng Y, Xiao CY, Ze rfas P, Cooperman S,
Eck haus M, Rouault T, Mishra L and Deng CX : AroleofSMAD4in
iron metabolism through the p ositive r egulation of hepicidin
exp ression. Cell Metabolism 2005, 2:399409.
347. Falzacappa MVV, Spasi c MV, Kessler R, Stolte J, Hentze MW and
Muckenthaler MU: STAT3 mediates hepatic hepcidin expres-
sion and its inflammatory stimulation. Blood 2007,
109:353358.
348. Pietrangelo A, Dierssen U, Valli L, Garuti C, Rump A, Corradini E,
Ernst M , Klein C and Trautwein C: STAT3 is required for IL-6-
gp1 30-dependent activation of hepcid in in vivo. Gastroenterol
2007, 132:294300.
349. Choi SO, Cho YS, Kim HL and Park JW: ROS mediate the
hypoxic repression of the hepcidin gene by i nhibiting C/EBP
alp ha and STAT-3. Biochem Biophys Res Comm 2007, 356:312317.
350. Flanagan JM, Truksa J, Peng HF, Lee P and Beutler E: In vivo imaging
of hepcidin promoter stimul ation by iron and inflammation.
Blood Cells Mols Dis 2007, 38:253257.
351. Détivaud L, Nemeth E, Boudjema K, Turlin B, Troadec MB,
LeroyerP,RopertM,JacquelinetS,CourselaudB,GanzT,
Brissot P and Loréal O: Hepcidin levels in huma ns are
cor related with hepatic iron stores, hemoglobin levels, and
hepatic function. Blood
2005, 106:746748.
352. Weizer-Stern O, Adamsk y K, Amariglio N, Rachmilewitz E, Breda L,
Rivella S and Rechavi G: mRNA expression of iron regulatory
gen es in beta-thalassemia intermedia and beta-thalassemia
major mouse models. Amer J Hematol 2006 , 81:479 483.
353. Weizer-Stern O, Adamsky K, Amariglio N, Levin C, Koren A,
Breuer W, Rachmilewitz E , Breda L, Rivella S, Cabantchik ZI and
Rechavi G: Downregulation of hepcidin and haemojuvelin
exp ression in th e hepatocyte cell-line HepG2 induced by
thalassaemic sera. Br J Haematol 2006, 135:129138.
354. Weizer-Stern O, Adamsky K, Margalit O, As hur-F abian O, Givol D,
Amariglio N and Rechavi G: Hepcidin, a key regulator of iron
metabolism, is transcriptionally activated by p53. Br J
Haematol 2007, 138:25362.
355. Sullivan JL: Macrophage iron, hepcid in, and atherosclerotic
pla que stability. Exp Biol Med (Maywood) 2007, 232:101420.
356. Adamsky K, W eizer O, Amariglio N, Breda L, Harmelin A, Rivella S,
Rachmilewitz E and Rechavi G: Decreased hepcidin mRNA
expression in thalassemic mice. Br J Haematol 2004,
124:123124.
357. Flower DR, North AC and Sansom CE: The lipocalin protein
family: structural and sequence overview. Biochim Biophys Ac ta
2000, 1482:924.
358. Kjeldsen L, Johnsen AH, Sengelov H and Borregaard N: Isolation
and primary structure of NGAL, a novel protein associated
with human neutrophil gelatinase. JBiolChem1993,
268:10425 32.
359. Stoesz SP a nd Gould MN: Overexpre ssion of neu-related
lipocalin (NRL) in neu-initiated but not ras or chemi cally
initiated rat mammary carcinomas. Oncogene 1995,
11:223341.
360. Kjeldsen L, Cowland JB and Borregaard N: Human n eutrophil
gelatinase-associated lipocalin and homologous p roteins in
rat and mouse. Biochim Biophys Acta 2000, 1482:272
83.
361. Liu Q, Ryon J and Nilsen-Hamilton M: Uterocalin: a mouse acute
phase protein e xpressed in the uterus around birth. Mol
Reprod Dev 1997, 46:50714.
362. Goetz DH, Holmes MA, Borregaard N, Bluhm ME, Raymond KN and
Strong RK: The neutrophil lipocalin NGAL is a bacteriostatic
agentthatinterfereswithsiderophore-mediated iron
acquisition. Mol Cell 2002, 10:103343.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 33 of 79
(page number n ot f or citation purposes)
363. Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK,
Akira S and Aderem A: Lipocalin 2 mediates an innate immune
response to bacterial infection by sequestrating ir on. Nature
2004, 432:91721.
364. Fluckinger M, Haas H, Merschak P, Glasgow BJ and Redl B: Human
tear lipocalin exhibits antimicrob ial activity by scavenging
microbial siderophores. Antimicrob Agents Chemother 2004,
48:336772.
365.HolmesMA,PaulseneW,JideX,RatledgeCandStrongRK:
Siderocalin (Lcn 2) also binds carboxymycobactins, poten-
tially defending against mycobacteri al infections through
iron sequestration. Structure 2005, 13:2941.
366. Berger T, Togawa A, Duncan GS, Elia AJ, Y ou-Ten A, Wak eham A,
Fong H E, Cheung CC and Mak TW: Lipocalin 2-d efici ent mice
exh ibit increased sensiti vity to Escherichia coli infection but
not to ischemi a-reperfusion injury. P roc Natl A cad Sci USA 2006,
103:18349.
367. Borregaard N and Cowland JB: Neutrophil gelatinase-associated
lipocalin, a siderophore-binding eukaryotic protein. Biometals
2006, 19:211215.
368. Y ang J, Goetz D, Li JY, Wang W, Mori K, Setlik D, Du T, Erdjument-
Bromage H, Tempst P, Strong R and Barasch J: An iron delivery
pathway mediated by a lipocalin. Mol Cell 2002, 10:104556.
369. Coles M, Diercks T, Muehlenweg B, Bartsch S, Zolzer V,
Tschesche H and Kessler H: The solution structure and
dynamics of human neutrop hil gelatinase-associated lipoca-
lin. JMolBiol1999, 289:13957.
370. T schesche H, Zolzer V, Triebe l S and Bartsch S: The human
neutrophil lipocalin supports the allosteric activation of
matrix metalloproteinases. Eur J Biochem 2001, 268:19181928.
371.YanL,BorregaardN,KjeldsenLandMosesMA:The high
mol ecular 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. JBiolChem2001, 276:3725865.
372. D evarajan P, Mishra J, Supavekin S, Patterson LT and Steven Po tter S:
Gene expression in early ischemic renal injury: clues
towards pathogenesis, biomarker discovery, and novel
therapeu tics. Mol Genet Metab 2003, 80:36576.
373. Mishra J, Ma Q, Prada A, Mitsnefes M, Z ahedi K, Yang J, Barasch J and
Devarajan P: Identification of neutrophil gelatina se-associated
lipocalin as a novel early urinary biomarker for ischemic
renal injury. J Am Soc Nephrol 2003, 14:253443.
374. Mishra J, Dent C, Tarabishi R, Mitsnefes MM, Ma Q, Kelly C, Ruff SM,
Zah edi K, Shao M, Bean J, Mori K, Barasch J and Devarajan P:
Neutrophil gelatinase-associated lipocalin (NGAL) as a
biomarker for acute renal injury a fter cardiac surgery. Lancet
2005, 365:12318.
375. Mori K, Lee HT, Rapoport D, Drexler IR, Foster K, Yang J, Schmidt-
Ott KM, Che n X, Li JY, Weiss S, Mishra J, Cheema FH, Markowitz G,
Suganami T, Sawai K, Mukoyama M, Kunis C, D'Agati V, Devara jan P
and Barasch J: Endocytic del ivery of lip occalin-siderophore-
iron complex rescues the kidney from ischemia-reperfusion
injury. Journal of Clinical Investigation 2005, 115:610621.
376. Herget-Rosenthal S: One step for ward in the early detection of
acute re nal failure. Lancet 2005, 365:12051206.
377. T rachtman H, Christen E, Cnaan A, Patri ck J, Mai V, Mish ra J, Jain A,
Bullington N and Devarajan P: Urinary neutrophil gelatinase-
associated lipocalcin in D+HUS: a n ovel marker of renal
injury. Pediatric Nephrology 2006, 21:989 994.
378. D ent CL, Ma Q, Dastrala S, Bennet t M, Mitsnefes MM, Barasch J and
Devarajan P: Plasma neutrophil gelatinase-associated lipoca-
lin predicts acute kidney injury, morbidity and mortal ity
aft er pediatric cardiac surgery: a prospective uncontrolled
cohort s tudy. Critical Care 2007, 11:R127.
379. Devarajan P: Update on mechanisms of ischemic acute kidney
injury. Journal of the American Society of Nephrology 2006,
17:15031520.
380. Schmid t-Ott KM, Mori K, Kalandadze A, Li JY, Paragas N, Nicholas T,
Devarajan P a nd Barasch J: Neutrophil gelatinase-associated
lipocalin-mediated iron traffic in kidney epithelia. Curr Opin
Nephrol Hypertens 2006, 15:4429.
381.Schmidt-OttKM,MoriK,LiJY,KalandadzeA,CohenDJ,
Devarajan P and Barasch J: Dual action of neutrophil gelati-
nase-associated lipocalin. J Am Soc Nephrol 2007, 18:40713.
382. Mori K and Nakao K: Neut rophil gelatinase-associated lipoca-
lin as the real-time indicator of active kidney damage. Kidney
Int 2007, 71:967
70.
383. Mitsnefes MM, Kathman TS, Mishra J, Kartal J, Khoury PR,
Nickolas TL, Barasch J and Devarajan P: Serum neutrophil
gelatinase-associated lipocalin as a marker of renal function
in children with chronic kidney di sease. Pediatr Nephrol 2007,
22:1018.
384. Zappitelli M, Washburn KK, Arikan AA, Loftis L, Ma Q, Devarajan P,
Parikh CR and Goldstein SL: Urine neutro phil gelatinase-
associated lipocalin is an early marker of acute kidney
injury in critically ill children: a prospective cohor t s tudy.
Critical Care 2007, 11:.
385. Molitoris BA, Melnikov VY, Okusa MD and Himmelfarb J: Technol-
ogy Insight: biomarker development in acute kidney injury
what can we anticipate? Nat Clin Pr act Nephrol 2008, 4:15465.
386. Nickolas TL, O'Rourke MJ, Yang J, Sise ME, Canetta PA, Barasch N,
Buchen C, Khan F, Mori K, Giglio J, Devarajan P and 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, 148:8109.
387. Malyszko J, Bachorzewska-Gajewska H, Sitniewska E, Malys zko JS,
Poniatowski B and Dobrzycki S: Serum neutrophil gelatinase-
associated lip ocalin as a marker o f rena l function in non-
dia betic patients with stage 24 chronic kidney disease. Ren
Fail 2008, 30:6258.
388. Devarajan P: NGAL in acute kidney injury: from serendipity to
utility. Am J Kidney Dis 2008, 52:3959.
389. Devarajan P: Neutrophil gelatinase-associated lipocalin
(NGAL): a new marker of kidney disease. Scand J Clin Lab
Invest Suppl 2008, 241:8994.
390. D evire ddy LR, Gazin C, Zhu X and Green MR: Acell-surface
receptor for lipocalin 24p3 selectively mediates apoptosis
and iron uptake. Cell 2005, 123:1293305.
391. Richardson DR: 24p3 and its receptor: dawn of a new iron age?
Cell 2005, 123:11757.
392. Hvidberg V, Jacobsen C, Strong RK, Cowland JB, Moestrup SK and
Borregaard N: The endocytic receptor megalin binds the iron
tra nsporting n eutro phil-gelatinase -associated lipocalin with
high affinity and mediates its cellular uptake. FEBS Lett 2005,
579:7737.
393. R oudke nar MH, Kuwahara Y, Baba T, Roushandeh AM, Ebishima S,
Abe S , Ohkubo Y and Fukumot o M: Oxidative stress induced
lipocalin 2 gene expression: Addressing its expression under
the harmful conditions. Journal of Radiation Research 2 007,
48:3944.
394. R oudke nar MH, Hal abian R, Ghasemipour Z, Roushandeh AM,
Rouhbakhsh M, Nekogoftar M, Kuwahara Y, Fukumoto M and
Shokrgozar MA: Neutrophil Gelatinase-associated Lipocalin
acts as a protective factor against H
2
O
2
toxicity. Arch Med Res
2008, 39:5606.
395. Gueler F, Gwinner W, Schwarz A and Haller H: Long-term effects
of acute ischemia and reperfusion injury. Kidney International
2004, 66:523527.
396. Sikorski EM, Hock T, Hill-Kapturczak N and Agarwal A: The story
so fa r: molecular regulation of the heme oxy genase-1 gene
in renal injury. Am J Physiol 2004, 286:F425F441.
397. Shah S: Role of iron in progressive renal disease. Am J Kidney Dis
2001, 37:S303.
398. Aigner F, Maier HT, Schwelberger HG, Wallnofer EA, Amberger A,
Obrist P, Berger T, Mak TW, Maglione M, Margreiter R,
Sch neeberger S and Troppmair J: Lipocalin-2 regula tes the
inflammatory response during ischemia and reperfusion of
the transplanted heart. Amer J Transplantation 2007, 7:779788.
399. Abergel RJ, Moore EG, Strong RK and Raymond KN: Microbial
eva sion of the immune system: structural modifications of
enteroba ctin impair siderocalin recognition. J Am Chem Soc
2006, 128:10998 9.
400. Fischbach MA, Lin H, Zhou L, Yu Y, Abergel RJ, Liu DR,
Raymond KN, Wanner BL, Strong RK, Walsh CT, Aderem A and
Smith KD: The pathogen-associated iroA gene cluster med-
iates bacterial evasion of lipocalin 2. Proc Natl Acad Sci USA
2006, 103:16502 7.
401. Abergel RJ, Clifton MC, Pizarro JC, Warner JA, Shuh DK, Strong RK
and Raymond KN: The siderocalin/enterobactin interaction: a
link between mammalian immunity and bacterial iron
transport. JAmChemSoc2008, 130:1152434.
402. Cowland JB and Borregaard N: Molecular characterization and
pattern of tissue expression of the gene for neutrophil
gelatinase-associated lipocalin from humans. Genomics 1997,
45:1723.
403. Xu S and Venge P: Lipocalins as biochemical markers of
disease. Biochim Biophys Acta 2000, 1482:29 8307.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 34 of 79
(page number n ot f or citation purposes)
404. Cowland JB, Sorensen OE, Sehested M and Borregaard N:
Neutrophil gelatinase-associated lipocalin is up-regulated
in human epithelial cells by IL-1 beta, but n ot by TNF-alpha.
J Immunol 2003, 171:66309.
405. Cowland JB, Muta T and Borregaard N: IL-1beta-specific up-
regulation of neutrophil gelatinase-asso ciated lipoc alin is
controll ed by IkappaB-zeta. J Immuno l 2006, 176:555966 .
406. Elneihoum AM, Falke P, Axelsson L, Lundb erg E, Lindgarde F and
Ohlsson K: Leukocyte activation detected by increased
pla sma levels of inflammatory mediators in patients with
ischemic cerebrovascular diseases. Stroke 1996, 27:17348.
407. Elneihoum AM, Falke P, Hedblad B, Lindgarde F and Ohlsson K:
Leukocyte activation in atherosclerosis: correlation wit h
risk factors. Atherosclerosis 1997, 131:7984.
408. Falke P, Eln eihoum AM an d Ohlsson K: Leukocyte activation:
relation to cardiovascular mortality after cerebrovascular
ischemia. Cerebrovasc Dis 2000, 10:97101.
409. Hemdahl AL, Gabrielsen A, Zhu C, Eriksson P, Hedin U, Kastrup J,
Thoren P and Hansson GK: Expression of neutrophil gelatinase-
associated lipocalin in atherosclerosis and myocardial
infarction. Arterioscler Thromb Vasc Biol 2006, 26:136 42.
410.StoeszSP,FriedlA,HaagJD,LindstromMJ,ClarkGMand
Gould MN: Heterogeneous expression of the lipocalin
NGAL in primary breast cancers. Int J Cancer 1998, 79:56572.
411. T ong Z, Wu X, Ovcharenko D, Zhu J, Chen CS a nd Kehrer JP:
Neutrophil gelatinase-associated lipocalin as a survival
factor. Biochem J 2005, 391:441 8.
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 and Guha S:
Neutrophil gelatinase-associated lipocalin: a novel suppres-
sor of invasion and angiogenesis in pancreatic ca ncer. Cancer
Res 2008, 68 :61008.
413. Weinberg ED: Iron out-of-balance: a r isk fac tor for acute and
chr onic disea ses. Hemoglobin 2008, 32:11722.
414. CESDI: Confidential Enquiry into Stillbirths and Deaths in
Infancy. 5th Annual Report Maternal and Child Health Research
Consortium; 1998.
415. LaMarca BD, Gilbert J and Granger JP: Recent progress toward
the understanding of the pathophysiology of hypertension
during preecl ampsia. Hypertension 2008, 51:9828.
416. R edman CW and Sargent IL: Placental debris, oxidative stre ss
and pre-eclampsia. Placenta 2000, 21:597602.
417. DiFederico E, Genbacev O and Fisher SJ: Preeclampsia is
associated with widespr ead apoptosis of placental cytotro-
phoblasts within the uterine wall. Am J Pathol 1999,
155:293301.
418. Leung DN, Smith SC, To KF, Sahota DS and Baker PN: Increased
pla cental apoptosis in pregnancies complicated by pree-
clampsia. Am J Obstet Gynecol 2001, 184:124950.
419. Huppertz B a nd Kingdom JC: Apoptosis in the trophoblastrole
of apoptosis in placental morphogenesis. J Soc Gynecol Investig
2004, 11:35362.
420. Harris LK, Keogh RJ, Wareing M, Bak er PN, Cartwright JE, Aplin JD
and Whitley GS: Invasive trophoblasts stimulate vascular
smooth muscle cell apoptosis by a fas ligand-dependent
mechanism. Am J Pathol 2006, 169:186374.
421. Heazell AE, Moll SJ, Jones CJ, Baker PN and Crocker IP: Formation
of syncytial knots is increased by hyperoxia, hypoxia and
reactive oxygen species. Placenta 2007, 28(Suppl A):S3340.
422. Keogh RJ, Harris LK, Freeman A, Baker PN, Aplin JD, Whitley GS
and Cartwright JE: Fetal-de rived tro phoblast u se t he apoptot ic
cytokin e tumor ne cr osi s factor-alpha-related apoptosis-
inducing ligand to induce smooth muscle cel l death. Circ
Res 2007, 10 0:83441.
423. Heazell AE, Lacey HA, J ones CJ, H uppertz B, Baker PN and
Crocker IP: Effects of oxygen on cell tu rnover and e xpression
of regulators of apoptosis in human placental trophoblast.
Placenta 2008, 29:17586.
424. Combs CA, Katz MA, Kitzmiller JL and Brescia RJ: Experimental
preeclam psia pr oduced by chronic constriction of t he lower
aorta: validation with longitudinal blood pressure measure-
ments in conscious rhesus monkeys.
Am J Obstet Gynec ol 1993,
169:21523.
425. Wisdom SJ, Wilson R, McKillop JH and Walker JJ: Antioxidant
systems in normal pregnancy and in pregnancy-induced
hypertension. Am J Obs Gynecol 1991, 165:17011704.
426. Genbacev O, Joslin R, Damsky CH, Polliotti BM and Fisher SJ:
Hypoxia alters early gestation human cytotrophoblast
differentiation/invasion in vitro and models the pl acental
defects t hat occur in preeclampsia. J Clin Invest 1996,
97:54050.
427. Be ny o DF, Mi le s TM and Conra d KP: Hypoxia stimulates
cytokine production by villous explants from the human
placenta. J Clin Endocrinol Me tab 1997, 82:15828.
428. Conrad KP and Benyo DF: Placental cytokines and the
pathogenesis of preeclampsia. Am J Reprod Immunol 1997,
37:2409.
429. Lefevre G, Berkane N, Uzan S and Etienne J: Preeclampsia and
oxygen fr ee radicals. Annales De Biologie Clinique 1997,
55:443450.
430. Conrad KP, Miles TM and Benyo DF: Circulating levels of
immunoreactive cytokines in women with pr eeclampsia. Am
J Reprod Immunol 1998, 40:102 11.
431. D avidge ST: Oxidative stress and altered endothelial cell
function in preeclampsia. SeminReprodEndocrinol1998,
16:6573.
432. Mise H, Sagawa N, Matsumoto T, Yura S, Nanno H, Itoh H, Mori T,
Mas uzaki H, Hosoda K, Ogawa Y and Nak ao K: Augmente d
pla cental production of leptin in pr eeclampsia: possible
involvement of placental hypoxia. JClinEndocrinolMetab1998,
83:32259.
433. Walker JJ: Antio xidants and inflammatory cell r esponse in
preeclam psia. SeminReprodEndocrinol1998, 16:4755.
434. W a l sh SW: Maternal-plac ental i nteractions of oxidative
stress and antioxidan ts in preeclampsia. Seminars in Reproduc-
tive Endocrinology 1998, 16:93
104.
435. Caniggia I, Grisaru-Gravnosky S, Kuliszewsky M, Post M and Lye SJ:
Inhibition of TGF -beta 3 restores the invasive capability of
extravil lous trophoblasts in preeclamptic pr egnancies. JClin
Invest 1999, 103:164150.
436. Hubel CA: Oxidative stress in the pathogenesis of pree-
clampsia. Proc Soc Exp Biol Med 1999, 222:22235.
437. R inehart BK, Terrone DA, Lagoo-Deenadayalan S, Barber WH,
Hale EA, Martin JN Jr and Bennett WA: Expression of the
pla cental cytokines tumor necrosis factor alpha, interleukin
1be ta, and interleukin 10 is increased in preeclampsia. Am J
Obstet Gynecol 1999, 181:915 20.
438. Caniggia I, M ostachfi H, Winter J, Gassmann M, Lye SJ, Ku liszewski M
and Post M: Hypoxia-inducible facto r-1 mediates the biologi-
cal effects of oxygen on human trophobl ast differentiation
through TGFbeta(3). J Clin Invest 2000, 105:577 87.
439. Kharb S: Total free radical trapping antio xidant poten tial i n
pre-eclampsia. Int J Gynaecol Obstet 2000, 69:236.
440. T álosi G, Endreffy E, Túri S and Németh I: Molecular and genetic
aspects of preeclampsia: State of the art. Mol Genet Metab
2000, 71:565572.
441. Walsh SW, Vaughan JE, Wang Y and Roberts LJ 2nd: Placental
isoprostane is significantly increased in preeclampsia. FASEB J
2000, 14:128996.
442. Wolf M, Kettyle E, Sandler L, Ecker JL, Roberts J and Thadhani R:
Obesity and preeclampsia: the potential role of inflamma-
tion. Obstet Gynecol 2001, 98:75762.
443. Benyo DF, Smarason A, Redman CW, Sims C and Conrad KP:
Expression of inflammatory cytokines in placentas from
womenwithpreeclampsia.J Clin Endocrinol Metab 2001,
86:250512.
444. Hung TH, Skepper JN and Burton GJ: In vitro ischemia-
reperfusion in jury in term human placenta as a model for
oxidative stress in pathological pregnancies. Am J Pathol 2001,
159:103143.
445. pez-Jaramillo P, Casas JP and Serrano N: Preeclamp sia: from
epidemiological observations to molecular mechanisms.
Braz J Med Biol Res 2001, 34:122735.
446. Malek A, Sager R and Schneider H: Effect of hypoxia, oxidati ve
stress and li popolysacch arides on the release of prostaglan-
dins and cytokines from human term placental expla nts.
Placenta 2001, 22(Suppl A):S4550.
447. Gitto E, Reite r RJ, Karbownik M, Tan DX, Gitto P, Barberi S and
Barberi I: Causes of oxidative stress in the pre- and perinatal
period. Biology of the Neonate 2002, 81:146157.
448. Granger JP, Alexander BT, Llinas MT, Bennett WA and Khalil RA:
Pathophysiology of preeclampsia: linking placental ische-
mia/hypoxia with m icrovascular dysfunction. Microcirculation
2002, 9:14760.
449. Roberts JM and Lain KY: Recent Insights into the pathogenesis
of pre-eclampsia. Placenta 2002, 23:35972.
450. Vaughan JE and Walsh SW: Oxidative stress reproduces
pla cental abnormaliti es of preecl ampsia. Hypertens P reg 2002,
21:205223.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 35 of 79
(page number n ot f or citation purposes)
451. Burton GJ and Jau niaux E: Placental oxidative stress: from
miscarriage to preeclampsia. J Soc Gynecol Investig 2004,
11:342352.
452. Connors N and Merrill D: Antioxidants for prevention of
preterm delivery. Clin Obs G ynecol 2004, 47: 822832.
453. R aijmakers MT, Peters WH, Steegers EA and Po ston L: Amino
thiols, detoxific ation and oxidative stress in pre-eclampsia
and other disorders of pregnancy. Curr Pharm Des 2005,
11:71134.
454. R edman CW an d Sargent IL: Pre-eclampsia, the placenta and
the matern al systemic inflammator y response a review.
Placenta 2003, 24(Suppl A):S217.
455. Downing JW, Ramasubramanian R, Johnson RF, Minzter BH,
Pas chall RL, Sundell HW, Engelhardt B and L ewis R: Hypothesis:
selective phosphodiesterase-5 inhibition improves outcome
in preeclampsia. Med Hypotheses 2004, 63:105764.
456. Myatt L and Cui XL: Oxidative stress in the placenta. Histochem
Cell Biol 2004, 122:369382.
457. Touyz RM and Schiffrin EL: Reactive oxygen sp ecies in vascular
biology: implications in hypertension. Histochem Cell Biol 2004,
122:33952.
458. Touyz RM: R eactive oxygen species, vascular oxidative stress,
and redox signaling in hypertension: what is the clinical
significance? Hypertension 2004, 44:24852.
459. T sukahara H, Ohta N, Sato S, Hiraoka M, Shukunami K, Uchiyama M,
Kawakami H, Sekine K and Mayumi M: Concentrations of
pentosidine, an advanced glycation end-product, in umbili-
cal cor d blood. Free Radic Res 2004, 38:691 5.
460. Bdolah Y, Karumanchi SA and Sachs BP: Recent advances in
understanding of preeclampsia. Croat Med J 2005, 46:72836.
461. Biondi C, Pavan B, Lunghi L, Florini S and Vesce F: The role and
modulation of the oxidative balance in pregnancy. Curr
Pharmaceut Design 2005, 11:20752089.
462. Engin-Üstün Y, Üstün Y, Kamaci M and Şekeroğlu R: Maternal
serum ceruloplasmin in preectampsia. Int J Gynecol Obs 2005,
89:5152.
463. Gupta S, Agarwal A and Sharma RK: The role of placental
oxidative stress and lipid peroxidation in preeclampsia.
Obstet Gynecol Surv 2005, 60:80716.
464. Noris M, Perico N and Remuzzi G: Mechanisms of disease: pre-
eclampsia. Nature Clin Pract Nephrol 2005, 1:98114.
465. Redman CW and Sargent IL: Latest advances in understanding
preeclam psia. Science 2005, 308:15921594.
466. Roberts JM and Gammill HS: P reeclampsia: recent insights.
Hypertension 2005, 46:12439.
467. R odrigo R, Parra M, Bosco C, Fernandez V, Barja P, Guajardo J and
Messina R: Pathophysiological basis for th e prophylaxis of
preeclam psia through early supplementation with antiox-
idant vitamins. Pharmacology & Therapeutics 2005, 107:177197.
468. Scholl TO, Leskiw M, Chen X, Sims M and Stein TP: Oxidative
stress, diet, and the etiology of preeclampsia. Am J Clin Nutr
2005, 81:13906.
469. Soleymanlou N, Jurisica I, Nevo O, Ietta F, Zhang X, Zamudio S,
Post M and Caniggia I: Molecul ar evidence of placental hypoxia
in preeclampsia. J Clin Endocrinol Metab 2005, 90:42994308.
470. Spinnato JA and Livingston JC: Prevention of pre eclampsia with
antioxidants: Evidence from randomized trials. Clinical
Obstetrics and Gynecology 2005, 48:416429.
471. Blackb urn S : F ree radicals in perinatal and neonatal care, part
2 Oxidative stress du ring the perinatal and neonatal
period. Journal of Perinat al & Neonatal Nursing 2006,
20:125127.
472. Borzychowski AM, Sa rgent IL and Redman CW: Inflammation and
pre-eclampsia. Semin Fetal Neonatal Med 2006, 11:30916.
473. Hauguel-de Mouzon S and Guerre-Millo M: The placenta c ytokine
network and inflammatory signals. Placenta 2006, 27:794 8.
474. P erk in s A V : Endogenous ant i-oxi dant s i n pregnancy an d
preeclam psia. Aus New Zeal J Obs Gynaecol 20 06, 46:7783.
475. R ogers MS, Wang CC, Tam WH, Li CY, Chu KO and Chu CY:
Oxidative stress in midpregnancy as a predictor of gesta-
tional hypertension and pre-eclampsia. BJOG 2006,
113:10539.
476. Sankaralingam S, Arenas IA, Lalu MM and Davidge ST: Preeclamp-
sia: current understanding of the molecular basis of vascular
dysfunct ion. Expert Rev Mol Med 2006, 8:120.
477. Sargent IL, Borzychowski AM and Redman CW: Immunoregula-
tion in normal pregnanc y and pre-eclampsia: an overview.
Reprod Biomed Online 2006, 13:6806.
478. Sharma JB, Sharma A, Bahadur A, Vimala N, Satyam A and Mittal S:
Oxidative stress markers and antioxidant levels in normal
pregnancy and pre-eclampsia. Int J Gynecol Obs 2006, 94:2327.
479. Webster RP, Brock man D and Myatt L: Nitration of p38 MAPK in
the placenta: association of nitration wit h reduced catalytic
activity of p38 MAPK in pre-eclampsia. Molecular Human
Reproduction 2006, 12:677685.
480. Burdon C, Mann C, Cindrova-Dav ies T, Ferguson-Smith AC and
Burton GJ: Oxidat ive stress and the in duction of cycloox-
ygenase enzymes and apoptosis in the murine placenta.
Placenta 2007, 28:72433.
481. Crocker I: Gabor Than Award Lecture 2006: pre-eclampsia
and villous trophoblast turnover: perspectives and possibi-
lities. Pla centa 2007, 28(Suppl A):S413.
482. Keelan JA and Mitchell MD: Placent al cytokines and preeclamp-
sia. Front Biosci 2007,
12:270627.
483. Milczarek R, Sokolowska E, Hallmann A and Klimek J: The
NADPH- and iron- dependent lipid peroxidation in human
pla cental microsomes. Molecular and Cellular Biochemistry 2007,
295:105111.
484. Schiessl B: Inf lammatory response in preecl ampsia. Mol Aspects
Med 2007, 28:2109.
485. Shah TJ and Walsh SW: Activation of NF-kappaB and
exp ression of COX-2 in association with neutrophil infiltra-
tion in systemic vascula r tis sue of women with preeclamp-
sia. Am J Obstet Gynecol 2007, 196:48 e18.
486. Walsh SW: Obesity: a risk factor for preeclampsia. Trends
Endocrinol Metab 2007, 18:36570.
487. Zamudio S: High-altitude hypoxia and preeclampsia. Frontiers in
Bioscience 2007, 12:29672977.
488. Gilbert JS, Ryan MJ, LaMarca BB, Sedeek M, Murphy SR and
Granger JP: Pathophysiology of hypertension durin g pree-
clampsia: linking placental ischemia with endothelial dys-
function. Am J Physiol Heart Circ Physiol 2008, 294:H54150.
489. R obinson NJ, Wareing M, Hudson NK, Blankley RT, Baker PN,
Aplin JD and Crocker IP: Oxygen and the liberation of placental
factors responsible for vascular compromise. Lab Invest 2008.
490. Entman SS, Richardson LD and Killam AP: Elevated serum ferritin
in the altered ferro kinetics of toxemia of pregnancy. AmJ Obs
Gynecol 1982, 144:418 422.
491. Entman SS and Richardson LD: Clinical applications of the
altered iron kinet ics of toxemia of pregnan cy. Am J Obs Gynecol
1983, 146:568574.
492. Entman SS, Richardson LD and Killam AP: Altered ferrokinetics in
toxemia of pregnancy a possible in dicator of dec reased red
cell survival . Cl in Exp Hypertens B 1983, 2:171178.
493. Entman SS, Kambam JR, Bradley CA and Cousar JB: Increased
levels of carboxyhemoglobin and serum iron as an indicator
of increased red cell turnover in preeclampsia. Am J Obstet
Gynecol 1987, 156:116973.
494. Samuels P, Main EK, Mennuti MT and Gabbe SG: The origin of
increased serum iron in pregnancy-induced hypertension.
Am J Obstet Gynecol 1987, 157:7215.
495. Lindheimer MD and Katz AI: Preeclampsia Patho -physiology,
dia gnosis, and management. Annual Review of Medicine 1989,
40:233250.
496. R aman L, Pawashe AB and Yasodhara P: Hyperferritinemia in
pregnancy induced hypertension and eclampsi a. J Postgrad
Med 1992, 38:657.
497. Das S S, Dhall GI, Dhall K and Dash S: Significance of serum iron
levels as a biochemical marker in pregnancy-induced
hypertension. Int J Gynecol Obs 1994, 45:39.
498. Hubel CA, Kozlov AV, Kagan VE, Evans RW, Davidge ST,
McLaughlin MK and Roberts JM: Decreased transferrin and
increased transferrin saturation in sera of women with
preeclam psia: implications for oxidative stress. Am J Obstet
Gynecol 1996, 175:692 700.
499. Sizoo BB, Paarlberg MM, Bouman AA and Dekker GA: The role of
serum iron levels in diagnostic hypertensive disorders in
pregnancy. Hypertension in Pregnancy 1997, 16:425 433.
500. Vitoratos N, Salamalekis E, Dalamaga N, Kassanos D and Creatsas G:
Defective antioxidant mechanisms via changes in serum
ceruloplasmin and total iron binding capacity of serum in
women with pr e-eclampsia. Eur J Obstet Gynecol Reprod Biol 1999,
84:637.
501. Lao TT, Tam KF and Chan LY: Third trimester iron status and
pregnancy outcome in non-anaemic women; pregnancy
unfavour ably affected by maternal iron excess. Hum Reprod
2000, 15:18438.
502. Waterstone M, Bewley S and Wolfe C: Incidence and predictors
of severe obstetric morbidity: case-control study. BMJ 2001,
322:108993.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 36 of 79
(page number n ot f or citation purposes)
503. Mello G, Parre tti E, Cioni R, Lagozio C, Mealli F and Pratesi M:
Individual longit udinal patterns in biochemical and hemato-
logical markers for the early prediction of pre-eclampsia.
J Matern Fetal Neonatal Med 2002, 11:939.
504. Casanueva E and Viteri FE: Iron and oxidative stress in
pregnancy. JNutr2003, 133:1700S1708S.
505. Hubel CA, Bodnar LM, Many A, Harger G, Nes s RB and Roberts JM :
Nonglycosylated ferritin predominates in the circulation of
women wi th preeclampsia but not intrauterine growth
restriction. Clin Chem 2004, 50:948951.
506. Norwitz ER, Tsen LC, Park JS, Fitzpatrick PA, Dorfman DM,
Saade GR, Buhimschi CS and Buhimschi IA: Discriminatory
proteomic biomarker analysis identifies free hemoglobin in
the cerebros pinal fluid of women with severe preecla mpsia.
Amer J Obs Gynecol 2005, 193:957964.
507. Scholl TO: Iron status during pregnancy: setting the stage for
mother and infant. Am J Clin Nutr 2005, 81:1218S1222S.
508. Serdar Z, Gür E and Develioğlu O: Serum iron and copper
status and oxidative stress in severe and mild preeclampsia.
Cell Biochem Funct 2006, 24:20915.
509. Smith TG and Robbins PA: Iron, pre-eclamps ia and hypoxia-
inducibl e factor. Bjog 2007, 114:15812.
510. Ziaei S, Nor rozi M, Faghihzadeh S and Jafarbegloo E: A randomised
pla cebo-controlled trial to determine the effect of ir on
supplementation on pregnancy outcome in pregnant
women with haemoglobin > or = 13.2 g/dl. BJOG 2007,
114:6848.
511. Guller S, Buhimschi CS, Ma YY, Huang ST, Yang L, Kuczynski E,
Zambrano E, L ockwood CJ and Buhimschi IA: Placental expres-
sion of ceruloplasmin in pregnancies complicated by severe
preeclam psia. Lab Invest 2008, 88:105767.
512.GamblingL,DanzeisenR,GairS,LeaRG,CharaniaZ,SolankyN,
Joory KD, Srai SKS and McArdle HJ: Effect of iron deficiency on
pla cental transfer of iron and expression of iron transport
proteins in vivo and in vitro. Biochem J 2001, 356:
883889.
513. Freeman DJ, McManus F, Brown EA, Cherry L, Norrie J, Ramsay JE,
Clark P, Walker ID , Sattar N and Gre er IA: Short- and long-term
changes in plasma inflammatory markers associated with
preeclam psia. Hypertension 2004, 44:70814 .
514. Bodnar LM, Ness RB, Harger GF and Roberts JM: Inflammation
and triglyc erides partially mediate the effect of prepreg-
nancy body ma ss index on the risk of preeclampsia. Am J
Epidemiol 2005, 162:1198206.
515. Braekke K, Holthe MR, Harsem NK, Fagerhol MK and Staff AC:
Calprotectin, a marker of inflammation, is elevated in the
maternal but not in the fetal circulation in preeclampsia. Am
JObstetGynecol2005, 193:227 33.
516. Hu W, Wang H, Wang Z, Huang H and Dong M: Elevat ed serum
levels of interleukin-15 and interleukin-16 in preeclampsia.
J Reprod Immunol 2007, 73:166 71.
517. Keelan JA and Mitchell MD: Cytokines, hyp oxia, and pree-
clampsia. J Soc Gynecol Investig 2005, 12:3857.
518. Elovitz MA: Anti-inflammatory interventions in pregnancy:
now and the future. Semin Fetal Neonatal Med 2006, 11:32732.
519. Patern oster DM, Fantinato S, Stella A, Nanhorngue KN, Milani M,
Plebani M, N icolini U and Girolami A: C-reactive protein in
hypertensive disorders in pregnancy. Clin Appl Thromb Hemost
2006, 12:3307.
520. D orrepaal CA, Berger HM , Benders M, vanZoerenGrobben D,
VanDeBor M and VanBel F: Nonprotein-bound iron in post-
asphyxia l reperfusion injury of the newborn. Pediatrics 1996,
98:883889.
521. Goldenberg RL, Tamura T, DuBard M, Johnston KE, Copper RL and
Neggers Y: Plasma ferritin and pregnancy outcome. Am J Obs
Gynecol 1996, 175:13561359.
522. T amura T, Goldenberg RL, Johnston KE, Cliver SP and Hickey CA:
Serum ferritin: A predictor of early spontaneous preterm
delivery. Obs Gynecol 19 96, 87:360 365.
523. Buhimschi IA, Buhimschi CS and Weiner CP: Protective effect of
N-acetylcys teine against fetal death and preterm labor
induced by maternal inflammation. American Journal of Obstetrics
and Gynecology 2003, 188:203208.
524. Ciccoli L, Rossi V, Leoncini S, Signor ini C, Paffetti P, Bracci R,
Buonocore G and Comporti M: Iron release in erythrocytes and
pla sma non protein-bound iron in hypoxic and non hypoxic
newborns. Free Radical Research 2003, 37:5158.
525. Buhimschi IA, Saade GR, Chwalisz K and Garfield RE: The nitric
oxide pathway in pre-eclampsia: pathophysiological implica-
tions. Human Reproduction Update 1998, 4:2542.
526. Milman N: Iron and pregnancy a delicate balance. Annals of
Hematology 2006, 85:559 565.
527. Milman N: Iron prophylaxis in pregnancy-general or indivi-
dual and in which dose? Annals of Hematology 2006, 85:821828.
528. Lachili B, Hininger I, Faure H, Arnaud J, Richard MJ, Favier A and
Roussel AM: Increa sed lipid peroxidation in pregnant women
aft er iron and vitamin C supplementation. Biol Trace Elem Res
2001, 83:10310.
529. D evrim E, Tarhan I, Ergüder IB and Durak I: Oxidant/antioxidant
status of placenta, blood, and cord blood samples from
pregnant women supplemented with iron. J Soc Gynecol Investig
2006, 13:5025.
530. Keyes LE, Armaza JF, Niermeyer S, Vargas E, Young DA an d
Moore LG: Intrauterine growth restriction, pr eeclampsia,
and intrauterine mortality at high altitude in Bolivia. Pediatric
Research 2003, 54:20 25.
531. Teramo KA, Hiilesmaa VK, Schwartz R, Clemons GK and
Widness JA: Amniotic fluid and cord plasma erythropoietin
levels in pregnan cies complicated by preeclampsia, preg-
nancy-induced hyper tension and chronic hypertension. J
Perinat Med 2004, 32:2407.
532. Liang ST, Wong VC, So WW, Ma HK, Chan V and Todd D:
Homozygous alpha-thalassaemia: clinical presentation,
dia gnosis and management. A review of 46 cases. Br J Obstet
Gynaecol 1985,
92:6804.
533. Tungwiwat W, Fucharoen S, Fucharoen G, Ratanasiri T and
Sanchaisuriya K: Development and application of a real-time
quantitative PCR for prenatal detection of fetal alpha(0)-
thalassemia from maternal plasma. AnnNYAcadSci2006,
1075:1037.
534. Senden IP, de Groot CJ, Ste egers EA, Bertina RM and Swinkels DW:
Preeclampsia and the C282Y mutation in the hemochro -
matosis (HFE) gene. Clin C hem 2004, 50:9734.
535. Quek L and Thein SL: Molecular therapies in beta-thalassae-
mia. British Journal of Haematology 2007, 136:353365.
536. Knox KS and Bake r JC: Genome-wide express ion profiling of
placentas in the p57Kip2 model of pre-eclampsia. Mol Hum
Reprod 2007, 13:25163.
537. Suire S, Stewart F, Beauchamp J and Kennedy MW: Uterocalin, a
lipocalin provisioning the preattachment equi ne conceptus:
fat ty acid and retinol binding properties, and structural
characterization. Biochem J 2001, 356:36976.
538. R yon J, Bendickson L and Nilsen-Hamilton M: High expression in
involuting reproductive tissues of uterocalin/24p3, a lipoca-
lin and acute phase protein. Biochem J 2002, 367:2717.
539. Nilsen-Hami lton M, Liu Q, Ryon J, Bendickson L, Lepont P and
Chang Q: Tissue involution and the acute phase response. Ann
N Y Acad Sci 2003, 995:94108.
540. D 'Anna R, Baviera G, Giordano D, Tod arell o G, Corrado F and
Buemi M: Second trimester neutrophil gelatinase-associated
lipocalin as a potential prediagnostic marker of preeclamp-
sia. Acta Obstet Gynecol Scand 2008, 14.
541. Coughl an MT, P ermezel M, Georgiou HM and Rice GE: Repression
of oxidant-induced nuclear factor-kappaB activity mediates
pla cental cytokine responses in gestational diabetes. JClin
Endocrinol Metab 2004, 89:358594.
542. Coughl an MT, Vervaart PP, Permezel M, Georgiou HM and Rice GE:
Altered placental oxidative stress status in gestational
dia betes mellitus. Placenta 2004, 25:
7884.
543. King JC: Maternal obesity, metabolism and pregnancy out-
comes. Annu Rev Nutrition 2006, 26:271291.
544. Mazar RM, Srinivas SK, Sammel MD, Andrela CM and Elovitz MA:
Metabolic score as a novel approach to assessing pre-
eclampsia risk. Am J Obstet Gynecol 2007, 197:411 e15.
545. R ademacher TW, Gumaa K and Sc ioscia M: Preeclampsia, insulin
signalling and immunological dysfunction: a fetal, maternal
or placental disorder? J Reprod Immunol 2007, 76:7884.
546. Hales CN and Ozanne SE: For Debate: Fetal and early postnatal
growth restriction lead to diabetes , the metabolic syndrome
and renal failure. Diabetologia 2003, 46:10131019.
547. McMillen IC and Robinson JS: Developmental origins of the
metabolic syndrome: prediction, plastic ity, and program-
ming. Physiol Rev 2005, 85:571 633.
548. Williams PJ, Gumaa K, Scioscia M, Redman CW and
Rademacher T W: Ino sitol phospho glycan P-type in preeclamp-
sia: a novel marker? Hypertension 2007, 49:849.
549. West IC: Radicals and oxidative str ess in diabetes. Diabet Med
2000, 17:17180.
550. Lipinski B: Pathophysiology of oxidative stress in diabetes
mellitus. J Diabetes Complications 2001, 15:20310.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 37 of 79
(page number n ot f or citation purposes)
551. Maddux BA, See W, Lawrence JC Jr, Goldfine AL, Goldfine ID and
Evans JL: Protec tion against oxidative stress-induced insulin
resistan ce in rat L6 muscle cells by mircomolar concentra-
tions of alpha-lipoic acid. Diabetes 2001, 50:40410.
552. Evans JL, Maddux BA and Goldfine ID: The molecu lar basis for
oxidative stress-induced insulin resis tance. Antioxid Redox Signal
2005, 7:104052.
553. Houstis N, Rosen ED and Lander ES: Reactive oxygen species
have a causal role in multiple forms of insulin resistance.
Nature 2006, 440:944 8.
554. Meigs JB, Larson MG, Fox CS, Keaney JF Jr, Vasan RS and
Benjamin EJ: Ass ociation of oxidative stress, insulin res istance,
and diabetes risk phenotypes: the Framingham Offspring
Study. Diabetes Care 2007, 30:252935.
555. Shah S, Iqbal M, Karam J, Salifu M and McFarlane SI: Oxidative
stress, glucose metabolism, and the prevention of type 2
diabetes: Pat hophy siological insights. Antioxid Redox Signal
2007, 9:911929.
556. Choi S W, Benzie IF, Ma SW, Strain JJ and Hannigan BM: Ac ute
hyperglycemia and oxidative stress: direct cause and effect?
Free Radic Biol Med 2008, 44:121731.
557. Nishikawa T, Ede lstein D, Du XL, Yamagis hi S, Matsumura T,
Kan eda Y, Yor ek MA, Beebe D, Oates PJ, Hammes HP, Giardino I
and Brownlee M: Normalizing mitochondrial superoxide
production blocks three pathways of hyperglycaemic
damage. Natur e 2000, 404:78790.
558. Evans JL, Goldfine ID, Maddu x BA a nd Grodsky GM: Oxidative
stress and stress-activated signaling pathways: A unifying
hypothesis of type 2 diabetes. Endocrine Reviews 2002,
23:599622.
559. Niedowicz DM and Daleke DL: The role of oxidative stress in
dia betic complications. Cell Biochem Biophys 2005, 43:289330.
560. Kelley DE, He J, Menshikova EV and Ritov VB: Dysfunction of
mitochondria in human skeletal muscle in type 2 diabetes.
Diabetes 2002, 51:294450.
561. Inoue I, Katayama S, Takahashi K, Negishi K, Miyazaki T, Sonoda M
and Komoda T: Troglitazon e has a scavenging effect on
reactive oxygen species. Biochem Biophys Res Commun 1997,
235:1136.
562. Fukui T, Noma T, Mizushige K, Aki Y, Kimura S and Abe Y: Dietary
troglitazone decreases oxidative stress in early stage type II
dia betic rats. Life Sci 2000, 66:20439.
563. Bao Y, Jia RH, Yuan J and Li J: Rosiglitazone ameliorates diabetic
nephropathy by inhibiting reactive oxygen species and i ts
downstream-signaling pathways. Pharmacology 2007, 80:5764.
564. Jung TW, Lee JY, Shim WS, Kang ES, Kim SK, Ahn CW, Lee HC and
Cha BS: Rosiglitazone protect s human neuroblastoma SH-
SY5Y cells against MPP+ induced cytotoxicity via inhibition
of mitochondrial dysfunction and ROS production. JNeurolSci
2007, 253:5360.
565. Jung Y, Song S and Choi C: Peroxisome proliferator activate d
receptor gamma agonists suppress TNFalpha-induced
ICAM-1 expression b y end othelial cells in a manner
potentially dependent on inhibition of reactiv e oxygen
species. Immunol Lett 2008, 117:639.
566. Allahtavakoli M, Shabanzadeh AP, Sadr SS, Parviz M and
Djahangu iri 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, 33:10528.
567. Collino M, Aragno M, Mastrocola R, Benetti E, Gallicchio M,
Dia nzani C, Danni O, Thiemermann C and Fantozzi R: Oxidative
stress and inflammatory response evoked by transient
cerebral ischemia/reperfusion: effects of the P PAR-alpha
ago nist WY14643. Free Ra dic Biol Med 20 06, 41:579 89.
568. Collino M, Aragno M, Mastrocola R, Gallicchio M, Rosa AC,
Dia nzani C, Danni O, Thiemermann C and Fantozzi R: Modulation
of the oxidative stress and inflammatory r esponse by PPAR-
gamma agonists in the hippocampus of rats exposed to
cerebral ischemia/reperfusion. Eur J Pharmacol 2006, 530:7080.
569. Allahtavakoli M, Shabanzadeh A, Roohbakhsh A and Pourshanazari A:
Combination therapy of rosiglitazone, a peroxisome pro-
liferator-activated receptor-gamma ligand, and NMDA
receptor antagonist ( MK-801) on experimental emb olic
stroke i n rats. Basic Clin Pharmaco l Toxicol 2007, 101:30914.
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,
Sch ork NJ, Schmid-Schon bein GW and O'Connor DT: C-reactive
protein, an 'intermedi ate phenotype' for inflammat ion:
human twin studies reveal heritability, associat ion with
blood pressure and the met abolic syndrome, and the
influence of common polymorphism at catecholaminergic/
beta-adrenerg ic pathway loci. J Hyperten s 2007, 25:32943.
571. Baynes JW and Thorpe SR: Role of oxidative stress in diabetic
complications: a new perspective on an old paradigm.
Diabetes 1999, 48:19.
572. Hu FB, Meigs JB, Li TY, Rifai N and Manson JE: Inflammatory
markers and risk of developing type 2 diabetes in women.
Diabetes 2004, 53:693700.
573. Pickup JC: Inflammation and activated innate immunity in the
pathogenesis of type 2 diabetes. Diabetes Care 2004, 27:81323.
574. Kolb H and Mandrup-Poulsen T: An immune origin of type 2
dia betes? Diabetologia 2005, 48:103850.
575. Lee YH and Pratley RE: The evolving role of inflammation in
obesity and the metabolic syndrome. Curr Diab Rep 2005,
5:705.
576. Tataranni PA and Ortega E: A burning question: does an
adipokine-induced activation of the immune system med-
iate the effect of overnutrition on type 2 diabetes? Diabetes
2005, 54:91727.
577. Wellen KE and Hotamisligil GS: Inflammation, stress, and
dia betes. JClinInvest2005, 115:11119.
578.AlexandrakiK,PiperiC,KalofoutisC,SinghJ,AlaverasAand
Kalofoutis A: Inflammatory process in type 2 diabetes: The
role of cytokines. AnnNYAcadSci2006, 1084:89117.
579. Chen H: Cellular inflammatory r esponses: novel insights for
obesity and insulin resistance. Pharmacol Res 2006, 53:469 77.
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 and
Tuomilehto J: Systemic immune mediators and lifestyle
changes in t he preventio n of type 2 diabetes: results from
the Finnish Diabetes Prevention Study. Diabetes 2006,
55:23406.
581. Hotamisligil GS: Inflammation and metabolic disorders. Nature
2006, 444:8607.
582. Kempf K , Rose B, Herder C, Kleophas U, Martin S and Kolb H:
Inflammation in metabolic syndrome and type 2 diabetes:
Impact of dietary glucose. AnnNYAcadSci2006, 1084:3048.
583. Neels J G and Olefsky J M: Inflamed fat: what starts the fire? JClin
Invest 2006, 116:335.
584. Shoelson SE, Lee J and Goldfine AB: Inflammation and insulin
resistan ce. J Clin Invest 2006, 116:1793801.
585. Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K,
CharoI,LeibelRLandFerranteAWJr:CCR2 modulates
inflammatory and metabolic effects of high-fat feeding.
J Clin Invest 2006, 116:11524.
586. Zozulinska D and Wierusz-Wysocka B: Type 2 diabetes mellitus
as inflammatory disease. Diabetes Research and Clinical Practice
2006, 74:S12S16.
587. Pradhan A: Obesity, metabolic syndrome, and type 2
dia betes: inflammatory basis of glucose metabolic disord ers.
Nutr Rev 2007, 65:S1526.
588. Shoelson SE, Her rero L and Naaz A: Obesity, inflamm ation, and
insulin resistance. Gastroen terology 2007, 132:21692180.
589. T horand B, Baumert J, Kolb H, Meisinger C, Chambless L, Koenig W
and Herder C: Sex differences in the prediction of type 2
dia betes by inflammatory markers: res ults from the MON-
ICA/KORA Augsburg case-cohort study, 19842002. Diabetes
Care 2007 , 30:85460.
590. Wei Y, Chen K, Whaley-Connell AT, Stump CS, Ibdah JA and
Sowers JR: Skeletal muscle insulin resistance: role of
inflammatory cytokines and reactive o xygen species. Am J
Physiol Regul Integr Comp Physiol 2008, 294:R673
80.
591. Lao TT, Chan PL and Tam KF: G estational diabetes mellitus in
the last trimester a feature of maternal iron exce ss? Diabet
Med 2001, 18:21823.
592. Lao TT, Tse KY, Chan LY, Tam KF and Ho LF: NBsAg carrier
status and the association between gestational diabetes with
increased serum ferritin concentration in Chinese wo men.
Diabetes Care 2003, 26:30113016.
593. Chen X, Scholl TO and Stein TP: Association of elevated serum
ferritin levels and the risk of gestational diabetes mellitus in
pregnant women: The Camden study. Diabetes Care 2006,
29:107782.
594. Oberley LW: Free radicals and diabetes. Free Radic Biol Med
1988, 5:11324.
595. Wolff SP: Diabetes mellitus and free radic als. Free radicals,
tra nsition metals and oxidative stress in the aetiology of
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 38 of 79
(page number n ot f or citation purposes)
diabetes mellitus and complications. Br Med Bull 1993,
49:64252.
596. T uomainen TP, Nyyssonen K , Salonen R, Tervahauta A, Korpela H,
Lakka T, Ka plan GA and Salonen JT: Body iron stores are
associated with serum insulin and blood g lucose concentra -
tions. Population study in 1,013 eastern Finnish men. Diabetes
Care 1997 , 20:4268.
597. Salonen JT, Tuomainen TP, Nyyssonen K, Lakka HM and
Punnonen K: Relation between iron st ores and non-insulin
dependent diabetes in men: case-control study. BMJ 1998,
317:727.
598. Fernández-Real JM, López-Bermejo A and Ricart W: Cross-talk
between iron metabolism a nd dia betes. Diabetes 2002,
51:234854.
599. Fernández-Real JM and Ricart W: Insulin resistance and chronic
cardiovascular inflammatory syndrome. Endocr Rev 2003,
24:278301.
600. Wilson JG, Li ndquist JH, Grambow SC, Crook ED and Maher JF:
Pot entia l role of increased iron s tores in diabetes. Am J Med
Sci 2003, 325:3329.
601. Lee DH, Folsom AR and Jacobs DR Jr: Dietary iron intake and
Type 2 diabetes incidence in postmenopausal women: the
Iowa Women's Health Study. Diabetologia 2004 , 47:18594.
602. Kazi TG, Afridi HI, Kazi N, Jamali MK, Arain MB, Jalbani N and
Kandhro GA: Copper, chromium, manganese, iron, nickel,
and zinc levels in biological samples of diabetes mellitus
patients. Biol Trace Elem Res 2008, 122:118.
603. R ajpat hak SN, Crandall JP, Wylie-Rosett J, Kabat GC, Rohan TE and
Hu FB: The role of iron in type 2 diabetes in humans. Biochim
Biophys Acta 2008 in press.
604. Adams PC, Kertesz AE and Valberg LS: Clinical presentation of
hemochromatosis: a changing scene. Am J Med 1991, 90:445 9.
605. Kaye TB, Guay AT and Simonson DC: Non-insulin-dependent
dia betes mel litus and ele vated serum ferritin level. J Diabetes
Complic 1993, 7:246249.
606. Fernández-Real JM, Ricart-Engel W, Arroyo E, Balanca R, Casamit-
jana-Abella R, Cabrer o D, Fernandez-Castaner M a nd Soler J:
Serum ferritin as a component of the insulin resistance
syndrome. Diabetes Care 1998, 21:6268.
607. Ford ES and Cogswell ME: Diabetes and serum ferritin
concentration among U.S. ad ults. Diabetes Care 1999,
22:197883.
608. Jiang R, Manson JE, Meigs JB, Ma J, Rifai N and Hu FB: Body iron
stores in relation to risk of type 2 d iabet es in apparently
hea lthy women. JAMA 2004, 291:7117.
609. Mojiminiyi OA, Marouf R and Abdella NA: Body iron stor es in
relation to the metabolic syndrome, glycemic control and
complications in female patients with type 2 diabetes. Nutr
Metab Cardiovasc Dis 2008, 18:55966.
610. Jehn ML, Guallar E , Clark JM , Couper D, Duncan BB, Ballantyne CM,
Hoogeveen RC, Harri s ZL and Pa nkow JS: A prospective study of
pla sma ferritin level and incident diabetes: the Athero-
sclerosis Risk in Communities (ARIC) Study. Am J Epidemiol
2007, 165:104754.
611. Fernández-Real JM, Moreno JM, Chico B, Lopez-Bermejo A and
Ricart W: Circulating visfatin is associated with parameters
of iron metabolism in subjects with altered glucose
tolerance. Diabetes Care 2007, 30:61621.
612. Wang Y, Lam KSL, Kraegen E W, Sweeney G, Zhang JL, Tso AWK,
Chow WS, Wat NMS, Xu JY , Hoo RLC and Xu AM: Lipocalin-2 is
an inflammatory marker closely associated with obesity,
insulin r esistance, and hyperglycemia in humans. Clinical
Chemistry 2007, 53:3441.
613. van Dam RM and Hu FB: Lipocalins and insulin resistance:
Etiological role of retinol-binding protein 4 and lipocalin-2?
Clinical Chemistry 2007, 53:57.
614. Hua NW, Stoohs RA and Facchi ni FS: Low iron status and
enhanced insulin sensitivity in la cto-ovo vegetarians. Br J Nutr
2001, 86:5159.
615. Liang Q, Carlson EC, Donthi RV, Kralik PM, Shen X a nd Epstein PN:
Overexpression of metallothionein reduces diabetic cardi-
omyopathy. Diabetes 2002, 51:17481.
616. Y e G, Metreveli NS, Ren J and Epstein PN: Metallothionein
prevents diabetes-induced deficits in cardiomyocytes by
inhibiting reactive oxygen species production. Diabetes 2003,
52:77783.
617. Cai L, Wang J, Li Y, Sun X, Wang L, Zh ou Z and Kang YJ: In hibit ion
of superoxide generation and associated nitros ative damage
is involved in metallothionein prevention of diab etic cardi-
omyopathy. Diabetes 2005, 54:1829 37.
618. Islam MS and Loots du T: Diabetes, metallothionein, and zinc
interact ions: a r eview. Biofactors 2007, 29:20312.
619. Li X, Cai L and Feng W: Diabetes and metallothionein. Mini Rev
Med Chem 2007, 7:7618.
620. Zheng Y, Li XK , Wang Y and Cai L: The role of zinc, copper and
iron in the pathogenesis of diabetes and diabetic complica-
tions: therapeutic effects by chelators. Hemoglobin 2008,
32:13545.
621. Lee DH, Liu DY, Jacobs DR Jr, Shin HR, Song K, Lee IK, Kim B and
Hider RC: Common presence of non-transferrin-bound iron
among patients with type 2 diabetes. Diabetes Care 2006,
29:10905.
622. Chittum HS and Champney WS: Erythromycin inhibits the
assembly of the large ribosomal subunit in growing
Escherichia coli cells. Curr Mi crobiol 1995, 30:2739.
623. Facchini FS, Hua NW and Stoohs RA: Effect of iron depletion in
carbohydrate-intolerant patients with clinical evidence of
nonalcoholic fatty liver disease. Gastroenterology 2002,
122:9319.
624. Machado M and Cortez-Pinto H: Nash, insulin resistance and
iron. Liver International 2006, 26:11591162.
625. Allerson CR, Cazzola M and Rouault T A: Clinical severity and
thermodynamic effects of iron-responsive element muta-
tions in her editary hyperferritinemia-cataract syndrome.
Journal of Biological Chemistry 1999, 274:2643926447.
626. Beau mont C, Leneu ve P, Devaux I, Scoaz ec JY, Ber thi er M,
Loi seau MN, Grandch amp B and Bonneau D: Mutation in the
Iro n-Responsi ve Element of the L-Ferritin Messenger-Rna in
a Family with Dominant Hyp erferritinemia and Cataract.
Nature Geneti cs 1995, 11:444446.
627. D andona P, Aljada A, Chaudhuri A, Mohanty P and Garg R:
Metabolic syndrome A comprehensive perspective based
on interactions between obesity, diabetes, and inflamma -
tion. Circulation 2005, 111:14481454.
628. Eckel RH, Grundy SM and Zimmet PZ: The metabolic syndrome.
Lancet 2005, 365:141528.
629. Roberts CK and Barnard RJ: Effects of exercise and diet on
chr onic disea se. JApplPhysiol2005, 98:330.
630. Nicols on GL: Metabolic syndrome and mitochondrial func-
tion: Molecular replacement and antioxidant supplements
to prevent membrane peroxidation and restore mitocho n-
drial function. Journal of Cell ular Biochemistry 2007, 100:13521369.
631. Y udkin JS: Insulin resistance and the metabolic syn drome or
the pit falls of epidemiology. Diabetologia 2007, 50:157686.
632. Jehn M, Clark JM and Guallar E: Serum ferritin and risk of the
metabolic syndrome in U.S. adults. Diabetes Care 2004,
27:24228.
633. Wrede CE, Buettner R, Bollheimer LC, Scholmerich J, Palitzsch KD
and Hellerbrand C: Associat ion b etween serum ferritin and
the insulin resistance syndrome in a representative popula-
tion. Eur J Endocrinol 2006, 154:333340.
634. Bozzini C, Girelli D, Olivieri O, Martinelli N, Bassi A, De Matteis G,
Ten uti I, Lotto V, Friso S, Pizzolo F and Corrocher R: Prevalence of
body iron excess in t he metabolic syndrome. Diabetes Care
2005, 28:20613.
635. Choi KM , Lee KW, Kim HY, Seo JA, Kim SG, Kim NH, Choi DS and
Baik SH: Association among serum ferritin, alanine amino-
transferase levels, and metabolic syndrome in Korean
postmenopausal women. Metabolism 2005, 54:15104.
636. Kurella M, Lo JC and Chertow GM: Metabolic syndrome and the
risk for chronic kidney disease among nondiabetic adults.
J Am Soc Nephrol
2005, 16:21342140.
637. den Boer M, Voshol PJ, Kuipers F, Havekes LM and Romijn JA:
Hepatic steatosis: a mediator of the metabolic syndrome.
Lessons f rom animal models. Arterioscler Thromb Vasc Biol 2004,
24:6449.
638. Mittra S, Bansal VS and Bhatnagar PK: From a glucocentric to a
lipocentric approach towards metabolic syndrome. Dr ug
Discov Today 2008, 13:2118.
639. R oberts CK, Barnard RJ, Sindhu RK, Jurczak M, Ehdaie A and
Vaziri ND: Oxidative stress and dysregulation of NAD(P) H
oxidase and antioxidant enzymes in diet-ind uced metabolic
syndrome. Metabolism 2006, 55:92834.
640. Booth FW, Gordon SE, Carlson CJ and Hamilton MT: Wa ging war
on modern c hronic di sease s: primary prevention through
exercise biology. JApplPhysiol2000, 88:77487.
641. Wild S, Roglic G, Green A, Sicree R and King H: Global prevalence
of diabetes: estimates for the year 2000 and projections for
2030. Diabetes Care 2004, 27:104753.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 39 of 79
(page number n ot f or citation purposes)
642. Kahn SE, Hull RL and Utzschneider KM: Mechanisms linking
obesity to insulin resistance and t ype 2 diabetes. Nature 2006,
444:8406.
643. Walker CG, Zariwala MG, Holness MJ and S ugden MC: Diet,
obesity and diabetes: a current update. Clin Sci (Lond) 2007,
112:93111.
644. Lecube A, Hernandez C, Pelegri D and Simo R: Factors
accounting for high ferritin levels in obesity. Int J Obes (Lond)
2008.
645. Fricker J, Lemoel G and Apfelbaum M: Obesity and iron status in
menstruating women. Am J Clin Nutrition 1990, 52:863866.
646. Farahani P, Chiu S, Bowlus CL, Boffelli D, Lee E, Fisler JS, Krauss RM
andWardenCH:Obesity in BSB mice is correlated with
exp ression of genes for iron homeostasis and leptin. Obesity
Research 2004, 12:191204.
647. Das UN: Is obesity an inflam matory condition? Nutrition 2001,
17:95366.
648. Hall JE, Crook ED, Jones DW, Wofford MR and Dubbert PM:
Mechanisms of obesity-associated cardiovascular and renal
disease. Am J Med Sci 2002, 324:127 37.
649. T rayhurn P and Wood IS: Ad ipokines: inflammation and the
pleiotropic role of white adipose tissue. Br J Nutr 2004,
92:34755.
650. Berg AH and Scherer PE: Adipose tissue, inflammation, and
cardiovascular disease. Circ Res 20 05, 96:939 49.
651. Hukshorn CJ, Lindeman JH, Toet KH, Saris WH, Eilers PH,
Westerterp-Plantenga MS and Kooistra T: Leptin and the
proinflammatory state associated with human obesity. J
Clin Endocrinol Metab 2004, 89:17738.
652. T oni R, Malaguti A, Castorina S, Roti E and Lechan R M: New
paradigms in neuroendocrinology: relationships between
obesity, systemic inflammation and the neuroendocrine
system. J Endocrinol Invest 2004, 27:1826.
653. Koh KK, Han SH and Quon MJ: Inflammatory markers and the
metabolic syndrome: insights from therapeutic interven-
tions. JAmCollCardiol2005, 46:197885.
654. Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H, Capeau J
and Feve B: Recent advances in t he relationship between
obesity, inflammation, and insulin resistance. Eur Cyt okine
Netw 2006, 17:412.
655. Kapiotis S, Holzer G, Schaller G, Haumer M, Widhalm H,
Weghuber D, Jilma B, Roggla G, Wolzt M, Widhalm K and
Wagner OF: A proinflammatory state is detectable in obese
children and is accompanied by functional and morp hologi-
cal vascular changes. Ar terioscler Thro mb Vasc Biol 2006,
26:25416.
656. T ilg H a nd Moschen AR: Adipocytokines: mediators linking
adipose tissue, inflammation and immunity. Nat Rev Immun ol
2006, 6:77283.
657. Vincent HK and Taylor AG: Biomarkers and potential mechan-
isms of obesity- induced oxidant stress in humans. Int J Obes
(Lond) 2006, 30:40018.
658. Wang B and Trayhurn P: Acute and prolonged effects of TNF-
alp ha on the expres sion and secretion of inflammation-
related adipokines by human adipocytes differentiated in
culture. Pflugers Arch 2006, 452:41827.
659. Chandalia M and Abate N: Metabolic complication s of obesity:
inflated or inflamed? J Diabetes Complications 2007, 21:12836.
660. Lumeng CN, Bodzin JL and Saltiel AR: Obesity induces a
phenotypic switch in adipose tissue macrophage polariza-
tion. JClinInvest2007, 117:17584.
661. Chen Y, Zhu J, Lum PY, Yang X, Pinto S , Macneil DJ, Zhang C,
Lam b J, Edwards S, Sieberts SK, Leonardson A, Castellini LW,
Wang S, Champy MF, Zhang B, Emilsson V, Doss S, Ghazalpour A,
Hor vath S, Drake TA, Lusis AJ and Schadt EE: Variations in DNA
elucidat e molecular networks that cause disease. Nature
2008, 452:429435.
662. Chung J, Nguyen AK, Henstridge DC, Holmes AG, Chan MH,
Mesa JL, Lancaster G I, Southgate RJ, Bruce CR, Duffy SJ, Horvath I,
Mestril R, Watt MJ, Hooper PL, Kingwell BA, Vigh L, Hevener A and
Febbraio MA: HSP72 protects against obesity-induced insulin
resistan ce. Proc Nat l Acad Sci USA 2008, 105:173944.
663. Emilsson V, Thorlei fsson G, Zhang B, Leonardson AS, Z ink F, Zhu J,
Carlson S, Helgason A, Walters GB, Gunnarsdottir S, Mouy M,
Steinthorsdottir V, Eiriksdotti r GH, Bjornsdottir G, Reynisdottir I,
Gudbjartsson D, Helgadottir A, Jonasdottir A , Jonasdottir A,
Sty rkars dottir U, Gr etarsdottir S, Magnusson KP, Stefansson H,
Fos sdal R, Kristjansson K, Gislason HG, Stefansson T, Leifss on BG,
Thorsteinsdottir U, Lamb JR, Gulcher JR, Reitman ML, Kong A,
Sch adt EE and Stefansson K: Genetics o f ge ne expression and its
effect on disease. Nature 2008.
664. Wärnbe rg J and Marcos A: Low-grade inflammation and the
metabolic syndrome in children and adolescents. Curr Op in
Lipidol 2008, 19:115.
665. Rajala MW and Scherer PE: Minireview: The adipocyte at the
cro ssroads of energy homeostasis, inflammation, and ather-
oscleros is. Endocrinology 2003, 144:376573.
666. V endrell J, Broch M, Vilarrasa N, Molina A, Gomez JM, Gutierrez C,
Simon I, Soler J and Richart C: Resistin, adiponectin, ghr elin,
leptin, and proinflammatory cytokines: relat ionships in
obesity. Obes Res 2004, 12:962 71.
667. Arner P: Insulin resistance in type 2 diabetes role of the
adipokines. Curr Mol Med 2005, 5:3339.
668. Hutley L and Prins J B: Fat as an endocrine organ: relationship
to the metabolic syndrome. Am J Med Sci 2005, 330:2809.
669. Kougias P, Chai H, Lin PH, Yao Q, Lumsden AB and Chen C: E ffects
of adipocyte-derived cytokines on endothelial functions:
implication of vascular disease. JSurgRes2005, 126:1219.
670. Lau DC, Dhillon B, Yan H, Szmitko PE and Verma S: Adipoki nes:
mol ecular links between obesity and ather oslcerosis. Am J
Physiol Heart Circ Physiol 2005, 288:H203141.
671. R osen ED a nd Spiegelman BM: Adipocytes as regulators of
energy balance and glucose homeostasis. Nature 2006,
444:84753.
672. Kim F, Pham M, Luttrell I, Bannerm an DD, Tupper J, Thaler J,
Hawn TR, Raines EW and Schwartz MW: Toll-like receptor-4
mediates vascular inflammation and insulin resistance in
diet-induced obesity. Circ Res 2007, 100:1589
96.
673. Shah PK: In nate immune pathway links obesity to insulin
resistan ce. Circ Res 2007, 100:15313.
674. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y,
Nakajima Y, Nakayama O, Makishima M, Matsuda M a nd
Shimomura I: Increased ox idative stress in obesity and its
imp act on metabol ic syndrome. J Clin Invest 2004, 11 4:175261.
675. Keaney JF, Larson MG Jr, Vasan RS, Wilson PW, Lipinsk a I, Corey D,
Mas saro JM, Sutherland P, V ita JA and Benjamin EJ: Obesity and
systemic oxidative stress: clinical correlates of oxidative
stress in the Framingham Study. Arterioscler Th romb Vasc Biol
2003, 23:4349.
676. Higdon JV and Frei B: Obesity and oxidative stress: a direct link
to CVD? Arterioscler Thromb Vasc Bio l 2003, 23:3657.
677. Whitworth JA: 2003 World Health Organization (WHO)/
International Society of Hypertension (ISH) statement on
management of hypertension. JHypertens2003, 21:1983 92.
678. Tu der RM a nd Voelke l NF: Pu lmonar y h ypertensi on a nd
inflammation. J L ab Clin Me d 1998, 132:1624.
679. Intengan HD and Schiffrin EL: Vascular remodeling in hyperten -
sion: roles of apoptosis, inflammation, and fibrosis. Hyperten-
sion 2001, 38:581 7.
680. Grundy SM: Inflammation , hypertension, and the metabolic
syndrome. JAMA 2003, 290:30002.
681. V irdis A and Schiffrin E L: Vascular inflammation: a role in
vascular disease in hypertension? Curr Opin Nephrol Hypertens
2003, 12:1817.
682. B o o s CJ and Li p GY: Elevated high-sensitive C-reactive
protein, large arterial stiffness and athe rosclerosis: a
relationship between infl ammation and hypertension?
J Hum Hy pertens 2005, 19:5113.
683. Varughese GI and Lip GY: Hypertension in patients with type-II
dia betes: relation to urinar y albumin excretion, endoth elial
function and inflamma tion.
J Hum Hyperten s 2005, 19:421 4.
684. Li JJ: Inflammation i n hypertension: primar y evidence. Chin
Med J (Engl) 2006 , 119:121521.
685. Savoia C and Schiffrin EL: Inflam mation in hypertension. Curr
Opin Nephrol Hypertens 2006, 15:1528.
686. Savoia C and Schiffrin EL: Vascular inflammation in hyperten-
sion and dia betes: molecular mechanisms a nd therapeutic
interventions. Clin Sci ( Lond) 2007, 112:37584.
687. Kannel WB, Brand N, Skinne r JJ Jr, Dawber TR an d McNamara PM:
The relation of adi posity to blood pressure and develop-
ment of hypertension. The Framingham s tudy. AnnInternMed
1967, 67:4859.
688. Mark AL, Correia M, Morgan DA, Shaffer RA and Haynes WG:
State-of-the-art-lecture: Obesity-induced hypertension: new
concepts from the emerging biology of obesity. Hypertension
1999, 33:53741.
689. Aizawa -Abe M, Ogawa Y, Masuzaki H, Ebihara K, Satoh N, Iwai H,
Matsuoka N, Hayashi T, Hosoda K, Inoue G , Yoshimasa Y and
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 40 of 79
(page number n ot f or citation purposes)
Nakao K: Pathophysiolo gical role of leptin in obesity-r elated
hypertension. J Clin Invest 2000, 105:124352.
690. Hall JE: The kidney, h ypertension, and obesity. Hypertension
2003, 41:62533.
691. Y anai H, Tomono Y, Ito K, Furutani N, Yoshida H and Tada N: The
underlying mechanisms for development of hypertension in
the metabolic sy ndrome. Nutr J 2008, 7:10.
692. Vaziri ND and Rodriguez-Iturbe B: Mechanisms of disease:
oxidative stress and inflamm ation in the pathogenesis of
hypertension. Nat Clin Pract Nephrol 2006, 2:58293.
693. Yakobson MG, Antonov AR, Golovatyuk AV, Efremov AV,
Markel AL and Yakobson GS: Iron content and parameters of
blood antioxidant activity in rats with hereditary arterial
hypertension dur ing experimental myocardial infarction. Bull
Exp Biol Med 2001, 132:10414.
694. Peterson JR, Sharma RV and Davisson RL: Reactive oxygen
species in the neuropathogenesis of hypertension. Curr
Hypertens Rep 2006, 8:23241.
695. Sullivan JL: Iron and the sex difference in heart-disease risk.
Lancet 1981, 1:1293 1294.
696. Sullivan JL: Misconceptions in the debate on the iron
hypothesis. Journal o f Nutritional Biochemistry 2001, 12:3337.
697. Sullivan JL: Stored iron and vascular reactivity. Arteriosclerosis
Thrombosis and Vascular Biology 2005, 25:15321535.
698. Sullivan JL: The big idea: the coxib crisis iron, aspirin and
hea rt disease risk revisite d. J R Soc Med 2007, 100:3469.
699. Meyers DG: The iron hypothesis: does iron play a role in
atherosclerosis? Transfusion 2000, 40:10231029.
700. Y uan XM and Li W: The iron hypothesis of atherosclerosis and
its clinical impact. Ann Med 2003, 35:57891.
701. Zheng H, Dimayuga C , Hudaihed A and Katz SD: Effect of
dex razoxane on homocysteine-induced endothelial dysfunc-
tion in normal subjects. Arterioscle r Thromb Vasc Biol 2002, 22:
E158.
702. Zheng H, Huang X, Zhang Q and Katz SD: Iron sucrose augments
homocysteine-induced endot helial dysfunction in normal
subjects. Kidney Int 2006, 69:679 84.
703. Sullivan JL: Is homocysteine an iron-dependent cardiovascular
risk factor? Kidney Int 2006, 69:642 4.
704. Clarke R: Homocysteine-lowering trials for prevention of
hea rt disease and str oke. Semin Vasc Med 2005, 5:21522.
705. Tuomainen TP, Punnonen K, Nyyssonen K and Salonen JT:
Association between body iron stores and the risk of acute
myocardial infarction in men. Circulation 1998, 97:14616.
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 and 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, 108:215469.
707. Oberg BP, McMenamin E, Lucas FL, McMonagle E, Morrow J,
Iki zler TA and Himmelfarb J: Increased prevalence of oxidant
stress and inflammation in pat ients with moderate to severe
chr onic kidney disease. Kidney Int 2004, 65:1009 16.
708. Vaziri ND: Roles of oxidative stress a nd antioxidant therapy
in chronic kidn ey disease a nd hypertension. Curr Opin Nephrol
Hypertens 2004, 13:939.
709. Abramson JL, Jurkovitz CT, Vaccarino V, Weintraub WS and
McClellan W: Chronic kidney disease, anemia, an d incident
stroke in a middle-aged, community-based population: the
ARIC Study. Kidney Int 2003, 64:610
5.
710. Smith JK, Carden DL, Grisham MB, Granger DN and Korthuis RJ:
Role of iron in postischemic microvascular injury. Am J Physiol
1989, 256:H14727.
711. Kukrej a RC and Hess ML: The oxygen free radical system: from
equations t hrough membrane protein i nteractions to
cardiovascular injury and protection. Cardiovasc Res 1992,
26:641655.
712. L e s nefsky E an d Y e J: Exo genous intracellular, but not
extracellular, iron augments myocardial reperfusion injury.
Amer J Physiol 1994, 266:H384H392.
713. Berenshtein E, Mayer B, Goldberg C, Kitrossky N and Chevion M:
Patterns of mobilization of copper and iron following
myocardial ischemia: Pos sible pr edictive criteria for tissue
injury. J Mol Cell Cardiol 19 97, 29:30253034.
714. Horwitz LD, Sherman NA, Kong YN, Pike AW, Gobin J,
Fennessey PV and Horwitz MA: Lipophilic siderophores Myco-
bacterium tuberculosis prevent cardiac reperfusion injury.
Pro c Natl Ac ad Sci 19 98, 95:52635268.
715. Horwitz LD and Rosenthal EA: Iron-mediated cardiovascular
injury. Vasc Med 1999, 4:939.
716. Pucheu S, Coudray C, Tresallet N, Favier A and Deleiris J: Effect of
iron overload in the isolated ischemic and reperfused rat
heart. Cardiovascular Drugs and Therapy 1993, 7:701711.
717. Amersi F, Dulkanchainun T, Nelson SK, Farmer DG, Kato H, Zaky J,
Melinek J, Shaw GD, Kupiec-Wegli nski JW, Horwit z L D,
Horwitz MA and Busuttil RW: Nov el iron chelator in combina-
tion with a P-selectin antagonist prevents ischemia/reperfu-
sion injury in a rat liver model. Transplantation 2001,
71:112118.
718. Mamtani M and Kulkarni H: Influence of iron chelators on
myocardial iron and cardiac function in transfusio n-depen-
dent thalassaemia: a systematic review and meta-analysis. Br
J Haematol 20 08, 141:882 90.
719. Ravati A, Ahlemeyer B, Becker A, Klumpp S and Krieglstein J:
Preconditioning-in duced neuroprotection is mediated by
reactive oxygen species and activation of the transcript ion
factor nuclear factor-kappa B. Journal of Neurochemistry 2001,
78:909919.
720. Berenshtein E, Vaisman B, Goldberg-Langerman C, Kitrossky N,
Konijn AM an d Chevion M: Roles of ferritin and iron in ischemic
preconditioning of the heart. Mol Cell Biochem 2002,
234:283292.
721. Calvillo L, Latini R, Kajstura J, Leri A, Anversa P, Ghez zi P, Sal io M,
Cerami A and Brines M: Recombinant human erythropo ietin
protects the myocardium from ischemia-reperfusion injury
and promotes beneficial remodeling. Pro cNatl Acad Sci 2003,
100:48024806.
722. Parsa CJ, Matsumoto A, Kim J, Riel RU, Pascal LS, Walton G B,
Thompson RB, Pet rofsk i JA, Ann ex BH, Stamler JS and Koch WJ: A
novel protective effect o f erythropoietin in the infarcted
heart. JClinInvest2003, 112:9991007.
723. Engber ding N, Spiekermann S , Schaefer A, Heineke A, Wiencke A,
Muller M, Fuchs M, Hil fiker-Kleiner D, Hornig B, Drexler H and
Landmesser U : Allopurinol attenuates left ventricular remo-
deling and dysfunction after experimental myocardial
infarction: a new action for an old drug? Circulation 2004,
110:21759.
724. Reyes AJ: Cardiovascular dru gs and serum uric acid. Cardiovasc
Drugs Ther 2003, 17:397414.
725. D unn WB, Broadhurst DI, Sasalu D, Buch M, McDowell G, Spasic I,
Ellis DI, Brooks N, Kell DB and Neyses L: Serum metabolomics
reveals many novel metaboli c markers of heart failure,
including pseudouridine and 2-oxoglutarate. Metabolomics
2007, 3:413426.
726. Hokamaki J, Kawano H, Yoshimura M, Soejima H, Miyamoto S,
Kajiwara I, K ojima S, Sakamoto T, Sugiyama S, Hirai N, Shimomura H,
Nagayoshi Y, Tsujita K, Sh ioji I, Sasaki S and Ogawa H: Urinary
biopyrrins levels are elevated in relation to severity of heart
failure. J Am Coll Cardiol 2004, 43:18805.
727. Mozaffarian D, Nye R and Levy WC: Anemia predicts mortality
in severe heart failure: the prospective randomized amlo-
dipine survival evaluation (PRAISE). J Am Coll C ardiol 2003,
41:19339.
728. Felker GM, Adams KF Jr, Gattis WA and O'Connor CM: Anemia as
a risk factor and therapeutic target in heart failure. JAmColl
Cardiol 2004, 44:95966.
729. Bolger AP, Bartlett FR, Penston HS, O'Leary J, Pollock N,
Kaprielian R and Chapman CM: Intravenous iron alone for the
treatment of anemia in patients with chronic heart failure. J
Am Coll Cardiol 20 06, 48:12257.
730. Weiss G and Goodnough LT: Anemia of chr onic disease. NEnglJ
Med 2005, 352:101123.
731. Kunsch C and Medford RM: Oxidative stress as a regulator of
gen e expression in the vasculature. Circ Res 1999, 85:75366.
732. Ross R: Atherosclerosis: an inflammatory d iseas e. NEnglJMed
1999, 340:11526.
733. Blake GJ and Ridker PM: Nov el clinical markers of vascular wall
inflammation. Circ Res 2001, 89:76371.
734. Hansson GK: Immune mechanisms in atherosclerosis. Arter-
ioscler Thromb Vasc Biol 2001, 21:187690.
735. Forrester JS: Prevention of plaque rupture: a new paradigm of
therapy. AnnInternMed2002, 137:82333.
736. Himmelfarb J, Stenvinkel P, Ikizler TA and Hakim RM: The elephant
in uremia: oxidant stress as a unifying concept of cardiovas-
cular disease in uremia. Kidney Int 2002, 62:152438.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 41 of 79
(page number n ot f or citation purposes)
737. Libb y P: Inflammatio n in at herosclerosis. Nature 2002,
420:86874.
738. Libby P, Ridker PM and Maseri A: Inflammation and athero -
sclerosis. Circulation 2002, 105:113543.
739. Y oung JL, Libby P and Sch onbeck U: Cytokines in the pathogen-
esis of atherosclerosis. Thromb Haemost 2002, 88:55467.
740. Binder CJ, Chang MK, Shaw PX, Miller YI, Hartvigsen K, Dewan A
and Witztum J L: Innate and acquired immunity in atherogen-
esis. Nat Med 2002, 8:121826.
741. Altman R: Ris k factors in coronary atherosclero sis athero-
inflammation: the meeting point. Thromb J 2003, 1:4.
742. D wyer JH, Allayee H, Dwyer KM, Fan J, Wu H, Ma r R, Lusis AJ and
Mehrabia n M: Arachidonate 5-lipoxygenase promoter geno-
type, dietary arachidonic acid, and atherosclerosis. NEnglJ
Med 2004, 350:2937.
743. Forrester JS: C ommon ancestors: chronic pr ogressive dis-
eases have the same pathogenesis. Clin Cardiol 2004, 27:18690.
744. Paoletti R, Gotto AM Jr and Hajjar DP: Inflammation in
atherosclerosis and implications for therapy. Circul ation
2004, 109:III206.
745. R idker PM, Brown NJ, Vaughan DE, Harris on DG and Mehta JL:
Establis hed and emerging plasma biomarkers in the predic-
tion of first athero thrombotic events. Circulation 2004, 109
(Suppl 1):619.
746. van Oostr om A J, va n Wij k J and Cabezas MC: Lipaemia,
inflammation and athero sclerosis: novel opportunities in
the understanding and treatment of atheroscler osis. Drugs
2004, 64(Suppl 2):1941.
747. Willerson JT and Ridker PM: Inflammati on as a cardiovascular
risk factor. Circulation 2004, 109:II2
10.
748. Hansson GK: Inflammation, atherosclerosi s, and coronary
artery disease. NEnglJMed2005, 352:168595.
749. Madamanchi NR, Hakim ZS and Runge MS: Oxidative stress in
atherogenesis and arterial thrombosis: the disconnect
between cellular studies and clinical outcomes. JThromb
Haemost 2005, 3:25467.
750. Madamanchi NR, Vendrov A and Runge MS: Oxidative stress and
vascular disease. Arterioscler Thromb Vasc Biol 2005, 25:2938.
751. Mullenix PS, Andersen CA and Starnes BW: Atherosclerosis as
inflammation. Ann Vasc Surg 2005, 19:1308.
752. Nigro J, Osman N, Dart AM and Little PJ: I nsulin resistance and
atherosclerosis. Endocr Rev 2006, 27:242 59.
753. T aqueti VR, Mitchell RN and Lichtman AH: Protecting the pump:
controll ing myocardial inflammatory responses. Annu Rev
Physiol 2006, 68:6795.
754. T edgui A and Mallat Z: Cytokines in atherosclerosis: patho-
gen ic and re gulatory pathways. Physiol Rev 2006, 86:51581.
755. Grainger DJ: TG F-beta and atherosclerosis in man. Cardiovasc
Res 2007, 74 :21322.
756. Popa C, Netea MG, van Riel PL , Meer van der JW and Stalenhoef AF:
The role of TNF-alpha in chron ic inflammatory conditions,
intermediary metabolism, and cardiovascular risk. J Lipid Res
2007, 48:75162.
757. Schleicher E and Friess U: Oxidative stress, AGE, and athero-
sclerosis. Kidney Int Suppl 2007, S 1726.
758. Kibel A, Belovari T and Drenjančević-Perić I: Theroleof
tra nsferrin in atherosclerosis.
Med Hypotheses 2008, 70:7937.
759. Packard RR and Libby P: Inflammation in atherosclerosis: from
vascular biology to biomarker discovery and risk prediction.
Clin Chem 2008, 54:2438.
760. R ader DJ and Daughe rty A: Translating molecular discoveries
into new therapies for atherosclerosis. Nature 2008,
451:90413.
761. Tan KT and Lip GY: Imaging of the unstable plaque. Int J Cardiol
2008, 127:15765.
762. v an Leuven SI, F ranssen R, Kastelein JJ, Levi M, St roes ES and Tak PP:
Systemic inflammation as a risk factor for atherothrombo-
sis. Rheumatology (Oxford) 2008, 47:37.
763. Lusis AJ: Atherosclerosis. Nature 2000, 407:233241.
764. Stocker R and Keaney JF: Role of oxidative modificat ions in
atherosclerosis. Physiol Rev 2004, 84:13811478.
765. Smith C, Mitchinson MJ, Aruoma OI and Halliwell B: Stimulation of
lipid peroxidation and hydroxyl rad ical generation by the
contents of human atherosclerotic lesions. Biochem J 1992,
286:901905.
766. Ramakrishna G, Rooke TW and Cooper LT: Iron and peripheral
art erial disease: revisiting the iron hypothesis in a different
light. Vasc Med 2003, 8:203210.
767. Stadler N, Lindner RA and Davies MJ: Direct detection and
quantification of transition metal ions in human athero-
sclerotic plaques: eviden ce for the presence of elevated
levels of iron and copper. Arterioscler Thromb Vasc Biol 2004,
24:94954.
768. Wolff B, Volzke H, Ludemann J, Robinson D, Vogelgesang D,
Sta udt A, Kessler C, Dahm JB, John U and Felix SB: Associ ation
between high serum ferritin levels an d carotid athero-
sclerosis in the study of health in Pomerania (SHIP). Stroke
2004, 35:4537.
769. R oijers RB, Dutta RK , Mutsae rs PHA, Gijbels MJJ, de Winther MPJ,
de Goeij JJM and Vusse van der GJ: Spatial correlation of trace
elements with morpholo gical features of atherosclerotic
pla ques. Nuclear Instruments & Methods in Physics Research Section B-
Bea m In teractions with Materials and Ato ms 20 05, 231:239244.
770. Stocker R and Keaney JF: New insights on oxidative stress in the
artery wall. Journal of Thrombosis and Haemostasis 2005,
3:18251834.
771. Stanley N, Stadler N, Woods AA, Bannon PG and Davies MJ:
Concentrations of iron correlate with the extent of protein,
but not lipid, oxidation in advanced human at heros clerotic
lesions. Free Radical Biology and Medicine 2006, 40:16361643.
772. Watt F, Rajendran R, Ren MQ, Tan BKH and Halliwell B: A nuclear
microscopy study of trace elements Ca, Fe, Zn and Cu in
atherosclerosis. Nucl Instr Meth Phys Res B 2006, 249:646652.
773. Fernandes de Godoy M, Takakura IT, Machado RD, Grassi LV and
Nogueira PR: Serum ferritin and obstructive coronary artery
disease: Angiographic correlation. Arquivos Brasileiros De
Cardiologia 2007, 88:430433.
774. Rajendran R, Ren M, Ning P, Tan Kwong Huat B, Halliwell B and
Watt F: Promotion of atherogenesis by copper or i ronwhich
is more likely? Biochem Biophys Res Commun 2007, 353:610.
775. Kazi TG, Afridi HI, Kazi N, Jamali MK, Arain MB, Sarfraz RA,
Jalbani N, Ansari R, Shah AQ, Memon AU and Khandhro GA:
Distribu tion 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, 389:1149.
776. Sullivan JL: Iron in arterial plaque: A modifiable risk fac tor for
atherosclerosis. Biochim Biophys Acta 2008 in press.
777. Y uan XM and Li W: Iron involvement in multiple signaling
pathways of atherosclerosis: a revisited hypothesis. Curr Med
Chem 2008, 15:215772.
778. Matthews AJ, Vercellotti GM, Menchaca HJ, Bloch PH, Michalek VN,
Marker PH, Murar J and Buchwald H: Iron and atherosclerosis:
inhibition by the iron chelator deferiprone (L1). JSurgRes
1997, 73:3540.
779. Ponraj D, Makjani c J, Thong PSP, Tan BKH and Watt F: The onset
of atheroscler otic lesion formation in hypercholesterolemic
rabbits is delayed by iron depletion. FEBS Let t
1999,
459:218222.
780. Lee TS, Shiao MS, Pan CC and Chau LY: Ir on-deficient diet
reduces atherosclerotic lesions in ApoE-def icient mi ce.
Circulation 1999, 99:12221229.
781. R en M Q, Rajendran R, Pan N, Tan BKH, Ong WY, Watt F and
Halliwell B: The iron chelator desferrioxamine inhibits
atherosclerotic lesion development and decreases lesion
iron concentrations in the chole sterol-fed rabbit. Free Radical
Biology and Medicine 2005, 38:12061211.
782. Ferrara DE and Taylor WR: Iron chelation and vascular
function: in search of t he mechanisms. Arterioscler Thromb
Vasc Biol 2005, 25:22357.
783. Kiechl S, Willeit J, Egger G, Poewe W and Oberhollenzer F: Body
iron stores and the risk o f carotid atherosclerosis: prospec-
tive results from the Bruneck study. Circulation 1997,
96:33007.
784. de Valk B and Marx JJM: Iron, atherosclerosis, and ischemic
hea rt disease . Arch Internal Med 1999, 159:1542 1548.
785.ZacharskiLR,ChowB,LavoriPW,HowesPS,BellMR,
DiT ommaso MA, Carnegie NM, Bech F, Amidi M and Muluk S:
The Iron (Fe) and Atherosclerosis Study (FeAST): A pilot
study of reduct ion of body iro n stores in atherosclerotic
peripheral vascular disease. American Heart J ournal 2000,
139:337345.
786. Day SM, Duquaine D, Mundada LV, Menon RG, Khan BV,
Rajagopalan S and Fay WP: Chronic i ron administration
increases vascular oxidative s tress and accelerates arterial
thrombosis. Circulation 2003, 107:26012606.
787. Ren MQ, Watt F, Huat BTK and Halliwell B: Trace elemental
distributions in induced atherosclerotic lesions using
nuclear microscop y. Nuclear Instr Meth Phys Res B 2003,
210:336342.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 42 of 79
(page number n ot f or citation purposes)
788. Minqin R, Watt F, Huat BTK and Halliwell B: Correlation of iron
and zinc levels with lesi on depth in newly formed athero-
sclerotic lesions. Free Radical Biology and Medicine 2003,
34:746752.
789. Y ou SA, Archacki S R, Angheloiu G, Moravec CS, Rao S, Kinter M,
Topol EJ and Wang Q: Proteomic approach to coronary
atherosclerosis shows ferritin light chain as a significant
marker: evidence consistent with iron hypothesis in athero -
sclerosis. Physiol Genomics 2003, 13:2530.
790. Zacharski LR and Gerhard GS: A therosclerosis: a manifestation
of chronic iron toxicity? Vascular Medicine 2003, 8:153155.
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, 39:13051309.
792. You SA and Wang Q: Ferr itin in atherosclerosis. Clin Chim Acta
2005, 357:116.
793. Lapenna D, Pierdo menico SD, Ciofani G, Ucchino S, Neri M,
Giamberardino MA and Cuccurullo F: Association of body iron
stores with low molecular weight iron and oxidant damage
of human atherosclerotic plaques. Free Rad Biol Med 2007,
42:492498.
794. Marx JJ, Kartikasari AE and Georgiou NA: Can iron chelators
influence the progression of atherosclerosis? Hemoglobin 2008,
32:12334.
795. Paulsson J, Dadfar E, Held C, Jacobson SH and Lundahl J: Activation
of peripheral and in vivo transmigrated neutrophils in
patients with stable coronary artery disease. Atherosclerosis
2007, 192:32834.
796. R ooyakkers TM, Stroes ES, Kooistra MP, van Faassen EE, Hider RC,
Rabelink TJ and Marx JJ: Ferric saccharate induces oxygen
radical stress and endothel ial dysfunction in vivo. Eur J Clin
Invest 2002, 32(Suppl 1):916.
797. Duffy SJ, Biegelsen ES, Holbrook M, Russell JD, Gokce N,
Keaney JF Jr and Vita JA : Iron chelation improves endothelial
function in patients with coronary art ery disease. Circulation
2001, 103:2799
804.
798. Ishizaka N, Saito K, Mori I, Matsuzaki G , Ohno M and Nagai R: Iron
chelation suppresses ferritin upregulation an d attenuates
vascular dysfunction in the aorta of angiotensin II-infused
rats. Arterioscler Thromb Vasc Biol 2005, 25:22828.
799. Saito K, Ishizaka N, Aizawa T, Sata M, Iso-o N, Noiri E, Mori I,
Ohno M and Nagai R: Iron chelation and a free radical
scavenger suppress angiotensin II-induced upregulation of
TGF-beta1 in the heart. Am J Physiol Heart Ci rc Ph ysiol 2005, 288:
H183643.
800. Thomas SR, Schulz E and Keaney JF: Hydrogen peroxide
restrains endothelium-derived nitric oxide bioactivity
Role for iron-dependent oxidative stress. Free Radical Biology
and Medicine 2006, 41:681688.
801. Zacharski LR, Ch ow BK, Howes PS, Shamayeva G, Baron JA,
Dal man RL, Malenka DJ , Ozaki C K and Lavori PW: Reduction of
iron stores and cardiovascular outcomes in patients with
peripheral arterial disease A randomized controlled trial.
JAMA 2007, 297:603610.
802. Shiesh SC, Chen CY, Lin XZ, Liu ZA and Tsao HC: Melatonin
prevents pigment gallstone formation induced by bile duct
ligation in guinea pigs. Hepatology 2000, 32:45560.
803. Koppis etti S, Jenigiri B, Terron MP, Tengattini S, Tamura H, Flores LJ,
Tan DX and Reiter RJ: Rea ctive oxygen species and the
hypomotility of the gall b ladder as targets for the treatment
of gallstones with melatonin: a review. Dig Dis Sci 2008,
53:2592603.
804. R obinson JG: Models for describing relations among the
various statin drugs, low-density lipoprotein chol esterol
low ering, pleiotropic effects, and cardiovascular risk. Am J
Cardiol 2008, 101:100915.
805. Prentice RL, Langer RD, Stefanick ML, Howard BV, Pettinger M,
Anderson GL, Barad D, Curb JD, Kotchen J, Kuller L, Limacher M
and Wactawski-W ende J: Combined anal ysis of Women's
Health Initiative observational and clinical trial data on
postmenopausal hormone treatment and cardiovascular
disease. Am J Epidemiol 2006, 163:58999.
806. Couzin J: Cholesterol veers off script. Science 20 08, 322:2203.
807. Peterson RT:
Chemical biology and the limits of reduction-
ism. Nat Chem Biol 2008, 4:6358.
808. Weitz-Schmidt G, Welzenbach K, Br inkmann V, Kamata T, Kallen J,
Bruns C, Cottens S, Takada Y and Hommel U: Statins selectively
inhibit leukocyte function antigen-1 by binding to a novel
regulatory integrin site. Nat M ed 2001, 7:68792.
809. Arkin MR and Wells JA: Small-molecule inhibitors of protein-
protein interactions: progressing towards the dream. Nat Rev
Drug Discov 2004, 3:30117.
810. Bellosta S, Ferri N, Arnaboldi L, Bernini F, Paoletti R and Corsini A:
Pleiotro pic effects of statins in atherosclerosis and diabetes.
Diabetes Care 2000, 23:B72 U5.
811. Kwak B, Mulhaupt F, Myit S a nd Mach F: Statins as a new ly
recognized type of immunomodulator. Nat Med 2000,
6:1399402.
812. Albert MA, Danielson E, Rifai N and Ridker PM: Effect of statin
therapy on C-reactive protein levels: the pra vastatin
inflammation/CRP evaluation (PRINCE): a randomized
trial and cohort study. JAMA 2001, 286:6470.
813. LaRosa JC: Pleiotropic eff ects of statins and their clinical
significance. American Journal of Cardiology 2001, 88:291.
814. Takemoto M, Node K, Nakagami H, Liao YL, Grimm M, Takemoto Y,
Kitakaze M and Liao JK: Statins as antioxidant therapy for
preventing cardiac myocyte hypertrophy. Journal of Clinical
Investigation 2001, 108:14291437.
815. Liao JK: Beyond lipid lowering: the role of statins in vas cular
protection. Int J Cardiol 2002, 86:5 18.
816. McFarlane SI, Muniyappa R, Francisco R and Sowers JR: Clinical
review 145 Pleiotropic effects of statins: Lipid reduction
and beyond. J Clin En docrinol Metab 2002 , 87:14511458.
817. Undas A, Brozek J and Musial J: Anti-inflammatory and
antithrombotic effects of statins in the management of
cor onary art ery disease.
Clin Lab 2002, 48:287 96.
818. Bonett i PO, Lerman LO, Napoli C and Lerman A: Statin effects
beyond l ipid loweringare they clinically relevant? Eur Heart J
2003, 24:22548.
819. D ichtl W, Dulak J, Frick M, Alber HF, Schwarzacher SP, Ares MPS,
Nil sson J, Paching er O and Weidinger F: HMG- CoA reductase
inhibitors regulate inflammatory transcription factors in
human endothelial and vascular smooth muscle cells.
Arteriosclerosis Thrombosis and Vascular Biology 2003, 23:5863.
820. Mulhaupt F, Matter CM, Kwak BR, Pelli G, Veillard NR, Burger F,
Graber P, Luscher TF and Mach F: Statins (HMG-Co A reductase
inhibitors) reduce CD40 expression in human vascular cells.
Cardiovasc Re s 2003, 59:75566.
821. Wierzbicki AS, Poston R and Ferro A: The lipid and non-lipid
effects of statins. Pharmacol T her 2003, 99:9511 2.
822. D avignon J: Beneficial cardiovascular pleiotropic effects of
statins. Circulation 2004, 109:3943.
823. Halcox JP and Deanfield JE: Beyond the laboratory: clinical
implications for statin pleiotropy. Circulat ion 2004, 109:II428.
824. Liao JK: Statins: potent vascular anti-inflam matory agents. Int
J Clin Pract Suppl 2004, 418.
825. Schönbeck U and Libby P: Inflammat ion, immunity, and HMG-
CoA reductase inhibitors Statins as antiinflamma tory
agents? Circulation 2004, 109:1826.
826. Liao JK and Laufs U: Pleiotropic effects of statins. Annu Rev
Pharmacol Toxicol 2005, 45:89118.
827. Elrod JW and Lefer DJ: The effects of statins on endothelium,
inflammation and cardioprotection. Drug News Perspect 2005,
18:22936.
828. R obinson JG, Smith B, Maheshwari N and Schrott H: Pleiotropic
effects of statins: Benefit beyond cholesterol reduction? A
meta-regression analysis. JAmerCollCardiol2005, 46:18551862.
829. T subouchi H, Inoguch i T, Sonta T, Sato N, Sekiguchi N, Kobayashi K,
Sumimoto H, Utsumi H and Nawata H: Statin attenuates high
glucose-induced and diabetes-induc ed oxidative str ess in
vitro and in vivo evaluated by el ectron spin resonance
measurement. Free Radical Biology and Medicine 2005, 39:444452.
830. Beckma n JA and Creager MA: The nonlipid effects o f statins on
endothelial function. Trends Cardiovasc Med 2006, 16:15662.
831. Carloni S, Mazzoni E, Cimino M, De Simoni MG, Perego C, Scopa C
and Balduini W: Simvastatin reduces caspase-3 activation and
inflammatory markers induced by hypoxia-ischemia in the
newborn rat. Neurobiol Dis 2006, 21:11926.
832. Grimes DS: Are statins analogues of vitamin D? Lancet 2006,
368:836.
833. Paumel le R, Blanquart C, Briand O , Barbier O, Duhem C, Wo erly G,
Percevault F, Fruchart JC, Dombrowicz D, Glineur C and Staels B:
Acute antiinflammatory properties of statins involve per-
oxisome proliferator-activated receptor-alpha via inhibition
of the protein kinase C signaling pathway. Circ Res 2006,
98:3619.
834. Alegret M and Silvestre JS: Pleiotropic ef fects of statins and
related pharmacological experimental approaches. Timely
Top Med Cardiovasc Dis 2007, 11:E10.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 43 of 79
(page number n ot f or citation purposes)
835. Farooqui AA, Ong WY, Horrocks LA, Chen P and Fa rooqui T:
Comparison of biochemical eff ects of statins and fish oil in
brain: the battle of the titans. Brain Res Rev 2007, 56:44371.
836. Kuipers H F and Elsen van den PJ: Immunomodulation by statins :
inhibition of cholesterol vs. isoprenoid biosynthesis. Biomed
Pharmacother 2007, 61:400 7.
837. Lahera V, Goicoechea M, de Vinuesa SG, Miana M, de las Heras N,
Cachofei ro V and Luno J: Endothelial dysfunction, oxi dative
stress and inflammation in at heros clerosis: beneficial effects
of statins. Curr Med Chem 2007, 14:2438.
838. Paraskevas KI, Tzovaras AA, Briana DD and Mikhailidis DP:
Emerging indications for statins: a pluripotent family of
agents with several potential applications. Curr Pharm Des
2007, 13:362236.
839. T erblanche M, Almog Y, Rosenson R, Smith TS and Hackam DG:
Statins and sepsis: multiple modifications at multiple levels.
Lancet Infectious Diseases 2007, 7:358368.
840. Wang CY, Liu PY and Liao JK: Pleiotropic effects of stati n
therapy: molecular mechanisms and clinical results. Trends
Mol Med 2008, 14:3744.
841.WagnerBK,KitamiT,GilbertTJ,PeckD,RamanathanA,
Sch reiber SL, Golub TR and Mootha VK: Large-scale chemical
dissection of mitochondrial function. Nat Biotechnol 2008,
26:343351.
842. Libby P and Aikawa M: Stabilization of atherosclerotic plaques:
new mechanisms and clinical targets. Nat Med 2002,
8:125762.
843. Blake GJ and Ridker PM: Are statins anti-inflammatory? Curr
Control Trials Cardiovasc Med 2000, 1:161165.
844. D iomede L, Albani D, S ottoc orno M, Donati MB, Bianchi M,
Fruscella P and Salmona M : In vivo anti-inflammatory effect of
statinsismediatedbynonsterol mevalonate products.
Arterioscler Thromb Vasc Biol 2001, 21:132732.
845. Laufs U, Gertz K, Dirnagl U, Bohm M, Nickenig G and Endres M:
Ros uvastatin, a new HMG-CoA reductase inhibitor, upre-
gulates endothelial nitric oxide synthase and protects from
ischemic stroke in mice. Br ain Res
2002, 942:2330.
846. Neuhaus O, Strasser-Fuchs S, Fazekas F, Kieseier BC,
Niederwieser G, Hartung HP and Archelos JJ: Statins as
immunomodulators: comparison with interferon-beta 1b
in MS. Neurology 2002, 59:9907.
847. R asmussen LM, Hansen PR, Nabipour MT, Olesen P, Kristiansen MT
and Ledet T: Diverse effec ts of inhibition of 3-hydroxy-3-
methylglutaryl-CoA reduct ase on the expression of VCAM-
1 and E-selectin in endothelial cells. Biochem J 2001,
360:36370.
848. T akemoto M an d Liao JK: Pleiotropic effects of 3-hydroxy-3-
methylglutaryl coenzyme a reductase inhibitors. Arterioscler
Thromb Vasc Biol 2001, 21:17129.
849. Sukhova GK, Williams JK and Libby P: St atins reduce inflamma-
tion in atheroma of nonhuman primates independent of
effects on serum cholesterol. Arterioscler Thromb Vasc Biol 2002,
22:14528.
850. Scalia R and Stalker TJ: Microcirculation as a target for the anti-
inflammatory properties of statins. Microcirculatio n 2002,
9:43142.
851. Weitz-Schmidt G: Statins as anti-inflammatory agents. Trends
Pharmacol Sci 2002, 23:4826.
852. Y oussef S, Stuve O, Patarroyo JC, Ruiz PJ, Radosevich JL, Hur EM,
Bravo M, Mitchell DJ, Sobel RA, Steinman L and Zamvi l SS: The
HMG-CoA reductase inhibitor, atorva statin, promotes a
Th2 bias and reverses paralysis in central nervous system
autoimmune disease. Nature 2002, 420:7884.
853. Balk EM, Lau J, Goudas LC, Jordan HS, Kupelnick B, Kim LU and
Karas RH: Effects of statins o n nonlipid serum markers
associated with cardiovascular disease: a systematic review.
AnnInternMed2003, 139:67082.
854. Blanco-Colio LM, Tuñon J , Martin-Ventura JL and Egido J: Anti-
inflammatory and immunomodulatory effects of statins.
Kidney Int 2003, 63:1223.
855. Kwak BR, M ulhaupt F and Mach F: Atherosclerosis: anti-
inflammatory and immu nomodulatory activities of statins.
Autoimmun Rev 2003,
2:3328.
856. Mason JC: Statins and their role in vascular protection. Cl in S ci
(Lond) 2003, 105:25166.
857. Naidu BV, Wool ley SM, Farivar AS, Tho mas R, Fraga C and
Mulligan MS: Simvastatin ameliorates injury in an experi-
mental model of lung ischemia-reperfusion. J Thorac Cardiovasc
Surg 2003, 126:4829.
858. Pate GE, Tahir MN, Murphy RT and Foley JB: Anti-inflammatory
effects of statins in patients with aortic stenosis. J Cardiovasc
Pharmacol Ther 2003, 8:2016.
859. Shishehbor MH, Aviles RJ, Brennan ML, Fu XM, Goormastic M,
Pea rce GL, Gokce N, Keaney JF, Penn MS, Sprecher DL, Vit a JA and
Hazen SL: Association of nitrotyrosine levels wi th cardiovas-
cular disease and modulation by statin therapy. JAMA 2003,
289:16751680.
860. S tüve O, Youssef S, Dunn S, Slavin AJ, Steinman L and Zamvil SS:
The potential therapeutic role of statins in central nervous
system autoimmune disorders. Cell Mol Life Sci 2003,
60:248391.
861. StüveO,Prod'hommeT,SlavinA,YoussefS,DunnS,SteinmanLand
Zamvil SS: Statins and their potential targets in multiple
sclerosis therapy. Expert Opin Ther Targets 2003, 7:61322.
862. Stüve O, Youssef S, Steinman L and Z amvil SS: Statins as potential
therapeu tic agents in neuroinflammatory disorders. Curr
Opin Neurol 2003, 16:393401.
863. D urant R, Klouche K, Delbosc S, Morena M, Amigues L, Beraud JJ,
Canaud B and Cristol JP: Su peroxide anion overproduction in
sepsis: effects of vitamin e and simvastatin. Shock 2004,
22:349.
864. Endres M and Laufs U: Effects of statins on endothelium and
signaling mechanisms. St roke 2004, 35:270811.
865. Kunkel EJ, Dea M, Ebens A, Hytopoulos E, Melrose J, Nguyen D,
Ota KS, Plavec I, Wang Y, Watson SR, Butcher EC and Berg EL: An
integrative biology approach for analysis of drug action in
models of human vascular inflammation. FASEB J 2004,
18:1279
81.
866. McCarey DW, McInnes IB, Madhok R, Hampson R, Scherbakov O,
Ford I, Capell HA and Sattar N: Trial of Atorvastatin in
Rheumatoid Arthritis (TARA): double-blind, randomised
pla cebo-controlled trial. Lancet 2004, 363:201521.
867. R osenson RS: Statins in atherosclerosis: lipid-lowering agents
with antioxidant capabilities. Atherosclerosis 2004, 173:1 12.
868. McInnes IB, Mc Carey DW and Sattar N: Do statins offer
therapeu tic potential in inflamma tory arthritis? Ann Rheum
Dis 2004, 63:15357.
869. Steffens S and Mach F: Anti-inflammatory properties of statins.
Semin Vasc Med 2004, 4:41722.
870. Stoll LL, McCormic k ML, Denning GM and Weintraub N L: Anti-
oxidant effects of statins. Drugs Today (Barc) 2004, 40:97590.
871. Arnaud C, Braunersreuther V and Mach F: Toward immunomo-
dulatory and ant i-inflammatory propert ies of statins. Trends
Cardiovasc Me d 2005, 15:202 6.
872. Endres M: Statins and stroke. J Cereb Blood Flow Metab 2005,
25:1093110.
873. Gaugler MH, Vereycken-Holler V, Squiban C, Vandamme M,
Vozenin-Brotons MC and Benderitter M: Pravastatin limits
endothelial activation after irradiation and decreases the
resulting inflammatory and thrombotic responses. Radiat Res
2005, 163:47987.
874. Jain MK and Ridker PM: Anti-inflammatory effects of statins:
clinical evidence and basic mech anism s. Nat Rev Drug Discov
2005, 4:97787.
875. Macin SM, Perna ER, Farias EF, Franciosi V, Cialzet a JR, Brizuela M,
Medina F, Tajer C, Doval H and Badaracco R: Atorvastatin has an
imp ortant acu te 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.
876. Methe H, Kim JO, Kofler S, Nabauer M and Weis M: Statins
decrease Toll-like receptor 4 expression and downstream
signaling in hum an CD14+ monocytes.
Arterioscler Thromb Vasc
Biol 2005, 25:143945.
877. Schult z Johansen J, Harris AK, Rychly DJ and Ergul A: Oxidative
stress and the use of antioxidants in diabetes: linking basic
science to clinical practice. Cardiovasc Diabetol 2005, 4:5.
878. T onelli M, Sacks F, Pfeffer M, Jhangri GS a nd Curhan G: Biomarkers
of inflammation and progression of chronic kidney disease.
Kidney Int 2005, 68:23745.
879. Abeles AM and Pillinger MH: Statins as antiinflammatory and
immunomodulatory agents: a future in rheumatologic
therapy? Arthriti s Rheum 2006, 54:393407.
880. Endres M: Statins: potential new indications in inflam matory
conditions. Atheroscler Suppl 2006, 7:315.
881. Y ang J, Li XP, Zhao SP, Li J, Li JD and Xie XM: The effect of
different doses of fluvastatin on inflammatory markers in
the early phase of acu te cor onary syndrome. Clin Chim Acta
2006, 368:1837.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 44 of 79
(page number n ot f or citation purposes)
882. Ahn KS, Sethi G and Aggarwal BB: Simvastatin potentiates TNF-
alp ha-in duced 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, 178:250716.
883. Becker RC: Off-target properties of pharmacotherapy and
the importance of mechanistic investigations in early clinical
phase drug development. J Thromb Thrombolysis 2007, 23:15961.
884. Greenwood J and Mason JC: Statin s and the vascular endothe-
lial inflamma tory response. Trends Immunol 2007, 28:8898.
885. Li JJ, Zheng X and Li J: Statins may be beneficial for patients
with slo w coronary flow syndrome due to its anti-inflam-
matory property. Med Hyp otheses 2007, 69:333 7.
886. Sandek A, Utchill S and Rauchhaus M: T he endotoxin-lipoprotein
hypothesis an update. Archives of Medical Science 2007, 3:
S81S90.
887. Siffrin V, Brandt AU, Herz J and Zipp F: New insights into
adaptive immunity in chronic neuroinflammation. Adv
Immunol 2007, 96:140.
888. Souza-Costa DC, S andrim VC, Lopes LF, Ge rlach RF, Rego EM and
Tanus-Santos JE: Anti-inflammatory effects of atorvastatin:
modulation by the T-786C polymorphism in the endothelial
nitric o xide synthase gene. Atherosclerosis 2007, 193: 43844.
889. Hernández-Romero MdC, Argüelles S, Villaran RF, de Pablos RM,
Delgado-Cortés MJ, Santiago M, Herrera AJ, Cano J and Machado A:
Sim vastatin prevents the inflammatory process and the
dopaminergic degeneration induced by the intranigral
injection of lipopoly saccharide. J Neurochem 2008, 105:44559.
890. Paraskevas KI: Statin treatment for rheumatoid arthritis: a
promising novel indication. Clin Rhe umato l 2008, 27:2817.
891. Prinz V, Laufs U, Gertz K, Kronenberg G, Balkaya M, Leithner C,
Lindauer U and Endres M: Intravenous rosuvastatin for acute
stroke treatment: an animal study. Stroke 2008, 39:433
8.
892. Xu SZ, Zhong W, Watson NM, Dickerson E, Wake JD, Lindow SW,
Newton CJ and Atkin SL: Fluvastatin reduces oxidative damage
in human vascular endothelial cells by upregulating Bcl-2.
J Thromb Haemost 2008, 6:692700.
893. D ombrecht EJ, De Tollenaere CB, Aerts K, Cos P, Schuerwegh AJ,
Bridts CH, Van Offel JF, Ebo DG, St evens WJ and De Clerck LS:
Antioxidant effect of bisphosphonates and simvastatin on
chondrocyte lipi d peroxidation. Biochem Biophys Res Commun
2006, 348:45964.
894. Lee DH, Zacharski LR and Ja cobs DR: Compar ison of the serum
ferritin and percentage of transferrin saturation as exposure
markers of iron- driven oxidat ive stress-related disease
outcomes. American Heart Journal 2006, 151:.
895. Anderson LJ, Holden S, Davis B, Prescott E, Charrier CC, Bunce NH,
Firmin DN, Wonke B, Porter J, Walker JM and Pennell DJ:
Cardiovascular T2-sta r (T2*) magnetic resonance for the
early diagnosis of myocardial iron overload. Eur Heart J 2001,
22:21712179.
896. Yeoh-Ellerton S and Stacey MC: Iron and 8-isopr ostane levels in
acute an d chronic wounds. J Invest Dermatol 2003, 121:918 925.
897. Bergan JJ, Schmid-Schonbein GW, Smith PDC, Nicolaides AN,
Boisseau MR and Eklof B: Mechanisms of disease: Chronic
venous disease. New England Journal of Medicine 2006,
355:488498.
898. Zamboni P, Izzo M, Fogato L, Carandina S and Lanzara V: Urine
hemosid erin: a novel marker to assess the severity of
chronic venous disease. J Vasc Surg 2003, 37:1326.
899. Zamboni P: The Big Idea: Iron-dependent inflammation in
venous disease and proposed parallels in multiple sclerosis.
JRSocMed2006, 99:589593.
900. Lucas SM, Rothwell NJ and Gibson RM: The role of inflammation
in CNS injury and disease. Br J Pharmacol 2006, 147(Su ppl 1):
S23240.
901.LewénA,MatzPandChanPH:Free radical pathways in CNS
injury. J Neurotrauma 2000, 17:87190.
902. Que EL, Domaille DW and Chang CJ: Metals in neurobiology:
probing their chemistry and biology with molecular ima-
ging. Chem Rev 2008, 108:151749.
903. Kent TA, Soukup VM and Fabian RH: Heterogeneity affecting
outcome from acute stroke therapy: making reperfusion
worse. Stroke 20 01, 32:231827.
904. Emsley HC a nd Tyrrell PJ: Inflammation and infection in clinical
stroke. J Cereb Blood Flow Metab 2002, 22:1399419.
905. Patt A, Horesh IR, Berger EM, Harken AH and Repine JE: Iron
depletion or chel ation reduces ische mia/reperfusion -
induced edema in gerbil brains. JPediatrSurg1990, 25:2247.
906. Dávalos A, Fernandezreal JM, Ricart W, Soler S, Molins A, Planas E
and Genis D: Iron-related damage in acute ischemic stroke.
Stroke 1994, 25 :15431546.
907. Bishop GM and Robinson SR: Quantitative analysis of cell death
and ferritin expression in response to cortical iron: implica-
tions for hypoxia-ischemia and stroke. Brain Research 2001,
907:175187.
908. D emougeot C, Van Hoecke M, Bertrand N, Prigent-Tessier A,
Mossiat C, Beley A and Marie C: Cytoprotective efficacy and
mechanis ms of the liposoluble iron chelator 2,2 '-dipyridy l in
the rat photothrombotic ischem ic stroke model. J Pharm Exp
Therapeut 2004, 311:10801087.
909. Mehta SH, Webb RC, Ergul A, Tawak A and Dorrance AM:
Neuroprotection by tempol in a model of iron-induced
oxidative stress in acute ischemic stroke. Am J Physiol 2004,
286:R283R288.
910. Selim MH and Ratan RR: The role of iron neurotoxicity in
ischemic stroke. Ageing Researc h Re views 2004, 3:345353.
911. v an der ADL, Grobbee DE, Roest M, Marx JJM, Voorbij HA and
Schouw van der YT: Serum ferritin is a risk factor for stroke in
postmenopausal women. Stroke 2005, 36:16371641.
912. Hua Y, Keep RF, Hoff JT and Xi G: Brain i njury after
intracerebral hemorrhage: the role of thrombin and iron.
Stroke 2007, 38 :759
62.
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 and
Dávalos A: Increased body iron stores are associated with
poor outcome after thrombolytic treatment in acute
stroke. Stroke 2007, 38:90 95.
914. Slemmer JE, Shacka JJ, Sweene y MI and Weber JT: Antioxidants
and free ra dical s caven gers for the treatment of stroke,
traumatic brain injury and aging. Curr Med Chem 2008 ,
15:40414.
915.BakerK,MarcusCB,HuffmanK,KrukH,MalfroyBand
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, 284:21521.
916. Sun AY and Chen YM: Oxidative str ess and neurodegenerative
disorders. Journal of Biomedical Science 1998, 5:401414.
917. Pong K: Oxidative stress in neurodegenerat ive diseases:
therapeutic implications for superoxide dismutase
mimetics. ExpertOpiniononBiologicalTherapy20 03, 3:127139.
918. Bastianetto S and Quirion R: Natural antioxidants and neuro-
deg enerative diseases. Frontiers in Bioscience 2004, 9:34473452.
919. Warner DS, Sheng HX and Batinic-Haberle I: Oxidants, antiox-
ida nts and the ischemic brain. JExpBiol2004, 207:32213231.
920. Craft JM, Watterson DM and Van Eldik LJ: Neuroinflammation: a
potential therapeutic target. Expert O pin Ther Targets 2005,
9:887900.
921. Margaill I, Plotkine M and Lerouet D: Antioxidant strategies in
the treatment of stroke. Free Radical Biology and Medicine 2005,
39:429443.
922. Zhao BL: Natural antioxidants for neurodegenerative dis-
eases. Mol Neurobiol 2005, 31:283293.
923. Obrenovitch TP:
Molecular physiology of preconditioning-
induced brain tolerance to ischemia. Physiol Rev 2008,
88:21147.
924. Sirén AL, Fratelli M, Brines M, Goemans C, Casagrande S, Lewczuk P,
KeenanS,GleiterC,PasqualiC,CapobiancoA,MenniniT,
Heumann R, Cerami A, Ehrenreich H and Ghezzi P: Erythropoietin
prevents neuronal apoptosis after cerebral ischemia and
metabolic stress. Proc Natl A cad Sci USA 2001, 98:40449.
925.BrinesML,GhezziP,KeenanS,AgnelloD,deLanerolleNC,
Cerami C, Itri LM and Cerami A: Erythropoietin crosses the
blood-brain barrier to protect against experimental brain
injury. Proc Natl Acad Sci USA 2000, 97:1052631.
926. Digicaylioglu M and Lipton SA: Erythropoietin-mediate d neuro-
protection involves cross-talk between Jak2 and NF-kappaB
signalling cascades. Nature 2001, 412:6417.
927. D awson TM : Precondi tioning- mediated neuroprotection
through erythropoietin? Lancet 2002, 35 9:967.
928. Ehrenr eich H, Hasselb latt 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 and Sirén AL:
Erythropoietin therapy for acute stroke is bot h safe and
beneficial. Mol Med 2002, 8:495 505.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 45 of 79
(page number n ot f or citation purposes)
929. Leker R R and Shohami E: Cerebral ischemia and trauma-
different etiologies yet similar mechanisms: neuroprotec-
tive opportu nities. Brain Res Rev 2002, 39:5573.
930. Aydin A, Genc K, Akhisaroglu M, Yorukoglu K, Gokmen N and
Gonullu E: Erythr opoie tin exerts neuroprotective effect in
neonatal rat model o f hypoxic-ischemic brain injury. Brain
Dev 2003, 25:4948.
931. Kumral A, Ozer E, Yilmaz O, Akhisaroglu M, G okmen N, Duman N,
Ulukus C, Genc S and Ozkan H: Neuroprotective effect of
erythrop oietin on hypo xic-ischemic brain injury in neonatal
rats. Biol Neonate 2003, 83:224 8.
932. Matsus hita H, Johnston MV, Lan ge MS and Wilson MA: Protective
effect of erythropoietin in neonatal hypoxic ischemia in
mice. Neurorepor t 2003, 14:175761.
933. Grasso G, Sfacteria A, Cerami A and Brines M: Erythropoietin as a
tissue-protective cytokine in brain injury: what d o we know
and where do we go? Neuroscientist 2004, 10:938.
934. Sun Y, Zhou C, Polk P, Nanda A and Zhang JH: Mechanisms of
erythropoietin-induced brainprotectioninneonatal
hypoxia-ischemia rat model. JCerebBloodFlowMetab2004,
24:25970.
935. Wang CH, Liang CL, Huang LT, Liu JK, Hung PH, Sun A and Hung KS:
Single intravenous injection of naked plasmi d DNA encod-
ing erythropoietin provides neuroprotection in hypoxia-
ischemia rats. Biochem Biophys Res Commun 2004, 314:106471.
936. Bailey DM, Robach P, Thomse n JJ and Lundby C: Erythropoietin
depletes iron stores: antioxidant n europrotection for
ischemic stroke? St roke 2006, 37:2453.
937. Grasso G, Sfacteria A, Meli F, Passalacqua M, Fodale V, Buemi M,
Giambartino F, Iacopino DG and Tomase llo F : Theroleof
erythropoietin in neuroprotection: therapeutic perspec-
tives. Drug News Perspect 2007, 20:315 20.
938. McPherson RJ and Juul SE: Recent trends in erythropoietin-
mediated neuroprotectio n. Int J Dev Neurosci 2008, 26:103 11.
939. Sola A, Peng H, Rogido M and Wen TC:
Animal models of
neonatal stroke and response to e rythropoietin and cardio-
trophin-1. Int J Dev Neurosci 2008, 26:2735.
940. Erbayraktar S, Yilmaz O, Gokmen N and Brines M: Erythropoietin
is a mult ifunctiona l tissue-protective cytokine. Curr Hematol
Rep 2003, 2:46570.
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,
Sfa cteria A, Erbayraktar S, Erbayraktar Z, Gokmen N, Yilmaz O,
Cerami-Hand C, Xie QW , Coleman T, Cerami A and Brines M:
Derivatives of erythropoietin that are tissue protective but
not erythropoietic. Science 2004, 305:23942.
942. Arcasoy MO: The non-haematopoietic biolo gical effects of
erythrop oietin. Br J Haematol 2008, 141:1431.
943. Agnello D, Bigini P, Villa P, Mennini T, Cerami A, Brines ML and
Ghezzi P: Erythropoieti n exerts an anti-inflammatory effect
on the CNS in a model of experimental autoimmune
encephalomyelitis. Brain Res 2002, 952:12834.
944. Fahn S and Cohen G: The oxidant stress hy pothesis in
Parkinson's disease: evidence supporting it. Ann Neurol 1992,
32:80412.
945. Halliwell B: Reactive oxygen species and the centr al nervous
system. J Neurochem 1992, 59:16091623.
946. Coyle JT and Puttfarcken P: Oxidative s tress, glutamate, and
neurodegenerative disorders. Science 1993, 262:68 995.
947. Behl C, Davis JB, Lesley R and Schubert D: Hydrogen peroxide
mediates amyloid beta protei n toxicity. Cell 1994, 77:81727.
948. Good PF, Werner P, Hsu A, Olanow CW and Perl DP: Evidence of
neuronal oxidative damage in Alzheimer's disease. Am J
Pathol 1996, 149:218.
949. Jenner P and Olanow CW: Oxidative stress and the pathogen-
esis of Parkinson's disease. Neurology
1996, 47:S16170.
950. Simoni an NA and Coyle JT: Oxidative stress in neurodegen-
erative diseases. Annu Rev Pharmacol Toxico l 1996, 36:8310 6.
951. Y oritaka A, Hattori N, Uchida K, Tanaka M, Stadtman ER and
Mizuno Y: Immunohistochem ical detection of 4-hydroxyno-
nenal p rotein adducts in Parkinson disease. Proc Natl Acad Sci
USA 1996, 93:2696701.
952. Packer L, Tritschler HJ and Wessel K: Neuroprotection by the
metabolic antioxidant alpha-lipoic acid. Free Radic Biol Med
1997, 22:35978.
953. Gao HM, Liu B, Z hang W and Hong JS: Novel anti-inflammatory
therapy for Parki nson's disease. Trends Pharmacol Sci 2003,
24:395401.
954. Marian i E, Polidori MC , Cherubini A and Mecocci P: Oxidative
stress in brain aging, neurodegenerative and vascular
diseases: an overview. J Chromatogr B Analyt Technol Biomed Life
Sci 2005, 827:6575.
955. Zeevalk GD, Bernard LP, Song C, Gluck M and Ehrhart J:
Mit ochondrial inhibition and oxidative stress: reciprocating
players in neurodegeneration. Antioxid Redox Signal 2005,
7:111739.
956. Lin MT and Beal MF: Mitochondrial dysfunction and oxidative
stress in neurodegenerative diseases. Nature 2006,
443:78795.
957. T rushina E and McMurray CT: Oxidative stress and mitochon-
drial dysfunction in neurodegenerative diseases. Neuroscience
2007, 145:123348.
958. Benzi G and Moretti A: Are reactive oxygen species involved in
Alzheimer's disease? Neurobiol Aging 1995, 16:66174.
959. Hensley K, Hall N, Subramaniam R, Cole P, Harris M, Aksenov M,
Aksenova M, Gabbita SP, Wu JF and Carney JM, et al: Brain
regional correspondence between Alzheimer's disease
histopathology and biomarkers of protein oxidation. J
Neurochem 1995,
65:214656.
960. Markesbery WR: Ox idative stress hypothesis in Alzheimer's
disease. Free Radic Biol Med 1997, 23:13447.
961. Markesbery WR and Lovell MA: 4-hydroxynonenal, a product of
lipid peroxidation, is increased in the brain in Alzhei mer's
disease. Neurobiol Aging 1998, 19:336.
962. Cassarino DS and Bennett JP Jr : An evaluation of the role of
mitochondria in neurodegenerat ive diseases: mitochondrial
mutations and oxidative pathology, protective nuclear
responses, and cell death in n eurodegeneration. Brain Res
Brain Res Rev 1999, 29:125.
963. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM,
Cooper NR, Eikelenboom P, Emm erling M, Fiebich BL, Finch CE,
FrautschyS,GriffinWS,HampelH,HullM,LandrethG,LueL,
Mrak R, Mackenzie IR, McGeer PL, O'Banion MK, Pachter J,
Pas inetti G, Plata-Salaman C, Rogers J , Rydel R, Shen Y, Streit W,
Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Vee rhuis R,
Walker D, Webster S, W egrzyniak B, Wenk G and Wyss-
Coray T: Inflammation and Alzheimer's disease. Neurobiol
Aging 2000, 21:383421.
964. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK,
Jon es PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS,
Petersen RB and Smith MA: Oxidat ive damage is the earliest
event in Alzheimer disease. J Neuropathol Exp Neurol 2001,
60:759767.
965. McGeer PL and McGeer EG: Inflammation, autotoxicity and
Alzheimer disease. Neurobiol Aging 2001, 22:799809.
966. Butterfield DA and Lau derback CM: Lipid peroxidation and
protein oxidation in Alzheimer's disease brain: potential
causes and consequences involving amyloid beta-peptide-
associated fre e radical oxidative st ress. Free Radic Biol Med
2002, 32:105060.
967. Eikelenboom P, Bate C, Van Gool WA, Hoozemans JJ,
Rozemuller JM, Veerhuis R and Williams A: Neuroinflammation
in Alzheime r's disease and prion disease. Glia 2002, 40:2329.
968. Suh YH and Checl er F: Amyloid precursor protein, p resenilins,
and alpha-synuclein: molecular pathogenesis and pharma-
cological applications in Alzheimer's disease. Pharmacol Rev
2002, 54:469525.
969. McGeer EG and McGeer PL: Inflammatory processes in
Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry
2003, 27:7419.
970. Butterfield DA, Perluigi M and Sultana R: Oxidative st ress in
Alzheimer's disease brain: new insights from red ox proteo-
mics. Eur J Pharmac ol 20 06, 545:3950.
971. Jenner P: Oxidative stress in Parkinson's disease. Ann Neurol
2003, 53(Suppl 3):S26 36.
972. Nunomura A, Castellani RJ, Zhu X, Moreira PI, Perry G and
Smith MA: Involvement of oxidative stress in Alzheimer
disease. J Neuropathol Exp Neurol 2006, 65:63141.
973. Licastro F, Porcellini E, Caruso C, Lio D and Corder EH: Genetic
risk profiles for Alzheimer's dise ase: integration of APOE
genotype and variants that up-regulate inflammation.
Neurobiol Aging 2007, 28:1637 43.
974. Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia-Alloza M, de
Calignon A, Rozkalne A, Koenigsknecht-Talboo J, Holtzman DM,
Bacskai BJ and Hyman BT: Rapid appearance and local toxicity
of amyloid-beta plaques in a mouse model of Alzheimer's
disease. Nature 2008, 451:720 4.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 46 of 79
(page number n ot f or citation purposes)
975. Goodman L: Alzheimer's disease; a clinico-pathologic analysis
of twenty-thre e cases with a theory on pathogenesis. J Nerv
Ment Dis 1953, 118:97130.
976. Dexter DT, Wells FR, Lees AJ, Agid F, Agid Y, Jenner P and
Marsden CD: Increased nigral iron content and alterations in
other metal ions occurring in brain in Parkinsons disease.
J Neurochem 1989, 52:1830 1836.
977. R iederer P, Sofic E, Rausch WD, Schmidt B, Reynolds GP, Jellinger K
and Youdim MB: Transition metals, ferritin, glutathione, and
ascorbic acid in parkinsonian brains. J Neurochem 1989,
52:51520.
978. Jellinger K, Paulus W, Grundke-Iqbal I, Riederer P and Youdim MB:
BrainironandferritininParkinson'sandAlzheimer's
diseases. J Neural Transm Park Dis Dement Sect 1990, 2:32740.
979. O'Brien PJ: Molecu lar mechanisms of quinone cytotoxicity.
Chemico-Biol Interact 1991, 80:141.
980. Connor JR, Snyder BS, Beard JL, Fine RE and Mufson EJ: Regional
distribution of iron and iron-regulatory proteins in the brain
in aging and Alzheimers disease. JNeuroscieRes1992,
31:327335.
981.GoodPF,PerlDP,BiererLMandSchmeidlerJ:Selecti ve
accumulation of aluminum and iron in the neurofibrillary
tangles of Alzheimer's disease: a laser microprobe
(LAMMA) study. Ann Neurol 1992, 31:28692.
982. Mont gomery EB Jr: Heavy metals and the etiology of
Parkinson's disease and other movement disorders. Toxicol-
ogy 1995, 97:39.
983. Koeppen AH: The history of iron in the brain. JNeurolSci1995,
134(Suppl):19.
984. D eibel MA, Ehmann WD and Markesbery WR: Copper, iron, and
zin c imbalances in severely degenerated brain regions in
Alzheimer's disease: possible relation to oxidat ive stress.
JNeurolSci1996, 143:137 42.
985. Gorell JM, Jo hnson CC, Rybicki BA, Peterson EL, Kortsha GX,
Brown GG and Richardson RJ: Occupational expo sures to
metals as risk factors for Parkinson's disease. Neurology
1997,
48:6508.
986. Koeppen AH: A brief history of brain iron research. JNeurolSci
2003, 207:957.
987. Y oudim MBH, Benshachar D and Riederer P: The possible role of
iron in the etiopathology of Parkins ons disease. Movement
Disorders 1993, 8:112.
988. Gerlac h M, Benshachar D, Rieder er P and Youdim MBH: Altered
brain metabolism of iron as a cause of neuro degenerati ve
diseases. J Neurochem 1994, 63:793807.
989. tz ME, Künig G, Rieder er P and Youdim MBH: Oxidative stress
free radical productio n in neural degeneration. Pharmacol
Therapeut 1994, 63: 37 122.
990. Smith MA, Perry G , Richey PL, Sayre LM, Anderson VE, Beal MF and
Kowall N: Oxidative damage in Alzheimer's. Nature 1996,
382:120121.
991. LeVine SM: Iron deposits in multiple sclerosis and Alzhei-
mer's disease bra ins. Brain Research 1997, 760:298303.
992. Smith MA, Harris PLR, Sayre LM and Perry G: Iron accumulation
in Alzheimer disease is a source of redox-generated free
radicals. Proc Natl Acad Sci 1997, 94:98669868.
993.LovellMA,RobertsonJD,TeesdaleWJ,CampbellJLand
Markesbery WR: Copper, iron a nd zinc in Alzheimer's disease
senile p laques. JNeurolSci1998, 158:4752.
994. Bartzokis G, Cummings J, Perlman S, Hance DB and Mintz J:
Increased basal ganglia iron levels in Hu ntington disease.
Arch Neurol 1999, 56:56974.
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 and 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.
996. Jellinger KA: The role of iron in neurodegeneration
Prospects for pharmacotherapy of Parkinson's disease.
Drugs & Aging 1999, 14:115140.
997. Olanow CW and Tatton WG: Etiology an d pathogenesis of
Parkinson's diseas e. Annu Rev Neurosci 1999, 22:123 144.
998. Sayre LM, Perry G and Smith MA: Redox metals and neurode-
generative disease. Current Opinion in Chemical Biology 1999,
3:220225.
999. Bush AI: Metals and neuroscience. Curr Opin Chem Biol 2000,
4:18491.
1000. Chiueh CC, Andoh T, Lai AR, Lai E and Krishna G: Neuropro-
tective strategies in Parkinson's disease: protection against
progressive nigral damage induced by free radicals. Neurotox
Res 2000, 2:293310.
1001. Christen Y: Oxidative stress and Alzheimer disease. American
Journal of Clinical Nutrition 2000, 71:621S629S.
1002. Floor E: Iron as a vulnerability factor in nigrostriatal
dege neration in aging and Parkinson's di sease. Cell Mol Bi ol
(Noisy-le-grand) 2000, 46:70920.
1003. Smith MA, Rottkamp CA, Nunomura A, Raina AK and Perry G:
Oxidative stress in Alzheimer's disease. Biochim Biophys Acta
2000, 1502:13944.
1004. Varadarajan S, Yatin S, Aksenova M and Butterfield DA: Alzhei-
mer's amyloid beta-peptide-associated free radical oxida-
tive stress and neurotoxicity. J Struct Biol 2000, 130:184208.
1005. Berg D, Gerlach M, Youdim MBH, Double KL, Zecca L, Riederer P
and Becker G: Brain iron pathways and their relevance to
Parkinson's disease. J Neurochem 2001, 79:225236.
1006. Blum D, Torch S, Lambeng N, Nissou MF, Benabid AL, Sadoul R and
Verna JM: Molecular pathways involved in the neurotoxicity
of 6-OHDA, dopamine and MPTP: contribution to the
apoptotic theory in Parkinson's disease. Pr ogress in Neurobiol-
ogy 2001, 65:135172.
1007.CampbellA,SmithMA,SayreLM,BondySCandPerryG:
Mechanisms by which metals promote events connected to
neurodege nerati ve diseases. BrainResearchBulletin2001,
55:12513 2.
1008. Halliwell B: Role of free radicals i n the neurodegenerative
diseases Therapeutic implications for antioxidant treat-
ment. Drugs & Aging 2001, 18:685716.
1009. HaMai D, B ondy SC, Becaria A and Campbell A: The chemistry of
transition meta ls in relation to their potential role in
neurodegenerative processes. Curr Top Med C hem 2001,
1:54151.
1010. Rottkamp CA, Raina AK, Zhu XW , Gaier E, Bush AI, Atwood CS,
Chevion M, Perry G and Smith MA: Redox-active iron mediates
amyloid-beta toxicity. Free Rad Biol Med 2001, 30:447450.
1011. Sayre LM, Smith MA and Perry G: Chemistry and biochemistry
of oxidative stress in neurodegenerative diseas e. Current
Medicinal Chemistry 2001, 8:721738.
1012. Bharath S, Hsu M, Kaur D, Rajagopalan S and Andersen JK:
Glutathione, iron and Parkins on's disease. Biochem Pharmacol
2002, 64:103748.
1013. Bishop GM, Robinson SR, Liu Q, Perry G, Atwood CS and
Smith MA: Iron: a pathological mediator of Alzheimer
disease? Dev Neurosci 2002, 24:1847.
1014. Giasson BI, Ischiropoulos H, Lee VM and Troja nowski JQ: The
relationship between oxidative/nitrative stress and patho-
logical inclusions in Alzheimer's and Parkinson's diseases.
Free Radic Biol Med 2002, 32:126475.
1015. Gnjec A, Fonte JA, Atwood C and Martins R N: Transition metal
chelator therapy A potential treatment for Alzheimer's
disease? Frontiers in Bioscience 2002, 7:D1016D1023.
1016.PerryG,SayreLM,AtwoodCS,CastellaniRJ,CashAD,
Rottkamp CA and Smith MA:
The role of iron and copper in
the aetiology of neurodegenerative disorders Therapeu-
tic implications. CNS Drugs 2002, 16:339 352.
1017. Rao AV and Balachandran B: Role of oxidative stress an d
antioxidants in n eurod egenerative diseases. Nu tritional Neu-
roscience 2002, 5:291309.
1018. Burdo JR and Connor JR: Brain iron uptake and homeostatic
mechanisms: An overview. Bi ometals 20 03, 16:6375.
1019. Bush AI: The metallobiology of Alzhe imer's disease. Trends
Neurosci 2003, 26:207214.
1020. Finefrock AE, Bush AI and Doraiswamy PM: Current status of
metals as therapeutic targets in Alzheimer's disease. JAm
Geriatr Soc 2003, 51:11438.
1021. KeYandQianZM:Iron misregulation in the brain: a primary
cause of neurodegenerative disorders. Lancet Neurol 2003,
2:246253.
1022. Barnham KJ, Masters CL and Bush AI: Neurodegenerative
diseases and oxidative stress. Nat R ev Dru g Disco v 2004,
3:20514.
1023. Bondy SC, Lahiri DK, Perreau VM , Sharman KZ, Cam pbell A,
Zhou J and Sharman EH: Retardation of brain aging by chronic
treatment with melatonin. AnnNYAcadSci2004,
1035:197215.
1024. Casadesus G, Smith MA, Zhu X, Aliev G, Cash AD, Honda K,
Petersen RB and Perry G: Alzheimer disease: evidence for a
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 47 of 79
(page number n ot f or citation purposes)
central pathogenic role of iron-mediated reactive oxygen
species. J Alzheimers Dis 2004, 6:1659.
1025. Doraiswa my PM and F inef rock AE: Metals in our minds:
therapeutic implications for neurodegenerative disorders.
Lancet Neurology 2004, 3:431434.
1026. Gotz ME, Double K, Gerlach M, Youdim MBH and 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, 1012:193208.
1027. Huang X, Moir RD, Tanzi RE, Bush AI and Rogers JT: Redox-
active metals, oxidative stress, an d Alzheimer's disease
pathology. AnnNYAcadSci2004, 1012:15363.
1028. Kalivendi SV, Cunningham S, Kotamraju S, Joseph J, Hillard CJ and
Kalyanaraman B: alpha-synuclein up-regulation and aggrega-
tion during MPP+-induced apoptosis in neuroblastoma
cells Intermediacy of transferrin receptor iron and
hydrogen peroxide. Journal of Biological Chemistry 2004,
279:1524015247.
1029. Kaur D and Anders en J: Does cellular iron dysregulation play a
causative role in Parkinson's disease? Ageing Re s Rev 2004,
3:327343.
1030. Mattson MP: Metal-catalyzed disruption of membrane
protein and lipid signaling in the pathogenesis of neurode-
generative disorders. AnnNYAcadSci2004, 1012:3750.
1031. Mattson MP: Pathways towards and away from Alzheimer's
disease. Nature 2004, 430:6319.
1032. Milton NGN: Role of hydrogen peroxide in the aetiology of
Alzheimer 's disease Implications for treatment. Drugs &
Aging 2004, 21:81100.
1033. Moos T and Morgan EH: The metabolism of neuronal iron and
its pathogenic role in neurological disease Review. Ann NY
Acad Sci 2004, 10 12:
1426.
1034. Poon HF, Calabrese V, Scapagnini G and Butterfield D A: Free
radicals and b rain aging. Clin Geriatr Med 2004, 20:32959.
1035. Richardson DR: Novel chelators for central nervous system
disorders that involve alt erations in the metaboli sm o f iron
and other metal ions. AnnNYAcadSci2004, 1012:32641.
1036. Warner DS, Sheng H and Batinić-Haberle I: Oxidants, antiox-
idants and the ischemic brain. JExpBiol2004, 207:322131.
1037. Youdim MBH, Fridkin M and 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.
1038. Zecca L, Youdim MBH, Riederer P, Connor JR and Crichton RR:
Iron, brain ageing and neurodegenerative disorders. Nat Rev
Neurosci 2004, 5:863873.
1039. Zhu X, Raina AK, Lee HG, Casadesus G, Smith MA an d Perry G:
Oxidative stress signalling in Alzheimer's disease. Brain Res
2004, 1000:329.
1040. Calabrese V, Lodi R, Tonon C, D'Agata V, Sapienza M, Scapagnini G,
Mangiameli A, Pennisi G, Stella AMG and Butterfield DA:
Oxidative stress, mitochondrial dysfunction and cellular
stress response in Friedreich's ataxia. Journal of the Neurolo-
gical Sciences 2005, 23 3:145 162.
1041. Gaeta A and Hider RC: The crucial role of metal ions in
neurodegeneration: the basis for a promising therapeutic
strategy. Br J Pharma col 2005, 146:104159.
1042. Hald A and Lotharius J: Oxidative stress and inflammation in
Parkinson's disease: Is there a causal link? Exp Neurol 2005,
193:279290.
1043. Honda K, Smith MA, Zhu X, Baus D, Merrick WC, T artakoff AM,
Hattier T, Harris PL, Siedlak SL, Fujioka H, Liu Q, Moreira PI,
Miller FP, Nunomura A, Shimohama S and Perry G: Ribosomal
RNA in Alzheimer disease is oxidiz ed by bound redox-
active iron. JBiolChem2005, 280:20978
86.
1044. Ide-Ektessabi A and Rabionet M: The role of trace metallic
elements in neurodegenerative disorders: quantitative
analysis using XRF and XANES spectro scopy. Anal Sci
2005, 21:88592.
1045. Liu G, Garrett MR, Men P, Zhu XW, Perry G and Smith MA:
Nanoparticle and ot her metal chelation therapeutics in
Alzheimer disease. Biochimica Et Biophysica Acta-Molecular Basis of
Disease 2005, 1741:246252.
1046. Maynar d CJ, Bush AI, Masters CL, Cappai R and Li QX: Metals and
amyloid-beta in Alzheimer's disease. Int J Exp Pathol 2005,
86:14715 9.
1047. Ong WY and Farooqui AA: I ron, neuroinflammation, and
Alzheimer 's disease. J Alzheimers Dis 2005, 8:183200.
1048. Adlard PA and Bush AI: Metals and Alzheimer's disease.
J Alzheimers Dis 2006, 10:145 163.
1049. Aracena P, Aguirre P, Munoz P and Nunez MT: Iron and
glutathione at the crossroad of redox metabolism in
neurons. Biol Res 20 06, 39:15765.
1050. Berg D and Hochstrasser H: Iron metabolism i n parkinsonian
syndromes. Movement Disorder s 2006, 21:12991310.
1051. Collingwood J and Dobson J: Mapping and characteri zatio n of
iron compounds in Alzheimer's tissue. J Alzheimers Dis 2006,
10:21522 2.
1052. Fasano M, Bergamasco B and Lopiano L: Modifications of the
iron-neuromelanin system in Parkinson's disease. J Neuro-
chem 2006, 96:909916.
1053. Gaggelli E, Kozlowski H, Valensin D and Valensin G: Copper
homeostasis and neurodegenerative disorders (Alzhei-
mer's, prion, and Parkinson's diseases and amyotrophic
lateral sclerosis). Chem Rev 2006, 10 6:19952044.
1054. Gerlach M, Double KL, Youdim MBH and Riederer P: Potential
sources of increased iron in the substantia nigra of
parkinsonian patients. JNeuralTransmission
Suppl 2006,
133 142.
1055. Halliwell B: Oxidative stress and neurodegeneration: where
are we now? Journal of Neurochem istry 2006, 97:16341658.
1056. Huber A, Stuchbury G, Burkle A, Burne ll J a nd Munch G:
Neuroprotective therapies for Alzheimer's disease. Curr
Pharmaceut Des 2006 , 12:705717.
1057. Liu GJ, Huang WD, Moir RD, Vanderburg CR, Lai B, Peng ZC,
Tanzi RE, Rogers JT a nd Huang XD: Metal e xposure and
Alzheimer 's pathogenesis. J Struct Biol 2006, 155:4551.
1058. Lee DW, Andersen JK an d Kaur D: Iron dysreg ulation and
neurodegeneration The mole cular connection. Molecular
Interventions 2006, 6:8997.
1059. Mancuso M, Coppede F, Migliore L, Siciliano G and Murri L:
Mitochond rial dysfunction, oxidative stress and neurode-
generation. Journal of Alzheimers Disease 2006, 10:5973.
1060. Mosley RL, Benner EJ, Kadiu I, Thomas M, Boska MD, Hasan K,
Laurie C a nd Gendelman HE: Neuroinflammation, oxidative
stress, and the pathogenesis of Parkinson's disease. Clinical
Neuroscience Resear ch 2006, 6:26128 1.
1061. Quintana C, Bellefqih S, Laval JY, Guerquin-Kern JL, Wu TD, Avila J,
Ferrer I, Arranz R and Patino C: Study of the localization of
iron, ferriti n, and hemosiderin in Alzheimer's disease
hippocampus by analytical microscopy at the subcellular
level. J Struct Biol 2006, 153:4254.
1062.WhitnallMandRichardsonDR:Iron: a new target for
pharmacological intervention in neurodegenerative dis-
eases. Semin Pediatr Neurol 2006, 13:18697.
1063. Zhang HY: Same causes, same cures. Biochem Biophys Res
Commun 2006, 351:578581.
1064. Bartzokis G, Tishler TA, Lu PH, Villablanca P, Altshuler LL,
Carter M, Huang D, Edwards N and Mintz J: Brain ferritin iro n
may influence age- and gender-related risks of neurode-
generation.
Neurobiol Aging 2007, 28:41423.
1065. Bondy SC and Sharman EH: Melatonin and the aging brain.
Neurochem Int 2007, 50:57180.
1066. Cass WA, Gron din R, Andersen AH, Zhang Z, Hardy P A, Hus sey-
Andersen LK, Rayens WS, Gerhardt GA and Gash DM: Iron
accumulation in the striatum predicts aging-r elated declin e
in motor function in rhesus monkeys. Neurobiol Aging 2007,
28:25871 .
1067. Castellani RJ, Moreira PI, Liu G, Dobson J, Perry G, Smith MA and
Zhu X: Iron: the Redox-active center of oxidative stress in
Alzheimer disease. Neurochem Res 2007, 32:16405.
1068. Crouch PJ, White AR and Bush AI: The modulation of metal
bio-availability as a therapeutic strategy for the treatment
of Alzheimer's disease. FEBS J 2007, 274:377583.
1069. Donnelly PS, Xiao Z and Wedd AG: Copper and Alzheimer's
disease. Curr Opin Ch em Biol 2007, 11:12833.
1070. Gaasch JA, Lockman PR, Geldenhuys WJ , Allen DD and Schyf Van
der CJ: Brain iron toxicity: differential responses of astro-
cytes, neurons, and endoth elial cells. Neurochem Res 2007,
32:1196208.
1071. Ke Y and Qian ZM: Brain iron metabolism: neurobiology and
neurochemistry . Prog Neurob iol 2007, 83:14973.
1072. Mancus o C, Scapagnini G, Curro D, Stella AMG, De Marco C,
Butterfie ld DA and Calabres e V: Mitocho ndrial dysfunction,
free radical generation and cellula r stress response in
neurodegenerative disorders. Frontiers in Bioscience 2007,
12:11071123.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 48 of 79
(page number n ot f or citation purposes)
1073. Mandel S, Amit T, Bar-Am O and 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 Neurobio l 2007,
82:34860 .
1074. Molina-Holgado F, Hider RC, Gaeta A, Williams R and Francis P:
Metals ions and neurodegeneration. Biometals 2007,
20:63954 .
1075. Peters en RB, Nunomura A, Lee HG, Casadesus G, Perry G,
Smith MA and Zhu X: Signal transduction cascades associated
with oxidative stress in Alzheimer's disease. J Alzheimers Dis
2007, 11:14352.
1076. Reynolds A, Laurie C, Mosley RL and Gendelman HE: Oxidative
stress and the pathogenesis of neurodegenerative disor-
ders. Int Rev Neurobiol 2007, 82:297 325.
1077. Singh C, Ahmad I and Kumar A: Pesticides a nd metals induced
Parkinson's disease: involvement of free radicals and
oxidative stress. Cell Mol Biol (Noisy-le-grand) 2007, 53:1928.
1078. Stankiewicz J, Panter SS, Neema M, Arora A, Batt CE and Bakshi R:
Iron in chronic brain disorders: imaging and neurother-
apeutic implication s. Neurotherapeutics 2007, 4:371 86.
1079. Zhu X, Su B, Wang X, Smith MA and Perry G: Causes of
oxidative stress in Alzheimer disease. Cell Mol Life Sci 2007,
64:220210.
1080. Amit T, Avramovich-Tirosh Y, Youdim MB and Mandel S: Target-
ing multiple Alzheimer's disease etiologies with multi-
modal neuroprotective and neurorestorative iron
chelators. FASEB J 2008, 22:1296 305.
1081. Bush AI and Curtain CC: Twenty years of metallo-neurobiol-
ogy: where to now? Eur Biophys J 2008, 37:241 5.
1082. Drechsel DA and Patel M: Role of react ive oxygen species in
the neurotoxicity of environmental agent s implicated in
Parkinson's disease. Free Radic Biol Med 2008, 44:187386.
1083. Pankhu rst Q, Hautot D, Khan N and Dobson J: Inc reased levels
of magnetic iron compounds in Alzheimer's disease. J
Alzheimers Dis
2008, 13:4952.
1084.RogersJT,BushAI,ChoHH,SmithDH,ThomsonAM,
Friedlich AL, Lahiri DK, Leedman PJ , Hu ang X and 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, 36:12827.
1085. Silvestri L and Camaschella C: A potential pathogenetic role of
iron i n Alz heimer's Disease. JCellMolMed2008,
12:15481550.
1086. Snyder AM and Connor JR: Iron, the substantia nigra and
related neurological disorders. Biochim Biophys Acta 2008 in
press.
1087. Szczerbowska-Boruchowska M: X-ray fluorescence spectro-
metr y, an analytical tool in neurochemical research. X-Ray
Spectrometry 2008, 37:2131.
1088. Sayre LM, Perry G, Harris PLR, Liu YH, Schubert KA and Smith MA:
In situ oxidative catalysis by neurofibrillary tangles and
senile plaques in Alzheimer's disease: A central role for
bound transition metals. Jour nal of Neu rochemistry 2000,
74:27027 9.
1089. Vélez-Pardo C, Del Río MJ, Verschueren H, Ebinger G and
Vauquelin G: Dopamine and iron induce ap optosis in PC12
cells. Pharmacol Toxicol 1997, 80:7684.
1090. Kostoff RN and Briggs MB: Literature-Related Discovery
(LRD): potential treatments for Parkinson's disease. Tech-
nol. Forecast. Soc. Change 2007.
1091. LeVine SM and Cha krab arty A: Theroleofironinthe
pathogenesis of experimental allergic encephalomyelitis
and multipl e s clerosis. Redox-Active Metals in Neurological
Disorders, Annals of the New York Academy of Sciences 2004,
1012:252266.
1092. Kotze MJ, de Villiers JNP, Rooney RN, Grobbela ar JJ, Mansve lt EPG,
Bouwens CSH, Carr J, Stander I and du Plessis L: Analysis of the
NRAMP1 gene i mplicated in iron transport : Association
with multiple sclerosis and age effects. Blood Cells Molecules
and Diseases 2001, 27:4453.
1093. LeVine SM, Lynch SG, Ou CN, Wulser MJ, Tam E and Boo N:
Ferritin, transferrin and iron concent rations in the cere-
brospinal fluid of m ultiple sclerosis patients. Brain Research
1999, 821:511515.
1094. Valberg LS, Flanagan PR, Kertesz A and Ebers GC: Abnormalities
in Iron-Metabolism in Multiple-Sclerosis. Canadian Journal of
Neurological Sciences 1989, 16:184 186.
1095. Adams CWM: Perivascular Iron Deposition and Other
Vascular Damage in Multiple-Sclerosis. Journal of Neurology
Neurosurgery and P sychiatry 1988, 51:260265.
1096. Drayer B, Burger P, Hurwitz B, Dawson D and Cain J: Reduced
Signal Intensity on Mr Images of Thalamus and Putamen in
Multiple-Scler osis Increased Iron Content. American Journal
of Neuroradiology 1987, 8:413 419.
1097. Craelius W, Migdal MW, Luessenhop CP, Sugar A a nd Mihalakis I:
Iron Deposits Surrounding Multiple-Sclerosis Plaques.
Arch ives of Pathology & Laboratory Medicine 1982, 106:397399.
1098. Ge Y, Jensen JH, Lu H, Helpern JA, Miles L, Inglese M, Babb JS,
Herbert J and Grossman RI: Quantitative assessment of iron
accumulation in the deep gray matter of multiple sclerosis
by magnetic field correlation im aging. AJNR Am J Neuroradiol
2007, 28:163944.
1099. Kim NH, Choi JK, Jeong BH , Kim JI, Kwon MS, Carp RI and Kim YS:
Effect of transition metals (Mn, Cu, Fe) and deoxycholic
acid (DA) on the conversion of PrPC to PrPres. FASEB J
2005, 19:7835.
1100. Basu S, Mohan ML, Luo X, Kundu B, K ong Q and Singh N:
Modulation of proteinase K-resis tant prion protein in cells
and infectious brain homogenate by redox iron: implica-
tions for prion replication and disease pathogenesis. Mol Biol
Cell 2007, 18:3302 12.
1101. Lill R and Muhlenhoff U: Maturation of iron-sulfur proteins in
eukaryotes: mechanisms, connected processes, and dis-
eases. Annu Rev Biochem 2008, 77:669700.
1102. Cavadini P, O'Neill HA, Benada O and Isaya G: Assembly and
iron-binding properties of human frataxin, the protein
deficient in Friedreich ataxia. Hum Mol Genet 2002, 11:21727.
1103. Napier I, Ponka P and Richardson DR: I ron trafficking in the
mitochondrion: novel pathways revealed by disease. Blood
2005, 105:1867 74.
1104. O'Neill HA, G akh O, Park S, Cu i J, Mooney SM, Sampson M,
Ferreira GC and Isaya G: Assembly o f human frataxin is a
mechanism for detoxifying redox-active iron.
Biochemistry
2005, 44:53745.
1105. Gakh O, Park S, Liu G, Macomber L, Imlay JA, Ferreira GC and
Isaya G : Mitochondrial iron detoxification is a primary
function of frataxin that limits oxidative damage and
preserves cell longevity. Human Molecular Genetics 2006,
15:46747 9.
1106. Napoli E, Taroni F and Cortopassi GA: Frataxin, iron-sulfur
clusters, heme, ROS, and aging. Antioxid Redox Signal 2006,
8:50616.
1107. Babady NE, Carelle N , Wells RD, Rouault TA, Hirano M, Lynch DR,
Delatycki MB, Wilson RB, Isaya G and Puccio H: Ad vancements
in the pathophysiology of Friedreich's Ataxi a and new
prospects for treatments. Mol Genet Metab 2007, 92:2335.
1108.LuCandCortopassiG:Frataxin knockdown causes loss of
cytoplasmic iron-sulfur clu ster functions, redox alterations
and induction of heme transcripts. Arch Biochem Biophys 2007,
457:11122.
1109. Zanella I, Derosas M, Corrado M, Cocco E, Cavadini P, Biasiotto G,
Poli M, Verardi R and Arosio P: The effects of frataxin silencing
in HeLa cells a re rescued by t he expression of human
mitochondrial ferritin. Biochim Biophys Acta 2008, 1782:908.
1110. Anderson PR, Kirby K, Hilliker AJ and Phillips JP: RNAi-mediated
suppression of the mitochondrial iron chaperone, frataxin,
in Drosophila. Hum Mol Genet 2005, 14:3397405.
1111. Jo WJ, Loguinov A, Chang M, Wintz H, Nislow C, Arkin AP,
Giaever G and 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.
1112. Gibson TJ, Koonin EV, Musco G, Pa store A and Bork P:
Friedreich's ataxia protein: phylogenetic evidence for
mitochondrial dysfunction. Trends Neurosci 1996, 19:4658.
1113. Koutnikova H, Campuzano V, Foury F, Dolle P, Cazzalini O and
Koenig M: Studies of human, mouse and yeast homologues
indicate a mitochondrial function for frataxin. Nat Genet
1997, 16:345
51.
1114. Foury F: Human genetic diseases: A cross-talk between man
and yeast. Gene 1997, 195:110.
1115. Foury F and Cazzalini O: Deletion of the yeast homologue of
the human gene associated with Fr iedreich's ataxia elicits
iron accumulation in mitochondria. FEBS Lett 1997,
411:373377.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 49 of 79
(page number n ot f or citation purposes)
1116. Cavadini P, Gellera C, Patel PI and Isaya G: Human fra taxin
maintains mitochondrial iron homeostasis in Saccharo-
myces cerevisiae. Hum Mol Genet 2000, 9:252330.
1117. Becker E and Richardson DR: Frataxin: its role in iron
metabolism and the pathogenesis of Friedreich's ataxia.
Int J Biochem Cell Biol 2001, 33: 110.
1118. Foury F and Talibi D: Mitoch ondrial control of iron home-
ostasis. A genome wide analysis of gene expression in a
yeast frataxin-defi cient strain. JBiolChem20 01, 276:77628.
1119. De Freitas J, Wintz H, Kim JH, Poynton H, Fox T and Vulpe C:
Yeast, a model organism for iron and copper metabolism
studies. Biometals 2003, 16:185197.
1120. González-Cabo P , Váz quez-Manr ique RP, García-Gimeno MA,
Sanz P and Palau F: Frataxin interacts functionally with
mitochondrial electron trans port chain proteins. Hum Mol
Genet 2005, 14:20918.
1121. Irazusta V, Cabiscol E, Reve rter-Branchat G, R os J and Tamarit J:
Manganese is the link between frataxin and iron-sulfur
deficiency in the yeast model of Friedreich ataxia. JBiolChem
2006, 281:1222732.
1122. zquez-Manrique RP, González-Cabo P, Ros S, Aziz H, Baylis HA
and Palau F: Reduction of Caenorhabditis elegans frataxin
incr eases sens itivity to oxidative stress, reduces lifespan,
and causes lethality in a mitochondrial complex II mutant.
Faseb J 2006, 20:1724.
1123. Irazusta V, Moreno-Cer meno A, Cabiscol E, Ros J and Tamarit J:
Major ta rgets of iron-induce d protein oxidative damage in
frataxin-deficient yeasts are magnesium-bin ding proteins.
Free Radic Biol Med 2008.
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, D e Michele G, Filla A, De Frutos R, Palau F, Patel PI, Di
Donato S, Mandel JL, Cocozza S, Koe nig M and Pandolfo M:
Friedreich's ataxia: autosom al reces sive disease caused by
an intronic GAA triplet repeat expansion. Science 1996,
271:14237.
1125. Palau F: Friedreich's ataxia and frataxin: molecular genetics,
evolution and pathogenesis (Revie w). Int J Mol Med 2001,
7:5819.
1126. Patel PI and Isaya G: Fr iedreich ataxia: from GAA triplet-
repeat expansion to frataxin deficiency.
Am J Hum Genet 2001,
69:1524.
1127. Wilson RB: Frataxin and frataxin deficiency in Friedreich's
ataxia. JNeurolSci2003, 207:1035.
1128. Michael S, Petrocine SV, Qian J, Lamarche JB, K nutson MD,
Garrick MD and Koeppen AH: Iron and iron-responsive
proteins in the cardiomyopathy of Friedreich's ataxia.
Cerebellum 2006, 5:25767.
1129. tig A, Sidi D, Munnich A and Rustin P: Molecular insights into
Friedreich's ataxia and antioxidant-based therapies. Trends
Mol Med 2002, 8:2214.
1130. Boddaert N, Le Quan Sang KH, Rotig A, Leroy-Willig A, Gallet S,
Brunelle F, Sidi D, Thalabard JC, Munnich A and Cabantchik ZI:
Selective iron chelation in Friedreich ataxia: biologic and
clinical implications. Blood 2007, 110:4018.
1131. Sohn YS, Breuer W, Munnich A and Cabantchik ZI: Redistribu-
tion of accumulated cell iron: a modality of chelation with
therapeutic im plications. Blood 2008, 111:16909.
1132. Besarab A, Bolton WK, Browne JK, Egrie JC, Nissen son AR,
Okamoto DM, Schwab SJ and Goodkin DA: The effects of
normal as compared with low hematocrit values in patients
with cardiac disease who are receiving hemodialysis and
epoetin. NEnglJMed1998, 339:58490.
1133. Campuzano V, Montermini L, Lutz Y, Cova L, Hindelang C,
Jiralerspong S, Trottier Y, KishSJ,FaucheuxB,TrouillasP,
Authier FJ, Durr A, Mandel JL, Vescovi A, Pandolfo M and
Koenig M: Frataxin is reduc ed in F riedreich ataxia patients
and is a ssociated w ith mitocho ndrial membranes. Hum Mol
Genet 1997, 6:177180.
1134. Wong A, Yang J, Cavadini P, Gellera C, Lonnerdal B, Taroni F and
Cor topassi G: The Frie dreich's ataxia mutation con fers
cellular sensitivity to oxidant stress which is rescued by
chelators of iron and calcium and inhibitors of apoptosis.
Hum Mol Genet 1999, 8:42530.
1135. Park S, Gakh O, Mooney SM and Isaya G: The ferroxidase
activity of yeast frataxin. J Biol Chem 2002, 277:3858995.
1136. Sturm B, Bistrich U, Schranzhofer M, Sarsero JP, Rauen U, S cheiber-
Mojdehkar B, de Groot H, Ioannou P and Petrat F: Friedreich's
ataxia, no changes in mitochondrial labile iron in human
lymphoblasts and fibroblasts: a decrease in antioxidative
capacity? JBiolChem2005, 280:67 018.
1137. Anderson PR, Kirby K, Orr WC, Hilliker AJ and Phillips JP:
Hydrogen peroxide scavenging r escues frataxin deficiency
in a Drosophila model of Friedreich's ataxia. Proc Natl Acad Sci
USA 2008, 105:6116.
1138. Wilson RB and Roof DM: Respiratory deficiency due to loss of
mitochondrial DNA in yeast lacking the frataxin homo-
logue. Nat Genet 1997, 16:3527.
1139. Seznec H, Simon D, Bouton C, Reutenauer L, Hertzog A, Golik P,
Procaccio V, Patel M, Drapier JC, Koenig M and Puccio H:
Friedreich ataxia: the oxidative stress paradox. Hum Mol
Genet 2005, 14:46374.
1140. Visapää I, Fellman V, Vesa J, Dasvarma A, Hutton JL, Kumar V,
Payne GS, Makarow M, Van Coster R, Taylor RW, Turnbull DM,
SuomalainenAandPeltonenL:GRACILE syndrome, a lethal
metabolic disorder with iron overload, is caused by a point
mutation in BCS1L. Am J Hum Genet 2002, 71:86376.
1141. Fellman V, Lemmela S, Sajantila A, Pihko H and Jarvela I: Screening
of BCS1L mutations in severe neonatal disorders suspic ious
for mitochondrial cause. JHumGenet2008, 53:5548.
1142. McGeer PL and McGeer E G: Inflammatory processes in
amyotrophic lateral sclerosis. Muscle Nerve 2002, 26:459 70.
1143. Garber K: The elusive ALS genes. Science 2008, 319:20.
1144. Rosen DR, Siddique T, Patterson D, Figlewicz D A, Sapp P,
Hentati A, Donaldson D, Got o J, O'Regan JP and Deng HX, et al:
Mutations in Cu/Zn superoxide dismu tase gene are asso-
ciated with familial amyotrophic lateral sclerosis. Nature
1993, 362:5962.
1145. Kruman II, Pedersen WA, Springer JE and Mattson MP: ALS-linked
Cu/Zn-SOD mutation increases vulnerability of motor
neurons to excitotoxicity by a mechanism involving
incr eased oxidative stress and perturbed calcium home-
ostasis. Exp Neurol 1999, 160:2839.
1146. Bruijn LI, Miller TM and Cleveland D W: Unraveling the
mechanisms involved in motor neur on degeneration in
ALS. Annu Rev Neurosci 2004, 27:72349.
1147. Kasarskis EJ, Tandon L, Lovell MA and Ehmann WD: Aluminum,
calcium, and iron in the spinal cord of patients with
sporadic amyotrophic lateral sclerosis using laser microp-
robe mass s pectr oscopy: a preliminary study. JNeurolSci
1995, 130:2038.
1148. Carri MT, Ferri A, Cozzolino M, Calabrese L and Rotilio G:
Neurodegeneration in amyotrophic lateral sclerosis: the
role of oxidativ e stress and altered homeostasis of metals .
Brain Res Bull 2003, 61:365 74.
1149. Carri MT, Grignaschi G and Bendotti C: Ta rgets in ALS:
designing multidrug therapies. Trends Pharmacol Sci 2006,
27:26773 .
1150. Hughes KA and Reynolds RM: Evolutionary and mechan istic
theories of aging. Annual Review of Entomology 2005, 50:421445.
1151. Zacharski LR, Ornstein DL, Woloshin S and Schwartz LM:
Association of age, sex, and race with body iron stores in
adults: analysis of NHANES III data. Am Hea rt J 2000,
140:98104.
1152. Ritchie RF, Palomaki GE, Neveux LM, Navol otskaia O, Ledue TB
and 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, 16:24652.
1153. Ritchie RF, Palomaki GE, Neveux LM, Navol otskaia O, Ledue TB
and Craig WY: Reference distributions for serum iron and
transferrin saturation: a practical, simple, and clinically
relevant approa ch in a large cohort. J Clin Lab Anal 2002,
16:23745 .
1154. Liu JM, Hankinson SE, Stampfer MJ, Rifai N, Willett WC and M a J:
Body iron stores and their determinants in healthy
postmenopausal US women. Am J Clin Nutr 2003, 78:11607.
1155. Doulias PT, Vlachou C, Boudouri C, Kanavaros P, Sia mopoulos KC
and Galaris D: Flow cytometric estimation of 'labile iron
pool' in human white b lood cell s reve als a positive
association with ageing. Free Radic Res 2008,
42:2539.
1156. Fleming DJ, Tucker KL, Jacques PF, Dallal GE, Wilson PW and
Wood RJ: Dietary fa ctors associated with the risk of high
iron stores in the elderly Framingham Heart Study cohort.
Am J Clin Nutr 2002, 76:137584.
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, 76:118990.
1158. Cade JE, Moreton JA, O'Hara B, Greenwood DC, Moor J, Burley VJ,
Kukalizch K, Bishop D T and Wo rwood M: Diet and genetic
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 50 of 79
(page number n ot f or citation purposes)
factors associated with iron status in middle-aged women.
Am J Clin Nutr 2005, 82:81320.
1159. Guralnik JM, Eisenstaedt RS, Ferrucci L, Klein HG and
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, 104:22638.
1160. Price EA: Aging and erythropoiesis: current state of knowl-
edge . Blood Cells Mol Dis 2008, 41:15865.
1161. Sharman EH, Bondy SC, Sharman KG, Lahiri D, Cotman CW and
Perreau VM: Effects o f melatonin and age on gene expression
in mo use CNS using microarray analysis. Neurochemistry
International 2007, 50:336344.
1162. Kirkwood TBL: Evolution of Aging. Nature 1977, 270:301304.
1163. Beckman KB and Ames BN: The free radical theory of aging
matures. Physiol Rev 1998, 78:54781.
1164. Finkel T and Holbroo k NJ: Oxidant s, o xidat ive stress and the
biology of ageing. Nature 2000, 408:23947.
1165. Sohal RS, Mockett RJ and Orr WC: Current issues concerning
the role of oxidative stress in aging: a perspective. Results
Probl Cell Differ 2000, 29:4566.
1166. Cutler RG a nd Rodriguez H: Critical reviews of oxidative
stress and aging: advances in basic science, diagnostics and
intervention. World Scientific, Singapore 2002.
1167. Herndon LA, Schmeissner PJ, Dudaronek JM, Br own PA,
Listner KM, Sakano Y, Paupard MC, Hall DH and Driscoll M:
Stochastic and gene tic fa ctors influenc e tissue-speci fic
decline in ageing C. elegans. Nature 2002, 419:80814.
1168. Sohal RS, Mockett RJ and Orr WC: Mechanisms of aging: an
appraisal of the oxidative stress hypothesis. Free Radic Biol
Med 2002, 33:57586.
1169. Partridge L and Gems D: Mechanisms of ageing: public or
priv ate? Nat Rev Genet 2002, 3:16575.
1170. Hekimi S and Guarente L: Genetics an d the specif icity of the
aging process. Science 2003, 299:13514.
1171. Krabbe KS, Pedersen M and Bruunsgaard H: Inflammatory
mediators in the elderly. Exp Gerontol 2004, 39:68799.
1172. Longo VD and Finch CE: Evolutionary medici ne: from dwarf
model systems to healthy centenarians? Science 2003,
299:13426.
1173. McGeer PL and McGeer EG: Inflammation and the d egen-
erative diseases of aging. AnnNYAcadSci2004, 1035:10416.
1174. Afanas'ev IB: Free radical mechanisms of aging processes
under physiological conditions. Biogerontology 2005, 6:28390.
1175. Kirkwood TB: Understanding the odd science of aging. Cell
2005, 120:43747.
1176. Tuppo EE and A rias HR: Theroleofinflammationin
Alzheimer 's disease. Int J Biochem Cell Biol 2005, 37:289305.
1177. Partridge L and Gems D: Beyond the evolutionary theory of
ageing, from functional genomics to evo-gero. Trends Ecol
Evol 2006, 21:33440.
1178. Salvioli S, Capri M, Valensin S, Tieri P, Monti D, Ottaviani E and
Franceschi C: Inflamm-aging, cyt okines and aging: state of
the art, new hypotheses on the role of mitochondria and
new perspectives from systems biology. Curr Pharm Des 2006,
12:316171.
1179. Franco OH, Kirkwood TB , Powell JR, Catt M, Goodwin J,
Ordovas JM and Oud eraa van der F: Ten commandmen ts for
the future of age ing research in the UK: a vision for action.
BMC Geriatr 2007, 7:10.
1180. Kirkwood TB: A systemati c look at an old problem. Nature
2008, 451:6447.
1181. Harman D: Aging: a theory based on free radical and
radiation chemistr y. J Gerontol 1956, 11:298300.
1182. Ames BN, Shigenaga MK and Hagen TM: Oxidants, antioxidants,
and the degenerative diseases of aging. Proc Natl Acad Sci USA
1993, 90:791522.
1183. Martin G M, Austad SN and Johnson TE: Genetic analysis of
ageing: role of oxidative damage and environmental
stresses. Nat Genet 1996, 13:2534.
1184. Ishii N, Fujii M, Hartman PS, Tsuda M, Yasuda K, Se noo-Matsuda N,
Yanase S, Ayusawa D and Suzuki K: A mutation in succinate
dehydrogenase cytochrome b causes oxidative stress and
ageing in nematodes. Nature 1998, 394:69 47.
1185. Fukagawa NK: Aging: is oxidative stress a marker or is it
causal? Proc Soc Exp Biol Med 1999, 222:293 8.
1186. Johnson FB, Sinclair DA and Guarente L: Mo lecular biology of
aging. Cell 1999, 96:291302.
1187. Melov S, Ravenscroft J, Malik S, Gill MS, Walker DW, Clayton PE,
Wallace DC, Malfroy B, Doctrow SR and Lithgow GJ: Extension of
life-span with superoxide dismutase/catalase mimetics.
Science 2000, 289:15679.
1188. Hamilton ML, Van Remmen H, Drake JA, Yang H, Guo ZM,
Kewitt K, Walter CA and Richardson A: Does oxidative dam age
to DNA increase with age? Proc Natl Acad Sci USA 2001,
98:1046974.
1189. von Zglinicki T, rkle A and Kirkwood TBL: Stress, DNA
damage and a geing an integrative approach. Exp Gerontol
2001, 36:10491062.
1190. Barja G: Rate of generation of oxidative stress-related
damage and animal longevity. Free Radical Biology and Medicine
2002, 33:1167
1172.
1191. Bonilla E, Medina-Leendertz S and Diaz S: Extension of life span
and stress resistance of Drosophila melanogaster by long-
term supplementation with melatonin. Exp Gerontol 2002,
37:62938 .
1192. Golden TR, Hinerfeld DA and Melov S: Oxidative stress and
aging: beyond correlation. Aging Cell 2002, 1:11723.
1193. Hagen TM: Oxidative stress, redo x imbalance, and the aging
process. Antioxidants & Redox Signal ing 2003, 5:503506.
1194. Karanjawala ZE, Murphy N, Hinton DR, Hsieh CL and 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,
12:39740 2.
1195. Melov S: Animal models of oxidative stress, aging, and
therapeutic antioxidant interven tions. Int J Biochem Cell Biol
2002, 34:1395400.
1196. Barros MH, Ban dy B, Tahara EB and Kowaltowski AJ: Higher
respiratory activity decreases mitochondrial reactive oxy-
gen release and inc reases life span in Saccharomyces
cerevisiae. Journal of Biological Chemistry 2004, 279:4988349888.
1197. Gredilla R and Barja G: Minireview: the role of oxidative stress
in relation to caloric restriction and longevity. Endocrinology
2005, 146:3713 7.
1198. Blagosklonny MV: Aging and immortality: quasi-programmed
senescence and its pharmacologic inhibition. Cell Cycle 2006,
5:2087102.
1199. Chung HY, Sung B, Jung KJ, Zou Y and Yu BP: The molecular
inflammatory process in aging. Antioxid Redox Signal 2006,
8:57281.
1200. Sedensky MM and Morgan PG: Mitochondrial res piration and
reactive oxygen species in C. elegans. Exp Gerontol 2006,
41:95767 .
1201. Terman A and Brunk UT: Oxidative stress, acc umulation of
biological 'garbage', and aging. Antioxidants & Redox Signaling
2006, 8:197204.
1202. Yu BP and Chung H Y: Adaptive mechanisms to oxidative
stress during aging. Mechanisms o f Ageing and Development 2006,
127:436443.
1203. Chakravarti B and Chakravarti DN: Oxidative modification of
proteins: Age-related changes. Gerontology 2007, 53:128139.
1204. Kim SK: Common aging pathways in worms, flies, mice and
humans. JExpBiol2007, 210:160712.
1205. Kregel KC and Zhang HJ: An integrated view of oxidative
stress in aging: basic mechanisms, functional effects, and
pathological considerations. Am J Physiol Regul I ntegr Comp
Physiol 2007, 292:R1836.
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 and Cornall RJ: DNA re pair
is limiting for hae matopoietic stem cells during ageing.
Nature 2007, 447:68690.
1207. Sohal RS and Weindruch R: Oxidative stress, caloric restric-
tion, and aging. Science 1996, 273:5963.
1208. Pardon MC: Stress and ageing interactions: a paradox in the
context of shared etiological and physiopathological pro-
cesses. Brain Res Rev 2007, 54:25173.
1209. Killilea DW, Atamna H, Liao C and Ames BN: Iron accumulation
during cellular senescence in human fibroblasts in vitro.
Antioxid Redox Signal 2003, 5:50716.
1210. Hamerman D: Toward an understanding of frailty. Ann Int Med
1999, 130:945950.
1211. Docherty JR: Cardiovascular responses in aging a review.
Pharmacol Rev 1990, 42:103
125.
1212. Dröge W and Schipper HM: Oxidative stress and ab errant
signaling in aging and cognitive decline. Aging Cell 2007,
6:361370.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 51 of 79
(page number n ot f or citation purposes)
1213. Massie H R, Aiello VR and Banziger V: Iron accumulation and
lipid peroxidation in aging C57bl/6j mice. Exp Gerontol 1983,
18:27728 5.
1214. Ershler WB: Biological interactions of aging and anemia: A
focus on cytokines. J Amer Geriatr Soc 2003, 51:S18 S21.
1215. Massie HR, Aiello VR and Williams TR: Inhibition of iron
absorption prolongs the life span of Drosophila. Mech Agei ng
Dev 1993, 67:227237.
1216. Rattan SIS: Aging, anti-aging, and hormesis. Mechanisms of
Ageing and Development 2004, 125: 285289.
1217. Kenyon C: The plasticity of aging: insights from long-lived
mutants. Cell 2005, 120:44960.
1218. Lee CK, Klopp RG, Weindruch R and Prolla TA: Gene expression
profile of aging and its retardation by caloric restriction.
Science 1999, 285:13903.
1219. Bordone L and Guarente L: Calorie restriction, SIRT1 and
metabolism: understanding longevity. Nat Rev Mol Cell Biol
2005, 6:298305.
1220. Taguchi A, Wartschow LM and White MF: Brain IRS2 signaling
coordinates life span and nutrient homeostasis. Science 2007,
317:36972.
1221. Hulbert AJ, Pamplona R, Buffenstein R and Buttemer WA: Life and
death: metabol ic rate, membrane composition, and li fe
span of animals. Physiol Rev 2007, 87:1175213.
1222. Kennedy BK, Steffen KK and Kaeberlein M: Ruminations on
dietary restriction and aging. Cell Mol Life Sci 2007, 64:13238.
1223. Mair W and Dillin A: Aging and survival: the genetics of life
span extension by dietary restriction. Annu Rev Biochem 2008,
77:727
54.
1224. Piper MD and Partridge L: Dietary r estriction in Drosophila:
delayed aging or experimental artefact? PLoS Genet 2007, 3:
e57.
1225. Weindruch R and Sohal RS: Caloric intake and aging. New
England Journal of Medicine 1997, 337:986994.
1226. Lass A, Sohal BH, Weindruch R, Fo rster MJ and Sohal RS: Caloric
restriction prevents age-associated acc rual of oxidative
damage to mouse skeletal muscle mitochondria. Free Radical
Biology and Medicine 19 98, 25:10891097.
1227. Masoro EJ: Caloric restriction and aging: an update. Exp
Gero ntol 2000, 35:299305.
1228. Thrasivoulou C, Soubeyre V, Ridha H, Giuliani D, Giaroni C,
Michael GJ, Saffrey MJ and Cowen T: Reactive oxygen species,
dietary restriction and neurotrophic factors in age-related
loss of myenteric neurons. Aging Cell 2006, 5:24757.
1229. Walz D: Thermo dynamics of oxidat ion-reduction reactions
and its application to bioenergetics. Biochim Biophys Acta 1979,
505:279353.
1230. Cook CI and Yu BP: Iron accumulation in aging: modulation
by dietary restriction. Mechanisms of Ageing and Development
1998, 102:113.
1231. Reverter-Branchat G, Cabiscol E, Tamarit J and Ros J: Oxidative
damage to specific proteins in replica tive and chrono logi-
cal-aged Saccharomyces cerevisiae Common targets and
prevention by calorie restriction. JBiolChem2004,
279:3198331989.
1232. Raffaghello L, Lee C, Safdie FM, Wei M, Madia F, Bianchi G and
Longo VD: Starvation-dependent differential stress resis-
tance protects normal but not cancer cells against high-
dose chemotherapy. Proc Natl Acad Sci USA 2008, 105:821520.
1233. Anisimov VN: E ffects of ex ogenous melatonina review.
Toxicol Pathol 2003, 31:
589603.
1234. Poeggeler B: Me latonin, aging, and age-related diseases:
perspectives for prevention, inter vention, and therapy.
Endocrine 2005, 27:20112.
1235. Bonilla E, Medina-L eendertz S, Villa lobos V, Molero L and
Bohorquez A: P araquat-induced oxidative stress in Droso-
phila melanogast er: Effects of melatonin, glutathione,
serotonin, minocycline, lipoic acid and ascorbic acid.
Neurochemical Research 2006, 31:14251432.
1236. Salvemini D, Riley DP and Cuzzocre a S: SOD mimetics a re
coming of age . Nature Reviews Drug Discovery 2002, 1:367374.
1237. Lithgow GJ, Gill MS, Olsen A and Sampayo JN: Pharmacological
intervention in invertebrate aging. Ageing Res Rev 2005,
27:21322 3.
1238. Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS,
Ahringer J, Li H and Kenyon C: Genes that act downst ream of
DAF-16 to influence the lifespan of Caenorhabditis elegans.
Nature 2003, 424:27783.
1239. Dong MQ, Venable JD, Au N, Xu T, Park SK, Coc iorva D,
Johnson JR, Dillin A and Yates JR 3rd: Quantitative mass
spectrometry identifies insulin signaling targets in C.
eleg ans. Science 2007, 317:6603.
1240. Honda Y and Honda S: The daf-2 gene network for longevity
regulates oxidative stress resistance and Mn-superoxide
dismutasegeneexpressioninCaenorhabditis elegans. FASEB J
1999, 13:138593.
1241. Bauerova K and Bezek S: Role of reactive oxygen and nitrogen
species in etiopathogenesis of rheumatoid arthritis. General
Physiology and Biophysics 1999, 18:1520.
1242. Aruoma OI: Free radicals, oxidative stress, and antioxidants
in human health and disease. J Amer Oil Ch emists Soc 1998,
75:199
212.
1243. Mapp PI, Grootveld MC and Blake DR: Hypoxia, oxidative stress
and rheumatoid arthritis. British Medical Bulletin 1995,
51:41943 6.
1244. Gracy RW, Talent JM, Kong Y and Conrad CC: Rea ctive oxygen
species: the un avoid able environmental insult? Mut Res 1999,
428:1722.
1245. Henrotin YE, Bruckner P and Pujol JPL: The role of reactive
oxygen species in homeostasis and degradation of carti-
lage. Osteoarthritis and Cartilage 2003 , 11:747755.
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 :273280.
1247. Weinstein IM: A Correlative Study of the Erythrokinetics
and Disturbances in Iron Metabolism Associated with the
Anemia of Rheumatoid Arthritis. Blood 1959, 14:950966.
1248. Bentley DP an d Williams P: Serum ferritin concentration as an
index of storage iron in rheumat oid arthritis. JClinPathol
1974, 27:786788.
1249. Hansen TM, Hanse n NE, Birgens HS, Holund B and Lorenzen I:
Serum Ferritin and the Assessment of Iron-Deficiency in
Rheumatoid-Arthritis. Scandinavian Journal of Rheumatology
1983, 12:353359.
1250. Weber J, Werre JM, Julius HW and Marx JJ: Decreased iron
absorption in patients with active rheumatoid arthritis,
with and without iron deficiency. Ann Rheum Dis 1988,
47:4049.
1251. Vreugdenhil G, Wognum AW, Vaneijk HG and Swaak AJG:
Anemia in Rheumatoid-Arthritis theRoleofIron,
Vitamin-B12, and Folic-Acid Deficiency, and Erythropoie-
tin Responsiveness. Annals of the Rheu matic Diseases 1990,
49:9398.
1252. Muirden KD and Senator GB: Ir on in synovial membrane in
rheumatoid arthritis and other joint diseases. Ann Rheumat
Dis 1968, 27:38.
1253. Rowley D, Gutteridge JMC, Blake D, Farr M and Halliwell B: Lipid
peroxidat ion in rheumatoid arthritis
thiobarbituric acid-
reactive material and catalytic iron salts in synovial fluid
from rheumatoid patients. Clin Sci 1984, 66:691695.
1254. Biemond P, Swaak AJG, Vaneijk HG and K oster JF: Intraarticular
ferritin-bound iron in rheumatoid arthritis afactorthat
incr eases o xygen free radical- induced tissue des truction.
Arthritis and Rh eumatism 1986, 29:11871193.
1255. Lipiński P and Drapier JC: Interplay between ferritin metabo-
lism, reactive oxygen species and nitric oxide. JBiolInorg
Chem 1997, 2:559566.
1256. Biemond P, Swaak AJG, Vaneijk HG and Koster JF: Superoxide
Dependent Iron Release from Ferritin in Inflammatory
Diseases. Free Radical Biology and Medicine 1988, 4:185198.
1257. Biemond P, S waak AJG, Beindorff CM and Koster JF: Superoxide-
Dependent and Superoxide-Indep endent Mechanisms of
Iron Mobilization from Ferritin by Xanthine-Oxidase
Implications for Oxygen-Free-Radical-Induced Tissue
Destruction During Ischemi a and Inflammation. Biochemical
Journal 19 86, 239:169173.
1258. Biemond P, Swaak AJG, Penders JMA, Beindorff CM and Koster JF :
Superoxide Production by Polymorphonuclear Leukocyt es
in Rheumatoid-Arthritis and Osteoarthritis Invivo Inhibi-
tion by the Antirheumatic Drug Piroxicam Due to Inter-
ference with the Act ivation of the Nadph-Oxidase. Annals of
the Rheumatic Diseases 1986, 45:249255.
1259. Biemond P, Vaneijk HG, Swaak AJG and Koster JF: Iron
Mobilization from Ferritin by Superoxide Derived from
Stimulated Polymorphonuclear Leukocytes Possible
Mechanism in Inflammation Diseases. Journal of Clinical
Investigation 1984, 73:15761579.
1260. Mladĕnka P, Šimůnek T, bl M and Hrdina R: The r ole of
reactive oxyg en and nitrogen species in cellular iron
metabolism. Free Radic Res 2006, 40:263
72.
BMC Medical Genomics 2009, 2:2 http://www.biomedcentral.com/1755-8794/2/2
Page 52 of 79
(page number n ot f or citation purposes)
1261. Harris LR, Cake MH and Macey DJ: Iron release f rom ferritin
and its sensit ivity to superoxide ions differs among
vertebrates. Biochem J 1994, 301:38 5389.
1262. Fairburn K, Grootveld M, Ward RJ, Abiuka C, Kus M, Williams RB,
Winyard PG and Blake DR: Alpha-Tocopherol, Lipids and
Lipoproteins in Knee-Joint Synovial-Fluid and Serum from
Patients with Inflam matory Joint Disease. Clinic al Science
1992, 83:657664.
1263. Dombrecht EJ, Cos P, Berghe Vanden D, Van Offel JF,
Schuerwegh AJ, Bridts CH, Stevens WJ and De Clerck LS:
Selective in vitro antioxidant properties of bisphospho-
nates. Biochem Biophys Res Commun 2004, 314:675 80.
1264. Ostrakhovitch EA and Afanas'ev IB: Oxidative stress in
rheumatoid arthritis leukocytes: suppression by rutin and
other antioxidants and chelators. Biochem Phar macol 2001,
62:74374 6.
1265. Hazes JMW, Dijk mans BAC, Vandenbroucke JP, Devries RRP and
Cats A: Pregnancy and the risk of developing rheumatoid
arthritis. Arthritis and Rhe umatism 1990, 33:17701775.
1266. ØstensenMandHusbyG:A prospec tive clinical study of the
effect of pregnancy on r heumatoid a rthritis and ank ylosing
spondylitis. Arthritis and Rheumatism 1983, 26:11551159.
1267. Da Silva JAP and Spector TD: The role of pregnancy in the
course and etiology of rheumatoid arthritis. Clin Rheumat
1992, 11:189194.
1268. O'Keeffe ST: Restless legs syndrome Areview.Archives of
Internal Medicine 1996, 156:243248.
1269. Ondo W, Tan EK and Mansoor J: Rheumatologic serologies in
secondary restless legs syndrome. Movement Dis orders 2000,
15:32132 3.
1270. Trenkwalder C, Paulus W and Walters AS: Th e restless legs
syndrome. Lancet Neurology 2005, 4:465475.
1271. Feelders RA, Kuiper-Kramer EPA and van Eijk HG: Structure,
function and clinical significance of transferrin receptors.
Clinical Chemistry and Laboratory Medici ne 1999, 37:110.
1272. Ahluwalia N: Diagnostic utility of serum transferrin recep-
tors measurement in assessing iron status. Nutrition Reviews
1998, 56:133141.
1273. Frostegård J: SLE, atherosclerosis and cardiov ascul ar dis-
ease. JInternMed2005, 257:48595.
1274. Mascitelli L and Pezzetta F: High iron stores and cardiovascular
disease in systemic lupus erythematos us. JInternMed2005,
258:584.
1275. Harvey AM, Shulman LE, Tumulty PA, Conley CL and
Schoenrich EH: Systemic lupus erythematosus: review of
the literature and clinical analysis of 138 cases. Medicine
(Baltimore) 1954, 33:291 437.
1276. Johnson RT and Richardson EP: The neurological manifesta-
tions of systemic lupus erythematosus. Medicine (Baltimor e)
1968, 47:33769.
1277. Feng PH, Cheah PS and Lee YK: Mortality in systemic lupus
erythematosus: a 10-year review. Br Med J 1973, 4: 7724.
1278. Matthay RA, Schwarz MI, Petty TL, Stanford RE, G upta RC, Sahn SA
and Steigerwald JC: Pulmonary manifestations of systemic
lupus ery thematosus: review of twelve cases of acute lupus
pneumonitis. Medicine (Baltimore) 1975, 54:397409.
1279. O'Loug hlin S, Schroeter AL and Jordon RE: Chronic ur ticaria-
like lesions in systemic lupus erythematosus. A review of 12
cases. Ar ch Dermato l 1978, 114:879 83.
1280. Hoffman BI and K atz WA: The gastrointes tinal manifestations
of systemic lupus erythematosus: a review of the literature.
Semin Arthritis Rheum 1980, 9:23747.
1281. Reynolds JC, Inman RD , Kimberly RP, Chuong JH, Kovacs JE and
Walsh MB: Acute pancreat itis in syst emic lupus erythema-
tosus: report of twenty cases and a review of the literature.
Medicine (Baltimore) 1982, 61:2532.
1282. Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF,
Schaller JG, Talal N and Winchester RJ : The 1982 revised
criteria for the classification of systemic lupu s erythema-
tosus. Arthritis Rheum 1982, 25:12717.
1283. Levine SR, Joseph R, D'Andrea G and Welch KM: Migraine and
the lupus anticoagulant. Case reports and review of the
literature. Cephalalgia 1987, 7:939.
1284. Crowson AN and Magro C: The cutaneous pathology of lup us
erythematosus: a review. J Cutan Pathol 2001, 28:123.
1285. Karrar A, Se queira W and Block JA: Coronary artery disease in
systemic lupus erythematosus: A review of the literature.
Semin Arthritis Rheum 2001, 30:43643.
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,
57:134857.
1287. Zhang X, Jin M, Wu H, Nadasdy T, Nadasdy G, Harris N, Green-
Church K, Nagaraja H, Birmingham DJ, Yu CY, Hebert LA and
Rovi n BH: Biomarkers of lup us nephritis determined by
serial urine proteomics. Kidney Int 2008, 74:799807.
1288. Lim MK, Lee CK, J u YS, Cho YS, Lee MS, Yoo B and Moon HB:
Serum ferritin as a serologic marker of activity in systemic
lupus erythematosus. Rheumatol Int 2001, 20:8993.
1289. Abou-Raya A, el-Hallous D and Fayed H: 8-Isoprostaglandin F2
alpha: a potential i ndex of lipid peroxidation in systemic
lupus erythematosus. Clin In vest Med 2004, 27:30611.
1290. Cutolo M and Otsa K: Review: vitamin D, immunity and
lupus. Lupus 2008, 17:610.
1291. Li HF: Are statins analogues of vitamin D? Lanc et 2006,
368:12334.
1292. Wu-Wong JR, Nakane M, Ma J and Ju TR: Statins do not directly
activate vitamin D receptor. JThrombHaemost2007,
5:4156.
1293. Lawman S, Mauri C, Jury EC, Cook HT and Ehrenstein MR:
Atorvastatin inhibits autoreactive B ce ll activation and
delays lupus development in New Zealand black/white F1
mice. J Immun ol 2004, 173:76416.
1294. Jury EC, Isenberg DA, Mauri C and Ehrenstein MR: Atorvastatin
restores Lck expression and lipid raft-associated signa ling
in T cells from patients with systemic lupus erythematosus.
J Immunol 2006, 177:741622.
1295. Swanson DR: Undiscovered public knowledge. Library Quarterly
1986, 56:103118.
1296. Swanson DR, Smalheiser NR and Torvik VI: Ranking indirect
connections in literature-based discovery: The role of
medical subject he adings. J A mer Soc Inf Sci Technol 2006,
57:14271439.
1297. Andreadis AA, Hazen SL, Comhair SAA and Erzurum SC:
Oxidative and nitrosative events in asthma. Free Radical
Biology and Medicine 20 03, 35:213 225.
1298. Gree ne LS: Asthma and oxidant stress nutritional,
environmental, and genetic risk factors. JAmerCollNutrition
1995, 14:317324.
1299. Han JY, Takeshita K and Utsumi H: Noninvasive detection of
hydroxyl radical generation in lung by diesel exhaust
particles