Sei sulla pagina 1di 6

Joint Bone Spine 74 (2007) 324e329

http://france.elsevier.com/direct/BONSOI/

Review

Reactive oxygen species and superoxide dismutases:


Role in joint diseases
Valery Afonso, Romuald Champy, Dragoslav Mitrovic, Pascal Collin, Abderrahim Lomri*
INSERM U606, IFR-139, Paris 7 University, Lariboisie`re Teaching Hospital, 2 rue Ambroise Pare, 75475 Paris Cedex 10, France
Received 9 May 2006; accepted 1 February 2007
Available online 24 May 2007

Abstract
Reactive oxygen species (ROS) are produced in many normal and abnormal processes in humans, including atheroma, asthma, joint diseases,
aging, and cancer. The superoxide anion O2 is the main ROS. Increased ROS production leads to tissue damage associated with inflammation.
Superoxide dismutases (SODs) convert superoxide to hydrogen peroxide, which is then removed by glutathione peroxidase or catalase. Thus,
SODs prevent the formation of highly aggressive ROS, such as peroxynitrite or the hydroxyl radical. Experimental models involving SOD
knockout or overexpression are beginning to shed light on the pathophysiological role of SOD in humans. Although the antiinflammatory effects
of exogenous native SOD (orgotein) are modest, synthetic SOD mimetics hold considerable promise for modulating the inflammatory response.
In this review, we discuss new knowledge about the role of the superoxide anion and its derivates as mediators of inflammation and the
role of SODs and SOD mimetics as antioxidant treatments in joint diseases such as rheumatoid arthritis, osteoarthritis, and crystal-induced
arthropathies.
2007 Elsevier Masson SAS. All rights reserved.


Keywords: Reactive oxygen species; Superoxide anion; Superoxide dismutase; SOD mimetics; Arthritis; Rheumatoid arthritis; Crystal-induced arthropathies

1. Introduction
Reactive oxygen species (ROS) are normal by-products of
cellular metabolism. Overproduction of ROS and their derivatives occurs in a number of diseases [1]. Among ROS, the superoxide anion (O2) plays a pivotal role in inflammation,
particularly in patients with inflammatory joint disease [2].
The enzyme superoxide dismutase (SOD) neutralizes O2 by
transforming it into hydrogen peroxide (H2O2), thereby preventing the formation of highly aggressive compounds such
as peroxynitrite (ONOO) and hydroxyl radical (HO ).
In many joint diseases, proinflammatory factors such as cytokines and prostaglandins are released at sites of inflammation,
together with ROS [3] and nitric oxide (NO) [4]. These factors
are associated with very low SOD concentrations in joint
fluid [5]. Studies involving assays of nitrotyrosine residues in


* Corresponding author. Tel.: 33 149 956 328; fax: 33 149 958 452.
E-mail address: lomri@larib.inserm.fr (A. Lomri).

synovial tissues from patients with rheumatoid arthritis (RA)


[6] or exposure of chondrocytes to synthetic peroxynitrite in
vitro [7] have established that the combination of the superoxide
anion to NO causes cartilage damage. Further evidence of the
deleterious effects of O2 comes from a study in which intraarticular injections of native SOD (bovine orgotein) produced
greater clinical improvements than did intraarticular aspirin in
patients with RA involving the knee [8]. Experiments involving
SOD knockout and overexpression in mouse models of arthritis
have confirmed the ability of SOD to protect against the harmful
effects of O2 [9,10]. Several SOD mimetics have been developed as therapeutic tools for reducing inflammation while minimizing side effects [11].
SOD activity is a key component of the cellular antioxidant
armamentarium that protects cells and the extracellular matrix
(ECM) from the harmful effects of O2 and its derivatives. In
this review, we discuss new insights into the roles for ROS and
SOD in joint disease, as well as the potential therapeutic usefulness of SOD mimetics.

1297-319X/$ - see front matter 2007 Elsevier Masson SAS. All rights reserved.
doi:10.1016/j.jbspin.2007.02.002

V. Afonso et al. / Joint Bone Spine 74 (2007) 324e329

325

2. Reactive oxygen species


ROS are atoms or small molecules that have unpaired valence shell electrons. They readily accept another electron or
transfer their unpaired electron to another molecule. ROS
are normal by-products of cellular metabolism (Fig. 1). However, alterations in the amount and nature of released ROS occur in various disease states [12]. Reactivity varies widely
from one ROS to the next. Among ROS produced by living
cells, O2 is a proinflammatory compound that damages cells
and the ECM. For instance, O2 damages endothelial cells,
increasing the permeability of the microvasculature and promoting the migration of neutrophils to foci of inflammation.
O2 can be converted to other, more aggressive, ROS such
as the hydroxyl radical HO , which is produced when O2 interacts with free iron or copper ions. These ions are normally
present in tiny amounts in healthy individuals, as they are sequestered by specialized proteins such as ferritin, so that virtually no HO is produced. O2 can combine with NO to produce
peroxynitrite (ONOO), thereby modifying the bioavailability
of NO (Fig. 2). Hydrogen peroxide (H2O2) can diffuse in the
cytoplasm and cross cell membranes. Some ROS act as mediators that regulate cell functions such as proliferation and apoptosis, by influencing intracellular signaling pathways via
effects on gene expression [13]. Other ROS, including peroxyl
radicals (ROO ) and hydroxyl radical (HO ), are highly aggressive: they fragment DNA [14], lipid structures [15], and matrix
components [16]; and they cause oxidative changes to proteins
that result in chemical fragmentation or increased vulnerability to proteases [17,18].
ROS concentrations are governed by the balance between
the production of ROS and their elimination by antioxidants.
An appropriate balance is crucial to normal cell and tissue
function (Fig. 3). ROS are produced during many metabolic


Fig. 2. Superoxide anion (O2) and its derivates.




processes including mitochondrial respiration and enzyme


activities (cytochrome P-450, NADPH oxidase, myeloperoxidase, NO synthase, and xanthine oxidase) (Fig. 4). Antioxidant
enzymes that scavenge ROS are ubiquitously distributed in the
body; they include the superoxide dismutases (SODs), glutathione peroxidase, and catalase. In addition, hydrosoluble
antioxidants (glutathione, vitamin C, and uric acid) and liposoluble antioxidants (vitamin E, carotenoids, and bilirubin)
are particularly important for protecting cell membranes and
plasma lipoproteins. Antioxidants are substances that compete
effectively with other oxidizable substrates even when present
in low concentrations, thereby protecting other substrates from
the damaging effects of ROS.
3. The superoxide dismutases
SOD catalyses the dismutation of O2 to dioxygen (O2) and
H2O2:


O2 O2 2H / H2 O2 O2


H2O2 is eliminated by glutathione peroxidase or catalase.


Glutathione peroxidase also metabolizes the hydroperoxides
generated by peroxidation of polyunsaturated fatty acids (linoleic acid, linolenic acid, arachidonic acid). Alterations in the

Fig. 1. Pathways for the production and removal of reactive oxygen species.
GSH, glutathione; GSSG, oxidized glutathione; GSR, glutathione reductase;
XO, xanthine oxidase.

Fig. 3. Superoxide anion (O2) and superoxide dismutases (SODs): endogenous SODs are crucial for controlling O2 levels. In inflammatory conditions,
the amount of O2 produced exceeds the ability of SOD to remove O2.


V. Afonso et al. / Joint Bone Spine 74 (2007) 324e329

326

Fig. 4. Extra- and intracellular sources of reactive oxygen species. XO, xanthine oxidase; P-450, cytochrome P-450.

activity of these enzymes may lead to oxidative stress. The


biochemistry and molecular structure of three SOD isoforms
found in different body compartments have been characterized
in humans. Cu/Zn-SOD or SOD1 is found in the cytoplasm,
whereas EC-SOD or SOD3 is extracellular. Both isoforms
use copper and zinc as cofactors. Manganese is the cofactor
for SOD2, which is found in mitochondria. Excellent reviews
of many studies into the physiological functions and O2-controlling role of the three SOD isoforms are available [19,20].


3.1. Structure and function of SOD1


The gene encoding SOD1 is located on chromosome 21.
SOD1 is found in the cytoplasm, nucleus, and intermembrane
space of mitochondria. Studies of the human SOD1 promoter
have identified several binding sites for transcription factors
modulated by the redox status of the cell, including AP-1
and NF-kB. SOD1 overexpression extends the life span of
Drosophila melanogaster [21]. In mice, SOD1 overexpression
is associated with protection of cerebral tissues from damage
associated with ischemia or Parkinsons disease [22]. SOD1
knockout is not lethal in mice [23] but leads to a marked decrease in cell division in vitro [24], with diminished resistance
to O2. In humans, SOD1 mutations are responsible for neurodegenerative diseases (e.g., amyotrophic lateral sclerosis)
associated with oxidative damage [25]. In mice, SOD1 mutations are associated with increased apoptosis and oxidative
protein damage. SOD1 plays a central role in cell survival
and growth. Studies showing that SOD1 transcripts increase
in response to various stimuli such as heat shock, shear stress,
heavy metals, and oxidative stress indicate that this enzyme is
involved in cell responses to various sources of stress.
Little is known about factors that inhibit SOD1. We studied
the role of proinflammatory cytokines in inhibiting cellular
antioxidant defense mechanisms. TNF-a overproduction in inflammatory conditions is associated with increased oxidative


stress consistent with inhibition of antioxidant enzymes, most


notably decreased SOD1 expression. We showed that TNF-a inhibited SOD1 expression via an effect on the JNK/AP-1 proapoptotic signaling pathway [26]. TNF-a-induced repression of
SOD1 explains the presence of O2 and its derivatives in inflammatory conditions.


3.2. Structure and function of SOD2


The gene encoding SOD2 has been mapped to chromosome
6. SOD2 knockout in mice results in lethal cardiomyopathy
and neurodegeneration. SOD2 is found in mitochondria and
provides vital protection against ROS generated by hyperoxia.
SOD2 deficiency results in increased levels of mitochondrial
O2, which inhibit the respiratory chain complexes I and II
[27]. Numerous polymorphisms associated with reduced
SOD2 activity have been identified as risk factors for cardiomyopathy. SOD2 overexpression in transgenic animals was associated with a reduction in infarct size and with protection of
tissues against apoptosis [28].
Proinflammatory cytokines such as interleukin-1 (IL-1), IL4, IL-6, and TNF-a are potent SOD2 activators. The promoter
region involved in SOD2 activation contains sites that bind to
transcription factors belonging to the NF-kB, C/EBP, and NF1 families [29].


3.3. Structure and function of SOD3


The SOD3 gene is located on chromosome 4. SOD3 exhibits strong affinity for heparin and other proteoglycans in
the ECM and plasma membrane. It is found chiefly in extracellular compartments (plasma, lymph, cerebrospinal fluid, and
joint fluid) [30,31]. SOD3 protects cells and the ECM against
the harmful effects of O2 generated by activated neutrophils.
Inflammatory cells release proteases belonging to the proprotein convertase family (furin), which cleave the SOD3 from


V. Afonso et al. / Joint Bone Spine 74 (2007) 324e329

the ECM, exposing the matrix to damage from ROS and increasing plasma SOD3 levels [32]. The only SOD3 mutation
identified to date affects the heparin-binding site (R213G)
and increases the risk of cardiovascular disease [33].
4. Potential role of oxidative stress in joint disease
4.1. Oxidative stress and osteoarthritis
The cartilage matrix undergoes considerable alterations in
structure, molecular composition, and mechanical properties
during aging. Surface fibrillation, proteoglycan structure and
composition changes, and increased collagen breakdown are
examples of these alterations. The cartilage loses tensile resistance and stiffness. IL-1b, one of the most active factors involved in osteoarthritis [34], diminishes the expression of
type 2 collagen and aggrecan, and increases the expression
of metalloproteases (MMP) 1 and 3. IL-1b stimulates NO production, leading to the formation of peroxynitrite, which targets guanine repeats in DNA telomeres, explaining the link
between oxidative stress and telomere erosion [35]. Aging of
cartilage and chondrocytes may be central to the pathogenesis
and progression of osteoarthritis.
ROS are highly reactive compounds with a short half-life.
However, nitrotyrosine, formed when tyrosine is oxidized in
the presence of nitrous oxide (NOO), can serve as a measure
of oxidative damage in vivo. Presence of nitrotyrosine in cartilage was associated with older age and with osteoarthritis,
suggesting a role of oxidative stress in cartilage aging and degeneration [36]. Thus, oxidative stress affects chondrocyte
function in joint cartilage.
4.2. Oxidative stress and rheumatoid arthritis
Although the exact causes of RA are unknown, involvement of ROS is suspected. Several studies suggest a beneficial
effect of antioxidants such as vitamin E in RA. At foci of
inflammation, activation of T cells and macrophages leads to
a large increase in oxygen consumption, whose corollary is increased release of ROS.
In vitro, peroxynitrite formation is associated with decreased production of type 2 collagen and aggrecan and with
a diminished chondrocyte response to the growth factor IGF1. In addition, peroxynitrite increases the expression of
MMP-3 and MMP-13 and decreases the production and activity of the tissue inhibitors of MMPs (TIMPs) [3]. Taken in
concert, these changes lead to increased matrix breakdown.
TNF-a overproduction is thought to be the main contributor
to increased ROS release in patients with RA. TNF-a not only
causes cell damage, but also inhibits SOD1 [26] and SOD3
[37]. The introduction of TNF-a antagonists has proved
a breakthrough in the treatment of RA. Recent work reports
that infliximab protects extracellular SOD3 from inhibition
by TNF-a in vitro [38].
SOD3 accounts for 80% of the enzyme activity of joint
fluid [30]. Studies of joint fluid from patients with RA showed

327

a 50% decrease in SOD3 activity, which may increase the vulnerability of synovial tissue to ROS-induced damage.
4.3. Crystal-induced arthropathies
Microcrystals contribute to the development of degenerative
joint disease. Gout constitutes the archetype of crystal-induced
arthropathies. Sodium urate crystals present within the joint
space cause acute synovial inflammation and contribute in
the long term to cause cartilage breakdown and bone lesions.
Several proinflammatory cytokines (TNF-a, IL-1, IL-6, and
IL-8) are produced by synovial cells and recruit neutrophils
to the joint fluid [39]. These cytokines exert their effects via activation of intracellular signaling pathways. One of these pathways is Syk phosphorylation, which is central to neutrophil
activation, an event associated with O2 release [40]. Urate
crystal phagocytosis by neutrophils is associated with the release of lysosomal enzymes and the production of large
amounts of ROS, prostaglandins, and LTB4 leukotriene, which
are involved in activation of the inflammatory response.
Other crystals, such as basic calcium phosphate (BCP) crystals, have been found in joint fluid in a variety of situations
ranging from the asymptomatic joint to acute arthritis and destructive arthritis [41]. BCP crystals, of which apatite is the best
known, are found in bone as hydroxyapatite and carbonated apatite. They also include octacalcium phosphate, tricalcium
phosphate, and whitlockite. The presence of BCP crystals has
been reported in many degenerative joint diseases and may correlate with disease severity. BCP crystals have been identified
in symptomatic and asymptomatic extraarticular deposits.
Proinflammatory potency and responses of synovial fibroblasts
and chondrocytes vary across crystals. Octacalcium phosphate
crystals chiefly induce TNF-a production [42], which inhibits
SOD1, leading to ROS build-up. However, a balance exists in
these situations between the production of proinflammatory cytokines and the production of their inhibitors.
Calcium-containing microcrystals stimulate the production
of TNF-a by many cell types including monocytes, chondrocytes, and probably synovial fibroblasts. BCP crystals activate
these cells to produce MMP-1 and MMP-3 via the enzyme
protein-kinase C, as well as MMP-8 and MMP-13 [43]. We
found evidence that hydroxyapatite and carbonated apatite
may decrease SOD1 expression by inhibiting the NF-kB pathway (Champy et al., unpublished data). Other studies have established that microcrystals enhance TNF-a production and
the JNK/AP-1 signaling pathway, SOD1 repression by microcrystals may be mediated directly by TNF-a, the result being
O2 release in the joint cavity.


5. Beneficial effects of SOD and SOD


mimetics in joint disease
5.1. SOD in joint disease
SODs exert protective effects in animal models of ischemia
and inflammation [22]. In mice that are genetically deficient in
SOD3, both the severity of collagen-induced arthritis and the

V. Afonso et al. / Joint Bone Spine 74 (2007) 324e329

328

production of proinflammatory cytokines are increased [9].


SOD3 gene transfer via the subcutaneous route [10] or into
the knee decreased the severity of experimental arthritis in
rodents [44].
In humans, serum SOD3 levels correlated negatively with
disease activity [45]. Studies of orgotein in RA and osteoarthritis yielded conflicting results [8,46]. Native bovine SOD
was associated with adverse effects such as immune responses
to the enzyme, pain, and burning sensations. As a result, native
bovine SOD was removed from the market.
In aggregate, available data suggest a protective role of
SOD in inflammatory joint disease.
5.2. SOD mimetics and joint disease

Fig. 5. Protective effects of SOD on cartilage.

Because O2 dismutation by SOD affects the course of inflammation, and to overcome the problems encountered with
native bovine SOD, several groups have developed synthetic
low-molecular-weight compounds that mimic the effects of
SOD. Among the various families of SOD mimetics available
today [11], the most promising are nitroxides (tempol) and
Mn(II) pentaazamacrocyclic ligand (M40403). Tempol is a soluble derivate of tempo used in electron-spin-resonance spectroscopy to detect O2. Its low molecular weight allows it to
cross membranes. Tempol has shown beneficial effects in animal models of inflammation, hypertension, diabetes, and endothelial cell dysfunction [47]. Tempol attenuates O2 effects in
vitro, diminishes hydroxyl radical production, and decreases
the cytotoxic effects of hydrogen peroxide and peroxynitrite.
Tempol decreased tissue inflammation and damage in a study
of rats with collagen-induced arthritis [48].
Among manganese-based compounds, M40403 is characterized by excellent stability and a very high level of SOD activity
[49]. M40403 increases the rate of O2 conversion to O2 and
H2O2. It seems specific of O2 and does not react with hydrogen
peroxide, peroxynitrite, or NO. The potent antiinflammatory
effects of M40403 are related to O2 elimination, which prevents peroxynitrite formation and therefore nitration of tyrosine
residues in proteins. By diminishing the expression of adhesion
molecules such as ICAM or P-selectin, M40403 decreases the
influx of neutrophils to inflammatory sites. M40403 decreases
the release of proinflammatory agents such as TNF-a, probably
by blocking the expression of the transcription factor NF-kB
[44]. Beneficial effects of M40403 have been reported in rats
with collagen-induced arthritis [11,50].


formation leading to greater bioavailability of NO (Fig. 5),


decreased influx of neutrophils to sites of inflammation, and
decreased release of proinflammatory cytokines. Thus, medications that target ROS-induced damage to joint cartilage may
deserve to be included in therapeutic strategies designed to
combat the development and progression of inflammatory joint
disease.

Acknowledgments

We are grateful to the French Society for Rheumatology


(research grant 2004 awarded to R Champy) and to the nonprofit organization Association Rhumatisme et Travail (research grant 2005) for their financial support.

6. Conclusion
The body of available data points to several conclusions.
First, joint diseases are associated with large increases in the
production of ROS including O2, NO, and their derivates.
Second, TNF-a overproduction in joint disease substantially
diminishes the activity of SODs and other antioxidant
enzymes. SOD mimetics exert beneficial effects in various conditions including joint disease. O2 elimination via the administration of SOD mimetics may attenuate the inflammatory
process via three main mechanisms: decreased peroxynitrite


References
[1] Halliwell B, Gutteridge JM, Cross CE. Free radicals, antioxidants and
human disease. Where are we now? J Lab Clin Med 1992;119:598e620.
[2] Henrotin Y, Kurz B, Aigner T. Oxygen and reactive oxygen species in
cartilage degradation: friends or foes? Osteoarthritis Cartilage
2005;13:643e54.
[3] Henrotin YE, Bruckner P, Pujol JP. The role of reactive oxygen species in
homeostasis and degradation of cartilage. Osteoarthritis Cartilage
2003;11:747e55.
[4] Sakurai H, Kohsaka H, Liu MF, Higashiyama H, Hirata Y, Kanno K,
et al. Nitric oxide production and inducible nitric oxide synthase expression in inflammatory arthritides. J Clin Invest 1995;96:2357e63.
[5] Marklund SL, Bjelle A, Elmqvist LG. Superoxide dismutase isoenzymes
of the synovial fluid in rheumatoid arthritis and in reactive arthritides.
Ann Rheum Dis 1986;45:847e51.
[6] Sandhu JK, Robertson S, Birnboim HC, Goldstein R. Distribution of protein nitrotyrosine in synovial tissues of patients with rheumatoid arthritis
and osteoarthritis. J Rheumatol 2003;30:1173e81.
[7] Mathy-Hartert M, Martin G, Devel P, Deby-Dupont G, Pujol JP,
Reginster JY, et al. Reactive oxygen species downregulate the expression
of pro-inflammatory genes by human chondrocytes. Inflamm Res
2003;52:111e8.
[8] Goebel KM, Storck U, Neurath F. Intrasynovial orgotein therapy in rheumatoid arthritis. Lancet 1981;1:1015e7.
[9] Ross AD, Banda NK, Muggli M, Arend WP. Enhancement of collageninduced arthritis in mice genetically deficient in extracellular superoxide
dismutase. Arthritis Rheum 2004;50:3702e11.

V. Afonso et al. / Joint Bone Spine 74 (2007) 324e329


[10] Iyama S, Okamoto T, Sato T, Yamauchi N, Sato Y, Sasaki K, et al. Treatment of murine collagen-induced arthritis by ex vivo extracellular superoxide dismutase gene transfer. Arthritis Rheum 2001;44:2160e7.
[11] Salvemini D, Cuzzocrea S. Therapeutic potential of superoxide dismutase mimetics as therapeutic agents in critical care medicine. Crit Care
Med 2003;31(Suppl. 1):S29e38.
[12] Halliwell B, Gutteridge JM. Role of free radicals and catalytic metal ions
in human disease: an overview. Methods Enzymol 1990;186:1e85.
[13] Haddad JJ. Antioxidant and prooxidant mechanisms in the regulation of
redox(y)-sensitive transcription factors. Cell Signal 2002;14:879e97.
[14] Wiseman H, Halliwell B. Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer.
Biochem J 1996;313:17e29.
[15] Bruckdorfer KR. Lipid oxidation products and vascular function. Free
Radic Res 1998;28:573e81.
[16] Tiku ML, Gupta S, Deshmukh DR. Aggrecan degradation in chondrocytes is mediated by reactive oxygen species and protected by antioxidants. Free Radic Res 1999;30:395e405.
[17] Davies KJ. Protein damage and degradation by oxygen radicals. I. general aspects. J Biol Chem 1987;262:9895e901.
[18] Stadtman ER. Oxidation of proteins by mixed-function oxidation systems: implication in protein turnover, aging and neutrophil function.
Trends Biochem Sci 1986;11:11e2.
[19] Johnson F, Giulivi C. Superoxide dismutases and their impact upon
human health. Mol Aspects Med 2005;26:340e52.
[20] McCord JM, Edeas MA. SOD, oxidative stress and human pathologies:
a brief history and a future vision. Biomed Pharmacother 2005;59:
139e42.
[21] Orr WC, Mockett RJ, Benes JJ, Sohal RS. Effects of overexpression of
copperezinc and manganese superoxide dismutases, catalase, and thioredoxin reductase genes on longevity in Drosophila melanogaster. J Biol
Chem 2003;278:26418e22.
[22] Fujimura M, Morita-Fujimura Y, Noshita N, Sugawara T, Kawase M,
Chan PH. The cytosolic antioxidant copper/zinc-superoxide dismutase
prevents the early release of mitochondrial cytochrome c in ischemic
brain after transient focal cerebral ischemia in mice. J Neurosci
2000;20:2817e24.
[23] Reaume AG, Elliott JL, Hoffman EK, Kowall NW, Ferrante RJ,
Siwek DF, et al. Motor neurons in Cu/Zn superoxide dismutase-deficient
mice develop normally but exhibit enhanced cell death after axonal
injury. Nat Genet 1996;13:43e7.
[24] Huang TT, Yasunami M, Carlson EJ, Gillespie AM, Reaume AG,
Hoffman EK, et al. Superoxide-mediated cytotoxicity in superoxide dismutase-deficient fetal fibroblasts. Arch Biochem Biophys 1997;344:
424e32.
[25] Okado-Matsumoto A, Fridovich I. Amyotrophic lateral sclerosis: a proposed mechanism. Proc Natl Acad Sci U S A 2002;99:9010e4.
[26] Afonso V, Santos G, Collin P, Khatib AM, Mitrovic D, Lomri N, et al. TNFa down-regulates human Cu/Zn superoxide dismutase 1 promoter via JNK/
AP-1 signaling pathway. Free Radic Biol Med 2006;41:709e21.
[27] Morten KJ, Ackrell BA, Melov S. Mitochondrial reactive oxygen species
in mice lacking superoxide dismutase 2: attenuation via antioxidant treatment. J Biol Chem 2006;281:3354e9.
[28] Wheeler MD, Nakagami M, Bradford BU, Uesugi T, Mason RP,
Connor HD, et al. Overexpression of manganese superoxide dismutase
prevents alcohol-induced liver injury in the rat. J Biol Chem 2001;276:
36664e72.
[29] Kiningham KK, Xu Y, Daosukho C, Popova B, St Clair DK. Nuclear factor kappaB-dependent mechanisms coordinate the synergistic effect of
PMA and cytokines on the induction of superoxide dismutase 2. Biochem
J 2001;353:147e56.
[30] Marklund SL. Human copper-containing superoxide dismutase of high
molecular weight. Proc Natl Acad Sci U S A 1982;79:7634e8.
[31] Fattman CL, Schaefer LM, Oury TD. Extracellular superoxide dismutase
in biology and medicine. Free Radic Biol Med 2003;35:236e56.
[32] Enghild JJ, Thogersen IB, Oury TD, Valnickova Z, Hojrup P, Crapo JD.
The heparin-binding domain of extracellular superoxide dismutase is

[33]

[34]
[35]

[36]

[37]

[38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

329

proteolytically processed intracellularly during biosynthesis. J Biol


Chem 1999;274:14818e22.
Juul K, Tybjaerg-Hansen A, Marklund S, Heegaard NH, Steffensen R,
Sillesen H, et al. Genetically reduced antioxidative protection and increased ischemic heart disease risk: the Copenhagen City Heart Study.
Circulation 2004;109:59e65.
Martel-Pelletier J, Alaaeddine N, Pelletier JP. Cytokines and their role in
the pathophysiology of osteoarthritis. Front Biosci 1999;4:694e703.
Yudoh K, Nguyen T, Nakamura H, Hongo-Masuko K, Kato T,
Nishioka K. Potential involvement of oxidative stress in cartilage senescence and development of osteoarthritis: oxidative stress induces chondrocyte telomere instability and downregulation of chondrocyte
function. Arthritis Res Ther 2005;7:380e91.
Loeser RF, Carlson CS, Del Carlo M, Cole A. Detection of nitrotyrosine
in aging and osteoarthritic cartilage: correlation of oxidative damage with
the presence of interleukin-1beta and with chondrocyte resistance to insulin-like growth factor 1. Arthritis Rheum 2002;46:2349e57.
Stralin P, Marklund SL. Multiple cytokines regulate the expression of extracellular superoxide dismutase in human vascular smooth muscle cells.
Atherosclerosis 2000;151:433e41.
Adachi T, Toishi T, Takashima E, Hara H. Infliximab neutralizes the suppressive effect of TNF-alpha on expression of extracellular-superoxide
dismutase in vitro. Biol Pharm Bull 2006;29:2095e8.
Terkeltaub R, Zachariae C, Santoro D, Martin J, Peveri P, Matsushima K.
Monocyte-derived neutrophil chemotactic factor/interleukin-8 is a potential mediator of crystal-induced inflammation. Arthritis Rheum
1991;34:894e903.
Liu-Bryan R, Liote F. Monosodium urate and calcium pyrophosphate dihydrate (CPPD) crystals, inflammation, and cellular signaling. Joint Bone
Spine 2005;72:295e302.
McCarty DJ, Lehr JR, Halverson PB. Crystal populations in human synovial fluid. Identification of apatite, octacalcium phosphate, and tricalcium phosphate. Arthritis Rheum 1983;26:1220e4.
Prudhommeaux F, Schiltz C, Liote F, Hina A, Champy R, Bucki B, et al.
Variation in the inflammatory properties of basic calcium phosphate crystals according to crystal type. Arthritis Rheum 1996;39:1319e26.
Reuben PM, Brogley MA, Sun Y, Cheung HS. Molecular mechanism of
the induction of metalloproteinases 1 and 3 in human fibroblasts by basic
calcium phosphate crystals. Role of calcium-dependent protein kinase C
alpha. J Biol Chem 2002;277:15190e8.
Dai L, Claxson A, Marklund SL, Feakins R, Yousaf N, Chernajovsky Y,
et al. Amelioration of antigen-induced arthritis in rats by transfer of
extracellular superoxide dismutase and catalase genes. Gene Ther
2003;10:550e8.
Ugur M, Yildirim K, Kiziltunc A, Erdal A, Karatay S, Senel K. Correlation between soluble intercellular adhesion molecule 1 level and extracellular superoxide dismutase activity in rheumatoid arthritis: a possible
association with disease activity. Scand J Rheumatol 2004;33:239e43.
Mazieres B, Masquelier AM, Capron MH. A French controlled multicenter study of intraarticular orgotein versus intraarticular corticosteroids in
the treatment of knee osteoarthritis: a one-year followup. J Rheumatol
Suppl 1991;27:134e7.
Cuzzocrea S, Riley DP, Caputi AP, Salvemini D. Antioxidant therapy:
a new pharmacological approach in shock, inflammation, and ischemia/
reperfusion injury. Pharmacol Rev 2001;53:135e59.
Cuzzocrea S, McDonald MC, Mota-Filipe H, Mazzon E, Costantino G,
Britti D, et al. Beneficial effects of tempol, a membrane-permeable radical scavenger, in a rodent model of collagen-induced arthritis. Arthritis
Rheum 2000;43:320e8.
Muscoli C, Cuzzocrea S, Riley DP, Zweier JL, Thiemermann C,
Wang ZQ, et al. On the selectivity of superoxide dismutase mimetics
and its importance in pharmacological studies. Br J Pharmacol 2003;
140:445e60.
Cuzzocrea S, Mazzon E, di Paola R, Genovese T, Muia C, Caputi AP,
et al. Synergistic interaction between methotrexate and a superoxide dismutase mimetic: pharmacologic and potential clinical significance.
Arthritis Rheum 2005;52:3755e60.

Potrebbero piacerti anche