You are on page 1of 12

Cancer and Metastasis Reviews 23: 311–322, 2004.

# 2004 Kluwer Academic Publishers. Manufactured in The Netherlands.

Metabolic oxidation/reduction reactions and cellular responses to ionizing


radiation: A unifying concept in stress response biology

Douglas R. Spitz1*, Edouard I. Azzam2, Jian Jian Li3 and David Gius4
1
B180 Medical Laboratories, Free Radical and Radiation Biology Program, Department of Radiation
Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242; 2Department of
Radiology, New Jersey Medical School, Newark, New Jersey 07103; 3Radiation Biology, Division of
Radiation Oncology, City of Hope National Medical Center, Duarte, California 91010; 4Radiation Oncology
Branch, Radiation Oncology Sciences Program, Center for Cancer Research, National Cancer Institute, NIH,
Bethesda, Maryland 20892

Key words: oxidative stress, redox regulation, metabolism, signal transduction, thioredoxin, superoxide
dismutase, non-targeted effects, mitochondria, cancer, aging

Summary

Exposure of eukaryotic cells to ionizing radiation (IR) results in the immediate formation of free radicals
that last a matter of milliseconds. It has been assumed that the subsequent alterations in multiple
intracellular processes following irradiation is due to the initial oxidative damage caused by these free
radicals. However, it is becoming increasingly clear that intracellular metabolic oxidation/reduction (redox)
reactions can be affected by this initial IR-induced free radical insult and may remain perturbed for
minutes, hours, or days. It would seem logical that these cellular redox reactions might contribute to the
activation of protective or damaging processes that could impact upon the damaging effects of IR. These
processes include redox sensitive signaling pathways, transcription factor activation, gene expression, and
metabolic activities that govern the formation of intracellular oxidants and reductants. The physiological
manifestations of these radiation-induced alterations in redox sensitive processes have been suggested to
contribute to adaptive responses, bystander effects, cell cycle perturbations, cytotoxicity, heat-induced
radiosensitization, genomic instability, inflammation, and fibrosis. While a great deal is known about the
molecular changes associated with the initial production of free radicals at the time of irradiation, the
contribution of perturbations in redox sensitive metabolic processes to biological outcomes following
exposure to IR is only recently becoming established. This review will focus on evidence supporting the
concept that perturbations in intracellular metabolic oxidation/reduction reactions contribute to the
biological effects of radiation exposure as well as new concepts emerging from the field of free radical
biology that may be relevant to future studies in radiobiology.

I. Historical perspective and lipids that contribute to the biological effects


of radiation [1–5]. In the presence of O2, the
It has been known for many years that exposure to radiation-induced free radicals that are formed
ionizing radiation (IR) results in the formation of initially are thought to include hydroxyl radical
free radicals in living systems that are believed to (OH?), superoxide (O?2 ), and organic radicals (R?)
persist for milliseconds and result in oxidative [1–3]. Immediately upon irradiation in the pre-
damage to biomolecules such as DNA, proteins, sence of O2, reactions of these free radicals gives

*Corresponding author.
E-mail: douglas-spitz@uiowa.edu
312 Spitz et al.

rise to other reactive oxygen species including extract, store, and move electrons through com-
hydrogen peroxide (H2O2) and organic hydroper- plex biological structures via a series of biochem-
oxides (ROOH). These hydroperoxide species ical oxidation/reduction (redox) reactions
together with redox active metal ions (such as Fe involving protein catalysts has been hypothesized
and Cu), commonly found in biological matrices, to provide the essential ‘life force’ for maintaining
are also believed to contribute to radiation- metabolic homeostasis in mammalian cells [23].
induced oxidative damage via Fenton type reac- This metabolic strategy, while extremely effi-
tions [3,4,6]. It has been hypothesized that the free cient, does lead to the formation of reactive
radical-mediated covalent modifications resulting oxygen species (ROS) as byproducts. Ideally the
from oxidative damage to critical biomolecules four-electron reduction of O2 to form H2O in
during and immediately (within 5 milliseconds) Complex IV of the mitochondrial electron trans-
following irradiation result in most, if not all, of port chain allows for the efficient removal of
the biological effects of ionizing radiation. electrons from the system. The flow of electrons,
The most compelling evidence in favor of this through Complexes I–IV, pumps protons across
hypothesis has come from observations that the inner mitochondrial membrane creating a
manipulations of antioxidants (i.e. thiols, super- proton gradient that is utilized to drive ATP
oxide dismutases, hydroxyl radical scavengers, and production by Complex V. However, there is the
hydroperoxide metabolizing enzyme systems) at probability that 1-electron reductions of O2 to
the time of irradiation appear to alter the reactions form O?2 will occur proximal to Complex IV,
of free radicals (and reactive oxygen species) probably at Complex I (NADH-dehydrogenase)
leading to alterations in oxidative damage as well or Complex III (ubiquinone-cytochrome b) of the
as alterations in the biological effects of IR [3,4,6– electron transport chain [24]. Superoxide then
15]. In addition, following irradiation, cells and rapidly dismutes to form H2O2 either sponta-
tissues appear to respond by increasing the neously or through the catalytic function of
expression of cellular antioxidant defenses and superoxide dismutases [24,25]. It is estimated
this increased antioxidant capacity has been during normal respiration in mammalian mito-
hypothesized to be at least partially responsible chondria that as much as 1% of O2 consumption
for radiation-induced adaptive responses [16–20]. could result in O?2 and H2O2 formation [24]. In
Taken together, this growing body of evidence addition to mitochondrial production of ROS,
clearly points to a causal relationship between there are many cellular enzymes involved in wide
radiation-induced free radical production at the variety of oxidative metabolic processes that also
time of irradiation and the long-term biological have the potential to generate ROS including
responses seen following exposure. NADPH oxidase enzymes, myeloperoxidase,
xanthine oxidase, amino acid oxidases, cyto-
chrome P450 enzymes, and several peroxisomal
II. Metabolic oxidative stress enzymes including flavoprotein dehydrogenase
enzymes involved in b-oxidation of fatty acids
For many years it has been known that metabo- and glycollate oxidase [25].
lism in mammalian cells primarily derives energy In complex living systems, metabolic production
from the tightly controled biochemical oxidation of O?2 and H2O2 would be expected to result in
of substrates (i.e. carbohydrates, fats, and amino OH? formation via the transition metal catalyzed
acids) to obtain reducing equivalents (electrons) Haber–Weiss reaction [25]. In the presence of O2,
necessary for mitochondrial electron transport OH? would be expected to abstract hydrogen
chain mediated oxidative phosphorylation to atoms from critical biomolecules leading to the
produce ATP, with O2 acting as the terminal formation of R? and ROOH, resulting in oxidative
electron acceptor [21,22]. In addition, reducing damage. In the presence of the aforementioned
equivalents in the form of NADH or NADPH are highly reactive species, lipid peroxidation chain
essential for many cellular biosynthetic processes reactions would also be expected to increase [25–
involved in replication, cell division, and macro- 27]. Since lipid peroxidation is a self propagating
molecular synthesis [22]. Therefore, the ability to process [25–27] that results in the formation of
Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation 313

diffusible cytotoxic byproducts (i.e. lipid hydro- Metabolic oxidative stress and the subsequent
peroxides, epoxides, endoperoxides, and lipid deleterious consequences are amenable to interven-
aldehydes), it could dramatically amplify the initial tion and this fact has generated a great deal of
number of reactive species produced by metabolic interest in the biomedical research community.
oxidative stress as well as the diffusion distance of When a cell ‘senses’ (see section below for details of
these species [25–27]. In this fashion, the metabolic redox sensitive signaling) shifts in metabolic
production of O?2 and H2O2 in living systems oxidation-reduction reactions, it can respond by:
could result in a spectrum of free radical reactions (1) shifting metabolism away from pathways
and oxidative damage similar (although at a much resulting in reactive species formation, (2) increas-
lower rate and in different cellular compartments) ing metabolic antioxidant capacity, and (3) increas-
to that postulated for ionizing radiation. This ing pathways that repair oxidative damage to
concept has been invoked in the development of critical biomolecules. This can be accomplished
the Free Radical Theory of Oxygen Toxicity immediately by transient activation or inactivation
[28,29], the Free Radical Theory of Aging of redox sensitive proteins regulating these path-
[30,31], and the Free Radical Theory of Cancer ways and by the induction of gene expression
[32,33]. If free radical damage derived from pathways responsible for synthesizing new pro-
metabolism were responsible for the pathological teins. These pathways might also be manipulated
changes seen during aging, this theoretical con- via exogenous addition of: (1) small molecular
struct might also explain why many of the same weight dietary antioxidants and/or synthetic anti-
biological phenomena seen to increase as a oxidant enzymes, (2) substrates for the regenera-
function of aging (i.e. carcinogenesis, atherosclero- tion of reducing equivalents necessary for
sis, inflammatory diseases, fibrotic changes in antioxidant pathways, and (3) genetic manipula-
normal tissues) are also induced by exposure to tions to enforce over expression of antioxidant
ionizing radiation. enzymes can be accomplished to limit the effects of
The production of potentially deleterious proox- metabolic oxidative stress. In this regard, manip-
idants by oxidative metabolism is held in check by ulations of pathways governing metabolic oxida-
a complex and interdependent system of antiox- tive stress might also be utilized to decrease
idants in order to maintain the intracellular antioxidant capacity and/or increase prooxidant
environment in a highly reduced steady state that production to improve the cytotoxic response of
is believed to fluctuate between redox potentials cancer cells to therapeutic intervention. These
about  200 mV to  240 mV [34]. These antiox- various strategies are currently under investigation
idants include lipid- and water-soluble small for a wide variety of degenerative diseases asso-
molecular weight dietary antioxidants (i.e. vitamin ciated with aging, cancer, and inflammatory
E, vitamin C, lipoic acid, b-carotene, etc.), small diseases thought to involve metabolic oxidative
molecular weight peptides and cofactors, (i.e. stress [36–42].
glutathione, NADPH, pyruvate, thioredoxin, glu-
taredoxin, etc.) and antioxidant enzymes (i.e.
catalase, superoxide dismutases, glutathione per- III. Metabolic oxidation/reduction reactions and
oxidases, peroxiredoxins, glutathione transferases, cellular communication
etc.) [25,35]. When prooxidant production begins
to exceed the ability of the cellular antioxidant Reactive species formed via metabolic activity can
capacity to maintain the normal steady state redox also act as second messengers in cell signaling
potential, a condition of oxidative stress exists [35]. cascades leading to shifts in gene expression
If oxidative stress persists for prolonged periods, patterns [43–49]. It is thought that this type of
oxidative damage will accumulate in biomolecules. redox regulated signaling could function to co-
If the repair of oxidative damage cannot compen- ordinately regulate the availability of metabolic
sate for the increased production of oxidatively substrates with the orderly transition of cellular
damaged biomolecules then injury, mutagenesis, functions [33,50–56]. In general protein kinases are
carcinogenesis, accelerated senescence, and cell activated by oxidation reactions, while protein
death can occur. phosphatases and zinc finger proteins are inacti-
314 Spitz et al.

vated by oxidation reactions and transcription changes in the local flux of reactive species that
factor binding is increased by reduction reactions could be sensed by redox sensitive signaling
[43–60]. Redox sensitive changes in signaling molecules, leading to alterations in gene expression
proteins are for the most part reversible (in pathways that provide the newly synthesized
keeping with their proposed regulatory role). proteins necessary to transition between different
This property allows for the binary type ‘on-off’ non-equilibrium steady states.
switching believed to be necessary for the constant
readjustment of metabolic activity to match
changes in cellular function necessary to maintain IV. Metabolic oxidation/reduction reactions and
a viable non-equilibrium steady state in living cellular radiation response
systems.
Typically the changes in the steady state Clearly, radiation-induced production of free
intracellular redox environment that occur during radicals/ROS during the time of exposure con-
normal fluctuations in metabolism or environmen- tributes to oxidative damage as well as radiation
tal stress are sensed by the movement of electrons response. Scavenging the reactive species produced
to and from redox sensitive moieties (i.e. redox at the time of irradiation with antioxidants can
sensitive thiol residues and metal ions) on proteins mitigate some of the effects of IR-induced injury;
involved with the initiation of signal transduction. but this is clearly limited by the accessibility of the
These redox changes on specific proteins can result antioxidants to the site of IR-induced free radical
in altered conformation and/or activity that can production as well as the rate constants of the
initiate signaling cascades resulting in altered reactive species interacting with critical biomole-
binding affinity, functional activity, and/or gene cules. However, it is also becoming clear that if
expression. The concept that subtle changes in radiation damage includes disruption of critical
redox sensitive moieties alters protein function biomolecules governing the metabolic production
seems logical given that most of the energy and of prooxidant species, metabolic oxidative stress
reducing equivalents necessary for maintenance of can also contribute to the biological effects of IR
homeostasis and responses to stress are derived long after the time of exposure. As a result of this
from metabolic oxidation/reduction reactions. In new concept and the historical understanding of
this way subtle alterations in electron flow through how free radical production contributes to IR-
redox sensitive signaling circuitry, within and induced damage, a new model of how metabolic
between proteins, could provide a rapidly respon- oxidation/reduction reactions contribute to IR
sive, freely reversible, and direct mechanism to effects following exposure can now be considered.
coordinately couple metabolic activity and gene Two pioneering studies implicating oxidative
expression processes during transitions between stress in IR response were done by Oberley’s and
different non-equilibrium steady states necessary Petkau’s groups. Oberley’s group [3] found that
to maintain living systems. modifying intracellular or extracellular SOD
ROS or reactive nitrogen species (RNS) derived activity in bacteria reduced the oxygen enhance-
from nitric oxide (?NO), another biologically ment ratio implying that superoxide was at least
relevant free radical produced enzymatically by partially responsible for the mechanisms leading to
NO-synthases [61,62], could be essential mediators the well-known effects of oxygen on radiosensiti-
of this type of redox regulation. Similar to O?2 , zation. In addition, Petkau et al. [7] were the first
and H2O2, ?NO is diffusible and levels of its to show that IV injection of 35 mg/g body weight
production are intimately linked to the levels of CuZn SOD, 2 and 4 h following 6 or 8 Gy whole
metabolic activity. The diffusible nature of these body x-irradiation, significantly protected Swiss
species coupled with their reactivity make them mice from lethality at 30 days. These studies
logical candidates to initiate redox regulatory clearly showed that alterations in a metabolic
cascades allowing for communication between enzyme with superoxide scavenging capability
different cellular compartments. In this way, following radiation could result in radioprotection
changes in electron flux through metabolic path- and implied that superoxide production (from
ways in one cellular compartment could lead to some source) following IR was participating in
Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation 315

radiation-induced injury. These types of studies substrate availability, activity changes, etc.).
have continued with more recent observations that Therefore, the target size for causing radiation-
active SOD enzymes, SOD mimetic compounds induced metabolic oxidative stress is very large
and in some instances catalase can lead to which fulfills one requirement for explaining the
inhibition of the deleterious effects induced by self-perpetuating process of radiation-induced
IR in a wide variety of in vitro and in vivo studies genomic instability [75].
ranging from transformation assays, bystander
effects, normal tissue damage associated with
inflammatory responses, and fibrosis [12–14,18– V. Genomic instability and metabolic oxidative
20,63–69]. Furthermore recent studies using mea- stress
surements of ROS and RNS derived from ?NO
have also discovered that radiation exposure can It is well established that chronic metabolic
induce increases in the metabolic production of oxidative stress can induce genomic instability
these species for several minutes and hours post and it has been suggested that mitochondria may
irradiation [70–72]. These same authors provided participate in this process [33]. The major mito-
evidence that the source of this increase in chondrial ETC complexes proposed to be involved
radiation-induced prooxidant production could with prooxidant production that are currently
involve mitochondria and may be related to under investigation include Complex I (or NADH
radiation-induced alterations in MAP kinase dehydrogenase), Complex II (succinate dehydro-
activation [70–72]. Finally, targeted irradiation of genase), and Complex III (ubiquinone-cytochrome
the cytoplasm, where many metabolic redox c oxidoreductase) [24,74]. The probability that
reactions occur, has been shown to induce muta- univalent reductions of O2 to form O?2 will occur
tions in nuclear DNA and manipulations of free at any of these complexes during mitochondrial
radical scavenging capability will modify radia- ETC activity would be expected to be governed by
tion-induced nuclear DNA damage under these the residence time of the electrons on subunits in
conditions [73]. These results support the hypo- the electron transport chains (ETCs) that are
thesis that radiation-induced damage to cytoplas- accessible to O2, as well as the rate constants for
mic metabolic pathways that result in free radical the forward and reverse reactions of the reduced
production can contribute to heritable changes in electron transport chain components with O2 [76].
the nuclear genome. The growing number of these Mitochondrial ETC Complexes I–V are made up
types of findings seems to provide strong support of 89 discrete protein subunits, 13 of which are
for the hypothesis that metabolic sources of O?2 coded for in mitochondrial DNA (mtDNA) and
and H2O2 (and possibly ?NO) contribute to 76 of which are coded for in genomic DNA in the
radiation effects following exposure. nucleus (nuDNA) [77]. A complex series of gene
The specific metabolic reactions responsible for expression and protein biosynthetic processes must
alterations in ROS (or RNS) production following be coordinately regulated to result in the proper
radiation exposure is an area of considerably more stoichiometric assembly of these complexes in the
controversy and appears to vary between model inner mitochondrial membrane [77]. It is possible
systems indicating that multiple different path- that mtDNA and/or nuDNA coding for mito-
ways are probably involved. In general, the chondrial ETC subunits as well as the biochemical
proposed metabolic sources of prooxidant produc- machinery necessary for the proper expression and
tion fall into two categories that include compo- assembly of ETC proteins is a primary target of
nents of mitochondrial electron transport chains oxidative damage during radiation exposure. In
[24,74] and oxidoreductase enzymes [25]. The fact this possibility has already been suggested for
activity of different metabolic pathways contribut- oxidative damage during the aging process [78–80].
ing to formation of these reactive species could be If radiation exposure induces disruptions in the
impacted upon by hundreds and perhaps thou- proper assembly and/or function of mitochondrial
sands of different radiation-induced perturbations ETCs (either by causing mutations or directly
in structure, function, and gene expression path- altering the protein structure/activity), an increase
ways (i.e. mutations, direct damage, effects on in residence time and/or accessibility of reduced
316 Spitz et al.

components of the ETCs to O2 could result in an of glucose-6-phosphate catalyzed by glucose-6-


increase in the probability of one-electron reduc- phosphate dehydrogenase (G6PD) [11,84]. Pentose
tions of O2 to form O?2 and H2O2. The resulting Cycle induction following oxidative stress and/or
increased fluxes of O?2 and H2O2 could then lead IR is thought to provide reducing equivalents, in
to a condition of metabolic oxidative stress that the form of NADPH, necessary for biosynthetic
could continue to cause further oxidative damage and repair processes. NADPH may also play a
to critical biological structures long after radiation role as a cofactor in the reduction of hydroper-
exposure. If radiation-induced damage (and/or oxides via the action of glutathione peroxidases
damage caused by metabolic oxidative stress), and peroxiredoxins [11,45]. In this regard radia-
resulted in mutations to either mtDNA and/or tion and oxidative stress induced increases in
nuDNA coding for genes required for the proper glucose metabolism through the Pentose Cycle
assembly of ETCs (or other prooxidant producing are thought to represent an immediate metabolic
enzyme systems), this aberrant condition of meta- response to stress in an attempt to offset increases
bolic oxidative stress could also become a heritable in oxidative damage with increases in protective
trait. Therefore, this mechanism could potentially and reparative processes.
contribute to radiation-induced genomic instabil- Immediately following IR exposure, several
ity that persists for many cell generations as well as signal transduction pathways (i.e. ERK1/2, JNK,
in the progeny from irradiated animals. p38, ATM, etc.) as well as transcription factors
In support of this hypothesis, persistent radia- (i.e. AP1, NF-kB, GADD153, p53, etc.) are
tion-induced genomic instability, alterations in activated resulting in the transcription of down-
growth, and signaling have already been reported stream genes thought to be involved in the
in cultured cells, bone marrow cells from radiation response [20,69–72,85–90]. The exact
irradiated animals, and the offspring of irradiated mechanisms responsible for sensing radiation
animals (reviewed in Morgan [75]). Also chronic induced free radical production leading to the
exposure to either exogenously applied H2O2 or activation of these signaling and gene expression
endogenous generation of ROS induced by pathways are not clearly understood. However,
hyperoxia has been shown to induce genomic given that many of the aforementioned signaling
instability and gene amplification [81]. Further- and gene expression pathways have been reported
more genomically unstable cells in culture (as to be sensitive to changes in intracellular oxida-
well as bone marrow cells from irradiated tion/reduction reactions [20,57,69–72,85–90], it is
animals) have also been shown to demonstrate tempting to hypothesize that radiation-induced
alterations in parameters indicative of oxidative changes in intracellular metabolic redox reactions
stress as well as mitochondrial dysfunction could be initiating events leading to activation of
[82,83]. Future studies using these model systems signal transduction, transcription factors, and
should be able to rigorously address the involve- gene expression.
ment of metabolic oxidative stress resulting from One of the potential links between IR-induced
damage to mitochondrial (or enzymatic) oxida- changes in oxidative metabolism and activation
tive metabolism in radiation-induced genomic of signaling leading to alterations in transcription
instability. factor activation, is the Pentose Cycle. This
would be expected, since NADPH is the source
of electrons for the reduction of thioredoxin,
VI. Radiation-induced alterations in metabolic which in the reduced form, is known to be
oxidation/reduction reactions and signal involved with transcription factor activation [43–
transduction 45,87,89]. In an elegant series of experiments, it
was shown that thioredoxin reductase appears to
One of the most well characterized metabolic pass electrons to thioredoxin, resulting in thio-
alterations observed immediately following IR redoxin nuclear translocation and a subsequent
exposure (and other oxidants) is increased Pentose interaction with redox factor-1 leading to the
Cycle activity leading to the regeneration of activation of the AP1 transcription factor [87,89].
NADPH from NADPþ coupled to the oxidation It is tempting to speculate that radiation-induced
Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation 317

increases in Pentose Cycle activity may alter the VII. Metabolic oxidative stress and radiation-
availability of NADPH to donate electrons to induced clastogenic factors
thioredoxin reductase leading to the initiation of
this signaling cascade. However, no data directly Clastogenic factors are secreted by injured cells
supporting this hypothesis is currently available. and are capable of diffusing significant distances
Since thioredoxin has been implicated in many causing genotoxic damage in cells that were never
different transcription factor activation pathways, exposed to the original stressing agent [98]. It has
this mechanism may also contribute to other been known for many years that radiation
radiation induced effects in addition to AP1 exposure is one of the agents capable of causing
activation. the release of clastogenic factors (reviewed in
Another possible way that radiation-induced Morgan [75]). While the precise identity of
changes in Pentose Cycle activity may be linked to radiation-induced clastogenic factors are not
radiation-induced signal transduction and bystan- known, active agents found in preparations of
der effects is via the activity of NADPH oxidase clastogenic factors isolated following exposure to
enzymes. NADPH oxidases are flavin containing other insults are thought to include inosine
enzymes that have several different subunits and triphosphate, lipid peroxidation products, and
are known to be broadly expressed in both cytokines such as tumor necrosis factor ([98],
phagocytic cells associated with inflammatory TNF). The cell injury mediated by clastogenic
responses, as well as non-phagocytic normal cells factors is also inhibited by SOD, implicating
including fibroblasts and smooth muscle cells [91– superoxide in the genotoxic effects [98]. Further-
96]. NADPH oxidases are also thought to be more, it has been shown that TNF is capable of
involved with signaling pathways leading to inducing metabolic oxidative stress in target cells
proliferation as well as fibrogenic responses [91– as well as inducing a superoxide-scavenging
96]. NADPH oxidase enzymes take electrons from enzyme located in mitochondria (MnSOD, [99]).
NADPH and produce O?2 and H2O2. Since the In addition over expression of MnSOD renders
NADPH oxidase enzymes are dependent on cells resistant to TNF [99], again implicating
NADPH to form ROS, changes in Pentose Cycle mitochondrial superoxide production in the bio-
activity could potentially impact upon the levels of logical effects of this cytokine.
ROS produced by these enzymes by affecting Since radiation exposure and metabolic oxida-
substrate availability. Exposure of smooth muscle tive stress are capable of initiating lipid peroxida-
cells and fibroblasts to H2O2-mediated oxidative tion reactions as well as cytokine release [100–102],
stress is known to lead to increases in non- it is tempting to hypothesize that these agents may
phagocytic NADPH oxidase activity [92] that be involved in radiation-induced clastogenesis as
contributes to H2O2-induced cell injury [92]. In well as bystander effects. Furthermore, if radiation
addition, several studies have suggested that induces persistent metabolic oxidative stress by
inhibitors of flavin containing enzymes such as one of the mechanisms discussed earlier resulting
NADPH oxidase are capable of inhibiting radia- in the formation of clastogenic factors via a redox
tion-induced signal transduction and injury in cells regulated process, these clastogens might be
adjacent to irradiated cells ([69], reviewed in significant contributors to persistent genomic
Azzam et al. [97]) suggesting that these enzymes instability. From this theoretical construct, it
may be involved in radiation-induced bystander seems clear that several phenomena involved in
effects. NADPH oxidase enzymes are also believed delayed radiation effects including inflammation,
to be mediators of inflammatory injury, mutagen- fibrosis, carcinogenesis, bystander effects, and
esis, growth disturbances, and transformation in a genomic instability could involve metabolic oxida-
wide variety of biologically relevant pathologies tive stress and the subsequent formation of
including tumor promotion and vascular injury as clastogenic factors. This would seem to be a fertile
well as normal cell signaling [91–97]. As such, they area for future studies, since the biological effects
represent excellent candidates for studies of radia- of clastogens have been reported to be amenable to
tion-induced effects attributable to metabolic manipulation by exogenous administration of
oxidative stress [91–97]. antioxidants ([98], reviewed in Morgan [75]).
318 Spitz et al.

VIII. Metabolic oxidative stress and radiation- reduce 14-3-3 zeta, GADD153, Cyclin A and
induced adaptive responses Cyclin B1 mRNAs in cells where MnSOD over
expression was independent of NF-kB [20]. This
For many years it has been observed that sub- study was the first to provide clear evidence that a
lethal or slightly toxic doses of radiation or redox sensitive transcription factor induced by
oxidative stress induce adaptive responses that radiation (NF-kB) was capable of regulating the
render cells resistant to further treatment by a expression of MnSOD, that in turn was capable of
second more cytotoxic challenge with the same or increasing the expression of genes that participate
similar damaging agent [103–108]. During the in radiation-induced adaptive responses. These
intervening time between the pretreatment and results also support the speculation that redox
challenge doses, signal transduction is induced sensitive signal transduction and a specific gene
leading to alterations in gene expression of expression profile following radiation may play a
proteins governing metabolic processes that are role in radiation-induced adaptive responses by
believed to protect the biological system from the rendering cells resistant to metabolic oxidative
second challenge dose. In this regard, several stress derived from mitochondrial metabolism.
enzymes thought to perform antioxidant func-
tions, such as MnSOD, proteins involved in
glutathione metabolism, and epoxide hydolase IX. Concluding remarks
[16–20,109,110] are induced following radiation,
suggesting a role in radiation inducible adaptive This review has presented some of the evidence
responses [18–20,109]. Furthermore, as noted ear- supporting the concept that metabolic oxidative
lier, several studies have suggested that in some stress derived from mitochondrial and enzymatic
systems, SOD administration provides some mea- sources of prooxidant production can contribute
sure of radiation protection against metabolic to the deleterious effects of IR as well as IR-
oxidative stress even after radiation exposure [7]. induced signaling and adaptive responses. It would
Recently is was confirmed that MnSOD is over appear from the number and persistence of these
expressed in radioresistant variants isolated from reports that in the new millennium many discov-
MCF-7 human carcinoma cells following exposure eries will likely arise from the pursuit of studies to
to fractionated ionizing, again supporting the more clearly define (1) the precise sources of this
concept that this mitochondrial superoxide scaven- radiation-induced metabolic oxidative stress, (2)
ging enzyme might play a role in radiation- the oxidants and reactive byproducts that are
inducible adaptive responses [20]. produced, (3) the relative contribution of these
The causal relationship between MnSOD and damaging agents to deleterious effects of radiation
resistance was confirmed when radioresistance in in animals and humans, and (4) the factors that
cells exposed to fractionated IR was reduced govern the mitigation of these effects that are
following expression of antisense MnSOD [20]. amenable to manipulation in clinical settings. It is
These cells also over expressed a group of stress also likely that much can be learned from the
responsive signaling proteins including p21, Myc, experiences of the free radical biology community
14-3-3 zeta, Cyclin A, Cyclin B1, and GADD153. that could potentially be applied to the study of
Radiation-induced expression of these six genes radiation-induced metabolic oxidative stress and
was suppressed in fibroblasts from MnSOD knock- in turn, the information gained from the study of
out mice (  /  ) as well as in radioresistant cells radiation effects on metabolic oxidative stress may
that expressed antisense MnSOD. The essential role also be relevant to the basic biology of aging.
of NF-kB in the adaptive response was demon-
strated by inhibiting NF-kB nuclear translocation
using mutant IkB and this manipulation also Acknowledgments
inhibited radioresistance as well as reducing steady
state levels of MnSOD, 14-3-3 zeta, GADD153, DRS, EIA, and JJL are supported by grants from
Cyclin A and Cyclin B1 mRNA [20]. In contrast, the Department of Energy (DE-FG02-02ER63447
mutant IkB was unable to inhibit radioresistance or and DE-FG02-03ER63634) and the Intramural
Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation 319

Program at the NIH/NCI supports DG. The radiation-induced injury. Free Radic Biol Med 33(6): 857–
authors would like to thank Drs. Larry W. 863, 2002
15. Ayene IS, Stamato TD, Mauldin SK, Biaglow JE, Tuttle
Oberley, Garry R. Buettner, Prabhat C. Goswami, SW, Jenkins SF, Koch CJ: Mutation in the glucose-6-
Frederick E. Domann, and William F. Morgan for phosphate dehydrogenase gene leads to inactivation of Ku
helpful discussions and encouragement. DNA end binding during oxidative stress. J Biol Chem
277: 9929–9935, 2002
16. Oberley LW, St. Clair DK, Autor AP, Oberley TD:
Increase in manganese superoxide dismutase activity in
the mouse heart after X-irradiation. Arch Biochem
References Biophys 254: 69–80, 1987
17. Summers RW, Maves BV, Reeves RD, Arjes LJ, Oberley
1. Altman KI, Gerber GB, Okada S: Radiation Biochem- LW: Irradiation increases superoxide dismutase in rat
istry. Academic Press, New York, 1970 intestinal smooth muscle. Free Radic Biol Med 6: 261–
2. Arena V: Ionizing Radiation and Life. The C.V. Mosby 270, 1989
Company, St. Louis, 1971 18. Kim SG, Nam SY, Kim CW, Kim JH, Cho CK, Yoo SY:
3. Oberley LW, Lindgren AL, Baker SA, Stevens RH: Enhancement of radiation-inducible hepatic glutathione-
Superoxide ion as the cause of the oxygen effect. Radiat S-transferases Ya, Yb1, Yb2, Yc1, and Yc2 gene expres-
Res 68: 320–328, 1976 sion by oltipraz: Possible role in radioprotection. Mol
4. Biaglow JE, Mitchell JB, Held K: The importance of Pharmacol 51(2): 225–233, 1997
peroxide and superoxide in the X-ray response. Int J 19. Shimizu T, Iwanaga M, Yasunaga A, Urata Y, Goto S,
Radiat Oncol Biol Phys 22: 665–669, 1992 Shibata S, Kondo T: Protective role of glutathione
5. Hall EJ: Radiobiology for the Radiologist. Lippincott synthesis on radiation-induced DNA damage in rabbit
Williams and Wilkins, Philadelphia, 2000 brain. Cellular & Molecular Neurobiology 18: 299–310,
6. Chiu SM, Xue LY, Friedman LR, Oleinick NL: Copper 1998
ion-mediated sensitization of nuclear matrix attachment 20. Guo G, Yan-Sanders Y, Lyn-Cook BD, Wang T, Tamae
sites to ionizing radiation. Biochemistry 32: 6214–6219, D, Ogi J, Khaletskiy A, Li Z, Weydert C, Longmate JA,
1993 Huang T-T, Spitz DR, Oberley LW, Li JJ: Manganese
7. Petkau A, Chelack WS, Pleskach SD: Protection of post- superoxide dismutase-mediated gene expression in radia-
irradiated mice by superoxide dismutase. Int J Radiat Biol tion-induced adaptive responses. Mol Cell Biol 23: 2362–
29: 297–299, 1976 2378, 2003
8. Biaglow JE, Clark EP, Epp ER, Morse-Guadio M, Varnes 21. Mitchell P: Keilin’s respiratory chain concept and its
ME, Mitchell JB: Nonprotein thiols and the radiation chemiosmotic consequences. Science 206: 1148–1159, 1979
response of A549 human lung carcinoma cells. Int J 22. Lehninger AL: Lehninger Principals of Biochemistry.
Radiat Biol 44: 489–495, 1983 Worth Publishers Inc., New York, NY, 2000
9. Biaglow JE, Varnes ME, Clark EP, Epp ER: The role of 23. Szent-Györgyi A: Electronic Biology and Cancer. Marcel
thiols in cellular response to radiation and drugs. Radiat Dekker Inc., New York, NY, 1976
Res 95: 437–455, 1983 24. Boveris A, Cadenas E: Production of superoxide radicals
10. Mitchell JB, Russo A: The role of glutathione in radiation and hydrogen peroxide in mitochondria. Superoxide
and drug induced cytotoxicity. Br J Cancer 55: S96–S104, Dismutase:Volume II, L.W. Oberley, ed. CRC Press
1987 Inc., Boca Raton, Florida, 1982
11. Tuttle SW, Varnes ME, Mitchell JB, Biaglow JE: 25. Halliwell, Gutteridge: Free Radicals in Biology and
Sensitivity to chemical oxidants and radiation in CHO Medicine. Oxford University Press Inc., New York, NY,
cell lines deficient in oxidative pentose cycle activity. Int J 1989
Radiat Oncol Biol Phys 22: 671–675, 1992 26. Esterbauer HH, Zollner H, Schaur RJ: Aldehydes formed
12. St.Clair DK, Wan XS, Oberley TD, Muse KE, St.Clair by lipid peroxidation: Mechanisms of formation, occur-
WH: Suppression of radiation-induced neoplastic trans- rence, and determination. Membrane Lipid Oxidation.
formaton by overexpression of mitochondrial superoxide CRC Press Inc., Boca Raton, Florida, pp. 240–268,
dismutase. Mol Carcinogenesis 6: 238–242, 1992 1990
13. Epperly MW, Epstein CJ, Travis EL, Greenberger JS: 27. Yamamoto Y, Niki E: Role of antioxidants in lipid
Decreased pulmonary radiation resistance of manganese peroxidation. Membrane Lipid Oxidation. CRC Press
superoxide dismutase (MnSOD)-deficient mice is cor- Inc., Boca Raton, Florida, pp. 286–301
rected by human manganese superoxide dismutase-plas- 28. Gerschman R, Gilbert DL, Nye SW, Dwyer P, Fenn WO:
mid/Liposome (SOD2-PL) intratracheal gene therapy. Oxygen poisoning and X-irradiation: A mechanism in
Radiat Res 154: 365–374, 2000 common Science 119: 623–626, 1954
14. Vujaskovic Z, Batinic-Haberle I, Rabbani ZN, Feng QF, 29. Jamieson D: Oxygen toxicity and reactive oxygen
Kang SK, Spasojevic I, Samulski TV, Fridovich I, metabolites in mammals. Free Radic Biol Med 7: 87–
Dewhirst MW, Anscher MS: A small molecular weight 108, 1989
catalytic metalloporphyrin antioxidant with superoxide 30. Harman D: Aging: A theory based on free radical and
dismutase (SOD) mimetic properties protects lungs from radiation chemistry. J Gerontol 2: 298–300, 1957
320 Spitz et al.

31. Finkel T, Holbrook NJ: Oxidants, oxidative stress and the 48. Xiao J, Biaglow JE, Chae-Park HJ, Jin J, Tuel-Ahlgren L,
biology of aging. Nature 408: 239–247, 2000 Myers DE, Burkhardt AL, Bolen JB, Uckun FM: Role of
32. Oberley LW, Oberley TD, Buettner GR: Cell division in hydroxyl radicals in radiation-induced activation of lyn
normal and transformed cells: The possible role of tyrosine kinase in human B-cell precursors. Leukemia &
superoxide and hydrogen peroxide. Med Hypotheses 7: Lymphoma 22(5–6): 421–430, 1996
21–42, 1981 49. Sun Yi, Oberley LW: Redox regulation of transcriptional
33. Spitz DR, Sim JE, Ridnour LA, Galoforo SS, Lee YJ: activators. Free Radic Biol Med 21: 335–348, 1996
Glucose deprivation-induced oxidative stress in human 50. Lee YJ, Galoforo SS, Berns CM, Chen JC, Davis BH, Sim
tumor cells: A fundamental defect in metabolism? Ann JE, Corry PM, Spitz DR: Glucose deprivation-induced
NY Acad Sci 899: 349–362, 2000 cytotoxicity and alterations in mitogen-activated protein
34. Schafer FQ, Buettner GR: Redox environment of the cell kinase activation are mediated by oxidative stress in
as viewed through the redox state of the glutathione multidrug-resistant human breast carcinoma cells. J Biol
disulfide/glutathione couple. Free Radic Biol Med 30: Chem 273: 5294–5299, 1998
1191–1212, 2001 51. Blackburn RV, Spitz DR, Liu X, Galoforo SS, Sim JE,
35. Sies H: Oxidative stress: Oxidants and antioxidants. Exp Ridnour LA, Chen JC, Davis BH, Corry PM, Lee YJ:
Physiol 82: 291–295, 1997 Metabolic oxidative stress activates signal transduction
36. Finkel T, Holbrook NJ: Oxidants, oxidative stress and the and gene expression during glucose deprivation in human
biology of aging. Nature 408(6809): 239–247, 2000 tumor cells. Free Radic Biol Med 26: 419–430, 1999
37. Oberley LW: Anticancer therapy by overexpression of 52. Lee YJ, Galoforo SS, Sim JE, Ridnour LA, Choi J,
superoxide dismutase. Antioxidants & Redox Signaling Forman HJ, Corry PM, Spitz DR: Dominant-negative
3(3): 461–472, 2001 Jun N-terminal protein kinase (JNK-1) inhibits metabolic
38. Gonzalez-Zulueta M, Ensz LM, Mukhina G, Lebovitz oxidative stress during glucose deprivation in human
RM, Zwacka RM, Engelhardt JF, Oberley LW, Dawson breast carcinoma cells. Free Radic Biol Med 28: 575–584,
VL, Dawson TM: Manganese superoxide dismutase 2000
protects nNOS neurons from NMDA and nitric oxide- 53. Goswami PC, Sheren J, Albee LD, Parsian AJ, Sim JE,
mediated neurotoxicity. J Neuroscience 18(6): 2040–2055, Ridnour LA, Higashikubo R, Hunt CR, Spitz DR: Cell
1998 cycle coupled variation in Topoisomerase IIa mRNA is
39. Milgram NW, Head E, Muggenburg B, Holowachuk D, regulated by the 30 -untranslated region: Possible role of
Murphey H, Estrada J, Ikeda-Douglas CJ, Zicker SC, redox sensitive protein binding in mRNA stability. J Biol
Cotman CW: Landmark discrimination learning in the Chem 275: 38384–38392, 2000
dog: Effects of age, an antioxidant fortified food, and 54. Song JJ, Rhee JG, Suntharalingam M, Walsh SA, Spitz
cognitive strategy. Neuroscience & Biobehavioral Reviews DR, Lee YJ: Role of glutaredoxin in metabolic oxidative
26(6): 679–695, 2002 stress: Glutaredoxin as a sensor of oxidative stress
40. Klein EA, Thompson IM, Lippman SM, Goodman PJ, mediated by H2O2. J Biol Chem 277(48): 46566–46575,
Albanes D, Taylor PR, Coltman C: SELECT: The 2002
selenium and vitamin E cancer prevention trial. Urol 55. Menon SG, Sarsour EH, Spitz DR, Ryuji Higashikubo,
Oncol 21: 59–65, 2003 Zhang H, Strum M, Goswami PC: Redox regulation of
41. Martin A: Antioxidant vitamins E and C and risk of the G1 to S transition in the mouse embryo fibroblast cell
Alzheimer’s disease. Nutrition Reviews 61(2): 69–73, 2003 cycle. Cancer Res 63: 2109–2117, 2003
42. Bulger EM, Maier RV: An argument for Vitamin E 56. Watson WH, Pohl J, Montfort WR, Stuchlik O, Reed
supplementation in the management of systemic MS, Powis G, Jones DP: Redox potential of human
inflammatory response syndrome. Shock 19(2): 99–103, thioredoxin-1 and identification of a second dithiol/
2003 disulfide motif. J Biol Chem 278: 33408–33415, 2003
43. Ueda S, Masutani H, Nakamura H, Tanaka T, Ueno M, 57. Hainaut P, Mann K: Zinc binding and redox control of
Yodoi J: Redox control of cell death. Antioxidants & p53 structure and function. Antioxidants & Redox
Redox Signaling 4(3): 405–414, 2002 Signaling 3(4): 611–623, 2001
44. Nakamura H, Nakamura K, Yodoi J: Redox regulation of 58. Kroncke KD: Zinc finger proteins as molecular targets for
cellular activation. Ann Rev Immunol 15: 351–369, 1997 nitric oxide-mediated gene regulation. Antioxidants &
45. Rhee SG, Chang TS, Bae YS, Lee SR, Kang SW: Cellular Redox Signaling 3(4): 565–575, 2001
regulation by hydrogen peroxide. J Am Soc 59. Wilcox DE, Schenk AD, Feldman BM, Xu Y: Oxidation
Nephrology of zinc-binding cysteine residues in transcription factor
46. Claiborne A, Mallett TC, Yeh JI, Luba J, Parsonage D: proteins. Antioxidants & Redox Signaling 3(4): 549–564,
Structural, redox, and mechanistic parameters for 2001
cysteine-sulfenic acid function in catalysis and regulation. 60. Webster KA, Prentice H, Bishopric NH: Oxidation of zinc
Adv Protein Chem 58: 215–276, 2001 finger transcription factors: Physiological consequences.
47. Claiborne A, Yeh JI, Mallett TC, Luba J, Crane EJ, Antioxidants & Redox Signaling 3(4): 535–548, 2001
Charrier V, Parsonage D: Protein-sulfenic acids: Diverse 61. Ignarro LJ: Nitric oxide as a unique signaling molecule in
roles for an unlikely player in enzyme catalysis and the vascular system: A historical overview. J Physiol &
redox regulation. Biochemistry 38(47): 15407–15416, 1999 Pharmacol 53(4 Pt 1): 503–514, 2002
Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation 321

62. Lancaster JR: A tutorial on the diffusibility and reactivity chondria. Free Radic Biol Med 29(supplement 1): S21,
of free nitric oxide. Nitric Oxide 1(1): 18–30, 1997 2000
63. Borek C, Troll W: Modifiers of free radicals inhibit in 77. Cecchii G: Function and structure of complex II of the
vitro the oncogenic actions of X-rays, bleomycin, and the respiratory chain. Annu. Rev. Biochem. 72: 77–109, 2003
tumor promoter 12-O-tetradecanoylphorbol 13-acetate. 78. Hamilton ML, Van Remmen H, Drake JA, Yang H, Guo
Proc Natl Acad Sci USA 80(5): 1304–1307, 1983 ZM, Kewitt K, Walter CA, Richardson A: Does oxidative
64. Delanian S, Baillet F, Huart J, Lefaix JL, Maulard C, damage to DNA increase with age? Proc Natl Acad Sci
Housset M: Successful treatment of radiation-induced USA 98(18): 10469–10474, 2001
fibrosis using liposomal Cu/Zn superoxide dismutase: 79. Lu CY, Lee HC, Fahn HJ, Wei YH: Oxidative damage
Clinical trial. Radiother Oncol 32: 12–20, 1994 elicited by imbalance of free radical scavenging enzymes is
65. Lefaix JL, Delanian S, Leplat JJ, Tricaud Y, Martin M, associated with large-scale mtDNA deletions in aging
Nimrod A, Baillet F, Daburon F: Successful treatment of human skin. Mutation Res 423(1–2): 11–21, 1999
radiation-induced fibrosis using Cu/Zn-SOD and Mn- 80. Beckman KB, Ames BN: Mitochondrial aging: Open
SOD: An experimental study. Int J Radiat Oncol Biol questions. Ann NY Acad Sci 854: 118–127, 1998
Phys 35: 305–312, 1996 81. Hunt CR, Sim JE, Featherstone T, Golden W, Von Kapp-
66. Greenberger JS, Epperly MW, Gretton J, Jefferson M, Herr C, Hock RA, Gomez RA, Parsian AJ, Spitz DR:
Nie S, Bernarding M, Kagan V, Guo HL: Radioprotective Genomic instability and catalase gene amplification
gene therapy. Current Gene Therapy 3(3): 183–195, induced by chronic exposure to oxidative stress. Cancer
2003 Res 58: 3986–3992, 1998
67. Kang SK, Rabbani ZN, Folz RJ, Golson ML, Huang H, 82. Clutton SM, Townsend KM, Walker C, Ansell JD,
Yu D, Samulski TS, Dewhirst MW, Anscher MS, Wright EG: Radiation-induced genomic instability and
Vujaskovic Z: Overexpression of extracellular superoxide persisting oxidative stress in primary bone marrow
dismutase protects mice from radiation-induced lung cultures. Carcinogenesis 17(8): 1633–1639, 1996
injury. Int J Radiat Oncol Biol Phys 57(4): 1056–1066, 83. Limoli CL, Giedzinski E, Morgan WF, Swarts SG, Jones
2003 GD, Hyun W: Persistent oxidative stress in chromoso-
68. Zhao W, Spitz DR, Oberley LW Robbins MEC: Redox mally unstable cells. Cancer Res 63(12): 3107–3111, 2003
modulation of the pro-fibrogenic mediator plasminogen 84. Varnes ME: Inhibition of pentose cycle of A549 cells by 6-
activator inhibitor-1. Cancer Res 61: 5537–5543, 2001 aminonicotinamide: Consequences for aerobic and
69. Azzam EI, de Toledo SM, Spitz DR, Little JB: Oxidative hypoxic radiation response and for radiosensitizer action.
metabolism modulates signal transduction and micro- NCI Monographs (6): 199–203, 1988
nucleus formation in bystander cells from a-particle- 85. Dent P, Yacoub A, Fisher PB, Hagan MP, Grant S:
irradiated normal human fibroblasts. Cancer Res 62: MAPK pathways in radiation responses. Oncogene
5436–5442, 2002 22(37): 5885–5896, 2003
70. Leach JK, Black SM, Schmidt-Ullrich RK, Mikkelsen 86. Watters DJ: Oxidative stress in ataxia telangiectasia.
RB: Activation of constitutive nitric-oxide synthase Redox Report 8(1): 23–29, 2003
activity is an early signaling event induced by ionizing 87. Wei SJ, Botero A, Hirota K, Bradbury CM, Markovina S,
radiation. J Biol Chem 277(18): 15400–15406, 2002 Laszlo A, Spitz DR, Yodoi J, Gius D: Thioredoxin
71. Leach JK, Van Tuyle G, Lin PS, Schmidt-Ullrich R, nuclear translocation and interaction with redox factor-1
Mikkelsen RB: Ionizing radiation-induced, mitochondria- activates the AP-1 transcription factor in response to
dependent generation of reactive oxygen/nitrogen. Cancer ionizing radiation. Cancer Res 60: 6688–6695, 2000
Res 61(10): 3894–3901, 2001 88. Bradbury CM, Locke JE, Wei SJ, Rene LM, Karimpour
72. Mikkelsen RB, Wardman P: Biological chemistry of S, Hunt C, Spitz DR, Gius D: Increased activator protein
reactive oxygen and nitrogen and radiation-induced signal 1 activity as well as resistance to heat-induced radio-
transduction mechanisms. Oncogene 22(37): 5734–5754, sensitization, hydrogen peroxide, and cisplatin are inhib-
2003 ited by indomethacin in oxidative stress-resistant cells.
73. Wu LJ, Randers-Pehrson G, Xu A, Waldren CA, Geard Cancer Res 61(8): 3486–3492, 2001
CR, Yu Z, Hei TK: Targeted cytoplasmic irradiation with 89. Karimpour S, Lou J, Lin LL, Rene LM, Lagunas L, Ma
alpha particles induces mutations in mammalian cells. X, Karra S, Bradbury CM, Markovina S, Goswami PC,
Proc Natl Acad Sci USA 96(9): 4959–4964, 1999 Spitz DR, Hirota K, Kalvakolanu DV, Yodoi J, Gius D:
74. Boveris A: Mitochondrial production of superoxide Thioredoxin reductase regulates AP-1 activity as well as
radical and hydrogen peroxide. Adv Exp Med Biol 78: thioredoxin nuclear localization via active cysteines in
67–82, 1977 response to ionizing radiation. Oncogene 21: 6317–6327,
75. Morgan WF: Non-targeted and delayed effects of 2002
exposure to ionizing radiation: II. Radiation-induced 90. Heinloth AN, Shackelford RE, Innes CL, Bennett L, Li L,
genomic instability and bystander effects in vivo, clasto- Amin RP, Sieber SO, Flores KG, Bushel PR, Paules RS:
genic factors and transgenerational effects. Radiat Res ATM-dependent and -independent gene expression
159: 581–596, 2003 changes in response to oxidative stress, gamma irradia-
76. Buettner GR, Ng C, Oberley LW, Rodgers VG, Schafer tion, and UV irradiation. Radiat Res 160(3): 273–290,
FQ: Does MnSOD influence H2O2 production in mito- 2003
322 Spitz et al.

91. Suh YA, Arnold RS, Lassegue B, Shi J, Xu X, Sorescu D, induced tumor necrosis factor-alpha expression: Clinical
Chung AB, Griendling KK, Lambeth JD: Cell transfor- application of transcriptional and physical targeting of
mation by the superoxide-generating oxidase Mox1. gene therapy. Lancet Oncol 3(11): 665–671, 2002
Nature 401(6748): 79–82, 1999 102. Tribble DL, Krauss RM, Chu BM, Gong EL, Kullgren
92. Li W-G, Miller FJ, Zhang HJ, Spitz DR, Oberley LW, BR, Nagy JO, La Belle M: Increased low density
Weintraub NL: H2O2-induced O2-production by a non- lipoprotein degradation in aorta of irradiated mice is
phagocytic NAD(P)H oxidase causes oxidant injury. inhibited by preenrichment of low density lipoprotein
J Biol Chem 276: 29251–29256, 2001 with alpha-tocopherol. J Lipid Res 41(10): 1666–1672,
93. Bokoch GM, Knaus UG: NADPH oxidases: Not just for 2000
leukocytes anymore! Trends in Biochemical Sciences 103. Shadley JD, Afzal V, Wolff S: Characterization of the
28(9): 502–508, 2003 adaptive response to ionizing radiation induced by low
94. Cai H, Griendling KK, Harrison DG: The vascular doses of X rays to human lymphocytes. Radiat Res 111(3):
NAD(P)H oxidases as therapeutic targets in cardiovas- 511–517, 1987
cular diseases. Trends in Pharmacological Sciences 24(9): 104. Wolff S: The adaptive response in radiobiology: Evolving
471–478, 2003 insights and implications. Environmental Health Perspec-
95. Ohshima H, Tatemichi M, Sawa T: Chemical basis of tives 106(Suppl 1): 277–283, 1998
inflammation-induced carcinogenesis. Arch. Biochem 105. Waldren CA: Adaptive response induced by low levels of
Biophys 417(1): 3–11, 2003 radiation. Summary and comments. Human & Experi-
96. Sorescu D, Griendling KK: Reactive oxygen species, mental Toxicology 18(7): 452–453, 1999
mitochondria, and NAD(P)H oxidases in the development 106. Spitz DR, Dewey WC, Li GC: Hydrogen peroxide or heat
and progression of heart failure. Congestive Heart Failure shock induces resistance to hydrogen peroxide in Chinese
8(3): 132–140, 2002 hamster fibroblasts. J Cell Physiol 131: 364–373, 1987
97. Azzam EI, Toledo SM, Little JB: Oxidative metabolism, 107. Sullivan SJ, Roberts RJ, Spitz DR: Replacement of media
gap junctions and ionizing radiation-induced bystander in cell culture alters oxygen toxicity: Possible role of lipid
effect. Oncogene 22: 7050–7057, 2003 aldehydes and glutathione transferases in O2 toxicity.
98. Emerit I, Garban F, Vassy J, Levy A, Filipe P, Freitas J: J Cell Physiol 147: 427–433, 1991
Superoxide-mediated clastogenesis and anticlastogenic 108. Sullivan SJ, Oberley TD, Roberts RJ, Spitz DR: A stable
effects of exogenous superoxide dismutase. Proc Natl O2-resistant cell line: Role of lipid peroxidation by-
Acad Sci USA 93(23): 12799–12804, 1996 products in O2-mediated injury. Am J Physiol (Lung
99. Wong GH, Elwell JH, Oberley LW, Goeddel DV: Cell Mol Physiol) 262: L748–L756, 1992
Manganous superoxide dismutase is essential for cellular 109. Lee AK, Cho CK, Kim MS, Kim SG: Enhanced
resistance to cytotoxicity of tumor necrosis factor. Cell expression of microsomal epoxide hydrolase and glu-
58(5): 923–931, 1989 tathione S-transferase by imidazole correlates with the
100. Xu Y, Greenstock CL, Trivedi A, Mitchel RE: Occupa- radioprotective effect. Res Commun Mol Path Pharmacol
tional levels of radiation exposure induce surface expres- 108(3–4): 155–165, 2000
sion of interleukin-2 receptors in stimulated human 110. Park WY, Hwang CI, Im CN, Kang MJ, Woo JH, Kim
peripheral blood lymphocytes. Radiat Environ Biophys JH, Kim YS, Kim JH, Kim H, Kim KA, Yu HJ, Lee SJ,
35(2): 89–93, 1996 Lee YS, Seo JS: Identification of radiation-specific
101. Weichselbaum RR, Kufe DW, Hellman S, Rasmussen responses from gene expression profile. Oncogene 21(55):
HS, King CR, Fischer PH, Mauceri HJ: Radiation- 8521–8528, 2002

You might also like