ArticlePDF AvailableLiterature Review

Abstract and Figures

Xanthine oxidoreductase (XOR) is the enzyme that catalyzes the oxidation of hypoxanthine to xanthine and xanthine to uric acid and is widely distributed among species. In addition to this housekeeping function, mammalian XOR is a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various pathways. This review intends to address the physiological and pathological roles of XOR-derived oxidant molecules. The cytocidal action of XOR products has been claimed in relation to tissue damage, in particular damage induced by hypoxia and ischemia. Attempts to exploit this activity to eliminate unwanted cells via the construction of conjugates have also been reported. Moreover, different aspects of XOR activity related to phlogosis, endothelial activation, leukocyte activation, and vascular tone regulation, have been taken into consideration. Finally, the positive and negative outcomes concerning cancer pathology have been analyzed because XOR products may induce mutagenesis, cell proliferation, and tumor progression, but they are also associated with apoptosis and cell differentiation. In conclusion, XOR activity generates free radicals and other oxidant reactive species that may result in either harmful or beneficial outcomes.
This content is subject to copyright. Terms and conditions apply.
Review Article
Xanthine Oxidoreductase-Derived Reactive Species:
Physiological and Pathological Effects
Maria Giulia Battelli, Letizia Polito, Massimo Bortolotti, and Andrea Bolognesi
Alma Mater Studiorum-University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine (DIMES),
General Pathology Unit, Via S. Giacomo 14, 40126 Bologna, Italy
Correspondence should be addressed to Letizia Polito; letizia.polito@unibo.it
Received  September ; Accepted  November 
Academic Editor: Tanea T. Reed
Copyright ©  Maria Giulia Battelli et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
Xanthine oxidoreductase (XOR) is the enzyme that catalyzes the oxidation of hypoxanthine to xanthine and xanthine to uric acid
and is widely distributed among species. In addition to this housekeeping function, mammalian XOR is a physiological source of
superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of variouspathways.
is review intends to address the physiological and pathological roles of XOR-derived oxidant molecules. e cytocidal action of
XOR products has been claimed in relation to tissue damage, in particular damage induced by hypoxia and ischemia. Attempts
to exploit this activity to eliminate unwanted cells via the construction of conjugates have also been reported. Moreover, dierent
aspects of XOR activity related to phlogosis, endothelial activation, leukocyte activation, and vascular tone regulation, have been
taken into consideration. Finally, the positive and negative outcomes concerning cancer pathology have been analyzed because
XOR products may induce mutagenesis, cell proliferation, and tumor progression, but they are also associated with apoptosis and
cell dierentiation. In conclusion, XOR activity generates free radicals and other oxidant reactive species that may result in either
harmful or benecial outcomes.
1. Introduction
e enzyme xanthine oxidoreductase (XOR) has a wide
distribution throughout living organisms and is highly con-
served in prokaryotic, plant, and animal species (reviewed in
[]). XOR is a dimeric metalloavoprotein comprising two
identical subunits of approximately  kDa each, including
one molybdenum-containing molybdopterin cofactor (Mo-
co) and one avin adenine dinucleotide (FAD) cofactor, as
well as two nonidentical iron-sulfur redox centers. e purine
oxidation occurs at the Mo-co site, while the FAD site is the
oxidized nicotinamide adenine dinucleotide (NAD+)andO
2
reduction sites. e electron ux moves between the Mo-
co and FAD cofactors through the two iron-sulfur clusters
(reviewed in []).
XOR catalyzes the oxidation of hypoxanthine to xanthine
andxanthinetouricacid,whicharethelasttwostepsof
purine catabolism in the highest primates. XOR has the
rate-limiting function of generating irreversible products,
thus precluding the salvage pathway of purine nucleotides.
Additionally, dierent endogenous metabolites and various
xenobiotics can be oxidized by XOR. Uric acid and its
oxidized derivatives may exert prooxidant activity, mainly
within the cell; however, it has in vivo antioxidant activity,
mainly in body uids. is scavenger action is supposed to
provide an evolutionary advantage to primates that lost their
uricaseactivityviamutationandacquiredacrucialdefense
against oncogenesis by free radicals [].
XOR is highly regulated at both the transcriptional and
posttranslational levels. XOR activity is present in all mam-
maliantissueanduids,although,inmostofthem,itis
expressed at very low levels because the human XOR gene
is usually subjected to a repressing regulation at the tran-
scriptional level []. e highest XOR levels are expressed
in liver, intestine, kidney, and lactating mammary gland
epithelial cells and in vascular endothelial cells (reviewed in
[]). XOR expression may be increased by various stimuli,
such as hormones, growth factors, inammatory cytokines,
and low oxygen tension. At the posttranslational level, XOR
Hindawi Publishing Corporation
Oxidative Medicine and Cellular Longevity
Volume 2016, Article ID 3527579, 8 pages
http://dx.doi.org/10.1155/2016/3527579
Oxidative Medicine and Cellular Longevity
is modulated with both quantitative and qualitative changes
initsactivity.XORproteinmaybeproducedindemolybdo-
and/or desulfo-forms, which are inactive in xanthine catalysis
at the Mo-co site, although they can oxidize the reduced
nicotinamide adenine dinucleotide (NADH) at FAD site.
ese defective XOR forms are present in varying percentages
in milk and could be reactivated with the reinsertion of the
lackingatomsattheactivesite.XORactivitywasobservedto
increase in response to hypoxia without changes in the levels
of mRNA or enzyme protein, indicating a posttranslational
regulation of XOR (reviewed in []). However, the most
peculiar modulation of XOR activity in mammals consists of
the conversion from the dehydrogenase to the oxidase form.
is transition occurs in various pathological conditions
(reviewed in []).
In all organisms, XOR is present in its constitutively active
dehydrogenase form, whereas, only in mammals, the NAD+-
dependent xanthine dehydrogenase (XDH, EC ...) can
be converted to the oxidase form (XO, EC ...) through
sulydryl group oxidation or limited proteolysis []. XO
delivers electrons directly to molecular oxygen (O2), thus
generating the reactive oxygen species (ROS), superoxide
anion (O2), and hydrogen peroxide (H2O2),viaaone-
electron and a two-electron reduction, respectively. is gives
rise to the hydroxyl radical (HO) in the presence of iron
via the Haber-Weiss and Fenton reactions. e percentage
of divalent versus univalent electron transfer to O2and the
relative quantities of O2and H2O2generated by XO are
dependent upon O2tension, pH, and purine concentration.
us, under normal physiological conditions, H2O2is the
major reactive product derived from the XO-catalyzed O2
reduction. H2O2formation is further favored when both
the O2levels and pH are reduced, such as under ischemic
and/or hypoxic conditions (reviewed in []). Under hypoxic
conditions, these ROS can also be produced by XDH, which,
at the FAD site, can oxidize NADH. Hypoxia-mediated acidic
pH and low O2tension lessen the nitric oxide (NO) formation
by NO synthase and increase its potential to uncouple and
produce O2. ese conditions reduce XOR anity for
xanthine while increasing anity for nitrites, which compete
with xanthine at the Mo-co site and can be reduced to NO.
Under the same conditions the amount of O2formation
by XOR is sucient to react with NO and generate reactive
nitrogen species (RNS), particularly peroxynitrite (ONOO).
Both free radicals, such as O2,HO
, and NO, and nonrad-
ical forms, such as H2O2and ONOO,haveanoxidizing
eect, thereby contributing to oxidative stress (reviewed in
[]).
e generation of these oxidants may be only partially
blocked by allopurinol, which inhibits the Mo-co site in a
competitive manner but does not inhibit the catalytic activity
at the FAD site. All together, these products are responsible
for XOR cytotoxic and proinammatory activities and for
pro- and antitumorigenic eects, in both physiological and
pathological conditions. e various XOR functions are
dependent on (i) the level of ROS production, as in the case
of cytotoxic eects; (ii) the type of the prevalent product,
for instance, NO in the presence of high nitrate level; (iii)
the specicity of dierent cell types, such as phagocytes
in inammation; (iv) the level of XOR gene expression, in
particular in cancer.
2. Cytotoxicity of Xanthine
Oxidoreductase Products
XOR cytotoxicity received much attention during the second
half of last century, together with the circumstances of the
conversion from XDH to XO. An elevated XO/XDH activity
ratio has been reported in dierent pathological conditions,
which were characterized by tissue damage and cell necrosis.
Inparticular,theXDHtoXOshiwasobservedinavariety
of hypoxic/ischemic conditions (reviewed in []), including
organ transplantation (reviewed in []). In such circum-
stances, any reoxygenation/reperfusion could increase the
supply of oxygen for the formation of oxidants, but it was not
strictly required. Additionally, the conversion from XDH to
XO was not necessary for ROS generation, as discussed above,
especially in the presence of low oxygen tension that favors
the NADH oxidase activity of XOR. However, the formation
of XOR-derived ROS was indicated as the causal agent of the
injury or, at least, of the damage amplication, although more
thanonesourceofROScouldbeimplicated(reviewedin
[]).
e mechanism of ROS cytotoxicity is attributed to
peroxidation of membrane lipids, DNA damage, and protein
oxidation, which impair mitochondrial function and lead to
apoptosis (reviewed in []) (Figure (a)). Indeed, DNA dam-
age and the consequent loss of cloning eciency occurred
in a Burkitt lymphoma-derived cell line via XOR activity
through the production of ROS []. Apoptosis and necrosis
were induced to proliferating human lymphocytes by XOR-
derived oxidative stress, which was prevented by catalase [].
Additionally, oxidative DNA damage, consequent to the ROS
generated by XOR activity, provoked cell death in a nasopha-
ryngeal carcinoma cell line []. Accordingly, XOR-derived
ROS caused DNA double-strand breaks that were associated
with p function/expression and caspase-dependent apop-
tosis in primary human lung microvascular endothelial cells
that were exposed to cigarette smoke extract [].
e oxidative stress could be utilized to eliminate un-
wanted cells, particularly cancer cells. An attempt to take
advantage of the cytotoxicity of XOR products was performed
by conjugating the XOR protein to monoclonal antibodies,
with the intent of delivering XOR activity to the antigen-
bearing cell. XOR-containing conjugates recognizing B lym-
phocyte antigens were prepared with the purpose of autol-
ogous bone marrow graing. ese conjugates selectively
killed B lymphoma cell lines [] without reducing normal
myeloid clonogenic eciency [] and were eective in
bone marrow purging from malignant B lymphocytes [].
XOR immunotargeting was also studied in an experimen-
tal model to eliminate T lymphocytes from bone marrow
for heterologous transplantation []. e cytotoxicity and
selectivity of conjugated XOR were enhanced by the addition
of chelated iron that potentiates the free radical formation
(reviewed in []). e ecacy of XOR activity was proven
in conditions that were very similar to the ex vivo treatment
for bone marrow purging from multiple myeloma cells, with
Oxidative Medicine and Cellular Longevity
Apoptosis
ROS
XOR
Membrane
Protein
DNA
(Necrosis)
(a)
Antibody
PEG
Hematological tumors Solid tumors
Specic delivery Enhanced delivery
(b)
F : Pharmaceutical applications of xanthine oxidoreductase (XOR) cytotoxicity. (a) Mechanisms of ROS cytotoxicity: ROS induce
peroxidation of membrane lipids, DNA damage, and protein oxidation and lead to cell death, mainly via apoptosis through impaired
mitochondrial function (reviewed in []). (b) XORwas conjugated to carriers for the experimental elimination of specic target cells. Selective
cell killing was obtained by conjugating XOR to an antibody that was able to specically deliver reactive oxygen species (ROS) to target cells
[]. Enhanced ROS delivery to solid tumors was achieved by XOR conjugation to polyethylene glycol (PEG) [].
a XOR/antibody conjugate or with a free monoclonal anti-
body followed by a XOR/anti-antibody conjugate. Both direct
and indirect methods induced a prevalence of apoptotic death
over necrosis in malignant B lymphocytes [] (Figure (b)).
To improve the ROS delivery eciency to solid tumors,
XOR was conjugated to polyethylene glycol [], which (i)
confers superior in vivo pharmacokinetic characteristics by
increasing the blood half-life of the enzyme; (ii) counteracts
the aspecic adhesiveness of XOR to the vascular inner sur-
face; and (iii) concentrates XOR in cancer tissues by exploit-
ing the enhanced permeability and retention eect of macro-
molecules and lipids in solid tumors (reviewed in [])
(Figure (b)).
3. Proinflammatory Activity of Xanthine
Oxidoreductase Products
e evolution of XOR from the highly conserved dehydroge-
nase to the interconvertible mammalian oxidase form confers
to its enzyme activity a new role of producing physiologic
signal transduction that is mediated by ROS as secondary
messengers (reviewed in [, ]).
XOR activity is known to be upregulated in response
to inammatory cytokines [], which induce the XDH to
XO transition and also increase the XOR level in plasma
[], supporting the hypothesis that XOR is a component of
the innate immune system (reviewed in []). Indeed, XOR
has been implicated in the defense against infectious diseases
because of its capability of activating the cellular phlogistic
response at various levels (reviewed in []). XOR-derived
ROS promote leukocyte-endothelial cell interactions by
increasing the adhesion of phagocytes []. ey also induce
the production of cytokines [], thus amplifying the inam-
matory response, and chemotactic factors [], which cause
the accumulation of polymorphonuclear granulocytes in the
microvasculature []. e bactericidal activity of XOR may
contribute to the oxygen-dependent cell killing during leuko-
cyte phagocytosis through ROS and ONOOproduction
[]. e antibacterial properties of XOR suggest that its
abundanceinmilkcouldhavetheroleofanaturalantibiotic,
representing one of the reasons to encourage breastfeeding by
mothers [] (Figure (a)).
e usually very low XOR serum level in humans may
become more elevated in pathological circumstances that
causetissuedamageandthereleaseofXORfromcells
into the bloodstream. Circulating XOR is converted to the
oxidase form and binds to endothelial cells, even at distant
sites, inducing proinammatory signaling or even remote
organ injury (reviewed in []). e proinammatory activity
exerted by the XOR-derived ROS may aect the microvascu-
lar lining by inducing endothelium permeabilization, which
begins both the physiological cascade of immune response
and the pathological events that induce atheromatous
plaque formation (reviewed in []) (Figure (b)). e XOR
Oxidative Medicine and Cellular Longevity
Inammation
Angiogenesis
Tissue repair
ROS
Adhesion
Chemotaxis
XOR
Cytokine release
ROS/RNS production
RNS
Endothelial activation
Leucocyte activation
Bactericidal activity
XOR serum level
XO/XDH activity
XOR expression
(a)
rombosis
Athe ros clerosis
Vascular tone regulation
ROS
Permeabilization
NO
XOR
Endothelial dysfunction
(b)
F : Prophlogistic action of reactive oxygen (ROS) and nitrogen (RNS) species. (a) Interferon and other cytokines increase xanthine
oxidoreductase gene (XOR) expression as well as the conversion of xanthine dehydrogenase (XDH) to xanthine oxidase (XO) and XOR serum
level (reviewed in []). XOR-derived ROS and RNS mediate the endothelial and phagocytic cell activation that is functional in antibacterial
defense (reviewed in []). (b) XOR products induce endothelial permeabilization and dysregulation of vascular tone, which may lead to
thrombosis and atherosclerosis (reviewed in []).
products together with the oxidants generated by NAD(P)H
oxidase and NO synthase may also modulate another endo-
thelial cell function, the regulation of arteriolar tone via NO
production, which has local and systemic vasodilating activ-
ity and causes XOR inhibition (Figure (b)). NO is produced
by endothelial NO synthase that is inhibited either by ROS or
under hypoxic conditions. In these circumstances, NO gener-
ation is assured by the nitrite reductase activity of both XOR
and NAD(P)H oxidase, which undergo reciprocal activation
by generating O2(reviewedin[]).Astheactivitiesof
these enzymes are interdependent in the endothelium, the
nal outcome is the result of a physiopathological balance
amongst their activities. us, it is not surprising that both
XOR activity and its inhibition by allopurinol may induce
endothelial dysfunction and promote platelet aggregation, as
well as aggravating hypertension and cardiovascular diseases
[].
In patients with coronary disease, the treatment with
the angiotensin receptor blocker losartan reduced the endo-
thelium-bound XOR activity and XOR inhibition with oxy-
purinol improved endothelium-dependent vasodilation, sug-
gesting that endothelial dysfunction in coronary disease
is at least in part dependent on angiotensin II-dependent
endothelial XOR activation []. In patients with metabolic
syndrome, XOR inhibition by allopurinol reduces myeloper-
oxidase and malondialdehyde blood levels, while increasing
theow-mediateddilation,suggestingthatXOR-induced
oxidative stress contributes to endothelial vasomotor dys-
function[].eunderlyingmechanismissupposedtobe
the reduced bioavailability of NO due to the reaction of NO
with O2(reviewed in []). However, in grade  drug-
na¨
ıve hypertensive subjects a dietary nitrate load reduces
systolic and diastolic blood pressure. is eect is related to
an increased NO generation, which is signicantly attenu-
ated by allopurinol and is associated with higher levels of
erythrocytic XOR expression and nitrite reductase activity
in hypertensive patients in comparison to normotensives
volunteers []. e eects of ROS generated by human XOR
on cardiovascular disease have been detailed in two recent
publications (reviewed in [, ]).
XORmayproduceROSandNO,whicharebothrequired
for the formation of normal granulation tissue and wound
healing. In vitro keratinocytes and endothelial cell prolif-
eration and migration were increased by H2O2and nitrite.
XOR expression was upregulated shortly aer wounding at
the wound edge. Locally applied allopurinol, as well as a
tungsten-enriched diet that drastically lowered XOR activity,
signicantly delayed wound healing in mice. e eect was
reversed and angiogenesis improved with the topical H2O2
administration, strongly suggesting that XOR contributes to
wound repair [].
4. Pro- and Antitumorigenic Activity of
Xanthine Oxidoreductase Products
In both experimental and clinical pathology, the level of XOR
expression was oen found to be higher or lower in cancer
tissues compared with the corresponding normal tissue or to
Oxidative Medicine and Cellular Longevity
Promotion
Dierentiation
Transformation
Metastasization
Progression
Angiogenesis Apoptosis
PROS
CONS
CANCER
F : Cancer pathogenesis: ambiguousrole of xanthine oxidore-
ductase (XOR). XOR-derived ROS may activate genes responsible
for each phase of cancer development (reviewed in []) as well as
genes that promote antioncogenic activities (reviewed in []).
the normal tissues bordering cancerous tissues (reviewed in
[]). In particular, XOR expression and activity in neoplastic
human tissues have been recently addressed and discussed
together with the XOR role in dierentiation and oncogenesis
(reviewedin[]).Moreover,XORproductshavebeenasso-
ciated with both the process of oncogenesis ([], reviewed in
[]) and its prevention ([], reviewed in []) (Figure ).
e level of XOR activity was higher than normal and
that of paraoxonase l, a free radical scavenger enzyme, was
lower in the serum of patients with various cancer illnesses
[].Alowactivityofvariousoxidativeenzymes,inparticular
XOR,hasbeenreportedtocorrelatewithcellproliferation
in dierent settings, including cancer, and a hypothesis has
been formulated that a low level of free radicals may stimulate
cancer cell growth []. XOR can also confer a cancer-
promoting action through the above-discussed proinam-
matory activities of ROS and RNS.
e analysis of a vast cohort of women followed for 
years showed a dose-dependent risk of breast cancer by
alcohol consumption [] and a mechanism involving XOR-
derived ROS has been proposed for the pathogenesis of this
cancer. XDH is expressed at high levels by mammary epithe-
lium,particularlyinrelationtolactation,andcanproduce
ROS by oxidizing ethanol [] as well as acetaldehyde and
NADH, which are generated by alcohol catabolism. ROS can
be responsible for DNA damage, mutagenesis, and neoplastic
transformation, especially in aged breast tissue with high iron
levels and low antioxidant levels (reviewed in [, ]).
XOR is an upstream regulator of various molecules with
transduction signal functions in dierent pathways, which
may result in either pro- or antitumorigenic signaling.
In human lung microvascular endothelial cells, XOR was
shown to increase the expression of the tumor suppressor
protein p, which is very oen mutated and deactivated in
humancancer.XORinducedoxidativestress,DNAdamage,
and the ROS-dependent upregulation of p protein, with the
consequent activation of the caspase enzymatic cascade and
apoptosis [].
In T-L murine cells, the ROS produced by the NADH-
oxidizing activity of XOR were able to stimulate the acti-
vation of peroxisome proliferator-activated receptor-gamma
(PPAR-𝛾), which belongs to the nuclear hormone receptor
superfamily. is ligand-activated intracellular transcrip-
tion factor has antiproliferative and antioncogenic activities
because it can favor cell dierentiation and inhibit angiogen-
esis [].
XOR-derived ROS can modulate the expression of the
inammation mediator, cyclooxygenase- (COX-), by either
increasing or decreasing its expression. e XOR-dependent
COX- expression in newborn mice was essential for regular
kidney development, and the lack of XOR was associated
with renal hypoplasia and dysplasia []. Additionally, XOR
depletion in primary renal epithelial cells induced positive
immunostaining for mesenchymal cell type markers and the
lack of reactivity to E-cadherin associated with cell morphol-
ogy changes from a cuboidal to myobroblastic shape, which
indicated epithelial to mesenchymal transition []. However,
a high XOR level in human mammar y epithelium lowered the
COX- and matrix metalloprotease expression levels, which
are crucial for cell migratory activity and thus for tumor
progression and ability of metastasis formation [].
XOR-generatedoxidantscanturnonthenuclearfactor
kappa-light-chain-enhancer of activated B cells (NF-𝜅B) in
rat liver both during ischemia [] and in type  diabetes [].
NF-𝜅B is a transcription factor that is usually activated during
chronic inammation and in cancer, where it promotes the
production of immunological cytokines and the expression
of a set of antiapoptotic genes.
In U-MG cells, derived from human brain, chemically
induced hypoxia increased XOR activity and the level of
XOR-derived ROS, which upregulated hypoxia-inducible
factor- alpha (HIF-𝛼) []. is transcription factor is over-
expressed in hypoxia and induces angiogenesis, as well as can-
cer invasion, thus contributing to both tumor development
and progression.
5. Conclusions
Mammal XOR is the end product of a complicated evolu-
tionary process leading to a hyperregulated enzyme with low
specicity and highly versatile activity. In mammals, XOR
has acquired many functions through the production of ROS,
NO, and RNS, whereby it is involved in the triggering of
key biological cell pathways and in the regulation of several
physiological and pathological conditions. For these reasons,
XOR represents the two faces of free radicals, which can
have either negative or positive eects. XOR-derived RNS
andROSmayhaveacytotoxiceect.isactivitymaybe
responsiblefortissuedamageinhypoxia/reoxygenationand
ischemia/reperfusion injury. However, this cytotoxic eect
can be pharmacologically exploited to obtain selective can-
cerous cell killing by conjugating XOR to a specic antibody.
Oxidative Medicine and Cellular Longevity
XOR activity increases during infectious diseases and its cyto-
toxic action is useful for the defenses against bacteria. Addi-
tionally, XOR-derived NO and ROS have proinammatory
activity because they regulate endothelial functions, by both
increasing the permeability of vascular lining and modulating
the arteriolar tone. For this reason, XOR has been implicated
in hypertension, cardiovascular diseases, and atherosclerosis.
XOR-derived ROS are also involved in cancer pathogenesis
because they may promote neoplastic transformation by
activating target genes with prophlogistic, antiapoptotic, and
proliferative actions. Moreover, they favor the progression to
malignancy by inducing angiogenesis and cell migration. On
the other hand, XOR products may activate the expression
of the proapoptotic protein p and of transcription factors
belonging to the nuclear hormone receptor superfamily with
antitumorigenic and antiproliferative activity, promoting cell
dierentiation and inhibiting angiogenesis.
Highlights
(i) XOR-derived ROS, NO, and RNS have proinamma-
tory and bactericidal activities.
(ii) XOR products may be cytotoxic in many circum-
stances.
(iii) XOR products modulate endothelial function and
arteriolar tone.
(iv) XOR products may induce mutagenesis, cell prolifer-
ation, and tumor progression.
(v) XOR products are associated with apoptosis and cell
dierentiation.
Abbreviations
COX-: Cyclooxygenase-
FAD: Flavin adenine dinucleotide
HO:Hydroxylradical
H2O2: Hydrogen peroxide
Mo-co: Molybdenum-containing molybdopterin
cofactor
NAD+: Oxidized nicotinamide adenine dinucleotide
NADH: Reduced nicotinamide adenine dinucleotide
NF-𝜅B: Nuclear factor kappa-light-chain-enhancer
of activated B cells
NO: Nitric oxide
ONOO:Peroxynitrite
RNS: Reactive nitrogen species
ROS: Reactive oxygen species
O2:Molecularoxygen
O2:Superoxideanion
XDH: Xanthine dehydrogenase
XO: Xanthine oxidase
XOR: Xanthine oxidoreductase.
Conflict of Interests
e authors declare that there is no conict of interests
regarding the publication of this paper.
Acknowledgment
is work was supported by the Pallotti Legacies for Cancer
Research.
References
[] D. A. Parks and D.N. Granger, “Xanthine oxidase: biochemistry,
distribution and physiology,” Acta Physiologica Scandinavica,
vol. , no. , pp. –, .
[] R. Hille and T. Nishino, “Xanthine oxidase and xanthine dehy-
drogenase,e FASEB Journal,vol.,no.,pp.,.
[] B.N.Ames,R.Cathcart,E.Schwiers,andP.Hochstein,“Uric
acid provides an antioxidant defense in humans against oxi-
dant- and radical-caused aging and cancer: a hypothesis,Pro-
ceedings of the National Academy of Sciences of the United States
of America, vol. , no. , pp. –, .
[] P. Xu, P. LaVallee, and J. R. Hoidal, “Repressed expression of
the human xanthine oxidoreductase gene. E-box and TATA-
like elements restrict ground state transcriptional activity,” e
JournalofBiologicalChemistry, vol. , no. , pp. –,
.
[] A. Kooij, “A re-evaluation of the tissue distribution and physiol-
ogy of xanthine oxidoreductase,Histochemical Journal,vol.,
no.,pp.,.
[] C. E. Berry and J. M. Hare, “Xanthine oxidoreductase and
cardiovascular disease: molecular mechanisms and pathophys-
iological implications,e Journal of Physiology,vol.,no.,
pp.,.
[] A. Boueiz, M. Damarla, and P. M. Hassoun, “Xanthine oxi-
doreductase in respiratory and cardiovascular disorders,e
American Journal of Physiology—Lung Cellular and Molecular
Physiology,vol.,no.,pp.LL,.
[] E. Della Corte and F. Stirpe, “e regulation of rat liver xanthine
oxidase. Involvement of thiol groups in the conversion of the
enzyme activity from dehydrogenase (type D) into oxidase
(type O) and purication of the enzyme,Biochemical Journal,
vol. , no. , pp. –, .
[] N. Cantu-Medellin and E. E. Kelley, “Xanthine oxidoreductase-
catalyzed reactive species generation: a process in critical need
of reevaluation,Redox Biology,vol.,no.,pp.,.
[] R. Harrison, “Structure and function of xanthine oxidoreduc-
tase: where are we now?” Free Radical Biology and Medicine,vol.
,no.,pp.,.
[] M. G. Battelli, A. Bolognesi, and L. Polito, “Pathophysiology of
circulating xanthine oxidoreductase: new emerging roles for a
multi-tasking enzyme,Biochimica et Biophysica Acta,vol.,
no.,pp.,.
[] C. Li and R. M. Jackson, “Reactive species mechanisms of
cellular hypoxia-reoxygenation injury,American Journal of
Physiology: Cell Physiology,vol.,no.,pp.CC,.
[] A. H. Bhat, K. B. Dar, S. Anees et al., “Oxidative stress, mito-
chondrial dysfunction and neurodegenerative diseases; a mech-
anistic insight,” Biomedicine & Pharmacotherapy,vol.,pp.
–, .
[] M. Chiricolo, P. L. Tazzari, A. Abbondanza, A. Dinota, and M.
G. Battelli, “Cytotoxicity of, and DNA damage by, active oxygen
species produced by xanthine oxidase,FEBS Letters,vol.,
no. , pp. –, .
[] M. G. Battelli, S. Musiani, P. L. Tazzari, and F. Stirpe, “Oxida-
tive stress to human lymphocytes by xanthine oxidoreductase
activity,Free Radical Research,vol.,no.,pp.,.
Oxidative Medicine and Cellular Longevity
[] C.-C. Huang, K.-L. Chen, C. H. A. Cheung, and J.-Y. Chang,
Autophagy induced by cathepsin S inhibition induces early
ROS production, oxidative DNA damage, and cell death via
xanthine oxidase,FreeRadicalBiologyandMedicine,vol.,
pp.,.
[] B. S. Kim, L. Serebreni, O. Hamdan et al., “Xanthine oxidore-
ductase is a critical mediator of cigarette smoke-induced endo-
thelial cell DNA damage and apoptosis,Free Radical Biology
and Medicine,vol.,pp.,.
[] M. G. Battelli, A. Abbondanza, P. L. Tazzari et al., “Selective
cytotoxicity of an oxygen-radical-generating enzyme conju-
gated to a monoclonal antibody,Clinical and Experimental
Immunology, vol. , no. , pp. –, .
[] P. L. Tazzari, M. G. Battelli, A. Abbondanza et al., “Targeting of
a plasma cell line with a conjugate containing xanthine oxidase
and the monoclonal antibody B,Tran spl antat ion,vol.,
no. , pp. –, .
[] A. Dinota, P. L. Tazzari, A. Abbondanza, M. G. Battelli, M.
Gobbi, and F. Stirpe, “Bone marrow purging by a xanthine oxi-
dase-antibody conjugate,Bone Marrow Transplantation,vol.,
no. , pp. –, .
[] M. G. Battelli, A. Abbondanza, P. L. Tazzari, A. Bolognesi, R. M.
Lemoli, and F. Stirpe, “T lymphocyte killing by a xanthine-oxi-
dase-containing immunotoxin,Cancer Immunology, Immun-
otherapy,vol.,no.,pp.,.
[] S. J. Dixon and B. R. Stockwell, “e role of iron and reactive
oxygen species in cell death,” Nature Chemical Biology,vol.,
no.,pp.,.
[] M. G. Battelli, L. Polito, F. Fal`
a et al., “Toxicity of xanthine oxi-
doreductase to malignant B lymphocytes,Journal of Biological
Regulators & Homeostatic Agents,vol.,no.-,pp.,
.
[] T. Sawa, J. Wu, T. Akaike, and H. Maeda, “Tumor-targeting
chemotherapy by a xanthine oxidase-polymer conjugate that
generates oxygen-free radicals in tumor tissue,Cancer Re-
search,vol.,no.,pp.,.
[] J.Fang,T.Seki,andH.Maeda,“erapeuticstrategiesbymod-
ulating oxygen stress in cancer and inammation,” Advanced
Drug Delivery Reviews,vol.,no.,pp.,.
[] H. Sauer, M. Wartenberg, and J. Hescheler, “Reactive oxygen
species as intracellular messengers during cell growth and
dierentiation,Cellular Physiology and Biochemistry,vol.,no.
, pp. –, .
[] A. Meneshian and G. B. Bulkley, “e physiology of endothelial
xanthine oxidase: from urate catabolism to reperfusion injury
to inammatory signal transduction,Microcirculation,vol.,
no. , pp. –, .
[] S. Page, D. Powell, M. Benboubetra et al., “Xanthine oxi-
doreductase in human mammary epithelial cells: activation in
response to inammatory cytokines,Biochimica et Biophysica
Acta—General Subjects,vol.,no.,pp.,.
[] A. Agarwal, A. Banerjee, and U. C. Banerjee, “Xanthine oxidore-
ductase: a journey from purine metabolism to cardiovascular
excitation-contraction coupling,Critical Reviews in Biotechnol-
ogy,vol.,no.,pp.,.
[] H. M. Martin, J. T. Hancock, V. Salisbur y, and R. Harrison, “Role
of xanthine oxidoreductase as an antimicrobial agent,Infection
and Immunity,vol.,no.,pp.,.
[] M. Suzuki, M. B. Grisham, and D. N. Granger, “Leukocyte-
endothelial cell adhesive interactions: role of xanthine oxidase-
derived oxidants,Journal of Leukocyte Biology,vol.,no.,
pp. –, .
[] R. Shenkar and E. Abraham, “Plasma from hemorrhaged
mice activates CREB and increases cytokine expression in
lung mononuclear cells through a xanthine oxidase-dependent
mechanism,American Journal of Respiratory Cell and Molecu-
lar Biology, vol. , no. , pp. –, .
[] W. F. Petrone, D. K. English, K. Wong, and J. M. McCord, “Free
radicals and inammation: superoxide-dependent activation of
a neutrophil chemotactic factor in plasma,Proceedings of the
National Academy of Sciences of the United States of America,
vol. , no. , pp. –, .
[] M. J. M¨
uller, B. Vollmar, H.-P. Friedl, and M. D. Menger, “Xan-
thine oxidase and superoxide radicals in portal triad cross-
clamping-induced microvascular reperfusion injury of the
liver,Free Radical Biology and Medicine,vol.,no.,pp.
, .
[] E.Tubaro,B.Lotti,C.Santiangelli,andG.Cavallo,“Xanthine
oxidase: an enzyme playing a role in the killing mechanism
of polymorphonuclear leucocytes,Biochemical Pharmacology,
vol. , no. , pp. –, .
[] C. R. Stevens, T. M. Millar, J. G. Clinch, J. M. Kanczler, T. Bod-
amyali, and D. R. Blake, “Antibacterial properties of xanthine
oxidase in human milk,e Lancet,vol.,no.,pp.
, .
[]M.G.Battelli,L.Polito,andA.Bolognesi,“Xanthineoxi-
doreductase in atherosclerosis pathogenesis: not only oxidative
stress,Atherosclerosis, vol. , no. , pp. –, .
[] H. Cai and D. G. Harrison, “Endothelial dysfunction in car-
diovascular diseases: the role of oxidant stress,Circulation
Research,vol.,no.,pp.,.
[] N. Cantu-Medellin and E. E. Kelley, “Xanthine oxidoreductase-
catalyzed reduction of nitrite to nitric oxide: insights regarding
where, when and how,Nitric Oxide,vol.,pp.,.
[] U. Landmesser, S. Spiekermann, C. Preuss et al., “Angiotensin
II induces endothelial xanthine oxidase activation: role for
endothelial dysfunction in patients with coronary disease,
Arteriosclerosis, rombosis, and Vascular Biology,vol.,no.
, pp. –, .
[] O. Yiginer, F. Ozcelik, T. Inanc et al., “Allopurinol improves
endothelial function and reduces oxidant-inammatory en-
zyme of myeloperoxidase in metabolic syndrome,Clinical
Research in Cardiology,vol.,no.,pp.,.
[] S. M. Ghosh, V. Kapil, I. Fuentes-Calvo et al., “Enhanced
vasodilator activity of nitrite in hypertension: critical role for
erythrocytic xanthine oxidoreductase and translational poten-
tial,Hypertension,vol.,no.,pp.,.
[] M. C. Madigan, R. M. McEnaney, A. J. Shukla et al., “Xanthine
oxidoreductase function contributes to normal wound healing,
Molecular Medicine,vol.,no.,pp.,.
[] G. Weber, “Biochemical strategy of cancer cells and the design
of chemotherapy: G.H.A. Clowes memorial lecture,Cancer
Research,vol.,no.,pp.,.
[] M. G. Battelli, L. Polito, M. Bortolotti, and A. Bolognesi, “Xan-
thine oxidoreductase in cancer: more than a dierentiation
marker,Cancer Medicine,Inpress.
[] I. A. Shmarakov and M. M. Marchenko, “Xanthine oxidase
activity in transplantable Guerin’s carcinoma in rats,Vo p r osy
Onkologii,vol.,no.,pp.,.
[] K. Balamurugan, “HIF- at the crossroads of hypoxia, inam-
mation, and cancer,International Journal of Cancer,.
[] M. A. Fini, D. Orchard-Webb, B. Kosmider et al., “Migratory
activity of human breast cancercel ls ismo dulated by dierential
Oxidative Medicine and Cellular Longevity
expression of xanthine oxidoreductase,” Journal of Cellular
Biochemistry, vol. , no. , pp. –, .
[] M. Valko, C. J. Rhodes, J. Moncol, M. Izakovic, and M. Mazur,
“Free radicals, metals and antioxidants in oxidative stress-
induced cancer,Chemico-Biological Interactions,vol.,no.,
pp. –, .
[] Z. Q. Samra, S. Pervaiz, S. Shaheen, N. Dar, and M. A. Athar,
“Determination of oxygen derived free radicals producer (xan-
thine oxidase) and scavenger (paraoxonasel) enzymes and lipid
parameters in dierent cancer patients,Clinical Laboratory,
vol. , no. -, pp. –, .
[] A. S. Sun and A. I. Cederbaum, “Oxidoreductase activities in
normal rat liver, tumor-bearing rat liver, and hepatoma HC-
,Cancer Research, vol. , no. , pp. –, .
[] S. A. Smith-Warner, D. Spiegelman, S.-S. Yaun et al., “Alcohol
and breast cancer in women: a pooled analysis of cohort
studies,eJournaloftheAmericanMedicalAssociation,vol.
, no. , pp. –, .
[]G.D.Castro,A.M.A.DelgadodeLayo,M.H.Costantini,
and J. A. Castro, “Cytosolic xanthine oxidoreductase mediated
bioactivation of ethanol to acetaldehyde and free radicals in rat
breast tissue. Its potential role in alcohol-promoted mammary
cancer,To x i c olog y ,vol.,no.,pp.,.
[]R.M.Wright,J.L.McManaman,andJ.E.Repine,“Alcohol-
induced breast cancer: a proposed mechanism,Free Radical
Biology and Medicine, vol. , no. -, pp. –, .
[] R. G. Dumitrescu and P. G. Shields, “e etiology of alcohol-
induced breast cancer,” Alcohol,vol.,no.,pp.,.
[] K. J. Cheung, I. Tzameli, P. Pissios et al., “Xanthine oxidore-
ductaseisaregulatorofadipogenesisandPPAR𝛾activity,Cell
Metabolism,vol.,no.,pp.,.
[]T.Ohtsubo,I.I.Rovira,M.F.Starost,C.Liu,andT.Finkel,
“Xanthine oxidoreductase is an endogenous regulator of cy-
clooxygenase-,Circulation Research, vol. , no. , pp. –
, .
[] T. Ohtsubo, K. Matsumura, K. Sakagami et al., “Xanthine oxi-
doreductase depletion induces renal interstitial brosis through
aberrant lipid and purine accumulation in renal tubules,
Hypertension,vol.,no.,pp.,.
[] N.Matsui,I.Satsuki,Y.Moritaetal.,“Xanthineoxidase-derived
reactive oxygen species activate nuclear factor kappa B during
hepatic ischemia in rats,e Japanese Journal of Pharmacolog y,
vol. , no. , pp. –, .
[] M. Romagnoli, M.-C. Gomez-Cabrera, M.-G. Perrelli et al.,
“Xanthine oxidase-induced oxidative stress causes activation of
NF-𝜅B and inammation in the liver of type I diabetic rats,Free
Radical Biology and Medicine,vol.,no.,pp.,.
[] C. E. Griguer, C. R. Oliva, E. E. Kelley, G. I. Giles, J. R. Lancaster
Jr., and G. Y. Gillespie, “Xanthine oxidase-dependent regulation
of hypoxia-inducible factor in cancer cells,Cancer Research,
vol.,no.,pp.,.
... Xanthine oxidase (XO), a crucial enzyme in the purine breakdown pathway, has been linked to elevated uric acid production. XO catalyses the conversion of hypoxanthine and xanthine to uric acid, and generates reactive oxygen species (ROS) such as hydrogen peroxide and superoxide radicals [4]. The generation of these ROS has been associated with the potential onset of several comorbidities, such as hypertension, diabetes, cardiovascular and renal diseases. ...
... Total ion chromatograms of positive and negative electrospray ionization of A. hierochuntica methanolic leaves extract. The methanolic leaves extract of A. hierochuntica was subjected to LC-4 QTOF-MS analysis. MassHunter software reported analysis of qualitative and quantitative data, depicting a mass screening spectrum of the total ion chromatogram. ...
Preprint
Full-text available
There is growing interest in the discovery of novel xanthine oxidase inhibitors for gout prevention and treatment with fewer side effects. This study aimed to identify the xanthine oxidase (XO) inhibitory potential and drug-likeness of the metabolites present in the methanolic leaves extract of Anastatica (A.) hierochuntica L. using in vitro and in silico models. The extract-derived metabolites were identified by liquid chromatography-quadrupole time-of-flight mass spectrometry (LC-QTOF-MS). Molecular docking predicted the XO inhibitory activity of the identified metabolites and validated the best scored in vitro XO inhibitory activities for experimental verification as well as predictions of their anticancer, pharmacokinetic and toxic properties, oral bioavailability, and endocrine disruption using SwissADMET, PASS, ProTox-II and Endocrine Disruptome web servers. A total of 12 metabolites with a majority of flavonoids were identified. Rutin, quercetin and luteolin flavonoids demonstrated the highest ranked docking score of -12.39, -11.15 and -10.43, respectively, while the half-maximal inhibitory concentration (IC50) values of these metabolites against XO activity were 11.35 µM, 11.1 µM and 21.58 µM, respectively. In addition, SwissADMET generated data related to physicochemical properties and drug-likeness of the metabolites. Similarly, PASS, ProTox-II and Endocrine Disruptome prediction models stated the safe and potential use of these natural compounds. However, in vivo studies are necessary to support the development of prominent and promising therapeutic use of A. hierochuntica methanolic leaves extract-derived metabolites as XO inhibitors for the prevention and treatment of hyperuricemic and gout patients. Furthermore, the predicted findings of the present study open a new paradigm for these extract-derived metabolites by revealing novel oncogenic targets for the potential treatment of human malignancies.
... Xanthine oxidoreductase (XOR) is another potential source of ROS in the vascular endothelium. Dependent on the form of XOR, the enzymatic reaction is accompanied by NADH production (xanthine dehydrogenase, XDH) or superoxide and hydrogen peroxide generation (xanthine oxidase, XO) [3,55]. XDH to XO conversion is stimulated by H 2 O 2 and this can be prevented by NO [56]. ...
Article
Full-text available
Multiple common cardiovascular comorbidities produce coronary microvascular dysfunction. We previously observed in swine that a combination of diabetes mellitus (DM), high fat diet (HFD) and chronic kidney disease (CKD) induced systemic inflammation, increased oxidative stress and produced coronary endothelial dysfunction, altering control of coronary microvascular tone via loss of NO bioavailability, which was associated with an increase in circulating endothelin (ET). In the present study, we tested the hypotheses that (1) ROS scavenging and (2) ETA+B-receptor blockade improve myocardial oxygen delivery in the same female swine model. Healthy female swine on normal pig chow served as controls (Normal). Five months after induction of DM (streptozotocin, 3 × 50 mg kg⁻¹ i.v.), hypercholesterolemia (HFD) and CKD (renal embolization), swine were chronically instrumented and studied at rest and during exercise. Sustained hyperglycemia, hypercholesterolemia and renal dysfunction were accompanied by systemic inflammation and oxidative stress. In vivo ROS scavenging (TEMPOL + MPG) reduced myocardial oxygen delivery in DM + HFD + CKD swine, suggestive of a vasodilator influence of endogenous ROS, while it had no effect in Normal swine. In vitro wire myography revealed a vasodilator role for hydrogen peroxide (H2O2) in isolated small coronary artery segments from DM + HFD + CKD, but not Normal swine. Increased catalase activity and ceramide production in left ventricular myocardial tissue of DM + HFD + CKD swine further suggest that increased H2O2 acts as vasodilator ROS in the coronary microvasculature. Despite elevated ET-1 plasma levels in DM + HFD + CKD swine, ETA+B blockade did not affect myocardial oxygen delivery in Normal or DM + HFD + CKD swine. In conclusion, loss of NO bioavailability due to 5 months exposure to multiple comorbidities is partially compensated by increased H2O2-mediated coronary vasodilation.
... XO aids in the metabolism of uric acid, producing superoxide ions, H 2 O 2 , and NO as byproducts under physiological circumstances (Battelli et al. 2016). These byproducts have been reported to exert cytocidal effects and are associated with tissue damage in the liver and kidney. ...
Article
Full-text available
Sodium fluoride (NaF) ingestion has several detrimental effects in humans and rodents. NaF mechanisms of toxicity include perturbation of intracellular redox homeostasis and apoptosis. Betaine (BET) is a modified amino acid with anti-inflammatory, antioxidant, and anti-apoptotic properties. This study investigates BET’s effect on NaF-induced hepatorenal toxicities in rats. Experimental rats (n = 30) were randomly assigned to groups (n = 6) and treated by gavage for 28 days. Group I (2 mL of distilled water), Group II (NaF: 9 mg/kg) alone, Group III: (BET: 100 mg/kg), Group IV: (NaF: 9 mg/kg and BET 1: 50 mg/kg), and Group V: (NaF: 9 mg/kg and BET 2: 100 mg/kg). Our findings revealed significantly (p < 0.05) increased hepatic transaminase activities alongside creatinine and urea levels following NaF-alone treatment in addition to increased oxidative status, lipid peroxidation, reactive oxygen and nitrogen species, decreased superoxide dismutase, catalase, glutathione-s-transferase, glutathione peroxidase, glutathione, and total sulfhydryl groups. The reduced levels of nuclear factor erythroid 2-related factor-2 and the activities of heme oxygenase-1, thioredoxin, and thioredoxin reductase in NaF-alone treated rats equally compromised cellular molecular responses to oxidative stress. Also, NaF increased (p < 0.05) hepatorenal inflammatory biomarkers-nitric oxide, interleukin-10, myeloperoxidase, and xanthine oxidase. Furthermore, caspase-3 and caspase-9 were increased (p < 0.05) in rats treated with NaF alone. Contrastingly, BET was observed to alleviate the harmful effects of NaF. Treatment with BET mitigated NaF-induced oxido-inflammatory responses and apoptosis in the experimental rat’s hepatorenal system. The study demonstrates the potential of BET to abate NaF-induced hepatorenal toxicity.
... Adenosine is produced during conditions of limited oxygen availability to confer tissue protection [46] and also acts as an endogenous activator of the cellular antioxidant defense system for cytoprotection during ischemic cell injuries [47,48]. Xanthine oxidase, the enzyme that catalyzes the last two steps in purine metabolism [49], is an enzyme that generates ROS, participates in oxidative stress [50] and has been found to be 10-20-fold higher than that found in healthy liver tissue [51]. Also, increased ADA has been linked to hepatic dysfunction [52]. ...
Article
Full-text available
Liver diseases have gained increasing attention due to their substantial impact on health, independently as well as in association with cardio-metabolic disorders. Studies have suggested that glutathione and adenosine assist in providing protection against oxidative stress and inflammation while glucocorticoid (GC) therapy has been associated with chronic inflammatory disorders, even in pregnancy. The implications of Glucocorticoid exposure on maternal health and fetal growth is a concern, however, the possible role of glutathione and adenosine has not been thoroughly investigated. The study therefore hypothesize that exposure to glucocorticoids leads to depletion of hepatic glutathione and adenosine levels, contributing to oxidative stress and tissue injury. Additionally, we aim to investigate whether the effects of glucocorticoids on hepatic health are pregnancy dependent in female rats. Twelve Pregnant and twelve age-matched non-pregnant rats were used for this study; an exogenous administration of glucocorticoid (Dex: 0.2 mg/kg) or vehicle (po) was administered to six pregnant and six non-pregnant rats from gestational day 14 to 19 or for a period of 6 days respectively. Data obtained showed that GC exposure led to a decrease in hepatic glucose-6-phosphate dehydrogenase, glutathione peroxidase, GSH/GSSG ratio and adenosine content in both pregnant and non-pregnant rats. In addition, increased activities of adenosine deaminase and xanthine oxidase, along with increased production of uric acid and increased levels of lactate dehydrogenase, aspartate aminotransferase, alanine transferase, alkaline phosphatase and gamma-glutamyl transferase were observed. In summary, the study indicates that GC-induced liver damage is underlined by depleted hepatic adenosine and glutathione levels as well as elevated markers of tissue inflammation and/or injury. Furthermore, the findings suggest that the effects of GC exposure on hepatic health are pregnancy independent.
... linked to oxidative stress [4][5]. The XO inhibitors have been investigated as potential drugs to block the biosynthesis of uric acid and have shown promise in anti-cancer therapies [6]. Allopurinol is a common drug for gout treatment, mainly included as an XO inhibitor to reduce uric acid levels. ...
Article
Full-text available
Xanthine oxidase (XO) is a crucial enzyme of the purine catabolism pathway, which catalyzes the reaction of hypoxanthine to xanthine and xanthine to uric acid. The high level of uric acid leads to gout, kidney disease, and several disorders. In this study, molecular docking was performed to investigate potential bioactive compounds from Thai medicinal plants that acted as XO inhibitors compared to commercial drugs. Among 30 bioactive compounds tested, 16 were classified as strong XO inhibitors. These compounds include asiatic acid, benzyl glucosinolate, beta-sitosterol, chlorogenic acid, curcumin, eupatorin, gamma-mangostin, hibiscitrin, lutein, nimbolide, piperine, quercetin, rosmarinic acid, rutin, sesamin, and vitexin. They exhibited binding affinity values ranging from -8.5 to -10.6 kcal/mol. Moreover, moderate XO inhibitors were identified with binding affinity of -6.2 to -8.0 kcal/mol, consisting of the 7 compounds of gallic acid, garcinia acid, gingerol, limonene, linalyl acetate, panduratin A, and scopoletin. As a result, Thai medicinal plants could serve as potential sources of bioactive compounds for further drug design for treating gout patients.
Article
Full-text available
Xanthine Oxidoreductase (XOR) is a ubiquitous, essential enzyme responsible for the terminal steps of purine catabolism, ultimately producing uric acid that is eliminated by the kidneys. XOR is also a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various physiological pathways, as well as contribute to the development and the progression of chronic conditions including kidney diseases, which are increasing in prevalence worldwide. XOR activity can promote oxidative distress, endothelial dysfunction, and inflammation through the biological effects of reactive oxygen species; nitric oxide and uric acid are the major products of XOR activity. However, the complex relationship of these reactions in disease settings has long been debated, and the environmental influences and genetics remain largely unknown. In this review, we give an overview of the biochemistry, biology, environmental, and current clinical impact of XOR in the kidney. Finally, we highlight recent genetic studies linking XOR and risk for kidney disease, igniting enthusiasm for future biomarker development and novel therapeutic approaches targeting XOR.
Article
Full-text available
Reactive oxygen species (ROS) serve as typical metabolic byproducts of aerobic life and play a pivotal role in redox reactions and signal transduction pathways. Contingent upon their concentration, ROS production not only initiates or stimulates tumorigenesis but also causes oxidative stress (OS) and triggers cellular apoptosis. Mounting literature supports the view that ROS are closely interwoven with the pathogenesis of a cluster of diseases, particularly those involving cell proliferation and differentiation, such as myelodysplastic syndromes (MDS) and chronic/acute myeloid leukemia (CML/AML). OS caused by excessive ROS at physiological levels is likely to affect the functions of hematopoietic stem cells, such as cell growth and self-renewal, which may contribute to defective hematopoiesis. We review herein the eminent role of ROS in the hematological niche and their profound influence on the progress of MDS. We also highlight that targeting ROS is a practical and reliable tactic for MDS therapy. Graphical Abstract
Article
Introduction: This paper highlights the relationship of inflammation and oxidative stress as damage mechanisms of Multiple Sclerosis (MS), considered an inflammatory and autoimmune disease. Development: The oxidative stress concept has been defined by an imbalance between oxidants and antioxidants in favor of the oxidants. There is necessary to do physiological functions, like the respiration chain, but in certain conditions, the production of reactive species overpassed the antioxidant systems, which could cause tissue damage. On the other hand, it is well established that inflammation is a complex reaction in the vascularized connective tissue in response to diverse stimuli. However, an unregulated prolonged inflammatory process also can induce tissue damage. Conclusion: Both inflammation and oxidative stress are interrelated since one could promote the other, leading to a toxic feedback system, which contributes to the inflammatory and demyelination process in MS.
Article
Full-text available
Human xanthine oxidoreductase (XOR) catalyzes the last two steps of purine catabolism and is present in two interconvertible forms, which may utilize O2 or NAD(+) as electron acceptors. In addition to uric acid, XOR products may comprise reactive oxygen and nitrogen species that have many biologic effects, including inflammation, endothelial dysfunction, and cytotoxicity, as well as mutagenesis and induction of proliferation. XOR is strictly modulated at the transcriptional and post-translational levels, and its expression and activity are highly variable in cancer. Xanthine oxidoreductase (XOR) expression has been negatively associated with a high malignity grade and a worse prognosis in neoplasms of the breast, liver, gastrointestinal tract, and kidney, which normally express a high level of XOR protein. However, the level of XOR expression may be associated with a worse outcome in cancer of low XOR-expressing cells, in relation to the inflammatory response elicited through the tissue damage induced by tumor growth. Xanthine oxidoreductase (XOR) has been implicated in the process of oncogenesis either directly because it is able to catalyze the metabolic activation of carcinogenic substances or indirectly through the action of XOR-derived reactive oxygen and nitrogen species. The role of uric acid is characterized by both oxidant and antioxidant action; thus, it is still debatable whether control of uricemia may be helpful to improve the outcomes of tumor illness.
Article
Full-text available
Mitochondria is one of the main source of oxidative stress (ROS), as it utilizes the oxygen for the energy production. ROS and RNS are normally generated by tightly regulated enzymes. Excessive stimulation of NAD(P)H and electron transport chain leads to the overproduction of ROS, results in oxidative stress, which is a good mediator to injure the cell structures, lipids, proteins, and DNA. Various oxidative events implicated in many diseases due to oxidative stress include alteration in mitochondrial proteins, mitochondrial lipids and mitochondrial DNA, Which in turn leads to the damage to nerve cell as they are metabolically very active. ROS/RNS at moderate concentrations also play roles in normal physiology of many processes like signaling pathways, induction of mitogenic response and in defense against infectious pathogens. Oxidative stress has been considered to be the main cause in the etiology of many diseases, which includes Parkinson's and Alzheimer diseases. Several PD associated genes have been found to be involved in mitochondrial function, dynamics and morphology as well. This review includes source of free radical generation, chemistry and biochemistry of ROS/RNS and mitochondrial dysfunction and the mechanism involved in neurodegenerative diseases.
Article
Full-text available
The complex cross-talk of intricate inter-cellular signaling networks between the tumor and stromal cells promotes cancer progression. Hypoxia is one of the most common conditions encountered within the tumor microenvironment that drives tumor progression. Most responses to hypoxia are elicited by a family of transcription factors called hypoxia-inducible factors (HIFs), which induce expression of a diverse set of genes that assist cells to adapt to hypoxic environments. Among the three HIF protein family members, the role of HIF-1 is well established in cancer progression. HIF-1 functions as a signaling hub to coordinate the activities of many transcription factors and signaling molecules that impact tumorigenesis. This mini review discusses the complex role of HIF-1 and its context-dependent partners under various cancer-promoting events including inflammation and generation of cancer stem cells (CSCs), which are implicated in tumor metastasis and relapse. In addition, the review highlights the importance of therapeutic targeting of HIF-1 for cancer prevention. This article is protected by copyright. All rights reserved. © 2015 UICC.
Article
Full-text available
Endothelial xanthine oxidoreductase (XOR) together with NAD(P)H oxidase and nitric oxide (NO) synthase plays a physiologic role in inflammatory signalling, the regulation of NO production and vascular function. The oxidative stress generated by these enzymes may induce endothelial dysfunction, leading to atherosclerosis, cardiovascular diseases and metabolic syndrome. XOR activity creates both oxidant and anti-oxidant products that are implicated in the development of hypertension, smoking vascular injury, dyslipidemia and diabetes, which are the main risk factors of atherosclerosis. In particular, uric acid may have a protective as well as a detrimental role in vascular alterations, thus justifying the multi-directional effects of XOR inhibition. Moreover, XOR products are associated with cell differentiation, leading to adipogenesis and foam cell formation, as well as to the production of monocyte chemoattractant protein-1 from arterial smooth muscle cells, after proliferation and migration. The role of XOR in adipogenesis is also connected with insulin resistance and obesity, two main features of type 2 diabetes. Copyright © 2014 The Authors. Published by Elsevier Ireland Ltd.. All rights reserved.
Article
Xanthine oxidoreductase is an enzyme which has the unusual property that it can exist in a dehydrogenase form which uses NAD⁺ and an oxidase form which uses oxygen as electron acceptor. Both forms have a high affinity for hypoxanthine and xanthine as substrates. In addition, conversion of one form to the other may occur under different conditions. The exact function of the enzyme is still unknown but it seems to play a role in purine catabolism, detoxification of xenobiotics and antioxidant capacity by producing urate. The oxidase form produces reactive oxygen species and, therefore, the enzyme is thought to be involved in various pathological processes such as tissue injury due to ischaemia followed by reperfusion, but its role is still a matter of debate. The present review summarizes information that has become available about the enzyme. Interpretations of contradictory findings are presented in order to reduce confusion that still exists with respect to the role of this enzyme in physiology and pathology.
Article
Influence of low-dosage pre-irradiation on xanthine oxidase levels in Guerin's carcinoma was studied with particular emphasis on free oxygen generation and free sulphohydrils in a fraction of proteins characterized by xanthine oxidase activity. Enhanced growth of tumor correlated with that of xanthine oxidase activity involving higher levels of enzymatic O-form due to thiol groups oxidation. Under similar conditions, xanthine oxidase and free oxygen generation levels were higher due to SH-group oxidation than in intact animals. Terminal stages involved dropping xanthine oxidase levels and enzymatic protein degradation.
Article
Objective.— To assess the risk of invasive breast cancer associated with total and beverage-specific alcohol consumption and to evaluate whether dietary and nondietary factors modify the association. Data Sources.— We included in these analyses 6 prospective studies that had at least 200 incident breast cancer cases, assessed long-term intake of food and nutrients, and used a validated diet assessment instrument. The studies were conducted in Canada, the Netherlands, Sweden, and the United States. Alcohol intake was estimated by food frequency questionnaires in each study. The studies included a total of 322647 women evaluated for up to 11 years, including 4335 participants with a diagnosis of incident invasive breast cancer. Data Extraction.— Pooled analysis of primary data using analyses consistent with each study's original design and the random-effects model for the overall pooled analyses. Data Synthesis.— For alcohol intakes less than 60 g/d (reported by >99% of participants), risk increased linearly with increasing intake; the pooled multivariate relative risk for an increment of 10 g/d of alcohol (about 0.75-1 drink) was 1.09 (95% confidence interval [CI], 1.04-1.13; P for heterogeneity among studies, .71). The multivariate-adjusted relative risk for total alcohol intakes of 30 to less than 60 g/d (about 2-5 drinks) vs nondrinkers was 1.41 (95% CI, 1.18-1.69). Limited data suggested that alcohol intakes of at least 60 g/d were not associated with further increased risk. The specific type of alcoholic beverage did not strongly influence risk estimates. The association between alcohol intake and breast cancer was not modified by other factors. Conclusions.— Alcohol consumption is associated with a linear increase in breast cancer incidence in women over the range of consumption reported by most women. Among women who consume alcohol regularly, reducing alcohol consumption is a potential means to reduce breast cancer risk.
Article
Chronic, nonhealing wounds result in patient morbidity and disability. Reactive oxygen species (ROS) and nitric oxide (NO) are both required for normal wound repair and derangements of these result in impaired healing. Xanthine oxidoreductase (XOR) has the unique capacity to produce both ROS and NO. We hypothesize that XOR contributes to normal wound healing. Cutaneous wounds were created in C57Bl6 mice. XOR was inhibited with dietary tungsten or allopurinol. Topical hydrogen peroxide (H2O2, 0.15%) or allopurinol (30ug) was applied to wounds every other day. Wounds were monitored until closure or collected at day 5 to assess XOR expression and activity, cell proliferation, and for histology. The effects of XOR, nitrite, H2O2, and allopurinol on keratinocyte (KC) and endothelial cell (EC) behavior were assessed. We identified XOR expression and activity in the skin and wound edges as well as granulation tissue. Cultured human KCs also expressed XOR. Tungsten significantly inhibited XOR activity and impaired healing with reduced ROS production with reduced angiogenesis and KC proliferation. The expression and activity of other tungsten sensitive enzymes were minimal in the wound tissues. Oral allopurinol did not reduce XOR activity or alter wound healing but topical allopurinol significantly reduced XOR activity and delayed healing. Topical H2O2 restored wound healing in tungsten fed mice. In vitro, nitrite and H2O2 both stimulated KC and EC proliferation and EC migration. These studies demonstrate for the first time that XOR is abundant in wounds and participates in normal wound healing through effects on ROS production.