You are on page 1of 24

Annual Reviews

www.annualreviews.org/aronline
Annu Rev. Pharmacol. Todcol. 19%. 36:83-106
Copyright 0 1996 by Annual Reviews Inc. All rights reserved

OXIDATIVE STRESS IN
NEURODEGENERATIVE
DISEASES
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

N. A. Sirnonianl and J. T. Coyle2


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

Departments of Neurology and 2Psychiatry,MassachusettsGeneral Hospital and


Harvard Medical School, Boston, Massachusetts02114

KEY WORDS: free radicals, Alzheimers disease, Parkinsons disease, amyotrophic lateral
sclerosis

ASSTRACT

Oxidative stress refers to the cytopathologic consequences of a mismatch be-


tween the production of free radicals and the ability of the cell to defend against
them. Growing data from experimentalmodels and human brain studies suggest
oxidative stress may play an importantrole in neuronal degeneration in diseases
such as Parkinsons disease, Alzheimers disease, and amyotrophiclateral scle-
rosis. Mitochondrial oxidative metabolism, nitric oxide, phospholipid metabo-
lism, and proteolytic pathways are potential sources of intracellular free radicals.
Alterationsin free radical defense systems may alsocontributeto oxidative stress.
A net increase in reactive oxygen species can produce damage to lipids, proteins,
and DNA and induce necrosis or apoptosis. Elucidating the pathways important
in the production of and defense from free radicals may be importantin devising
new pharmacologicstrategies to slow or halt neuronal degeneration.

Progressive degeneration of a subset of neurons is the pathologic hallmark of


adult-onset neurodegenerative disorders such as Alzheimers disease (AD),
Parkinsons disease (PD), and amyotrophic lateral sclerosis (ALS). Growing
evidence points to the involvement of free radicals in mediating neuronal death
in these diseases. Elucidation of the intracellular pathways involved in the
formation of free radicals in neurons may be important not only for under-
standing the pathophysiologic basis for neuronal death in these disorders, but
also for devising rational pharmacologic strategies to slow or prevent neuronal
degeneration. In this chapter, we review recent advances made in our under-
standing of the intracellular pathways involved in free radical generation and
how they may contribute to selective neuronal degeneration in these disorders.
83

0362-1642/96/0415-0083$08.00
Annual Reviews
www.annualreviews.org/aronline

84 SIMONIAN & COYLE

SOURCES OF FREE RADICALS


Free radicals are normal products of cellular aerobic metabolism (1-3). Su-
peroxide (Of)and hydroxyl (OH') species are the predominant cellular free
radicals. Hydrogen peroxide (HzOz)and peroxynitrite (ONOO-), although not
themselves free radicals, contribute importantly to the cellular redox state.
Together, these molecules are referred to as reactive oxygen species (ROS).
The major sources of ROS are mitochondrial oxidative metabolism, enzymatic
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

reactions involving mixed-function oxidation, and autoxidation of small mole-


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

cules. Mixed-function oxidation reactions occur in the cytoplasm, plasma and


nuclear membrane, endoplasmic reticulum, and peroxisomes. oi.. is formed
by leakage of high-energy electrons along the mitochondrial electron transport
chain and by a variety of cytosolic and membrane-bound enzymes, including
xanthine oxidase, the cytochrome P450 complex, and phospholipase Az
(PLA,). HzOzalso is produced along the electron transport chain, as well as
through autooxidationof small molecules and dismutation of Of by superoxide
dismutase (SOD) (Figure 1) (1). Though not itself reactive, HzOz, in the
presence of reduced metal via the Fenton reaction, forms the highly reactive
OH. (Figure 1) (2). ONOO-, formed by the reaction of nitric oxide (NO) with
w, is a highly reactive molecule that also breaks down to form OH. (Figure
1) (394).

CELLULAR DEFENSE SYSTEMS


An array of cellular defense systems exist to counterbalance ROS (1,3). These
include enzymatic and nonenzymatic antioxidants that lower the steady-state
concentrations of free radical species and repair oxidative cellular damage.
There are three forms of SOD: an extracellular and an intracellular copper/zinc
(Cu/Zn) SOD and a mitochondrial, manganese (Mn) SOD. All three convert
oi..to H202(Figure 1) (1,5). Most HzOzin the brain is removed by glutathione
peroxidase (GSH-Px), which uses it to oxidize reduced glutathione (GSH)
(Figure 1) (4,6, 7). GSH is the primary low-molecular-weight thiol in the
cytoplasm and is a major reserve for cysteine. GSH in conjunction with the
reductant NADPH can reduce lipid peroxides, free radicals, and H20,. GSH
is converted to glutathione disulfide (GSSG), which is reconverted to GSH by
glutathione reductase (GSH-Red). Catalase, which is found at very low levels
in the brain, also removes H202.The cyclic one-electron oxidation and re-
duction of endogenous quinone is also a source of intracellular Of.
NAD(P)H:Quinone reductase (QR), using reducing equivalents from NADH
or NADPH, reduces quinones to the less-toxic hydroquinones (8,9). Vitamin
E is the major lipid-soluble,chain-breaking antioxidant (IO). Transferrin binds
iron and reduces its participation in lipid peroxidation. Ceruloplasmin oxidizes
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVE STRESS 85
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

Figure I Reactions important in the production and defense from free radicals in neurons.
Abbreviations: superoxide (Of); superoxide dismutase. (SOD);hydrogen peroxide (HzOz); iron
@e); copper (Cu);hydroxyl radical (OH'); nitric oxide (NO'); peroxynitrite (ONOO-);glutathione
(GSH); glutathione peroxidase (GSH-Px); glutathione disulfide (GSSG); glutathione reductase
(GSH-Red).

iron without release of ROS and inhibits lipid peroxidation. Uric acid binds
iron and copper tightly, scavenges radicals, and inhibits lipid peroxidation.
Metallothionein and heme-oxygenase (HO- l), proteins induced by oxidative
stress, also have antioxidant functions.

OXIDATIVE STRESS
Oxidative stress refers to the cytologic consequences of a mismatch between
the production of free radicals and the ability of the cell to defend against
them. Oxidative stress can thus occur when the production of free radicals
increases, scavenging of free radicals or repair of oxidatively modified mac-
romolecules decreases, or both. This imbalance results in a build-up of oxida-
tively modified molecules that can cause cellular dysfunction and, for post-
mitotic cells such as neurons, cell death.
ROS, especially OH- and ONOO-, can produce functional alterations in
lipids, proteins, and DNA (1,2, 1 1) (Figure 2). The incorporation of molecular
oxygen into polyunsaturated fatty acids initiates a chain reaction in which
ROS-including OH-, HZOz,and peroxyl and alkoxy1 radicals-are formed
(3). Oxidative lipid damage, termed lipid peroxidation, produces a progressive
loss of membrane fluidity, reduces membrane potential, and increases perme-
ability to ions such as Caz+.
Annual Reviews
www.annualreviews.org/aronline

86 SIMONIAN & COYLE

NMDA AMPAIUA Voltage-gated


Receptor Receptor Calcium Channels

Nat; Cat+ Nat; Cat+ Cat+


Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

Figure 2 Intracellularpathways that contribute to oxidative stress in neurons. (See text fordetails.)
Abbreviations: phospholipase A2 (PLAz); phospholipids (PLs); arachidonic acid (AA); calcium-
and calmodulin-dependent protein kinase (CaMK); nitric oxide synthase (NOS); arginine (Arg);
nitric oxide (NO'); superoxide (Of);hydrogen peroxide (HzOz);hydroxyl radical (OH');
peroxynitrite (ONOO-); superoxide dismutase (SOD).
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVESTRESS 87

ROS can damage proteins (12-14). Redox cycling cations such as Fez+and
Cu*+can bind to cation-binding sites on proteins and, with the further attack
of H202and Of,change amine groups on amino acids into carbonyls. Oxida-
tive damage can activate or inactivate proteins. Oxidative damage to enzymes
can increase their susceptibility to proteolysis, which is involved in the regu-
lation of enzyme degradation and in the accumulation of altered forms of
enzymes during aging (13,15). ONOO can mediate oxidation by at least three
distinct pathways: (a) the hydrogen ion-catalyzed decomposition, to form OW
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

and nitrogen dioxidelike intermediate; (b) direct reaction with sulfhydryl


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

groups; and (c) a reaction with metal ions, to form a potent nitrating agent
(16). Nitration of tyrosine residues on proteins on the carbon adjacent to
oxygen is a stable product of the attack of ONOO- on proteins.
ROS can also damage DNA (17). OW modifies ribose phosphates and
pyrimidine nucleosides and nucleotides and reacts with the sugar phosphate
backbone of DNA to break strands. Hydroxylation of deoxyguanosineresidues
produces 8-hyroxydeoxyguanosine (8-OHDG), which can be used as a marker
of oxidative DNA damage (18-20). 8-OHDG concentrations increase with age
in the human brain, with levels of 8-OHDG in mitochondrial DNA (mtDNA)
10-fold higher than in nuclear DNA (nDNA) (21).

Apoptosis and Necrosis


Oxidative stress has been associated with both necrosis and apoptosis. Mor-
phologic and biochemical characteristics distinguish these two forms of cell
death (22, 23). In necrosis, a selective loss of membrane permeability occurs,
which results in swelling of organelles, loss of membrane depolarization, and
rupture of the plasma membrane. In apoptosis, a death promoting signal acti-
vates a program of cell death through either new protein and RNA synthesis
or derepression of an existing pathway (24, 25). Cells undergoing apoptosis
show shrinkage of the nucleus, condensation and fragmentation of chromatin,
and (often) DNA degradation by endonucleases into fragments in multiples of
18&200 base pairs (22).
Oxidative stress can induce necrotic cell death. Attack of free radicals on
lipids can disrupt membrane ion gradients and produce plasma membrane
rupture. For example, neurons from the hypothalamic cell line GT1-7 undergo
necrotic cell death following exposure to the GSH-depleting drug, buthionine
sulfoximine (26). Oxidative stress can also induce apoptotic cell death. Expo-
sure of B cells to H202 or to the redox cycling drug menadione induces
apoptosis (27). Cortical neurons grown in media low in cysteine, which de-
pletes GSH, also undergo apoptosis (28). Inhibition of SOD can induce apop
tosis in PC12 cells and in neurons in spinal cord cultures (29, 30). Oxidative
DNA damage is believed to be a trigger for apoptosis (3 1,32). Oxidized DNA
Annual Reviews
www.annualreviews.org/aronline

88 SIMONIAN & COYLE

activates a nuclear enzyme, poly(ADP ribose) polymerase (PAW), which is


involved in DNA repair (33). Proteolysis of PARP precludes its recruitment
to sites of DNA damage and induces apoptosis (34). Although the specific
factors that determine the mechanism of oxidative stress-induced neuronal
death remain poorly characterized, cellular phenotype, the predominant site of
oxidative damage (lipid versus DNA), and the intensity of the insult (high-ne-
crotic, low-apoptotic) may be important.
Growing evidence also exists that oxidative stress may be a mediator of
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

apoptosis induced by a variety of stimuli. Lipid peroxidation occurs in and


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

antioxidants protect against B cells undergoing apoptotic cell death following


interleukin-3 (IL-3) deprivation (27). Talley et a1 (35) reported that the anti-
oxidant N-acetylcysteine prevents tumor necrosis factor a-induced apoptosis
in neuroblastoma cells. Lastly, Greenlund et al (36) reported increases in
intracellulare following nerve growth factor removal and a delay in cell
death following SOD administration in sympathetic neurons undergoing apop-
tosis. Oxidative stress, however, is not essential for all apoptotic cell death.
Near-anaerobic conditions, in which the generation of ROS is absent or greatly
reduced, can inhibit apoptosis induced by ROS-generating agents but not by
other means (37).

Excitotoxicity
Glutamate is the major excitatory neurotransmitterin the brain. Three subtypes
of glutamatergic ionotropic receptors exist that are named for their pharma-
cologic agonists: N-methyl-D-aspartate (NMDA), and the non-NMDA agonists
quisqualate/a-amino-3-hydroxy-5-methyl-4-isox~olepropionicacid (AMPA)
and kainic acid (KA) (38) (Figure 2). The recent cloning of the genes encoding
the polypeptides that form the various glutamate receptors revealed a high
level of molecular heterogeneity, pharmacologic diversity, and biophysical
variation (39). Notably, it is now known that posttranscriptionalmRNA editing
transforms non-NMDA receptors from permitting the passage of Caz+ to sus-
taining only Na+ currents. Excitotoxicity refers to the excessive activation of
glutamate receptors that results in neuronal death. Excitotoxicity is important
in neuronal degeneration following acute insults such as hypoxia, ischemia,
and trauma (40, 41). Circumstantial evidence exists that excitotoxicity may
play a role in neurodegenerative diseases such as AD, ALS, PD, and Hunt-
ington's disease (HD). Although sodium influx through glutamate-gated ion
channels may mediate acute forms of neuronal degeneration, calcium appears
to be critical for delayed cell death induced by activation of NMDA and
non-NMDA receptors (42). Portrera-Cailliau et al(43) reported the occurrence
of both apoptotic and necrotic cell death in excitotoxic striatal lesions and in
HD striatum.
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVESTRESS 89

Growing data implicate oxidative stress as a mediator of excitotoxic cell


death (41, 44, 45). Following activation of NMDA and KA receptors, the
number of free radicals increases (46,47),oxidative damage to lipids occurs,
and antioxidants protect against cell death (44,48-51). These findings suggest
that glutamate receptor-linked processes may activate intracellular pathways
that produce free radicals, uncouple mitochondrial electron transport, depress
cellular defense systems, or a combination of these. In addition, free radicals
themselves can increase the release and decrease the reuptake of glutamate,
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

thus leading to increased glutamate in the synaptic cleft (52). This may lead
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

to a self-perpetuating cycle in which the activation of glutamate receptors


increases free radicals, which may lead to further receptor activation.

INTRACELLULAR PATHWAYS IMPORTANT IN FREE


RADICAL GENERATION

Recent advances have been made in the biochemistry and pharmacology of


specific intracellular pathways that may mediate oxidative stress in neurode-
generative diseases. Mitochondrial electron transport, NO-associated path-
ways, arachidonic acid (AA) metabolism, and protease activation may directly
or indirectly contribute to oxidative stress. We review evidence for involve-
ment of these pathways in both excitotoxic cell death and specific in vitro and
in vivo models of AD, PD, and ALS.

Mitochondrial Electron Transport Chain


Mitochondria are one of the major intracellular sources of free radicals (1).
Leakage of electrons along the electron transport chain, especially in the
ubiquinone B-cytochrome p region of complex III, results in the production
of ROS. Mitochondrial free radical production is increased by factors that
stimulate respiration, such as low concentrations of NAD-linked substrates,
succinate, ADP, and oxygen. Mitochondria are critical in buffering intracellu-
lar CaZ+(53), and the mitochondrial sequestration of Cat+can uncouple respi-
ration and produce metabolic acidosis (54). Dykens (55) recently demon-
strated that intact mitochondria generate OH. when exposed to pathologic
concentrations of Caz+ and ADP.
Mitochondria may be important in the induction of oxidative stress by
excitotoxins. Activation of glutamate-gated ion channels puts the mitochondria
at double jeopardy for generating ROS. First, the depolarization activates the
neuronal oxidative metabolism to replenish ATP utilized by the Na+ pumps.
Secondly, by increasing intracellular calcium, the sequestration of calcium
uncouples mitochondrial respiration. The increased production of ROS may
Annual Reviews
www.annualreviews.org/aronline

90 SIMONIAN & COYLE

damage mitochondrial proteins, DNA, and lipids, which may result in further
generation of ROS and energy depletion (Figure 2).
Circumstantial evidence exists that defects in mitochondrial energy meta-
bolism may cause neuronal degeneration in neurodegenerative diseases (56).
Inhibition of succinate dehydrogenase of the electron transport chain (complex
11) and Kreb's cycle by 3-nitropropionic acid (3-NP) can produce pathology
in the striatum similar to HD (57). Alterations in mitochondria energy meta-
bolism play a role in the degeneration of nigral neurons in PD. l-Methyl-4-
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

phenyl 1,2,3,6 tetrahyropyridine (MI"), a by-product of synthesis of the


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

designer drug isomeperidine, produces dopaminergic neuronal degeneration


and parkinsonian symptoms in humans and nonhuman primates (58). 1-Meth-
yl4phenylpyridinium (MPP+),produced by the catabolism of M I T P by mono-
amine oxidase B (MAO-B) in glia, is selectively and avidly taken up into
dopaminergic neurons by the dopamine transporter. Within dopaminergic neu-
rons, MPP+is concentrated by the electrochemical gradient into mitochondria.
MPP+ selectively inhibits complex I of the mitochondrial electron transport
chain and induces neuronal degeneration. Evidence exists that similar mito-
chondrial dysfunction may occur in idiopathic PD. Complex I defects have
been consistently found in individuals with PD. Reductions have been found
in the substantia nigra but not in other brain regions (59, 60). There are
conflicting reports on the existence of complex I defects outside the central
nervous system, such as in platelets (for review, see 61).
Evidence exists that excitotoxicity may play a critical role in cell death
induced by mitochondrialenergy failure. In vitro, inhibition of oxidative phos-
phorylation renders neurons vulnerable to what would otherwise be nontoxic
levels of glutamate (62). The reduction in cellular energy stores may cause a
reduction in membrane potential that relieves the voltage-sensitivemagnesium
block of NMDA receptors, thereby allowing persistent activation by endo-
genous glutamate (56). In support of this hypothesis, glutamate antagonists
have been found to reduce neuronal death induced by the mitochondrial toxins
3-NP and MPP+ (57, 63), although the latter finding has been contested (64).
Recent evidence suggests oxidative stress plays a crucial role in neuronal
degeneration induced by mitochondrial toxins. ROS are produced following
inhibition of the electron transport, and in particular, increases in Of are
observed with inhibition of complex I by MPP+ (65,66). U-78517F,a potent
inhibitor of iron-catalyzed lipid peroxidation, and DMSO, a OW scavenger,
can protect nigral neurons from MPP+ toxicity (67). Furthermore, transgenic
mice expressing a fivefold increase in Cu/Zn SOD are resistent to the neuro-
toxic action of M I " (68). Schulz et a1 (69) recently suggested a potential
role for nitric oxide in MPTP-induced neuronal degeneration after finding
evidence that nitric oxide synthetase (NOS) inhibitors prevent nigral degen-
eration in mice. Taken together, these results suggest that ROS generated from
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVESTRESS 91

the mitochondrialelectron transport chain and/or secondarily from intracellular


pathways linked to glutamate receptor activation may cause degeneration of
dopaminergic neurons by MPTP and MPP+.

Nitric Oxide
NO. is a stable free radical and a diffusable liposoluable second messenger
that activates guanylate cyclase (70). Under physiologic conditions, N O plays
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

a role in a variety of functions, including synaptogenesis, synaptic plasticity,


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

memory formation, cerebral blood flow, and neuroendocrine secretion. NO.


is formed from the conversion of L-arginine to L-citrulline by NOS. At least
three isoforms of NOS exist: the constitutively expressed, calcium-dependent
forms found in neurons and in endothelial cells; and an inducible, non-cal-
cium-dependent form, iNOS, found in macrophages, astrocytes, and microglia.
Elevation of neuronal intracellular calcium, through activation of the NMDA
receptor in particular, leads to the calcium- and calmodulin-dependentprotein
kinase-linked activation of NOS and NO. formation (71) (Figure 2).
N O has been hypothesized to be an important mediator of neuronal death
under pathologic conditions. NOS inhibition and removal of the NO precursor,
L-arginine, can reduce excitotoxic and hypoxic-ischemic neuronal cell death
(72-76). NO. reacts in biologic systems with oxygen, Of,and transition
metals (77). Reactions with metal- and thiol-containing proteins can lead to
alterations in enzyme activities or protein functions. For example, NO. can
attack iron-containingcomplexes in the electron transport chain that may result
in impaired energy metabolism. Hewett et a1 (78) recently found that NMDA
toxicity was potentiated by inducible NOS in mixed cortical-glial cell cultures,
suggesting a contribution of astrocytic NO. production in neuronal degenera-
tion (78).
The real culprit in NO-mediated toxicity may be ONOO-, which is formed
by the reaction of O$ with NO (Figure 1) (4, 11, 79). This reaction is three
times faster than the rate at which SOD scavenges Of but is normally mini-
mized by high SOD activity and low % and NO. concentrations. Under
conditions in which concentrations of N O and Of are increased andor con-
centrations of SOD are decreased, however, the formation of ONOO- may be
favored. ONOO- can react rapidly with proteins, lipids, and DNA and, unlike
OH or Of,is stable to diffuse up to several cell diameters. The attack of
ONOO- on proteins causes nitration of tyrosine residues. ONOO- causes
inhibition of complex II and IV of the electron transport chain and promotes
calcium efflux from mitochondria (80,81).
NO. and related species appear to play a dual role, having both detrimental
and protective effects. Under certain experimental conditions, NOS inhibition
and removal of the NO. precursor L-arginine is not only ineffectual, but at
Annual Reviews
www.annualreviews.org/aronline

92 SIMONIAN & COYLE

times potentates neuronal cell death (82-86). Although the exact reason(s) for
these conflicting results are unclear, recent studies suggest at least three pos-
sibilities. First, the redox state of the cell may determine the predominant
species and cellular effect of N O (79, 87). Under reducing conditions, N O
is preferentially formed and can react with 0-i. to produce the toxic ONOO-.
Under oxidizing conditions, the ion (NO) is preferentially formed and can
reduce excitatory neurotransmission through the Nh4DA receptor by binding
to the redox site and decreasing receptor activity (79, 88). Second, the per-
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

centage of NOS-containing neurons in primary cultures may influence toxicity


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

in vitro (89). NOS-containing neurons, such as cerebellar granule cells and a


subset of cortical neurons, appear to be insensitive to N O toxicity (85, 90).
Third, differential inhibition of neuronal versus endothelial NOS activities may
also contribute to the dual protective and potentiating role of nonselective NOS
inhibitors in vivo. Inhibition of endothelial NOS in addition to neuronal NOS
may alter cerebral blood flow and produce detrimental effects that overshadow
the protective effects of neuronal NOS inhibition. This hypothesis is supported
by recent data from mice transgenic for null mutations of neuronal NOS that
demonstrate that the absence of neuronal NOS protects against ischemic brain
injury, whereas the additional loss of endothelial NOS potentiates injury (91).

Arachidonic Acid Metabolism


AA, a 20-carbon unsaturated fatty acid, is a major component of excitable
membranes (92). It is liberated from membranes by activation of PLA, or
phospholipase C-diacyglycerol lipase or in the synthesis of platelet-activating
factor. One of the major functions of AA is to serve as a source of biologically
active lipids, such as prostaglandins, leukotrienes, and thromboxanes. AA,
through its ability to diffuse in and out of cells, serves as both an intra- and
intercellular messenger and plays a role in synaptic plasticity and long-term
potentiation in the brain (93).
Once formed, AA is further metabolized by 5- and 12-lipoxygenases and
cyclooxygenase (94). This metabolism is associated with the production of
ROS, since enzymes in phospholipid metabolism are mixed-function oxidases
that utilize molecular oxygen. In addition to the lipoxygenase and cyclooxy-
genase pathways, ROS can also be generated by the P450 system and by
cooxidation of AA with xanthine oxidase (XO). Bazans finding that hypoxic-
ischemic injury produces a rapid accumulation of free AA in the brain first
suggested a role for AA in neuronal death (95). It was subsequently found that
AA increases Of in brain slices and that the lipid peroxidation and neuronal
degeneration induced by AA can be prevented by vitamin E and lipoxygenase
inhibitors (96, 97).
In the brain, stimulation of glutamate receptors has been shown to increase
Annual Reviews
www.annualreviews.org/aronline
OXIDATIVE STRESS 93

the release of AA and its eicosanoid metabolites. Activation of NMDA recep-


tors on primary neurons in vitro results in activation of PLA,, a calcium-de-
pendent enzyme, and the release of arachidonic acid (98-100) (Figure 2). The
importance of AA in mediating toxicity by excitatory amino acids is supported
by the finding that the lipoxygenase inhibitor, nordihydroguaiaretic acid, re-
duces NMDA and KA toxicity in primary cultures of neurons (101, 102). A
relationship between NMDA receptor activation, ROS, and AA was demon-
strated by the use of electron paramagnetic resonance to detect free radical
species (47). Activation of NMDA receptors in cultured neurons induced an
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

increase in that was abolished by the PLA, inhibitor mepacrine. Taken


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

together, these results suggest that AA and metabolites may be a source of


damaging ROS following glutamate receptor activation.
The AA pathway may both promote and protect against neuronal degenera-
tion. AA reduces glutamate uptake into astrocytes, thereby increasing gluta-
mate concentrations in the synaptic cleft (103-105). Miller et al (106) have
reported that AA also potentiates current through NMDA receptors and am-
plifies increases in intracellular calcium. AA may thus exacerbate and/or
induce excitotoxicity by inhibiting glutamate uptake and raising intracellular
calcium concentrations (105,106). In contrast, AA has been reported to depress
synaptic transmission in striatal neurons and calcium channel currents, via
protein kinase C and ROS, in cerebellar granule cells. (107, 108). At low
concentrations, AA promotes neurite elongation (97). PLA,, the calcium-de-
pendent enzyme that catalyzes the release of AA following glutamate receptor
activation, itself reported to preferentially remove oxidized fatty acid from
membranes and thus may lower oxidative stress (109). These data thus suggest
that a balance between protective and toxic properties of AA pathways may
exist, although the specific conditionsthat favor protection over toxicity remain
poorly understood.

Protease Activation
Proteases play an important role in the normal turnover of proteins and in
structural changes in the nervous system, such as neurite outgrowth and mi-
gration (1 10). Mounting evidence also suggests that activation of proteases
may be an important downstream event in cell death induced by a variety of
stimuli (54,111). For example, a major proapoptotic gene identified in Caeno-
rhabdiris elegans, ced-3, encodes a putative cysteine protease related to the
mammalian interleukin-1honverting enzyme (ICE) (1 12). Expression of ICE
in mammalian cells induces apoptosis and inhibition of ICE by the cowpox
gene crmA can prevent degeneration of sympathetic neurons induced by nerve
growth factor deprivation (1 13, 114).
Proteases are important in the breakdown of oxidatively modified proteins
Annual Reviews
www.annualreviews.org/aronline

94 SIMONIAN & COYLE

(13, 115). The susceptibility of oxidatively modified proteins to protease deg-


radation may depend on the level of oxidative stress (1 16). For example, in
mammalian cells in vitro, low levels of ROS induce proteolysis, whereas high
levels decrease proteolysis, possibly due to the cross-linking, aggregation, and
decreased solubility of proteins. The free radical-scavenging enzymes SOD
and catalase are themselves vulnerable to oxidative damage and protease
degradation (1 15). The inactivation of these important defense enzymes could
potentiate cellular oxidative stress. In endothelial cells, protease inhibition
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

prevents free radical-mediated increases in intracellular calcium and cell death


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

(1 17).
Because calcium plays a central role in cell death in excitable cells, the
calciumdependent cysteine protease calpain has been investigated as a me-
diator of cell death in the nervous system (111, 118). Two types of calpain
exist, which differ in their sensitivity to calcium: Calpain I is activated by
calcium in the low micromolar range, whereas calpain I1 requires high micro-
molar concentrations. By binding to specific sites on the enzyme, calcium
induces autoproteolysis of calpain, thereby activating the protease. Calpain
proteolysis is limited in scope and results in alterations rather than destruction
of substrates. Activated calpain cleaves a selective subset of proteins that
includes cytoskeletal proteins, membrane proteins, and signal transduction
molecules.
Activation of the calciumdependent cysteine protease calpain has been
implicated in excitotoxic and in some types of apoptotic cell death (1 19, 120).
After NMDA and non-NMDA receptor activation, elevated intracellular cal-
cium causes autoproteolytic cleavage of calpain I and selective degradation of
calpain-sensitive substrates. These changes occur rapidly, which suggests that
proteolysis is not restricted to the terminal phases of cell death. In vitro,
neuronal death induced by hypoxia and AMPA receptor activation is reduced
by calpain inhibitors (76, 121, 122). In vivo, the proteolytic pattern of cy-
toskeletal protein degradation observed following transient ischemia and the
administration of NMDA and kainate matches that induced by calpain (1 19,
123, 124). Calpain inhibitors have been reported to prevent excitotoxic and
hypoxic-ischemia cell death in vivo (125-127). In cultures of cerebellar gran-
ule cells, however, a dissociation between calpain activation and excitotoxic
cell death has been seen, which may reflect alternative calcium-dependent
pathways in these cells (128, 129). Circumstantial evidence suggests that
calpain activation may be important in neuronal degeneration in AD. Increases
in activated forms of calpain as well as spectrin breakdown products have been
observed in brains from individuals with AD compared to aged controls and
those with HD (130).
Calpain activation has been linked to oxidative stress through XO (2). XO
is an enzyme that catalyzes the oxidation of hypoxanthine and xanthine to uric
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVE STRESS 95

acid, yielding Of.The enzyme exists primarily as the dehydrogenase form


(XD) in the brain and other tissues (131). Conversion to the oxidase form
results in the generation of ROS. This enzyme has been implicated in reper-
fusion tissue injury in experimental models (2). It had been hypothesized that
a calcium-dependent protease such as calpain would be a likely candidate to
mediate conversion of XD to XO during ischemia. In a purified preparation
of XD from rat liver, however, calpain I and II did not convert XD to XO,
suggesting that another protease may be important (132). Furthermore, the
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

restricted endothelial distribution of XO in the brain suggests that this pathway


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

may not be important in neuronal degeneration in neurodegenerative diseases


(133). Thus, despite evidence that both oxidative stress and calpain activation
are important in excitotoxicity, the link between the two is poorly understood.

NEURODEGENERATIVE DISEASES

Amyotrophic Lateral Sclerosis


ALS is characterized by progressive degeneration of motor neurons in the
spinal cord and brain stem. Multiple missense mutations in the cytosolic CuEn
SOD (SOD-1) gene have been identified in a subset of families with familial
ALS (FALS) (134). The majority of the mutations effect the structural back-
bone of the enzyme and not the active site directly (135). Reduced SOD activity
has been reported in red blood cells, lymphoblastoid cell lines and brain tissue
from many individuals with these mutations (135-38). Transfection experi-
ments, however, do not support a dominant negative mutation (138).
Several lines of evidence now suggest that motor neuron degeneration in at
least some of the FALS patients with SOD-1 mutations results from a gain of
function rather than from a loss of SOD-1 activity alone. First, dominantly
inherited mutations are usually gain of function mutations. Second, transgenic
mice that express high levels of the ALS mutant human SOD protein can
develop clinical and pathologic features of ALS with normal or elevated levels
of SOD activity (139, 140). Under these circumstances, the normal comple-
ment of mice SOD genes are present and expressed, so functional SOD activity
is actually increased. Third, lymphoblastoids from individuals with the G37R
mutation demonstrate normally active heterodimeric SOD activity (138).
Lastly, in transfected neuronal cell lines, FALS mutations were found to
promote apoptotic cell death despite retaining normal SOD activity, demon-
strating a dissociation between SOD1 activity and the induction of cell death
(141).
The gain of function that may arise from mutations in SOD-1 is unknown.
One hypothesis proposes that these mutations cause copper andor zinc toxicity
(142, 140). Normal SOD-1 binds copper and zinc very avidly. Mutations in
Annual Reviews
www.annualreviews.org/aronline

96 SIMONIAN & COYLE

SOD-1 may decrease binding of copper and zinc, leading to an increase in the
intracellular concentration of these free metals. Both zinc and copper can be
neurotoxic (143,144). Copper toxicity can induce oxidative stress presumably
through its reaction with H,Oz to produce OW. Demonstration of an elevation
of copper and/or zinc in individuals and transgenic mice with SOD-1 mutations
would support this hypothesis. An alternative hypothesis is that mutations in
SOD-I may increase nitration of tyrosine residues by ONOO-, which would
alter protein phosphorylation and signal transduction. ONOO- can react with
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

SOD-I to form an intermediate that nitrates tyrosine residues (145). Beckman


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

et a1 (146) have proposed that mutations in SOD-1 may disrupt the active site
of the enzyme and permit greater access of ONOO- to copper. This may lead
to increased nitration by ONOO- of critical cellular targets, such as neurofila-
ment proteins. Demonstration of an elevation of nitrotyrosine residues in
individuals and transgenic mice with SOD-I mutations would support this
hypothesis. Few studies to date have examined brains of individuals with ALS
for evidence of oxidative damage. In one study, an elevation in protein carbonyl
groups, a marker of oxidative protein damage, was found in the frontal cortex
of individuals with sporadic ALS (137). Further studies of oxidative cellular
damage in tissue from individuals with FALS and sporadic ALS and from the
SOD-I transgenic mice are needed.

Parkinson 's Disease


PD is characterized by loss of dopaminergic neurons in the substantia nigra
pars compacta. These neurons are believed to be particularly vulnerable to
oxidative stress because of the ROS normally generated in dopamine metabo-
lism (147). H202 is generated in the synthesis of dopamine by tyrosine hy-
droxylase, in its catabolism by monoamine oxidase, and by nonenzymatic
autoxidation. Nigral neurons contain neuromelanin, which can bind metals
such as iron and promote the reaction of H202 with reduced metals to form
the highly reactive OW.
Alterations in pro- and antioxidant molecules have been reported in post-
mortem tissue from individuals with PD. Increased total iron has been found
in the substantia nigra in PD (148-150). Iron could increase oxidative stress
by promoting the formation of OW from H202 via the Fenton reaction. Re-
ductions in glutathione levels in the nigra have been reported (148, 151-153).
These reductions were not detected in other neurodegenerative diseases in
which nigral cell loss occurs, suggesting they are specific to PD and not
secondary to cell loss alone. Decreases in glutathione have also been found in
the substantia nigra in individuals with incidental Lewy bodies at postmortem,
a potential marker of preclinical PD, suggesting that alterations in glutathione
are an early event (154). Reductions in glutathione levels could promote or be
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVE STRESS 97

a consequence of oxidative stress, or both. Because GSH is involved in the


detoxification of H,O,, reductions in GSH could result from increased con-
centrations of H2OZand in the presence of metals, the highly reactive OW.
The presence of lipid peroxidation and oxidative DNA damage further supports
the existence of oxidative stress in PD (155-157).
The hypothesis that oxidative stress is important in neuronal degeneration
in PD was tested in a large clinical study of vitamin E and deprenyl, an inhibitor
of MAO-B, in individuals with idiopathic PD (the DATATOP study) (158).
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

Deprenyl, but not vitamin E, delayed functional disability in early untreated


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

patients with PD. Deprenyl, by inhibiting MAO-B, potentially limits H20,


formation from the oxidative metabolism of dopamine and may also have
antioxidant properties independent of MAO-B inhibition (159). Deprenyl, by
increasing dopamine concentration, had symptomatic effects on parkinsonian
symptoms, which confounded the results of this study. Thus, at this time, it is
unknown whether deprenyl delays disability in individuals because of an
antioxidant and neuroprotective effect or because of a symptomatic effect of
the drug.

Alzheimers Disease
AD is characterized by degeneration of neurons, especially pyramidal neurons
in the hippocampus, entorhinal cortex, and neocortical areas and cholinergic
neurons in the median forebrain. One of the neuropathologic hallmarks of AD
is the deposition of extracellular aggregates of the P-amyloid peptides (AP).
The finding that AP can be toxic to neurons in vitro has led to the hypothesis
that neuronal degeneration in AD is due to AP toxicity (160). Recent in vitro
evidence suggests that oxidative stress may contribute to AP toxicity. Increases
in H202were detected in cells following exposure to AP, and both vitamin E
and catalase prevented cell death (161, 162). Inhibitors of NADPH oxidase
and other flavin oxidases were also found to be protective, whereas NOS,
cyclooxygenase, and lipoxygenase inhibitors were not. AP has also been re-
ported to induce the release of reactive NO? from microglial cells (163). Free
radical peptides that can inactivate oxidation-sensitive enzymes and initiate
lipid peroxidation have been detected in aqueous solutions of AP (164, 165).
These findings suggest that amyloid itself may generate free radicals that can
damage neurons.
Oxidative stress may contribute to aggregation of soluble AP into insoluble
plaques. Soluble AP has been found to aggregate in vitro by the addition of
metal-catalyzed oxidation systems (166). In addition, antioxidants inhibited
aggregation. These results suggest that the aggregation of AP induced by free
radical attack may be a primary event that leads to amyloid plaque formation.
Polymerization of tau, the major component of intracellular neurofibrillary
Annual Reviews
www.annualreviews.org/aronline

98 SIMONIAN & COYLE

tangles (NFTs) (another pathologic hallmark in AD) has also been associated
with oxidative stress. Tronosco et al(167) has demonstrated that oxidation of
tau in vitro induces dimerization and polymerization of the protein into fila-
ments. Increases in advanced glycation end products (AGEs), lipid and protein
glycation, and oxidation products that are formed upon exposure to reducing
sugars have been detected by immunoblotting and immunohistochemistry in
brains with AD (168, 169). AGEmodified proteins form cross-links that de-
crease solubility and are also a source of ROS (170). Using immunohistochem-
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

istry, Yan et al (169) found that AGEs were colocalized with NFTs in AD.
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

They also found that AGE-recombinant tau generates ROS when transfected
into neuroblastoma cells in vitro (169).
Increases in heme oxygenase-1 (HO-l), a protein induced by oxidative stress
and heat shock, have been detected by immunoblottingand immunohistochem-
istry in brains with AD (169, 171, 172). HO-I immunoreactivity was found in
NFT-bearing neurons, senile plaques, and astrocytes in brains with AD but not
in control brains. Increases in HO-I immunoreactivity were also associated
with NFTs in progressive supranuclear palsy, a degenerative disease of basal
ganglia and brain stem, suggesting that a common pathophysiologic mecha-
nism may underlie tangle formation.
Levels of oxidative cellular damage are increased in the brains of individuals
with AD. Elevated products of lipid peroxidation occur in vulnerable cortical
regions in brains with AD, and NFT-bearing neurons have enhanced im-
munoreactivity for malondialdehyde, a marker of lipid peroxidation (169,
173-175). A threefold increase in mitochondrial 8-OHDG, a marker of oxi-
dized DNA, has also been reported in brains from individuals with AD com-
pared to elderly controls (176). Protein oxidative damage is suggested by the
selective reduction in glutamine synthetase, an enzyme particularly sensitive
to mixed function oxidation reactions, in brains from individuals with AD
compared to age-matched control brains (177). These results provide circum-
stantial evidence that oxidative stress exists in AD.

F'UTURE DIRECTIONS
There is growing evidence that oxidative stress may play an important role in
neuronal degeneration in PD, ALS, and AD. What is not yet known is whether
oxidative stress is primary, resulting from a defect in cellular defense mecha-
nisms or enhanced free radical production; secondary; or part of a final com-
mon pathway in neuronal degeneration. What is particularly daunting about
attempts to identify the first causes in the pathology of oxidative stress is that
oxidative stress involves an expanding, self-perpetuating, and reinforcing se-
ries of metabolic events, which promote the generation of ROS and impair
potential protective mechanisms. Like a spreading wildfire, the site of the
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVE STRESS 99

initiating spark may be obscured in its terminal stages. Similarly, interventions


at many points in the sequence of events may restore, temporarily, the cells
ability to defend against the ROS load. Thus, an intervention, such as vitamin
E, may be acting at a site quite remote from the initiating event. For these
reasons, genetic models, such as the recently developed mice transgenic for
the human SOD-I mutations in FALS, will provide the most effective way of
identifying the proximate source of the ROS insult.
The remarkable cellular selectivity of these neurodegenerative disorders
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

emphasizes the highly localized effects of ROS within the cell and points to
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

a confluence of factors that may render certain neuronal populations uniquely


vulnerable. Furthermore, the highly localized effects of ROS within the cell
also suggest that pharmacologic interventions, to be most effective, must exert
their effects at the sites initially responsible for ROS generation. Nevertheless,
recent clinical and basic studies hold out the promise of therapeutic and even
preventive treatments directed at mitigating the effects of ROS for this group
of neurodegenerative disorders.

Any Annual Review chapter, as well 8 ~ any


3 article cited in an Annual Review chapter,
may be purchased from the Annual Reviews Preprints and Reprints service.
1-800-347-8007; 411259-5017; email: arpr8class.ow

Literature Cited
1. Freeman BA, Crapo JD. 1982. Free tion. Arch Biochem. Biophys. 216: 178-
radicals and tissue injury. Lab. Invesr. 85
47:412-26 9. Emster L. 1987. DT diaphorase: a his-
2. McCord JM. 1985.Oxygen-derived free torical review. Chem. Script. 27:l-13
radicals in postischemic tissue injury. 10. Halliwell B, Borish ET, Pryor WA,
N. Engl. J. Med. 312:159-63 Ames BN, Saul RL, et al. 1987. Oxygen
3. HalliweU B, Gutteridge JMC. 1985. radicals and human disease. Ann. Inr.
Oxygen radicals and the nervous system. Med. 107:526-45
Trends Neurosci. 622-26 11. Beckman J. 1994. Peroxynitrite versus
4. Beckman JS, Beckman TW, Chen J, hydroxyl radical: the role of nitric oxide
Marshall PA, Freeman BA. 1990. Ap- in superoxide-dependentcerebral injury.
parent hydroxyl radical production by Ann. NYAcad. Sei. 738382-87
peroxynitrite: implications for endothe- 12. Stadtman E. 1986. Oxidation of proteins
lial injury from nitric oxide and super- by mixed-function oxidation systems.
oxide. Proc. Natl. Acad Sei. USA 87: Trends Biol. Sei. 11: 11-1 2
1620-24 13. Davies KJA. 1987. Protein damage and
5. Fridovich I. 1975. Superoxide dismu- degradation by oxygen radicals. I. Gen-
tases. Science 44:147-59 eral aspects. J. Biol. Chem. 2629895-
6. Sinet PM, Heikkila RE, Cohen G. 1980. 901
Hydrogen peroxide production by rat 14. Stadtman E. 1993. Oxidation of free
brain in vivo. J. Neurochem. 341421- amino acids and amino acid residues in
28 proteins by radiolysis and by metal cata-
7. Meister A, Anderson M. 1983. Glu- lyzed reactions. Annu. Rev. Biochem.
tathione. Annu. Rev. Biochem. 52:711- 62:797421
60 15. Oliver C, Ahn BA, Moerman U,Gold-
8. Lind C, Hochstein P, Ernster L. 1982. stein S , Stadtman ER. 1987. Age-related
DT-diaphorase as a quinone reductase: changes in oxidized proteins. J. Biol.
a cellular control device against semi- Chem. 2625488-91
quinone and superoxide radical forma- 16. Beckman JS, Chen J, Ischiropoulos H,
Annual Reviews
www.annualreviews.org/aronline

100 SIMONIAN & COYLE

Crow JP. 1994. Oxidative chemistry of 30. Troy CM, Shelanski ML. 1994. Down-
peroxynitrite. Methods Engno1. 233: regulation of coppedzinc superoxide
22940 dismutase causes apoptotic death in
17. Brawn K. Fridovich I. 1981. DNA PC12 neuronal cells. Proc. Natl. Acad.
strand scission by enzymatically gener- Sei. USA 91:638447
ated oxygen free radicals. Arch. Bio- 31. Ward J. 1977. Molecular mechanisms
chem. Biophys. 206414-19 of radiation-induced damage to nucleic
18. Kasai H, Nishimura S. 1984. Hydroxy- acids. A&. Radar. Biol. 5:18 1
lation of deoxyguanosine at the C-8 32. Myers LS. 1980. Free radical damage
position by ascorbic acid and other re- of nucleic acids and their components.
ducing agents. Nucleic Acids Res. 12: In Free Radicals in Biology, ed. W
2137-45 Pryor, 4:95. New York Academic
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

19. Kasai H, Crain PF, Kuchino Y, Nish- 33. Schraufstatter IU, Hyslop PA, Hinshaw
imura S, Ootsuyama A, et al. 1986. DB, S p r a g RG, Sklar LA, et al. 1986.
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

Formation of 8-hydroxyguanine moiety Hydrogen-peroxide-induced injury of


in cellular DNA by agents producing cells and its prevention by inhibitors of
oxygen radicals and evidence for its poly(ADP-ribose) polymerase. Proc.
repair. Carcinogenesis 7: 1849-5 1 Natl. Acad. Sei. USA 8324908-12
20. Ames BN, Shigenaga MK, Hagen TM. 34. Nicholson DW, Ali A, Thormbeny NA,
1993. Oxidants, antioxidants, and the Vaillancourt JP, Ding CK, et al. 1995.
degenerative diseases of aging. Proc. Identification and inhibition of the.
Natl. Acad. Sei. USA 90:791>7922 ICWCED-3 protease necessary for
21. Mecocci P, MacGarvey U, Kaufman mammalian apoptosis. Nature 37637-
A, Koontz D, Shoffner J, et al. 1993. 43
Oxidative damage to mitochondrial 35. Talley AK, Dewhurst S, Parry SW, Dol-
DNA shows marked age-dependent in- lard SC, Gummuluru S, et al. 1995.
creases in human brain. Ann Neurol. Tumor necrosis factor alpha-induced
34:609-16 apoptosis in human neuronal cells: pro-
22. Wyllie AH, Kerr JF, Cume AR. 1980. tection by the antioxidant N-acetylcys-
Cell death: the significance of apoptosis. teine and the genes bcl-2 and c d .
Int. Rev. Cytol. 68:251-306 Mol. Cell. Biol. 15:2359-66
23. Schwartman RA, Cidlowski JA. 1993. 36. Greenlund L, Deckwerth T, Johnson E.
Apoptosis: the biochemistry and m e 1995. Superoxide dismutase delays
lecular biology of programmed cell neuronal apoptosis: a role for reactive
death. Endocrine Rev. 14:133-51 oxygen species in programmed neuronal
24. Raff MC, Barres BA, B u m JS, Ishizaki death. Neuron 14:303-15
Y, Jacobson MD. 1993. Programmed 37. Jacobson M, Raff M. 1995. Programmed
cell death and the control of cell sur- cell death and Bcl-2 protection in very
vival: lessons from the nervous system. low oxygen. Nature 374:814-16
Science 262:695-700 38. Watkins JC, Olverman HJ. 1987. Ago-
25. Steller H. 1995. Mechanisms of genes nists and antagonists for excitatory
and cellular suicide. Science 267 1445- amino acids. Trends Neurosci. 10265-
49 72
26. Kane DJ, Sarafh TA, Anton R, Hahn 39. Hollmann M, Heinemann S. 1994.
H, Gralla EB, et al. 1993. Bcl-2 inhi- Cloned glutamate receptors. Annu. Rev.
bition of neural death: decreased gen- Neurosci. 17:31-108
eration of reactive oxygen species. 40. Choi DW. 1988. Glutamate neurotoxic-
Science 2621274-77 ity and diseases of the nervous system.
27. Hockenbery DM, Oltvai ZN,Yin X-M, Neuron 1 :623-34
Milliman CL, Korsmeyer SJ. 1993. 41. Coyle JT, Puttfarcken P. 1993. Oxida-
Bcl-2 functions in an antioxidant path- tive stress, glutamate, and neurodegen-
way to prevent apoptosis. Cell 75:241- erative disorders. Science 262589-95
51 42. Choi DW. 1995. Calcium: still center-
28. Ratan RR, Murphy TH, Baraban JM. stage in hypoxic-ischemic neuronal
1994. Oxidative stress induces apoptosis death. Trendr Neurosci. 1858-60
in embryonic cortical neurons. J. Neuro- 43. Portera-Cailliau C, Hedreen J, Price D,
chem. 62:37679 Koliatsos V. 1995. Evidence for apop
29. Rothstein JD, Bristol LA, Hosler B, totic cell death in Huntingtons disease
Brown RH, Kuncl RW. 1994. Chronic and excitotoxic animal models. J. Neu-
inhibition of superoxide dismutase pro- rosci. 15:377547
duces apoptotic death of spinal neu- 44. Dykens JA, Stem A, Trenkner E. 1987.
rons. Proc. Natl. Acad Sei. USA 91: Mechanisms of kainate toxicity to cere-
4155-59 bellar neurons in vitro is analogous to
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVE STRESS 101

reperfusion tissue injury. J. Neurochem. 3-nitropopionic acid. J. Neurosci. 13:


49~1222-28 4181-92
45. Beal MF. 1992. Mechanisms of excite 58. Tipton KE, Singer TP. 1993. Advances
toxicity in neurologic diseases. FASEB in our understanding of the mechanisms
J. 6:3338-44 of the neurotoxicity of MPTP and re-
46. Sun AY, Cheng Y, Bu Q, Oldfield F. lated compounds. J. Neurochem. 61:
1992. The biochemical mechanisms of 1191-206
the excitotoxicity of kainic acid. Mol. 59. Schapira AHV, Cooper J, Dexter D,
Chem Neuropathol. 1751-63 Jenner P, Clark JB, et al. 1989. Mito-
47. Lafon-Cazal M, Pletri S, Culcasi M, chondrial complex I deficiencyin Park-
Bockaert J. 1993. NMDA-dependent su- inson's disease. J. Neurochem. 5 4
peroxide production and neurotoxicity. 823-27
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

Nature 364535-37 60. Schapira AHV, Mann VM, Cooper JM,


Miyamoto M, Murphy TH, Schnaar RL, Dexter D, Daniel SE, et al. 1990.
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

48.
Coyle JT. 1989. Antioxidants protect Anatomic and disease specificity of
against glutamate-induced cytotoxicity NADH-CcQ reductase (complex I) de-
in a neuronal cell line. J. Pharmacal. ficiency in Parkinson's Disease. J. Neu-
Exp, Ther. 250:1132-40 rochem. 55:2142-45
49. Monyer H, Hanley DM, Choi DW. 61. Bowling AC, Beal MF. 1995. Bioener-
1990. 21-Aminosteroids attenuate exci- getic and oxidative stress in neurode-
totoxic neuronal injury in cortical cell generative disease. Life Sei. 561151-71
cultures. Neuron 5:121-24 62. Novielli A, Reilly JA, Lysko PG, Hen-
5 0. Miyamoto M, Coyle J. 1990. Idebenone neberry RC. 1988. Glutamate becomes
attenuates neuronal degeneration in- neurotoxic via the N-methyl-D-aspartate
duced by intrastriatal injection of exci- receptor when intracellular energy lev-
totoxins. Exp. Neurol. 1083845 els are reduced. Brain Res. 451:205-12
5 1. Puttfarcken PS, Getz RL, Coyle JT. 63. Turski L, Bressler K, Rettig KJ, Losch-
1993. Kainic acid-induced lipid peroxi- mann PA, Wachtel H. 1991. Protection
dation: protection with butylated hy- of the substantianigra from MPP neu-
droxytoluene and U78517F in primary rotoxicity by N-methyl-waspartate.Na-
cultures of cerebellar granule cells. ture 349414-18
Brain Res. 624:22>32 64. Sonsalla PK, Zeevalk GD, Manzino L,
52. Volterra A, Trotti C, Tromba C, Floridi Giovanni A, Nicklas WJ. 1992. MK-
S , Racagni G. 1994. Glutamate uptake 801 fails to protect against the dopa-
inhibition by oxygen free radicals in rat minergic neuropathology produced by
brain cortical astrocytes. J. Neurochem. systemic I-methy14phenyl-l,2,3,6tetra-
142924-32 hydropyridine in mice or intranigral
53. White RJ, Reynolds U. 1995. Mitochon- I-methyl-4-phenylpyridiniumin rats.
dria and Na'/Ca2' exchange buffer glu- J. Neurochem. 58:197942
tamate-induced calcium loads in 65. Boveris A, Chance B. 1973. The mito-
cultured cortical neurons. J. Neurosci. chondrial generation of hydrogen per-
15: 1318-28 oxide. J. Biochem. 134:707-16
54. Wang GJ, Randall RD, Thayer SA. 66. Hasegawa E, Takeshiga K, Oishi T,
1994. Glutamate-induced intracellular Murai Y, Minikami S. 1990. I-methyl-
acidification of cultured hippocampal Cphenylpyridium induces NADH-&-
neurons demonstrates altered energy pendent superoxide formation and
metabolism resulting from Ca" loads. enhances NADH-dependent lipid per-
J. Neurophysiol. 722563-69 oxidation in bovine heart submitochon-
55. Dykens J. 1994. Isolated cerebral and drial particles. Biochem. Biophys. Res.
cerebellar mitochondria produce free Commun 170 1049-55
radicals when exposed to elevated Ca" 67. Wu R-M, Murphy DL, Chiueh CC.
and Na': implications for neurodegen- 1994. Protection of nigral neurons
eration. J. Neurochem. 63584-91 against MPP'-induced oxidative injury
56. Bed MF, Hyman BT, Koroshetz W. by deprenyl (selegilene), U-785 17F, and
1993. Do defects in mitochondrial en- DMSO. N. Trends Clin. Neuropharma-
ergy metabolism underlie the pathology C O ~ .8: 187-88
of neurodegenerative diseases? Trendr 68. Przedborski S , Kostic V, Jackson-Lewis
Neurosci. 16:125-3 1 V, Naini AB, Simonetti S, et al. 1992.
57. Beal MF,Brouillet E, Jenkins BG, Fer- Transgenic mice with increased C a n -
rante RJ, Kowall NW, et al. 1993. superoxide dismutase activity are resis-
Neurochemical and histologic charac- tant to N-methyl-Cphenyl- 1,2,3,6tetra-
terization of striatal excitotoxic lesions hydropyndine-induced neurotoxicity. J.
produced by the mitochondrial toxin Neurosci. 12:165&67
Annual Reviews
www.annualreviews.org/aronline

102 SIMONIAN & COYLE

69. Schulz JB, Matthews RT, Muqit MMK, oxide pathway on neuronal injury in-
Browne SE, Beal MF. 1995. Inhibition duced by L-glutamate or hypoxia. Bio-
of neuronal nitric oxide synthase by chem. Biophys. Res. Commun 181:
7-nitroindazde protects against M R P - 456-64
induced neurotoxicity in mice. J. Neuro- 83. Lerner-Natoli M, Rondouin G, Debock
chem. 64:936-39 F, Bockaert J. 1992. Chronic NO syn-
70. Lowenstein CJ,Snyder SH. 1992. Nitric thase inhibition fails to protect hippo-
oxide, a novel biologic messenger. Cell campal neurones against NMDA tox-
70705-7 icity. NeuroReport 3:1109-12
7 1. Bred DS, Snyder SH. 1989. Nitric oxide 84. Pauwels PJ, Leysen JE. 1992. Blockade
mediates glutamate-linked enhancement of nitric oxide formation does not pre-
of cGMP levels in the cerebellum. Proc. vent glutamate-induced neurotoxicity in
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

Nutl. Acad. Sci. US4 869030-33 neuronal cultures from rat hippocampus.
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

72. Nowicki JP, Duval D, Poignet A, Scat- Neurosci. Left. 143:27-30


ton B. 1991. Nitric oxide mediates 85. Puttfarcken PS, Lyons WE, Coyle JT.
neuronal death after focal ischemia in 1992. Dissociation of nitric oxide gen-
the rat. Eur. J. Phurmacol. 204:33940 eration and kainate-mediated neuronal
73. Dawson VL, Dawson TM, London ED, degeneration in primary cultures of rat
Bredt DS, Snyder SH. 1991. Nitric oxide cerebellar granule cells. Neurophunna-
mediates glutamate neurotoxicity in pri- cology 31:565-75
mary cortical cultures. Proc. Natl. Acud. 86. Lafon-Cazal M, Culcasi M, Gaven F,
Sei. USA 886368-71 Pietri S, Bockaert J. 1993. Nitric oxide,
74. Moncada C, Lekieffre D, Avrin B, superoxide and peroxynitrite: putative
Meldrum B. 1992. Effect of NO syn- mediators of NMDA-induced cell death
thetase inhibition on NMDA- and is- in cerebellar granule cells. Neumphar-
chemia-induced hippocampal lesions. mucology 32:1259-66
NeuroReport 3:530-32 87. Lei SZ, Pan Z-H, Aggarwal SK,Chen
75. Dawson VL, Dawson TM, Uhl GR, HSV, Hartman J, et al. 1992. Effect of
Snyder SH. 1993. Mechanisms of nitric nitric oxide production on the redox
oxidemediated neurotoxicity in primary modulatory site of the NMDA receptor-
brain cultures. J. Neurosci. 13265141 channel complex. Neuron 8: 1087-99
76. Brorson JR, Manzolillo PA, Miller R.I. 88. Manzoni 0, Prezeau L, Marin P,
1994. Ca2' entry via AMPAKA recep- Deshager S, Bockaert J, et al. 1992.
tors and excitotoxicity in cultured cere- Nitric oxideinduced blockade of
bellar Purkinje cells. J. Neurosci. 14: NMDA receptors. Neuron 8653-62
187-97 89. Bredt D, Snyder S. 1992. Nitric oxide,
77. Stamler JS. 1994. Redox signaling: ni- a novel neuronal messenger. Neuron
trosylation and related target interac- 83-11
tions of nitric oxide. Cell 78931-36 90. Koh J, Choi DW. 1988. Vulnerability
7 8. Hewett SJ, Csernansky CA, Choi DW. of cultured cortical neurons to damage
1994. Selective potentiation of NMDA- by excitotoxins: differential susceptibil-
induced neuronal injury following in- ity of neurons containing NADPH-dia-
duction of astrocyte INOS. Neuron 13: phorase. J. Neurosci. 8:2153-63
487-94 91. Huang 2, Huang PL, Panahian NP,Dal-
79. Lipton S, Choi Y-B, Pan Z-H, Lei SZ, kara T, Fishman MC, et al. 1994. Effects
Chen HV, et al. 1993. A redox-based of cerebral ischemia in mice deficient
mechanism for the neuroprotective and in neuronal nitric oxide synthase. Sci-
neurodestructive effects of nitric oxide ence 265: 1883435
and related nitroso-compounds. Nature 92. Bazan NG. 1989. Arachidonic acid in
364526-32 the modulation of excitable membrane
80. Packer MA, Murphy MP. 1994. Per- function and at the onset of brain dam-
oxynitrite causes calcium efflux from age. Ann. NY Acud. Sei. 559: 1-16
mitochondria which is prevented by cy- 93. Williams El, Errington ML, Lynch MA,
closporin A. FEBS Len. 345:23740 Bliss TVP. 1989. Arachidonic acid in-
81. Bolanos JP, Heales SIR, Land JM,Clark duces long-term activity-dependent en-
JB. 1995. Effect of peroxynitrite on the hancement of synaptic transmission in
mitochondrial respiratory chain: differ- the hippocampus. Nature 341:739-42
ential susceptibility of neurons and as- 94. Piomelli D, Greengard P. 1990. Lipoxy-
trocytes in primary culture. J. Neuro- genase metabolites of arachidonic acid
chem. 64:1965-72 in neuronal transmembrane signalling.
82. Demerle-Pallardy C, Lonchampt MO, Trends Pharmacol. Sei. 11:367-7 1
Chabrier PE, Braquet P. 1991. Absence 95. Bazan NG. 1970. Effects of ischemia
of implication of the L-argininehitric and electroconvulsive shock on free
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVE STRESS 103

fatty acid pool in the brain. Biochim. in cultured striatal neurons. Neuron 11:
Biophys. Acta 218:l-10 633-44
96. Chan P, Fishman R. 1980. Transient 109. van Kuijk FJ, Sevanian A, Handelman
formation of superoxide. radicals in GJ, Dratz EA. 1987. A new role for
polyunsaturated fatty acid-induced brain phospholipase A 2 protection of mem-
swelling. J. Neurochem. 351004-7 branes from lipid peroxidation damage.
97. Okuda S, Saito H, Katsuki H. 1994. Trends Biol. Sci. 1231-34
Arachidonic acid toxic and trophic ef- 110. Siman R. 1992. Proteolytic mechan-
fects on cultured hippocampal neurons. sisms for the neurodegeneration of
Neuroscience 63:691-99 Alzheimer's disease. Am.-NYAcad. Sci.
98. Dumuis A, Sebben M, Haynes H, Pin 674 193-202
P,Bockaert J. 1988. NMDA receptors 111. Saido T, Sorimachi H,Suzuki K. 1994.
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

activate the arachidonic acid cascade. Calpain: new perspectives in molecular


system in striatal neurons. Nature 336: diversity and physiologic-pathologic in-
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

68-70 volvement. FASEB J. 8:814-22


99. Sanfeliu C, Hunt A, Patell AJ. 1990. 112. Yuan J, Shaham S, Ledoux S, Ellis J,
Exposure to N-methyl-maspartate in- Horvitz J. 1993. The C. elegans cell
creases release of arachidonic acid in death gene ced-3 encodes a protein simi-
primary cultures of rat hippocampal lar to mammalian interleukin-lp-con-
neurons and not in astrocytes. Brain verting enzyme.. Cell 75:641-52
Res. 526:24148 113. Miura M, Zhu H, Rotello R, Hartwieg
100. Lazarewicz J, Wroblewski J, Costa E. EA, Yuan J. 1993. Induction of apop
1990. N-methyl-D-aspartate-sensitive tosis in fibrobIasts by IL-lp-converting
glutamate receptors induce calcium-me- enzyme, a mammalian homolog of the
diated arachidonic acid release in pri- C. elegans cell death gene ced-3. Cell
mary culturesof cerebellar granule cells. 75:1-20
J. Neurochem. 55:1875-81 114. Gagliardini V, Fernandez P-A, Lee
101. Rothman SM, Yamada KA, Lancaster R, Drexler H, Rotello R, et al. 1994.
N. 1993. Nordihydroguaiaretic acid at- Prevention of vertebrate neuronal
tenuates NMDA neurotoxicity-action death by the crmA gene. Science
beyond the receptor. Neuropharmacol- 263:826-28
ogy 321279-88 15. Salo DC,Pacifici RE, Sin SW, Giulivi
102. Dugan L, Choi D. 1994. Excitotoxicity, C, Davies KJA. 1990. Superoxide dis-
free radicals, and cell membrane mutase undergoes proteolysis and frag-
changes. Ann Neurol. 35:S17-S21 mentation following oxidative modifi-
103. Yu A, Chan P, Fishman R. 1986. Effect cation and inactivation. J. Biol. Chem.
of arachidonic acid on glutamate and 265: 11919-27
gamma-aminobutyric acid uptake in pri- 16. Grune T, Reinheckel T, Joshi M, Davies
mary cultures of rat cerebral cortical K. 1995. Proteolysis in cultured liver
astrocytes and neurons. J. Neurochem. epithelial cells during oxidative stress.
1986:47 J. Biol. Chem 2702344-51
104. Barbour B, Szatkowski M, Ingledew N, 117. Geeraerts M, Ronveaux-Dupal M-F, Le-
Attwell D. 1989. Arachidonic acid in- masters J, Herman B. 1991. Cytosolic
duces prolonged inhibition of glutamate free Ca2' and proteolysis in lethal oxi-
uptake into glial cells. Nature 342818- dative injury in endothelial cells. Am.
920 J. Physiol. 261:C889-96
105. Volterra A, Trotti D, Cassutti P, Tromba 118. Wang KKW, Yuen P-W. 1994. Calpain
C, Salvaggion A, et al. 1992. High inhibition: an overview of its therapeutic
sensitivity of glutamate uptake to ex- potential. Trends Pharmucol. Sci. 15:
tracellular free arachidonic acid levels 412-19
in rat cortical synaptosomes and astro- 119. Siman R, Noszek J. 1988. Excitatory
cytes. J. Neurochem. 5960Cb6 amino acid actiate calpain I and induce
106. Miller B, Sarantis M, Traynelis S, structural protein breakdown in vivo.
Attwell D. 1992. Potentiation of NMDA Neuron 1:279-87
receptor currents by arachidonic acid. 120. Squier M, Miller A, Malkinson A, Co-
Nature 355722-25 hen J. 1994. Calpain activation in apop
107. Keyser D, Alger B. 1990. Arachidonic tosis. J. Cell. Physiol. 159:229-37
acid modulates hippocampal calcium 121. Caner H, Collins J, Harris S, Kassell N,
currentsvia protein kinase C and oxygen Lee K. 1993. Attenuation of AMPA-in-
radicals. Neuron 5:545-53 duced neurotoxicity by a calpain inhibi-
108. Fraser D, Hoehn K, Weiss S, MacVicar tor. Brain Res. 607:354-56
B. 1993. Arachidonic acid inhibits so- 122. Rami A, Krieglstein J. 1993. Protective
dium currents and synaptic transmission effect of calpain inhibitors against neu-
Annual Reviews
www.annualreviews.org/aronline

104 SIMONIAN & COYLE

ronal damage caused by cytotoxic hy- tissue localization of human xanthine


poxia in vitro and ischemia in vivo. oxidase. Biochim. Biophys. Acta 11 64:
Brain Res. 609:67-70 327-30
123. Said0 T,Yokota M, Nagao S. Yamaura 134. Rosen DR, Siddique T, Patterson D,
I, Tani E, et al. 1993. Spatial resolution Figlewicz DA, Sapp P, et al. 1993.
of fodrin proteolysis in postischemic Mutations in CdZn superoxide dismu-
brain. J. Biol. Chem 268:2523M3 tase gene are associated with familial
124. Roberts-Lewis J, Savage M,Marcy V, amyotrophic lateral sclerosis. Nature
Pinsker L, Siman R. 1994. Immunolo- 36259-62
calization of calpain I-mediated spectrin 135. Deng H, Hentati A, Tainer J, Iqbal Z,
degradation to vulnerable neurons to the Cayabyab A, et al. 1993. Amyotrophic
ischemic gerbil brain. J. Neurosci. 14: lateral sclerosis and structural defects
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

3934-44 in Cu,Zn superoxide dismutase. Science


Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

125. Lee K, Frank S, Vanderlish P, Ami A, 26 1:1047-5 1


Lynch G. 1991. Inhibition of proteolysis 136. Robberecht W, Sapp P, Viaene M,
protects hippocampal neurons from is- Rosen D, McKenna-Yasek D, et al.
chemia. Proc. Natl. Acad. Sci. USA 88: 1994. Cu/Zn superoxide dismutase ac-
7233-37 tivity in familial and sporadic amyotro-
126. Hong S-C, Goto Y, Lanzino G, Soleau phic lateral sclerosis. J. Neurochem. 62:
S, Kassell N, et al. 1994. Neuroprotec- 384-87
tion with a calpain inhibitor in a model 137. Bowling AC, Schulz JB, Brown RH,
of focal cerebral ischemia. Stroke 25: Bed MF. 1993. Superoxide dismutase
663-69 activity, oxidative damage, and mito-
127. Bartus RT, Baker KL, Heiser AD, Saw- chondrial energy metabolism in familial
yer S, Dean RL, et al. 1994. Post- and sporadic amyotrophic lateral scle-
ischemic adminstration of AK275, a cal- rosis. J. Neumchem. 61:2322-25
pain inhibitor, provides substantial pro- 138. Borchelt D, Lee M, Slunt H,Guamieri
tection against focal ischemic brain M, Xu Z S , et al. 1994. Superoxide
damage. J. Cereb. Blood Flow Metab. dismutase. 1 with mutations linked to
14537-44 familial amyotrophic lateral sclerosis
128. Di Stasi A, Gallo V, Ceccarini M, possess significant activity. Proc. Natl.
Petrucci T. 1991. Neuronal fodrin pro- Acad. Sci. USA 91:8292-96
teolysis occurs independently of excita- 139. Gurney ME, Pu H, Chiu AY, Dal Canto
tory amino acid-induced neurotoxicity. MC, Polchow CY, et al. 1994. Motor
Neuron 6:445-54 neuron degeneration in mice that ex-
129. Manev H, Favaron M, Siman R, Gui- press a human CuZn superoxide dis-
dotti A, Costa E. 1991. Glutamate mutase mutation. Science 264 1772-74
neurotoxicity is independent of calpain 140. Wong P, Pardo C, Borchelt D, Lee M,
I inhibition in primary cultures of cere- Copeland N, et al. 1995. An adverse
bellar granule cells. J. Neurochem. 57: property of a familial ALS-linked SOD1
1288-95 mutation causes motor neuron disease
130. Saito K-I, Eke JS, Hamos JE, Nixon characterized by vacuolar degeneration
RA. 1993. Widespread activation of cal- of mitochondria. Neuron 14:1105-16
cium-activated neutral proteinase (cal- 141. Rabizadeh S, Gralla E, Borchelt DR,
pain) in the brain in Alzheimers Gwinn R, Valentine JS, et al. 1995.
disease: a potential molecular basis for Mutations associated with amyotrophic
neuronal degeneration. Proc. Natl. lateral sclerosis convert superoxide dis-
Acad. Sci. USA 902628-32 mutase from an antiapoptotic gene to a
131. Wajner M, Harkness R. 1989. Distribu- proapoptotic gene: studies in yeast and
tion of xanthine dehydrogenase and oxi- neural cells. Proc. Natl. Acad Sci. USA
dase activities in human and rabbit 92:3024-28
tissues. Biochim. Biophys. Acta 991:79- 142. Brown R. 1995. Amyotrophic lateral
84 sclerosis: recent insights from genetics
132. Starck K, Seubert P, Lynch G, Baudry and transgenic mice. Cell 80587-92
M. 1989. Proteolytic conversion of xan- 143. Sheinberg H. 1988. The neurotoxicity
thine dehydrogenase. to xanthine oxi- of copper. In Metal Neumtoxicology,
dase: evidence against a role for ed S Bondy, K Prasad, pp. 55-60. Boca
calcium-activated protease (calpain). Raton, n:CRC
Biochem. Biophys. Res. Commun. 165: 144. Koh J, Choi D. 1988. Zinc alters exci-
858-64 tatory amino acid neurotoxicity on cen-
133. Moriwaki Y, Yamamoto T, Suda M, tral neurons. J. Neumsci. 8:2164-71
Nasako Y, Takahashi S, et al. 1993. 145. Beckman J, Ischiropoulos H, Zhu L,
Purification and immunohistochemical van der Woerd M, Smith C, et al. 1992.
Annual Reviews
www.annualreviews.org/aronline

OXIDATIVE STRESS 105

Kinetics of superoxide dismutase and 158. Group PS. 1993. Effect of tocopherol
iron catalyzed nitration of phenolics by and deprenyl on the progression of dis-
peroxynitrite. Arch. Biochem. Biophys. ability in early Parkinsons disease. N.
29843%45 Engl. J. Med. 328:17643
146. Beckman J, Carson M, Smith C, Kop 159. Wu R-M, Chiueh C, Pert A, Murphy
penol W. 1993. ALS, SOD and per- D. 1993. Apparent antioxidant effect of
oxynitrite. Nature 364584 L-deprenyl on hydroxyl radical forma-
147. Olanow C, Arendash G. 1994. Metals tion and nigral injury elicited by MPP+
and free radicals in neurodegeneration. in vivo. Eur. J. Pharmacol. 243:24147
Curr. Opin. Neurol. 7:54&58 160. Yankner B, Dawes L, Fisher S, Villa-
148. Riederer P, Sofic E, Rausch W-D, Komaroff L, Oster-Granite M, et al.
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

Schmidt B, Reynolds G, et al. 1989. 1989. Neurotoxicity of a fragment of


Transition metals. ferritin, glutathione,
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

the amyloid precursor associated with


and ascorbic acid in Parkinsonian brains. Alzheimers disease. Science 245:417-
J. Neurochem. 52515-20 20
149. Dexter D, Carayon A, Vidailhet M, 161. BeN C, Davis J, Cole GM, Schubert D.
Ruberg M, Agid F, et al. 1990. De- 1992. Vitamin E protects nerve cells
creased ferritin levels in brain in Park- from amyloid P protein toxicity. Bio-
insons disease. J. Neurochem. 52: chem. Biophy. Res. Commun 186:944-
16-20 50
150. Dexter D, Carayon A, Javoy-Agid F, 162. BeN C, Davis J, Lesley R, Schubert D.
Agid Y, Wells F, et al. 1991. Alterations 1994. Hydrogen peroxide mediates
in the levels of iron, ferriten and other amyloid P protein toxicity. Cell 77:817-
trace metals in Parkinsons disease and 27
other neurodegenerative diseases affect- 163. Meda L, Cassatella M, Szendrei G,
ing the basal ganglia. Brain 114 1953- Otvos L, Baron P, et al. 1995. Activation
75 of microglial cells by P-amyloid protein
151. Perry T, Godin D, Hansen S. 1982. and interferon-y. Nature 374:647-50
Parkinsons Disease: a disorder due to 164. Hensley K, Carney JM, Mattson M,
nigral glutathione deficiency? Neurosci. Aksenova M, Harris M, et al. 1994. A
Lett. 33:305-10 model for P-amyloid aggregation and
152. Sofic E, Lange KW, Jellinger K, Ried- neurotoxicity based on free radical gen-
erer P. 1992. Reduced and oxidizied eration by the peptide: relevance to
glutathione in the substantia nigra of Alzheimers disease. Proc. Nail. Acad.
patients with Parkinsons disease. Neu- Sei. USA 91:3270-74
rosci. Lett 142:12&30 165. Butterfield D, Hensley K, Harris M,
153. Sian J, Dexter D, Lees A, Daniel S, Mattson M, Carney J. 1994. P-amyloid
Agid Y, et al. 1994. Alterations in glu- peptide free radical fragments initiate
tathione levels in Parkinsons disease synaptosomal lipoperoxidation in a se-
and other neurodegenerative disorders quence-specific fashion: implications to
affecting the basal ganglia. Ann. Neurol. Alzheimers disease. Biochem. Biophys.
36~348-55 Res. Commun. 2007 10-1 5
154. Dexter D, Sian J, Rose S, Hindmarsh 166. Dyrks T, Dyrks E, Hartmann T, Masters
J, Mann V, et al. 1994. Indices of oxi- C, Beyreuther K. 1992. Amyloidogenic-
dative stress and mitochondrial function ity of beta A4 and beta A4-bearing
in individuals with incidental lewy body amyloid protein precursor fragments by
disease. Ann Neurol. 35:3844 metal-catalyzed oxidation. J. Biol.
155. Dexter D, Carter C, Wells F, Javoy-Agid Chem. 267:18210-17
F. Agid Y. et al. 1989. Basal lipid 167. Troncoso J, Costello A, Watson A,
peroxidation in substantia nigra is in- Johnson G. 1993. In vitro polymeriza-
creased in Parkinsons disease. J. Neu- tion of oxidized tau into filaments. Brain
rochem. 52:38149 Res. 613:313-16
156. Jenner P, Dexter D, Sian J, Schapira A, 168. Smith M, Taneda S, Richey P, Miyata
Marsden C. 1992. Oxidative stress as a S, Yan S-D, et al. 1994. Advanced
cause of nigral cell death in Parkinsons Maillard reaction products are associ-
disease and incidental lewy body dis- ated with Alzheimers disease pathol-
ease. Ann. Neurol. 32382487 ogy. Proc. Natl. Acad Sei. USA 91:
157. Sanchez-Ramos A, Ames BN. 1994. A 57 10-14
marker of oxyradical-mediated DNA 169. Yan S-D, Chen X, Schmidt A-M, Brett
damage (8-hydroxy-2-deoxygunosine) J, Godman G, et al. 1994. Glycated tau
is increased in the nigrostriatum of Park- protein in Alzheimers disease: a mecha-
insons disease brain. Neurodegenera- nism for induction of oxidative stress.
tion 3: 197-204 Proc. Natl. Acad. Sci. USA 91:7787-91
Annual Reviews
www.annualreviews.org/aronline

106 SIMONIAN L COYLE

170. Yan S-D, Schmidt AM, Anderson GM, 174. Hajimohammadreza I, Brammer M.
Brett J, Mora R, et al. 1994. Enhanced 1990. Brain membrane fluidity and lipid
cellular oxidant stress by the interaction peroxidation in Alzheimers disease.
of advanced glycation end products with Neurosci. Len. 112:333-37
their receptorshinding proteins. J. Biol. 175. Palmer A, Burns M. 1994. Selective
Chem. 2699889-97 increase in lipid peroxidation in the
171. Smith M, Kutty R, Richey P, Yan S-D, inferior temporal cortex in Alzheimers
Stem D, et al. 1994. Heme oxygenase-1 disease. Brain Res. 645:338-42
is associated with the neurofibrillary 176. Mecocci P, MacGarvey U, Beal MF.
pathology of Alzheimers disease. Am. 1994. Oxidative damage to mitochon-
1. Parhol. 145:42-47 drial DNA is increased in Alzheimers
172. Schipper H, Cisse S, Stopa E. 1995. disease. Ann Neurol. 36:747-51
Annu. Rev. Pharmacol. Toxicol. 1996.36:83-106. Downloaded from www.annualreviews.org

Expression of heme oxygenase1 in the 177. Smith CD, Carney JM,Stark-Reed PE,
Access provided by Jawaharlal Nehru University on 01/07/15. For personal use only.

senescent and Alzheimer-diseased Oliver CN, Stadtman ER, et al. 1991.


brain. Am. Neurol. 37:758-68 Excess brain protein oxidation and en-
173. Subbarao K,Richardson J, Ang L. 1990. zyme dysfunction in normal aging and
Autopsy samples of Alzheimers cortex in Alzheimers disease. Proc. Natl.
show increased lipid peroxidation in Acad. Sci. US4 881054043
vitro. J. Neurochem. 55:34245

You might also like