You are on page 1of 16

Neuroscience 145 (2007) 1233–1248

REVIEW
OXIDATIVE STRESS AND MITOCHONDRIAL DYSFUNCTION
IN NEURODEGENERATIVE DISEASES
E. TRUSHINAa AND C. T. MCMURRAYa,b,c* Mutations with direct effects on MT Function: FRDA 1240
a
Department of Molecular Pharmacology and Experimental Therapeu- Features of disease 1240
tics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA Abnormal regulation of antioxidants in MT in FRDA 1241
b
Mutations with direct effect on oxidative DNA damage 1242
Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 DNA damage response as a common link 1242
First Street SW, Rochester, MN 55905, USA Therapeutic implications of oxidative damage 1242
c
Molecular Neuroscience Program, Mayo Clinic, 200 First Street SW, Acknowledgment 1243
Rochester, MN 55905, USA References 1243

Abstract—In recent years, it has become increasingly clear In humans, oxidative DNA damage is considered an im-
that mitochondrial dysfunction and oxidative damage are portant promoter of neurological diseases and aging
major contributors to neuronal loss. Free radicals, typically
(Singh, 2006; Poulsen, 2005). In the cell, reactive oxygen
generated from mitochondrial respiration, cause oxidative
damage of nucleic acids, lipids, carbohydrates and proteins. species (ROS) are generated primarily by mitochondria
Despite enormous amount of effort, however, the mechanism (MT) (Reddy, 2006). To ensure efficient removal of MT-
by which oxidative damage causes neuronal death is not well generated ROS, the inner mitochondrial membrane incor-
understood. Emerging data from a number of neurodegen- porates a number of free radical scavengers such as vita-
erative diseases suggest that there may be common features min E, ascorbate, and glutathione. Additionally, there is an
of toxicity that are related to oxidative damage. In this review, enzymatic removal of free radicals by superoxide dis-
while focusing on Huntington’s disease (HD), we discuss
mutase (SODs). These lines of defense insure that forma-
similarities among HD, Friedreich ataxia and xeroderma pig-
mentosum, which provide insight into shared mechanisms of tion of ROS as a by-product of respiration does not dam-
neuronal death. © 2006 Published by Elsevier Ltd on behalf of age the cell. The defense can be compromised by either
IBRO. genetic mutations leading to decreased activity of the an-
tioxidants as in some familial cases of amyotrophic lateral
Key words: neurodegeneration, mitochondria, oxidative dam- sclerosis (ALS) with mutation in CuZn SOD (Mariani et al.,
age, Huntington’s disease, Friedreich ataxia, xeroderma pig- 2005), or by increased radical production. In either case,
mentosum.
oxidative damage can impart harmful consequences to the
cell (Harman, 1956).
Contents
HD 1233 The mechanism by which oxidative damage causes
Features of HD pathophysiology 1234 neuronal death is poorly understood. Therefore, an unre-
Mhtt alters the functions of its interacting partners 1234 solved issue is whether mitochondrial defects are the pri-
MT are dysfunctional in HD patients and in mouse models mary cause of toxicity or a secondary response to the
for disease 1235 damage. In this review, we consider characteristics of
Possible mechanisms of MT dysfunction three classes of neurodegenerative diseases with differ-
and toxicity in HD 1238
ent, but overlapping links to oxidative damage: Hunting-
Mutations in mtDNA and nuclear DNA 1238
Defects in MT functions 1238 ton’s disease (HD), Friedreich ataxia (FRDA), and xero-
Lipid/membrane damage 1239 derma pigmentosum (XP) (Fig. 1). XP arises directly from
inability to remove some forms of oxidative DNA damage,
*Corresponding author. Tel: ⫹1-507-284-1597; fax: ⫹1-507-284-9111.
E-mail address: mcmurray.cynthia@mayo.edu (C. T. McMurray).
FRDA arises from defects in MT and promiscuous Fenton
Abbreviations: BDNF, brain-derived neurotrophic factor; CREB, cyclic chemistry, and HD is representative of an “aggregation”
AMP response element-binding protein; cybrid, cytoplasmic hybrid; disorder where oxidative damage and MT defects are
FRDA, Friedreich ataxia; HAP1, huntingtin-associated protein 1; HD, thought to cause and/or contribute to toxicity. The goal of
Huntington’s disease; HIP1, huntingtin interacting protein 1; Hippi,
huntingtin interacting protein-1 protein interactor; htt, huntingtin; the review is to gain insight into whether oxidative damage
InsP3R1, inositol (1,4,5)-triphosphate receptor; mhtt, mutant hunting- is a common denominator for neurodegeneration.
tin; MnSOD, manganese superoxide dismutase; MSN, medium spiny
neurons; MT, mitochondria; mtDNA, mitochondrial DNA; MT-PTP, HD
mitochondrial permeability transition pore; NER, nucleotide excision
repair; NPC, Niemann-Pick type C1; polyQ, polyglutamine; ROS, re- HD is a hereditary autosomal dominant disorder in which
active oxygen species; SDH, succinate dehydrogenase; SOD, super-
oxide dismutase; TCR, transcription-coupled repair; XP, xeroderma the underlying mutation is an expanded CAG repeat in
pigmentosum; YAC, yeast artificial chromosome. exon 1 of the coding region of the HD gene (Fig. 2A). The
0306-4522/07$30.00⫹0.00 © 2006 Published by Elsevier Ltd on behalf of IBRO.
doi:10.1016/j.neuroscience.2006.10.056

1233
1234 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

Fig. 1. Oxidative DNA damage in neurodegenerative disorders can be caused directly and/or indirectly through altered mitochondrial function. Three
hereditary diseases mediate oxidative DNA damage and neuronal death through different pathways. Expression of mhtt in HD can directly increase
production of ROS and subsequent oxidative DNA damage through defects in MT. In FRDA, diminished levels of frataxin, a mitochondrial iron-binding
protein, directly affect MT function and ROS production. Lack of frataxin allows unbound iron to participate in Fenton chemistry with subsequent
generation of toxic hydroxyl radicals. In XP, neuronal death is caused by mutations in nucleotide excision repair proteins that interfere with the ability
to remove and repair oxidative DNA damage.

CAG triplet encodes the amino acid glutamine in the gene high molecular weight complexes and inclusion bodies.
product called huntingtin (htt) (The Huntington’s Disease Thus, despite an incomplete understanding of toxic mech-
Collaborative Group, 1993). The mutant huntingtin (mhtt) anism, it is generally accepted that in HD alteration and/or
has elongated polyglutamine (polyQ) stretch in the N-ter- sequestration of cellular targets are likely to be early caus-
minal region (⬎37 units) that affects its interaction with ative events of the neuronal dysfunction and death (Lee
htt-binding proteins and makes them susceptible to aggre- and Kim, 2006; Michalik and Van Broeckhoven, 2003;
gation (Harjes and Wanker, 2003). The onset of HD usu- Ross and Poirier, 2004).
ally happens in the fourth and fifth decades of life. The
phenotype is characterized by loss of medium spiny neu- Features of HD pathophysiology
rons (MSN) in the striatum, cognitive deterioration and
motor dysfunction (van Dellen et al., 2005) (Fig. 2B). The Mhtt alters the functions of its interacting partners.
CAG expansion process is dynamic, and growth of the Proteins targeted by mhtt are presumably inactivated by it
CAG tract can occur in the germ cells and in somatic cells or assume an alternate conformation (Harjes and Wanker,
with age. The elongated polyQ tract generates mhtt with 2003; Li and Li, 2004) (Fig. 3). Indeed, mhtt modifies the
increasingly aberrant properties (Lee and Kim, 2006; functions of many cytoplasmic and nuclear proteins often
Michalik and Van Broeckhoven, 2003). In patients, the found in aggregates (Harjes and Wanker, 2003; Li and Li,
progressive toxicity of HD is manifested by a phenomenon 2004). The N-terminal, truncated form of mhtt containing
called anticipation where the onset of the disease inversely polyQ tract can bind to and interfere with nuclear transcrip-
correlates with the length of the polyQ tract (The Hunting- tion factors such as cyclic AMP response element-binding
ton’s Disease Collaborative Group, 1993). protein (CREB) (Sugars et al., 2004), CREB binding pro-
While mutation analysis and transgenic animal models tein (CBP) (Sugars et al., 2004; Rao and Finkbeiner, 2003;
for HD have unequivocally identified the expanded polyQ Steffan et al., 2000), p53 (Steffan et al., 2000; Sawa, 2001;
tract as key for toxicity (Lee and Kim, 2006; Michalik and Bae et al., 2005; Feng et al., 2006), co-repressor (Kegel et
Van Broeckhoven, 2003), the mechanism by which mhtt al., 2002), transcriptional activator Sp1 and TAFII130 (Du-
progressively kills brain cells is poorly understood. PolyQ nah et al., 2002; Li et al., 2002). Disruption of CREB in the
regions self-associate to form polar zippers, the ␤-strands MT matrix in neurons decreases the expression of a sub-
capable to assemble into sheets or barrels using hydrogen set of mitochondrial genes, including the ND5 subunit of
bonds and promote aggregation (Ross and Poirier, 2004; complex I, and down-regulates complex I-dependent mito-
Perutz, 1999). In HD-affected areas of the caudate and chondrial respiration. Thus, mhtt can sequester CREB or
cortex, immunochemical detection reveals that mhtt forms any of its interacting partners by direct binding in either
E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248 1235

Fig. 2. Expression of mhtt causes selective neuronal loss in striatum. (A) The HD gene has a polymorphic region consisting of the CAG repeats in
exon 1. In normal individuals, the number of CAG repeats is typically below 35. Growth of the CAG repeats in the coding region of HD gene produces
mutant protein with elongated polyQ region and aberrant functions. CAG expansion is progressive; the length of polyQ directly correlates with the
severity of HD and inversely with the onset of the disease. (B) Magnetic resonance image of normal (left) and HD brain (right). Expression of mhtt
causes neuronal loss in the striatum (caudate (C) and putamen (P)) and cortex as the disease progresses. Relative to the normal brain, substantial
neuronal loss is observed in the C and P, typical of an advanced stage (grade IV) of HD. Additionally, diseased brains are characterized by large
invaginations in the cortex and general brain atrophy. GP, globus pallidus: V, ventricle; hip, hippocampus.

nuclear or MT compartments (Shimohata et al., 2000; calcium load, which, in turn, renders striatal neurons more
Wyttenbach et al., 2001; Nucifora et al., 2001; Gines et al., susceptible to toxicity (Tang et al., 2004).
2003). Mhtt also has altered binding affinity to HIP1. In this
Accumulating data suggest that a particularly important case, binding of mhtt is less strong relative to its normal
effect of mhtt expression is disruption of microtubules counterpart. The weaker interaction of mhtt with HIP1
(Trushina et al., 2003) and inhibition of vesicular trafficking. shifts the equilibrium toward free HIP1, which can interact
Htt normally interacts with trafficking motors and clathrin- with associated factors such as Hippi (huntingtin interact-
interacting proteins including Huntingtin interacting protein ing protein-1 protein interactor) (Tang et al., 2004; Majum-
1 (HIP1), Huntingtin interacting protein-related (HIP1R), der et al., 2006). Hippi forms a heterodimeric complex with
and Huntingtin-associated protein 1 (HAP1) (Harjes and HIP1 and, under conditions of mhtt over-expression, the
Wanker, 2003; Li et al., 1998; Engelender et al., 1997; HIP1/Hippi complex activates caspase-8 and initiates a
Tukamoto et al., 1997; Waelter et al., 2001). Consistent cascade of events leading to cell death. Steps include
with these protein interactions, htt appears to be involved cleavage of death agonist protein Bid, release of cyto-
in both axonal trafficking and endocytosis, and inhibitory chrome c and apoptosis inducing factor from MT, activa-
effects of mhtt on axonal trafficking have been directly tion of caspase-9/caspase-6 and nuclear fragmentation
demonstrated in squid axoplasm (Szebenyi et al., 2003), in (Majumder et al., 2006). The fact that Hippi activates
Drosophila (Gunawardena et al., 2003) and in mice caspase-6 is significant since proteolytic cleavage of mhtt
(Trushina et al., 2004) (Fig. 4). For example, htt together at the caspase-6 site appears to be a critical intermediate
with HAP1 enhances microtubule-based trafficking of an of cell death (Graham et al., 2006).
important neuronal growth factor, brain-derived neurotro-
phic factor (BDNF), and expression of mhtt interferes with MT are dysfunctional in HD patients and in mouse
that process (Gauthier et al., 2004). Mhtt binds with higher models for disease. Perhaps, most relevant to oxidative
affinity to HAP1 relative to htt, and interferes with BDNF damage is the fact that mhtt directly impairs motility of MT.
trafficking by competing with HAP1 for microtubule binding Real-time imaging of primary neurons from animals ex-
(Gauthier et al., 2004). HAP1 can also serve as an adaptor pressing mhtt with 72 polyQ reveals that the movement of
between htt and inositol (1,4,5)-triphosphate receptor elongated particles that by size and morphology resemble
(InsP3R1) in lipid bilayers of MSN (Tang et al., 2003). In MT (1.9 – 4.5 ␮m) is inhibited. The identity of the elongated
the presence of mhtt, HAP1 sensitizes calcium release by particles as MT has been confirmed by tetramethylrho-
the InsP3R1 leading to the increase in the intracellular damine, methyl ester staining (Fig. 4). Indeed, the motility
1236 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

Fig. 3. Cellular dysfunction caused by mhtt expression. Multiple protein–protein interactions and biological processes are altered in cells expressing
mhtt. Wild-type htt interacts with a large number of proteins that directly or indirectly regulate MT function and biogenesis. Mhtt has altered binding
affinity with interacting partners commonly affecting their functions. Diagram represents proteins and cellular functions that are affected by expression
of mhtt and could have an impact on MT dysfunction. Defective MT respond by over-production of ROS, and additional damage to MT and DNA would
be predicted. A vicious cycle eventually lead to neuronal damage and cell death.

of MT was strikingly impaired, and MT slowed down and SDH subunits are found in the cerebral cortex and cere-
stopped frequently in primary striatal neurons from mice bellum, two regions less vulnerable to degeneration in HD
expressing mhtt. Remarkably, MT in neurons from HD patients (Benchoua et al., 2006).
animals travel distances 70% shorter than their normal In transgenic animal models for HD, MT trafficking
counterparts in the retrograde direction (Trushina et al., defects are observed early in the embryo and appear
2004). It is not yet clear whether immobilization of MT before other signs of toxicity (Trushina et al., 2004). How-
leads to their eventual dysfunction. However, MT that lose ever, MT dysfunction progresses with development and is
their capacity to traverse the neurite heighten glutamate evident in older tissues by at least two measures: (1) the
excitotoxicity and alter calcium handling (Chang et al., diminished ATP production, and (2) the increased lactate
2006), and inability to move could result in insufficient local levels (a signature of MT dysfunction). Both have been
delivery of ATP. associated with HD in humans and in mouse models. A
In postmortem tissue from HD patients, severe defi- decrease in succinate oxidation ranging from 39 to 59%
ciencies in mitochondrial complex II and III and, some- also occurs in the striatum of HD patients (Browne et al.,
times, complex IV were found in the caudate (Mann et al., 1997; Tabrizi et al., 1999), and elevated lactate levels
1990; Gu et al., 1996). Defects in complex II may be most correlate with CAG repeat length in HD occipital cortex and
important for toxicity. Expression of both full-length and striatum (Koroshetz et al., 1997; Jenkins et al., 1998).
N-terminal fragments of mhtt causes increased sensitivity These energetic defects have been observed even in
to complex II inhibitor 3-nitropropionic acid, while the ef- asymptomatic HD gene carriers (Jenkins et al., 1998).
fects of complex I inhibition do not induce cell death (Ruan Perhaps not surprising, HD-related MT dysfunction is also
et al., 2004; Benchoua et al., 2006). Inhibition of MT com- observed in other tissues with higher ATP consumption
plex II appears to impart dysfunction by reducing the levels such as skeletal muscles. Myoblasts from presymptomatic
of complex II/succinate dehydrogenase (SDH) subunits Ip and symptomatic HD subjects are characterized by cellular
and Fp. As might be expected in this circumstance, the abnormalities in vitro such as mitochondrial depolarization,
SDH activity of the complex is diminished. Thus, in vitro, cytochrome c release, and increased caspase-3, -8, and -9
treatment with 3-nitropropionic acid recapitulates MT de- activities. Although initially thought to exist exclusively in
fects observed in human striatum and associated with the brain, mhtt-containing inclusion bodies are present in
human disease. No major changes in the expression of differentiated myotubes (Ciammola et al., 2006). In HD
E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248 1237

Fig. 4. Expression of mhtt inhibits trafficking of vesicles and MT in primary striatal neurons. (Top) Schematic diagram of axonal trafficking in neurons.
MT and vesicles are transported toward (retrograde) or away (anterograde) from the cells body. These trafficking functions influence secretion, Golgi
processing, MT biology, and energetic. (Bottom) Real time imaging of MT in axons in striatal neurons from mice expressing full-length mhtt. For
experiments with MT trafficking in living neurons, cells were treated for 5 min with tetramethylrhodamine dye (TMRM) (Invitrogen Corporation,
Carlsbad, CA, USA). The experiments were performed using confocal microscope LSM 510 (Carl Zeiss Inc., Oberkochen, Germany) with a
Plan-Apochromat 100⫻ (1.4 na) oil objective. Ar/Kr laser was set up to 568 nm for excitation; emission was set up to 585 nm and greater. TMRM was
washed away with fresh F-12K medium prior to imaging. Images were taken every 1 s at highest scan speed (0.9 s) for 10 min. A total of 600 images
was recorded per cell. Trafficking was analyzed using LSM 510 software that allowed animation of 600 images into a “movie.” Representative striatal
neuron (transmitted image t⫽0 s) and MT movement is shown on fluorescent images (t⫽0, 12, and 21 s). Reduced MT trafficking could cause
insufficient energy supply to the axonal growing cones and synaptic sites contributing to the neuronal dysfunction and altered synaptic transmission.
Aggregation observed later with disease progression can also significantly contribute to the defective MT trafficking.

pre-symptomatic and symptomatic patients, the effects in have been useful markers of oxidative damage (Floyd et
muscles are accompanied by a decrease in complex I al., 1990). In the R6/1 transgenic model for HD, progres-
activity, a decrease in phosphocreatine concentrations at sion of the neurological phenotype correlates with a pro-
rest, and a reduction in ATP synthesis during exercise gressive increase in striatal lipid peroxidation (Perez-
(Lodi et al., 2000; Arenas et al., 1998; Saft et al., 2005). Severiano et al., 2000). Similarly, oxidative damage in
The energetic defects observed in humans are repli- mitochondrial DNA (mtDNA) occurs in the parietal region
cated in rodent models for disease. Similar to humans, of human HD brain, but not frontal cortex or cerebellum
systemic administration of the mitochondrial complex II consistent with the regional specificity of neuronal vulner-
inhibitor, 3-nitropropionic acid in rats, causes neurobehav- ability in HD (Polidori et al., 1999). Dysfunction in HD is
ioral and pathological abnormalities consistent with HD accompanied by a rise of p53 expression in brain cells
(Beal et al., 1991). In muscles of the R6/2 animal model of (Bae et al., 2005). P53 is a surveillance protein and tran-
HD, cytochrome c oxidase activity is reduced and abnor- scription factor that is “turned on” in response to DNA
mal complex I-dependent respiration of MT is observed damage. Thus, the up-regulation of p53 in animals and
(Gizatullina et al., 2006). Lactate is significantly increased cells strongly suggests that neuronal death may in part
in the striatum from adult yeast artificial chromosome involve an oxidative DNA damage response (Kruman et
(YAC) 72 mice relative to embryo (Trushina et al., 2004). al., 2004; Hazra et al., 2001; Bjelland and Seeberg, 2003).
These in vitro data are consistent with those observed in Consistent with that notion, treatment of HD animals and
patients, and together implicate MT damage as relevant to humans with anti-oxidants such as creatine (Ferrante et
the disease. al., 2000; Andreassen et al., 2001) and coenzyme Q (Fer-
If such a mechanism is pertinent in vivo, then an in- rante et al., 2002; Schilling et al., 2001) has been benefi-
crease in free radical production from defective MT might cial, although not curative, in preliminary clinical trials.
initiate DNA damage in mhtt expressing animals. Indeed, It seems clear that MT damage is an intermediate of
recent data indicate that this is the case. Over-expression toxicity and promotes the release of destructive hydroxyl
of free radical scavenger, Cu/Zn SOD1 reverses the oxi- radicals and loss of ATP production. Unfortunately, the
dative stress-induced proteasomal malfunction, aggrega- plethora of cellular effects due to mhtt expression leaves
tion, and cell death observed in HD (Goswami et al., 2006). unresolved the issue of whether MT damage is a primary
Increases in DNA lesions such as 8-hydroxy-2=-deox- or secondary event in toxicity. Ironically, MT is both the
yguanosine (8-OHdG) and the extent of lipid peroxidation major source of ROS production, but also the major target
1238 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

of ROS damage. Therefore, oxidative damage to the MT MT proteins from lymphoblast cell lines of these family
may destroy the special defense mechanisms that main- members revealed no detectable alterations. These data
tain functional integrity and protection against ROS. In MT, suggest that the maternally inherited mitochondrial ge-
superoxide is converted to the less reactive hydrogen per- nome does not play a crucial role in determining age of
oxide by the mitochondrial matrix enzyme manganese su- onset in HD (Irwin et al., 1989). Similar conclusions were
peroxide dismutase (MnSOD) (van Loon et al., 1986; reached using cytoplasmic hybrid (cybrid) cells. Cybrid
Guidot et al., 1993). Hydrogen peroxide levels are in turn cells lack their own MT. Thus, mtDNA from a human donor
regulated by glutathione peroxidase (Takahashi et al., can be reintroduced into these cells to test for functional
1987; Esposito et al., 2000) and catalase (Michiels et al., defects coded for by MT genome (King and Attardi, 1989).
1994) activity, although the latter is not normally found in However, cybrid cell lines containing mtDNA from HD pa-
the MT (Zhou and Kang, 2000). Any remaining hydrogen tients do not differ from control cybrid lines in electron
peroxide is free to react with available iron to produce transport chain, oxidative stress or calcium homeostasis
hydroxyl radicals. The small amount of superoxide that is assays (Swerdlow et al., 1999). While mitochondrial dys-
not removed by MnSOD remains dangerous and can lead function in HD does not appear to be due to inherited
to cell damage in two ways. One is to react with nitric oxide mutations in mtDNA, somatic mutations in mtDNA may
to form reactive peroxynitrite (Huie and Padmaja, 1993), a play the important role. A common mtDNA4977 deletion is
toxic species capable of inhibiting the respiratory complex markedly increased in the cortex of HD patients during
(Radi et al., 1994), as well as inactivating aconitase (Cas- progression of disease (Horton et al., 1995) likely due to
tro et al., 1994) and MnSOD by nitration of tyrosine resi- oxidative damage mediated by faulty MT. Somatic mtDNA
dues (MacMillan-Crow et al., 1998). Alternatively, super- mutations are particularly devastating to the striatal neu-
oxide can directly inactivate certain Fe-S proteins (Kuo et rons (Cantuti-Castelvetri et al., 2005), which accumulate
al., 1987; Gardner and Fridovich, 1991), particularly acon- more mtDNA point mutations than any other brain cells.
itase (Gardner et al., 1995), by stripping iron from the These data suggest that oxidative damage and accumu-
prosthetic group. The released iron becomes available and lation of mtDNA mutations could contribute to toxicity dur-
represents a secondary path by which superoxide can ing progression of disease.
influence rates of Fenton chemistry (Keyer and Imlay, In fact, the importance of the DNA damage response in
1996; McCormick et al., 1998). Mitochondrial impairment toxicity is supported by the fact that loss of p53 in mhtt
and oxidative stress are detected even in asymptomatic expressing animals suppresses neurodegeneration (Bae
HD carriers early in HD progression (Saft et al., 2005), and et al., 2005; Sawa, 2001; Feng et al., 2006). The response
htt itself may be an iron-binding protein (Hilditch-Maguire
to DNA damage must, at some level, be transmitted to MT
et al., 2000). Thus, it is difficult to determine what process
since p53 is a regulator of many MT genes. Interestingly,
is responsible for initiation of the pathological cascade in
the increase in nuclear DNA damage and up-regulation of
HD. Direct damage of DNA, aberrant MT functions, altered
p53 activity in neurons is accompanied by an abnormal
calcium responses, and damage to lipids are all potentially
flow of calcium ions across the MT membrane and in-
toxic. There is evidence to indicate that all contribute to
creased aggregate formation (Chang et al., 2006). Thus,
some extent to HD pathophysiology.
DNA damage and its detection by p53 appear to create a
Possible mechanisms of MT dysfunction feed-forward loop of toxicity that exacerbates the initial
and toxicity in HD effects of oxidative damage.

Mutations in mtDNA and nuclear DNA. MT are con- Defects in MT functions. Perhaps, the connection
trolled by two genomes: nuclear and mtDNA. Indeed, it has between oxidative DNA damage and mhtt arises directly
been definitively shown that mutations in either nuclear or from binding to MT. In cells expressing mhtt, the mitochon-
mtDNA can contribute to MT dysfunction and neurodegen- drial permeability transition pores (MT-PTP) respond to a
eration (Beal, 2005). The trinucleotide expansion, which lower Ca2⫹ threshold (Panov et al., 2002; Choo et al.,
underlies HD, does not directly affect the expression of 2004) (Fig. 5). In both humans and mouse models, the
proteins involved in oxidative phosphorylation, and, thus, observed dysfunction occurs in purified MT indicating that,
does not directly alter MT function. However, mtDNA is at least in part, the MT themselves are defective. Isolated
predicted to be particularly vulnerable to ROS-induced MT from lymphoblasts of HD patients, transgenic animals,
damage since it is located relatively close to the sites of and immortalized striatal cells derived from HD knock-in
ROS generation. Several experimental studies have ad- mice, consistently show more sensitivity in opening of the
dressed the relationship between mtDNA damage and MT-PTP in response to calcium. Moreover, HD cells are
dysfunction in HD, yet, to date, there is little evidence to also more sensitive to cyanide, a complex IV inhibitor, and
suggest that inherited mutations in mtDNA are a primary susceptible to apoptosis upon stress relative to normal
cause of disease. cells (Sawa et al., 1999; Panov et al., 2002). These defects
Scanning of mtDNA from members of the large Vene- correlate directly with gene dosage. Lymphoblasts from
zuelan family with 43 restriction endonucleases failed to HD patients heterozygous or homozygous for the mutation
reveal any differences in the mitochondrial genotype that have the same age of disease onset, however, homozy-
could account for the differences in age of onset between gous patients display a more aggressive phenotype with
the affected father and his progeny. In addition, analysis of enlarged MT with calcium-sensitized membrane potential.
E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248 1239

Fig. 5. Potential targets for ROS-generated oxidative damage in MT. MT-mediated neuronal dysfunction could be caused by disruption of the
mitochondrial membrane potential, sensitized MT-PTP, excitotoxin-induced Ca2⫹ influx, and diminished ATP production. In experimental models,
mitochondrial toxins (3-nitropropionic acid and malonate) impair respiratory enzyme activities and ATP generation and increase ROS. This is
consistent with impaired electron transport chain activities in HD. Superoxide radical (O2⫺) interacts with nitric oxide (NO) and produces peroxynitrite
(ONOO⫺). Both ROS and ONOO⫺ may then damage cellular macromolecules through nitrosylation, oxidation, and peroxidation, which can directly
contribute to neuronal injury. In addition, the release of cytochrome c from damaged/dysfunctional MT triggers the activation of apoptotic cascade and
release of initiator and executioner caspases (caspase-9 and -3), resulting in neuronal cell death. Calcium channels and ATP synthesis are also
affected.

Thus, early MT impairment at the basal level may affect the models (Fig. 5). Careful analyses of several mouse models
severity of HD progression (Squitieri et al., 2006). Ultra- using sensitive electrodes indicate that the sensitivity to
structural examination of MT in cortical biopsies from the calcium production is tissue-specific (Brustovetsky et al.,
patients with juvenile and adult-onset HD also display mor- 2005). MT in striatum appear to be more sensitive to
phological abnormalities (Browne and Beal, 2004). Due to calcium than are those in cortex, but there is no change
the sensitivity of the MT-PTP to calcium, it has been spec- in calcium sensitivity with age (Brustovetsky et al., 2005).
ulated that MT from HD patients might be less well In fact, the sensitivity to calcium load recovers with time in
equipped to buffer calcium influxes into cells. Indeed, mhtt- transgenic animals expressing mhtt. It has also been re-
expressing cells are sensitive to glutamate receptor stim- ported that expression of mhtt with 111 polyQ in clonal
ulation, and import of extracellular calcium ions into the striatal cells from knock-in HD mice causes significant
cytoplasm. Glutamate receptors are abundantly expressed reduction in ATP production without directly affecting MT
in the MSN, the most affected cell type in HD. In striatal complexes I–IV (Milakovic and Johnson, 2005). These
MSN from YAC transgenic mouse model for HDg20 (Zeron experiments appear to be in agreement with the data
et al., 2002; Tang et al., 2005), glutamate released from obtained in the pre-symptomatic or grade I HD patients
corticostriatal projection neurons stimulates NR1A/NR2B (Guidetti et al., 2001), and findings in patient fibroblasts,
N-methyl-D-aspartate receptor (NMDAR) and mGluR5 re- which demonstrate that ATP production declines in a linear
ceptors. However, HD MSN are sensitized by InsP3R1 to proportion with increasing polyQ length (Seong et al.,
activation by InsP3, and once the mitochondrial Ca2⫹ stor- 2005). Thus, ATP production and the sensitivity to calcium
age capacity is exceeded, the MT-PTP opens, leading to load may be separable. The discrepancy in the results
the release of cytochrome c into the cytosol and activation could be attributed either to the differences in the method-
of caspases-9 and -3. In support of this model, glutamate ological approaches, cell or tissue types, and/or ages of
blockers reduce glutamate-induced apoptosis in the animals used. A careful and systematic analysis may be
YAC128 mouse model for HD (Leavitt et al., 2006) and necessary to resolve these issues.
over-expression of full-length wild-type htt, which is known
to be neuroprotective (Cattaneo et al., 2001; Rigamonti et Lipid/membrane damage. Incubation of MT with
al., 2000; Bezprozvanny and Hayden, 2004; Leavitt et al., GST-fused polyQ tracts of pathological length results in a
2001), reduces excitotoxicity in response to glutamate reduction in membrane potential and an increase in depo-
stimulation. Therefore, mhtt may promote excitotoxic death larization (Panov et al., 2003). No inhibition of any electron
by raising the calcium level to a concentration that exceeds transfer chain (ETC) complexes, mitochondrial ATP syn-
the buffering capacity of the MT (Beal, 1992). thase or adenine nucleotide translocase activities has
There are, however, uncertainties with respect to both been observed in purified MT (Puranam et al., 2006). At
calcium sensitivity and ATP production in mhtt-expressing the same time, addition of antioxidants or cytochrome c
1240 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

prevents defects in isolated MT respiration caused by animals with a very short lifespan. Therefore, it is possible
polyQ proteins. Thus, toxic effect of polyQ proteins could that loss of cholesterol is a feature of dying cells. Further
be in facilitating the significant increase in the amount of analysis will be required to distinguish among the
ROS in isolated MT. The detailed mechanism of the inter- possibilities.
action behind these processes remains to be elucidated. Whether MT defects are the primary cause of toxicity
However, one of the possible mechanisms could include or a secondary response in HD remains an unresolved
the increased ROS production facilitated by membrane- issue. The number of protein interactions altered by mhtt
associated oxidative stress (MAOS). association makes it difficult to cleanly separate effects of
Brain MT have a higher concentration of lipids with altered function from the effects of oxidative damage (Fig.
polyunsaturated acyls, which are more sensitive to free 3). However, neurodegeneration in several genetic dis-
radical oxidation than any other lipids (Bolanos et al., eases arises more directly from MT dysfunction and oxi-
1997). Membrane lipid oxidation and oxidative modification dative DNA damage, and may shed light on the relative
of membrane proteins in MT can cause diminished ATP importance of oxidative damage in initiating neuronal
production with subsequent neuronal dysfunction (Matt- death.
son, 2004). Moreover, peroxidative damage to membrane
lipids is known to affect membrane viscosity and barrier MUTATIONS WITH DIRECT EFFECTS ON MT
function (Mattson, 2004). Recent data suggest that both htt FUNCTION: FRDA
and mhtt interact with lipid bilayers and MT membranes
(Kegel et al., 2000, 2005; Suopanki et al., 2006; Panov et Features of disease
al., 2002). There is little evidence that lipid peroxidation of FRDA is a hereditary recessive disorder directly associ-
MT by mhtt is a major feature of disease. However, the ated with MT dysfunction and oxidative damage that arises
potential effects could be severe. PolyQ expansion in mhtt from inappropriate Fenton chemistry. The disease affects
can alter its membrane localization (Kegel et al., 2005) and roughly one in 50,000 people, and is characterized by
can possibly interfere with the distribution and localization cardiac failure due to impaired ATP synthesis in cardiac
of antioxidants present at the MT membrane. Mhtt might muscles (Lodi et al., 2006; Vorgerd et al., 2000). Atrophy of
also interfere with other membrane-associated events, e.g. sensory and cerebellar pathways causes ataxia, deep sen-
vesicle fission/fusion or endocytosis. Recently, association sory loss and loss of tendon reflexes. Corticospinal degen-
of both htt and mhtt with brain lipids has been shown to eration leads to muscular weakness, hypertrophic car-
promote spontaneous aggregate formation in vivo (Suo- diomyopathy may contribute to disability and cause pre-
panki et al., 2006). A similar phenomenon was recently mature death. The first symptoms usually appear in
demonstrated for ␣-synuclein where binding to the plasma childhood, but age of onset may vary from infancy to
membrane changed its structure, primed oligomerization adulthood. Individuals suffering from FRDA have life
and accelerated fibrillogenesis (Cole et al., 2002; Jo et al., expectancy of 38 years on average (Gatchel and
2004). Thus, the interaction of proteins with membrane Zoghbi, 2005).
lipids could be of general importance in neurodegenerative The FRDA gene encodes a protein of 210 amino acids
disorders. called frataxin (Campuzano et al., 1996). In humans,
Consistent with lipid abnormalities, a defect in choles- frataxin mRNA is most abundant in the heart, spinal cord
terol metabolism has been linked to the MT dysfunction and developing brain. Increasing evidence suggests that
and neurodegeneration in Niemann-Pick type C1 (NPC) this protein directs the intra-mitochondrial synthesis of
(Huang et al., 2006; Yu et al., 2005). Impaired cholesterol iron-sulfur (Fe/S) clusters (Lill and Muhlenhoff, 2005; Rötig
transport in NPC neurons causes elevated cholesterol lev- et al., 1997; Puccio et al., 2001; Muhlenhoff et al., 2002)
els in MT membranes leading to decreases in MT mem- (Fig. 5). MT generate readily available energy equivalents
brane potential, ATP synthase activity, and markedly de- converting nutrients into ATP by oxidative phosphorylation.
creases the levels of ATP. Changes in the lipid composi- This process is dependent on Fe/S clusters, which are
tion of the MT membrane affect oxidative phosphorylation, essential parts of MT enzymes including aconitase and
cell survival, by increasing the passive proton permeability complexes I, II and III (Lill and Muhlenhoff, 2005). Although
and membrane fluidity (Daum, 1985; Hoch, 1992). In con- the exact function of frataxin is only emerging, it is thought
trast to other neurodegenerative disorders, in postmortem to be a mitochondrial iron-binding protein that prevents this
tissues of HD patients and in fixed tissues from the R6/2 metal from participating in Fenton chemistry and inappro-
HD mice, the levels and synthesis of cholesterol are mark- priate generation of hydroxyl radicals (Fig. 6). Since FRDA
edly reduced (Valenza et al., 2005). Given, the similarities is caused by frataxin deficiency, it follows that loss of
among the degenerative diseases with respect to MT dys- frataxin is likely to enhance these events.
function, it is perhaps unusual that cholesterol content in FRDA deficiency begins at the DNA level. Frataxin
HD differs from that of the others, which is typically ele- contains a GAA triplet repeat expansion in the first intron of
vated (Sjogren et al., 2006). It is possible that lipid metab- the gene (Wilson, 2003). The disease is dominant reces-
olism is differentially affected in HD. Alternatively, reduc- sive, and most FRDA patients are homozygous for the
tion of cholesterol in HD may depend on the progression of GAA expansion. Repeats in normal chromosomes contain
disease and/or the mouse model. Cholesterol reductions up to 40 triplets, while the disease allele can contain from
have been measured only in postmortem human tissue or 90 to thousands of GAA repeats (Gacy et al., 1998). In
E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248 1241

Fig. 6. Proposed consequences of frataxin deficiency in FRDA. A reduction in the levels of frataxin is predicted to affect mitochondrial iron metabolism,
with a decrease in the biosyntheses of heme and iron-sulfur clusters and a simultaneous increase in the labile iron pool. Labile Fe2⫹ no longer
detoxified by frataxin participates in Fenton chemistry leading to oxidative damage and progressive accumulation of ferric iron oxides.

about 2% of FRDA cases, however, one allele is expanded well as promotes oxidative damage to both nuclear and
and the other contains point mutations that alter the mtDNA (Karthikeyan et al., 2002, 2003). Similar to HD,
frataxin coding sequence (Cossee et al., 1999). In either ATP synthesis in skeletal muscles from FRDA patients is
case, affected individuals express little to no normal reduced after exercise, as measured by magnetic reso-
frataxin, which is essential for survival. Like HD, FRDA nance spectroscopy (Lodi et al., 1999), and ATP synthesis
shows the anticipation phenotype (Montermini et al., inversely correlates with the length of the GAA tract.
1997a,b), and triplet expansion in the disease alleles both
arise from DNA secondary structure intermediates. In HD, Abnormal regulation of antioxidants in MT in FRDA
the CAG repeats form an aberrant hairpin structure, which
lies in the coding sequence of the gene, and results in the As for MT dysfunction, the same Fe-S defects found in
HD protein with aberrant functions. In FRDA, the long GAA tissues from FRDA patients are present in yeast with def-
tract resides in an intron, and does not code for a protein. icits in respiratory complexes I, II and III, and aconitase
Rather, the GAA disease allele adopts a triple helical struc- (Rötig et al., 1997) (Fig. 5). Although oxidative damage is
ture (Gacy et al., 1998; Ohshima et al., 1998), which acts up regulated in FRDA, there is no induction of the detoxi-
as a roadblock to transcription, translation, or splicing of fying enzyme MnSOD. This is surprising since MnSOD is
the mRNA, and creates a functional knockout of the protein induced by a variety of factors that generate oxidative
(Sakamoto et al., 2001). stress, including proinflammatory cytokines such as tumor
As in HD, ROS-mediated damage and MT dysfunction necrosis factor (TNF) (Visner et al., 1990; Ono et al.,
are associated with FRDA. MT in these patients progres- 1992). These data suggest that part of the toxic mecha-
sively lose respiratory competence, and cells become hy- nism in FRDA involves disruption of a regulatory pathway
persensitive to hydrogen peroxide (Wong et al., 1999). for MnSOD. Under normal conditions, hydrogen peroxide
Consistent with this feature, FRDA patients have in- generation appears to be the rate-limiting factor in Fenton
creased plasma levels of lipid peroxidation (Emond et al., chemistry, and prevents stripping of Fe-S proteins. Thus,
2000), oxidative DNA damage products, and hydroxyl rad- MnSOD activity is inversely correlated with hydroxyl radi-
ical production (Schulz et al., 2000) (Fig. 5). The increases cal production (Borrello et al., 1996; Manna et al., 1998;
in oxidation products in FRDA appear to be related to im- Shi et al., 1999). Since free, soluble iron and iron bound in
paired efflux of iron out of MT (Radisky et al., 1999). MT in Fe-S clusters account for the majority of MT iron, loss of
FRDA patients accumulates iron more then 10-fold in ex- frataxin in FRDA cells is expected to increase the level
cess of wild type. Loss of respiratory competence requires of free iron as is observed (Tan et al., 2001). A number of
the presence of iron in the culture medium, and occurs recent studies indicate that frataxin has a role in heme
more rapidly as the iron concentration is increased. These synthesis, not just iron–sulfur cluster synthesis/repair (Al-
data suggest that permanent MT damage is the conse- Karadaghi et al., 2006). Frataxin appears to be a bifunc-
quence of iron toxicity and the elevation of oxidative dam- tional protein with the ability to bind ferrous iron and either
age is due to inappropriate Fenton chemistry. transfer it to other iron-binding proteins or detoxify it by
Neither Yfh1, the yeast homolog of frataxin, nor human converting it to a stable ferrihydrite mineral. The latter is
frataxin is regulated by iron levels, and their mRNAs do not inactive in ROS-generating reactions.
contain an iron responsive element. However, human There are parallels between HD and FRDA. Mhtt is
frataxin (Cavadini et al., 2002) and Yfh1p (Adamec et al., also reported to be an iron binding protein, and iron de-
2000) are iron-binding proteins. Yfh1 appears to form a posits have been detected in the brain of HD patients
high molecular weight complex in the presence of ferrous (Hilditch-Maguire et al., 2000). MnSOD induction in FRDA
ions (Adamec et al., 2000; Park et al., 2003). Thus, Yfh1p is controlled by AP-1 and ATF-1/CREB-1 activation (Bor-
and frataxin may protect iron in MT from contacts with free rello et al., 1992). CREB itself is a target of mhtt binding,
radicals, and act as a mitochondrial iron chaperone (Bul- and if inactivated by mhtt, then MnSOD pathways may be
teau et al., 2004). In the absence of frataxin, impaired affected. The subsequent damage to the respiratory com-
biosyntheses and transport processes may lead to iron plexes could lead to increased leakage of the superoxide
accumulation in a toxic, redox-active form (Fig. 6). Consis- radicals that would account for the inactivation of Fe-S
tent with this possibility, disruption of the frataxin homo- proteins and DNA damage. Thus, MT dysfunction and
logue in yeast increases oxidative stress (Gakh et al., overlap in some properties of HD and FRDA have raised
2006; Wong et al., 1999; Piemonte et al., 2001; Chantrel- the possibility that oxidative DNA damage might be a
Groussard et al., 2001) and sensitivity against oxidants, as common feature in neurodegenerative diseases and may
1242 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

directly contribute to neuronal toxicity. For FRDA, this hy- cross-links, bulky chemical lesions, cyclodeoxyadenosine,
pothesis is consistent with the fact that over-expression of thymine glycol, cyclopurines, and infrequently, 8-oxo-G
frataxin reduces intracellular accumulation of ROS, and (Bjelland and Seeberg, 2003). Other DNA repair pathways
prevents menadione-induced malignant transformation of can remove oxidative damage to DNA due to ROS, notably
fibroblasts (Shoichet et al., 2002). The loss of frataxin base excision repair. While some of these lesions can also
function in FRDA also elevates ROS in cells, and mito- be removed by NER, in vivo, the major defect in neurons of
chondrial dysfunction and a rise in oxidation is associated XP patients may be the inability to repair cyclopurines
with HD. (Hanawalt et al., 2003; Friedberg, 2003; Bootsma et al.,
2005). In any case, it appears that loss of repair of oxida-
tive DNA lesions alone can result in severe neurodegen-
MUTATIONS WITH DIRECT EFFECT ON
erative disease with early onset in the absence of addi-
OXIDATIVE DNA DAMAGE tional protein defects. Thus, the degree to which oxidative
A rise in oxidative DNA bases is observed in both HD and DNA damage accumulates may be linked to the onset of
FRDA. However, in both of these disorders a range of disease.
cellular processes is affected. In the former, mhtt can
interact and inactivate key proteins associated with cyto- DNA DAMAGE RESPONSE AS A COMMON
plasmic and nuclear events, while in the latter iron metab- LINK
olism appears to be altered. Thus, effects of faulty protein
The consequences of oxidative damage among neurode-
function cannot be cleanly separated from oxidative DNA
generative diseases are complex, and dysfunction can
damage, which, on its own, may be sufficient to elicit cell
occur at many levels. However, a common denominator is
death. The extent to which oxidative DNA damage contrib-
likely to be an oxidative DNA damage response, which
utes to cell death of neurons is not known. However,
appears to be sufficient by itself to promote neurodegen-
lessons can be learned from a group of disorders referred
eration. In the case of HD, mhtt can rescue the develop-
to as XP. XP arises from mutations in nucleotide excision
mental defects of htt deletion (White et al., 1997). Thus,
repair (NER) proteins, which are essential for removal of
mhtt must retain some of the functions of the normal pro-
some forms of oxidative DNA damage. NER is carried out
tein. Moreover, HD displays later onset than does XP.
by a number of proteins that operate in discrete steps:
Thus, failure to remove oxidative damage in XP may be
lesion recognition, single strand excision, lesion removal,
more toxic than the effects of a faulty HD protein in other-
and, with the help of polymerase, gap-filling synthesis
wise repair-competent neurons. Since repair of oxidative
(McMurray, 2005; Cleaver, 2005; Dip et al., 2004). NER is
lesions by either BER or NER occurs through a single
characterized by two types of repair pathways: removal of
strand break intermediate, oxidative stress may cause tox-
oxidative DNA damage found globally in the genome
icity through the accumulation of unrepaired single strand
(GGR), and oxidative DNA damage found in transcribed
breaks (El-Khamisy and Caldecott, 2006) and the possible
genes (transcription coupled repair (TCR)) (Cleaver, 2005;
generation of double strand breaks. These data, as a
Dip et al., 2004). GGR and TCR share most steps in the
whole, raise the testable hypothesis that DNA damage,
repair pathway, but differ in their recognition step. GGR is when unchecked in neurons, is a common factor that
initiated by recognition by XPC/HR23B or XPE/DDB2. In promotes toxicity.
contrast, TCR is initiated by two factors that are defective
in a form of XP called Cockayne’s syndrome (CS). CSA
THERAPEUTIC IMPLICATIONS OF OXIDATIVE
and CSB signal TCR by recognizing a stalled RNA poly-
DAMAGE
merase itself.
The NER pathway is associated with at least seven Based on the evidence, it might be predicted that anti-
complementary groups, XP (A–G), which represent muta- oxidants and/or inhibitors of excitotoxic damage might be
tions in specific NER proteins. The incidence of neurolog- therapeutic. However, to date, the majority of HD preclin-
ical abnormalities varies considerably. Neurodegeneration ical mouse trials designed to test the effect of different
is prominent in many XP patients but extremely rare in neuroprotective agents demonstrated limited success
patients with XPC or XPE. Since XPC and XPE are in- (Handley et al., 2006). Processes of free radical damage,
volved in the initiation of GGR, these data indicate that excitotoxicity and membrane integrity have been consid-
defects in TCR rather than GGR are connected to the ered. Beneficial agents include creatine (Ferrante et al.,
neurodegeneration phenotype. These data point to the 2000; Andreassen et al., 2001), coenzyme Q (Ferrante et
importance of keeping expressed genes lesion free. Oxi- al., 2002; Schilling et al., 2001), remacemide (Ferrante et
dative damage within expressed genes is more likely to be al., 2002; Schilling et al., 2001), riluzole (Schiefer et al.,
associated with neuronal toxicity, not unexpectedly, given 2002), a-lipoic acid (Andreassen et al., 2001) and essential
the fact that neurons cannot divide and may accumulate fatty acids (Clifford et al., 2002). Creatine treatment, for
unrepaired DNA damage over time. In XP patients, faulty example, is neuroprotective in R6/2 mice (Ferrante et al.,
NER and unrepaired oxidative DNA damage, on its own, 2000). Similarly, administration of creatine to HD patients
can result in death of neurons in the absence of additional for 2 years (10 g/day) prevented weight loss and, in some
insults from defective proteins. The spectrum of ROS- patients, caused an improvement in the scores for neuro-
mediated lesions repaired by NER includes intrastrand logical testing (Tabrizi et al., 2005). In an additional study,
E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248 1243

administration of creatine (8 g/day or 16 weeks) markedly Bjelland S, Seeberg E (2003) Mutagenicity, toxicity and repair of DNA
reduced serum levels of 8-hydroxy-2=-deoxyguanosine, an base damage induced by oxidation. Mutat Res 531:37– 80.
indicator of oxidative injury to DNA (Hersch et al., 2006). Bolanos JP, Almeida A, Fernandez E, Medina JM, Land JM, Clark JB,
Heales SJ (1997) Potential mechanisms for nitric oxide-mediated
Small molecules that inhibit oxidative damage, such as
impairment of brain mitochondrial energy metabolism. Biochem
cystamine and cysteamine, have improved survival of Soc Trans 25:944 –949.
mhtt-expressing cells (Mao et al., 2006) and animal mod- Bootsma D, Kraemer KH, Cleaver JE, Hoeijmakers JHJ (2005) Nucle-
els for HD (Borrell-Pages et al., 2006) by preventing MT otide excision repair syndromes: xeroderma pigmentosum, cock-
depolarization and increasing BDNF levels in the brain. For ayne syndrome, and trichothiodystrophy. In: The metabolic and
FRDA, Bayer 11–7082, a compound known to regulate molecular bases of inherited disease, Vol. 3 (Scriver C, Beaudet
activity of the mitogen-activated protein kinases, JNK-1 AL, Sly WS, eds), pp 137–150. McGraw-Hill Companies.
and p38 (Read et al., 1997), is capable of inhibiting path- Borrell-Pages M, Canals JM, Cordelieres FP, Parker JA, Pineda JR,
Grange G, Bryson EA, Guillermier M, Hirsch E, Hantraye P,
ways of iron-mediated MnSOD induction. The existence of
Cheetham ME, Neri C, Alberch J, Brouillet E, Saudou F, Humbert
a mechanism linking MnSOD regulation remains to be S (2006) Cystamine and cysteamine increase brain levels of BDNF
proven but represents a logical link by which cellular ROS in Huntington disease via HSJ1b and transglutaminase. J Clin
defenses respond to changes in MT iron states. Although Invest 116:1410 –1424.
none are curative, recent data continue to support benefi- Borrello S, De Leo ME, Landriscina M, Palazzotti B, Galeotti T (1996)
cial effects of antioxidants. As judged by dominant hered- Diethyldithiocarbamate treatment up regulates manganese super-
itary disorders, the data indicate that ROS are major con- oxide dismutase gene expression in rat liver. Biochem Biophys
tributors to neuronal toxicity, and there is continued hope Res Commun 220:546 –552.
Borrello S, De Leo ME, Wohlrab H, Galeotti T (1992) Manganese
that blocking ROS might be used to offset disease.
deficiency and transcriptional regulation of mitochondrial superox-
ide dismutase in hepatomas. FEBS Lett 310:249 –254.
Acknowledgment—This work was supported by the Mayo Foun- Browne SE, Beal MF (2004) The energetics of Huntington’s disease.
dation, the National Institutes of Health grants NS40738 (C.T.M.), Neurochem Res 29:531–546.
and GM 066359 (C.T.M.). We thank Kurt Johnson and Grazia Browne SE, Bowling AC, MacGarvey U, Baik MJ, Berger SC, Muqit
Isaya for reading the manuscript, and Grazia Isaya for provision of MM, Bird ED, Beal MF (1997) Oxidative damage and metabolic
Fig. 6. dysfunction in Huntington’s disease: selective vulnerability of the
basal ganglia. Ann Neurol 41:646 – 653.
Brustovetsky N, LaFrance R, Purl KJ, Brustovetsky T, Keene CD, Low
REFERENCES WC, Dubinsky JM (2005) Age-dependent changes in the calcium
sensitivity of striatal mitochondria in mouse models of Huntington’s
Adamec J, Rusnak F, Owen WG, Naylor S, Benson LM, Gacy AM, disease. J Neurochem 93:1361–1370.
Isaya G (2000) Iron-dependent self-assembly of recombinant Bulteau AL, O’Neill HA, Kennedy MC, Ikeda-Saito M, Isaya G, Szweda
yeast frataxin: implications for Friedreich ataxia. Am J Hum Genet N (2004) Frataxin acts as an iron chaperone protein to modulate
67:549 –562. mitochondrial aconitase activity. Science 305:242–245.
Al-Karadaghi S, Franco R, Hansson M, Shelnutt JA, Isaya G, Ferreira Campuzano V, Montermini L, Molto MD, Pianese L, Cossee M, Cav-
GC (2006) Chelatases: distort to select? Trends Biochem Sci alcanti F, Monros E, Rodius F, Duclos F, Monticelli A, Zara F,
31:135–142. Canizares J, Koutnikova H, Bidichandani SI, Gellera C, Brice A,
Andreassen OA, Dedeoglu A, Ferrante RJ, Jenkins BG, Ferrante KL, Trouillas P, De Michele G, Filla A, De Frutos R, Palau F, Patel PI,
Thomas M, Friedlich A, Browne SE, Schilling G, Borchelt DR,
Di Donato S, Mandel JL, Cocozza S, Koenig M, Pandolfo M (1996)
Hersch SM, Ross CA, Beal MF (2001) Creatine increase survival
Friedreich’s ataxia: autosomal recessive disease caused by an
and delays motor symptoms in a transgenic animal model of
intronic GAA triplet repeat expansion. Science 271:1423–1427.
Huntington’s disease. Neurobiol Dis 8:479 – 491.
Cantuti-Castelvetri I, Lin MT, Zheng K, Keller-McGandy CE, Betensky
Arenas J, Campos Y, Ribacoba R, Martin MA, Rubio JC, Ablanedo P,
RA, Johns DR, Beal MF, Standaert DG, Simon DK (2005) Somatic
Cabello A (1998) Complex I defect in muscle from patients with
mitochondrial DNA mutations in single neurons and glia. Neurobiol
Huntington’s disease. Ann Neurol 43:397– 400.
Aging 26:1343–1355.
Bae BI, Xu H, Igarashi S, Fujimuro M, Agrawal N, Taya Y, Hayward
Castro L, Rodriguez M, Radi R (1994) Aconitase is readily inactivated
SD, Moran TH, Montell C, Ross CA, Snyder SH, Sawa A (2005)
by peroxynitrite, but not by its precursor, nitric oxide. J Biol Chem
p53 Mediates cellular dysfunction and behavioral abnormalities in
Huntington’s disease. Neuron 47:29 – 41. 269:29409 –29415.
Beal MF (1992) Does impairment of energy metabolism result in Cattaneo E, Rigamonti D, Goffredo D, Zuccato C, Squitieri F, Sipione
excitotoxic neuronal death in neurodegenerative illnesses? Ann S (2001) Loss of normal huntingtin function: new developments in
Neurol 31:119 –130. Huntington’s disease research. Trends Neurosci 24:182–188.
Beal MF (2005) Mitochondria take center stage in aging and neuro- Cavadini P, O’Neill HA, Benada O, Isaya G (2002) Assembly and
degeneration. Ann Neurol 58:495–505. iron-binding properties of human frataxin, the protein deficient in
Beal MF, Ferrante RJ, Swartz KJ, Kowall NW (1991) Chronic quino- Friedreich ataxia. Hum Mol Genet 11:217–227.
linic acid lesions in rats closely resemble Huntington’s disease. Chang DT, Rintoul GL, Pandipati S, Reynolds IJ (2006) Mutant hun-
J Neurosci 11:1649 –1659. tingtin aggregates impair mitochondrial movement and trafficking
Benchoua A, Trioulier Y, Zala D, Gaillard MC, Lefort N, Dufour N, in cortical neurons. Neurobiol Dis 22:388 – 400.
Saudou F, Elalouf JM, Hirsch E, Hantraye P, Deglon N, Brouillet E Chantrel-Groussard K, Geromel V, Puccio H, Koenig M, Munnich A,
(2006) Involvement of mitochondrial complex II defects in neuronal Rötig A, Rustin P (2001) Disabled early recruitment of antioxidant
death produced by N-terminus fragment of mutated huntingtin. Mol defenses in Friedreich’s ataxia. Hum Mol Genet 10:2061–2067.
Biol Cell 17:1652–1663. Choo YS, Johnson GV, MacDonald M, Detloff PJ, Lesort M (2004)
Bezprozvanny I, Hayden MR (2004) Deranged neuronal calcium sig- Mutant huntingtin directly increases susceptibility of mitochondria
naling and Huntington disease. Biochem Biophys Res Commun to the calcium-induced permeability transition and cytochrome c
322:1310 –1317. release. Hum Mol Genet 13:1407–1420.
1244 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

Ciammola A, Sassone J, Alberti L, Meola G, Mancinelli E, Russo MA, frataxin that limits oxidative damage and preserves cell longevity.
Squitieri F, Silani V (2006) Increased apoptosis, huntingtin inclu- Hum Mol Genet 15:467– 479.
sions and altered differentiation in muscle cell cultures from Hun- Gardner PR, Fridovich I (1991) Superoxide sensitivity of the Esche-
tington’s disease subjects. Cell Death Differ 13:2068 –2078. richia coli 6-phosphogluconate dehydratase. J Biol Chem 266:
Cleaver JE (2005) Cancer in xeroderma pigmentosum and related 1478 –1483.
disorders of DNA repair. Nat Rev Cancer 5:564 –573. Gardner PR, Raineri I, Epstein LB, White CW (1995) Superoxide
Clifford JJ, Drago J, Natoli AL, Wong JY, Kinsella A, Waddington JL, radical and iron modulate aconitase activity in mammalian cells.
Vaddadi KS (2002) Essential fatty acids given from conception J Biol Chem 270:13399 –13405.
prevent topographies of motor deficit in a transgenic model of Gatchel JR, Zoghbi HY (2005) Diseases of unstable repeat expansion:
Huntington’s disease. Neuroscience 109:81– 88. mechanisms and common principles. Nat Rev Genet 6:743–
Cole NB, Murphy DD, Grider T, Rueter S, Brasaemle D, Nussbaum RL 755.
(2002) Lipid droplet binding and oligomerization properties of the Gauthier LR, Charrin BC, Borrell-Pages M, Dompierre JP, Rangone H,
Parkinson’s disease protein alpha-synuclein. J Biol Chem 277: Cordelieres FP, De Mey J, MacDonald ME, Lessmann V, Humbert
6344 – 6352. S, Saudou F (2004) Huntingtin controls neurotrophic support and
Cossee M, Durr A, Schmitt M, Dahl N, Trouillas P, Allinson P, Kostr- survival of neurons by enhancing BDNF vesicular transport along
zewa M, Nivelon-Chevallier A, Gustavson KH, Kohlschutter A, microtubules. Cell 118:127–138.
Muller U, Mandel JL, Brice A, Koenig M, Cavalcanti F, Tammaro A, Gines S, Seong IS, Fossale E, Ivanova E, Trettel F, Gusella JF,
De Michele G, Filla A, Cocozza S, Labuda M, Montermini L, Poirier Wheeler VC, Persichetti F, MacDonald ME (2003) Specific pro-
J, Pandolfo M (1999) Friedreich’s ataxia: point mutations and gressive cAMP reduction implicates energy deficit in presymptom-
clinical presentation of compound heterozygotes. Ann Neurol 45: atic Huntington’s disease knock-in mice. Hum Mol Genet 12:
200 –206. 497–508.
Daum G (1985) Lipids of mitochondria. Biochim Biophys Acta Gizatullina ZZ, Lindenberg KS, Harjes P, Chen Y, Kosinski CM, Land-
822:1– 42. wehrmeyer BG, Ludolph AC, Striggow F, Zierz S, Gellerich FN
Dip R, Camenisch U, Naegeli H (2004) Mechanisms of DNA damage (2006) Low stability of Huntington muscle mitochondria against
recognition and strand discrimination in human nucleotide excision Ca2⫹ in R6/2 mice. Ann Neurol 59:407– 411.
repair. DNA Repair (Amst) 3:1409 –1423. Goswami A, Dikshit P, Mishra A, Mulherkar S, Nukina N, Jana NR
(2006) Oxidative stress promotes mutant huntingtin aggregation
Dunah AW, Jeong H, Griffin A, Kim YM, Standaert DG, Hersch SM,
and mutant huntingtin-dependent cell death by mimicking pro-
Mouradian MM, Young AB, Tanese N, Krainc D (2002) Sp1 and
teasomal malfunction. Biochem Biophys Res Commun 342:
TAFII130 transcriptional activity disrupted in early Huntington’s
184 –190.
disease. Science 296:2238 –2243.
Graham RK, Deng Y, Slow EJ, Haigh B, Bissada N, Lu G, Pearson J,
El-Khamisy SF, Caldecott KW (2006) TDP1-dependent DNA single-
Shehadeh J, Bertram L, Murphy Z, Warby SC, Doty CN, Roy S,
strand break repair and neurodegeneration. Mutagenesis 21(4):
Wellington CL, Leavitt BR, Raymond LA, Nicholson DW, Hayden
219 –224.
MR (2006) Cleavage at the caspase-6 site is required for neuronal
Emond M, Lepage G, Vanasse M, Pandolfo M (2000) Increased levels
dysfunction and degeneration due to mutant huntingtin. Cell
of plasma malondialdehyde in Friedreich ataxia. Neurology 55:
125:1179 –1191.
1752–1753.
Gu M, Gash MT, Mann VM, Javoy-Agid F, Cooper JM, Schapira AH
Engelender S, Sharp AH, Colomer V, Tokito MK, Lanahan A, Worley
(1996) Mitochondrial defect in Huntington’s disease caudate nu-
P, Holzbaur EL, Ross CA (1997) Huntingtin-associated protein 1
cleus. Ann Neurol 39:385–389.
(HAP1) interacts with the p150Glued subunit of dynactin. Hum Mol
Guidetti P, Charles V, Chen EY, Reddy PH, Kordower JH, Whetsell
Genet 6:2205–2212.
WO Jr, Schwarcz R, Tagle DA (2001) Early degenerative changes
Esposito LA, Kokoszka JE, Waymire KG, Cottrell B, MacGregor GR, in transgenic mice expressing mutant huntingtin involve dendritic
Wallace DC (2000) Mitochondrial oxidative stress in mice lacking abnormalities but no impairment of mitochondrial energy produc-
the glutathione peroxidase-1 gene. Free Radic Biol Med 28:754 – tion. Exp Neurol 169:340 –350.
766. Guidot DM, McCord JM, Wright RM, Repine JE (1993) Absence of
Feng Z, Jin S, Zupnick A, Hoh J, de Stanchina E, Lowe S, Prives C, electron transport (Rho 0 state) restores growth of a manga-
Levine AJ (2006) p53 Tumor suppressor protein regulates the nese-superoxide dismutase-deficient Saccharomyces cerevi-
levels of huntingtin gene expression. Oncogene 25:1–7. siae in hyperoxia. Evidence for electron transport as a major
Ferrante RJ, Andreassen OA, Dedeoglu A, Ferrante KL, Jenkins BG, source of superoxide generation in vivo. J Biol Chem 268:
Hersch SM, Beal MF (2002) Therapeutic effects of coenzyme Q10 26699 –26703.
and remacemide in transgenic mouse models of Huntington’s dis- Gunawardena S, Her LS, Brusch RG, Laymon RA, Niesman IR,
ease. J Neurosci 22:1592–1599. Gordesky-Gold B, Sintasath L, Bonini NM, Goldstein LS (2003)
Ferrante RJ, Andreassen OA, Jenkins BG, Dedeoglu A, Kuemmerle S, Disruption of axonal transport by loss of huntingtin or expression of
Kubilus JK, Kaddurah-Daouk R, Hersch SM, Beal MF (2000) Neu- pathogenic polyQ proteins in Drosophila. Neuron 40:25– 40.
roprotective effects of creatine in a transgenic mouse model of Hanawalt PC, Ford JM, Lloyd DR (2003) Functional characterization of
Huntington’s disease. J Neurosci 20:4389 – 4397. global genomic DNA repair and its implications for cancer. Mutat
Floyd RA, West MS, Eneff KL, Schneider JE, Wong PK, Tingey DT, Res 544:107–114.
Hogsett WE (1990) Conditions influencing yield and analysis of Handley OJ, Naji JJ, Dunnett SB, Rosser AE (2006) Pharmaceutical,
8-hydroxy-2=-deoxyguanosine in oxidatively damaged DNA. Anal cellular and genetic therapies for Huntington’s disease. Clin Sci
Biochem 188:155–158. (Lond) 110:73– 88.
Friedberg EC (2003) DNA damage and repair. Nature 421:436 – 440. Harjes P, Wanker EE (2003) The hunt for huntingtin function: interac-
Gacy AM, Goellner GM, Spiro C, Chen X, Gupta G, Bradbury EM, tion partners tell many different stories. Trends Biochem Sci 28:
Dyer RB, Mikesell MJ, Yao JZ, Johnson AJ, Richter A, Melancon 425– 433.
SB, McMurray CT (1998) GAA instability in Friedreich’s ataxia Harman D (1956) Aging: a theory based on free radical and radiation
shares a common, DNA-directed and intraallelic mechanism with chemistry. J Gerontol 11:298 –300.
other trinucleotide diseases. Mol Cell 1:583–593. Hazra TK, Hill JW, Izumi T, Mitra S (2001) Multiple DNA glycosylases
Gakh O, Park S, Liu G, Macomber L, Imlay JA, Ferreira GC, Isaya G for repair of 8-oxoguanine and their potential in vivo functions. Prog
(2006) Mitochondrial iron detoxification is a primary function of Nucleic Acid Res Mol Biol 68:193–205.
E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248 1245

Hersch SM, Gevorkian S, Marder K, Moskowitz C, Feigin A, Cox M, cycle activation linked to neuronal cell death initiated by DNA
Como P, Zimmerman C, Lin M, Zhang L, Ulug AM, Beal MF, damage. Neuron 19:549 –561.
Matson W, Bogdanov M, Ebbel E, Zaleta A, Kaneko Y, Jenkins B, Leavitt BR, Guttman JA, Hodgson JG, Kimel GH, Singaraja R, Vogl
Hevelone N, Zhang H, Yu H, Schoenfeld D, Ferrante R, Rosas HD AW, Hayden MR (2001) Wild-type huntingtin reduces the cellu-
(2006) Creatine in Huntington disease is safe, tolerable, bioavail- lar toxicity of mutant huntingtin in vivo. Am J Hum Genet
able in brain and reduces serum 8OH2=dG. Neurology 66: 68:313–324.
250 –252. Leavitt BR, Raamsdonk JM, Shehadeh J, Fernandes H, Murphy Z,
Hilditch-Maguire P, Trettel F, Passani LA, Auerbach A, Persichetti F, Graham RK, Wellington CL, Raymond LA, Hayden MR (2006)
MacDonald ME (2000) Huntingtin: an iron-regulated protein essen- Wild-type huntingtin protects neurons from excitotoxicity. J Neuro-
tial for normal nuclear and perinuclear organelles. Hum Mol Genet chem 96:1121–1129.
9:2789 –2797. Lee ST, Kim M (2006) Aging and neurodegeneration. Molecular mech-
Hoch FL (1992) Cardiolipins and biomembrane function. Biochim Bio- anisms of neuronal loss in Huntington’s disease. Mech Ageing Dev
phys Acta 1113:71–133. 127:432– 435.
Horton TM, Graham BH, Corral-Debrinski M, Shoffner JM, Kaufman
Li SH, Cheng AL, Zhou H, Lam S, Rao M, Li H, Li XJ (2002) Interaction
AE, Beal MF, Wallace DC (1995) Marked increase in mitochondrial
of Huntington disease protein with transcriptional activator Sp1.
DNA deletion levels in the cerebral cortex of Huntington’s disease
Mol Cell Biol 22:1277–1287.
patients. Neurology 45:1879 –1883.
Li SH, Gutekunst CA, Hersch SM, Li XJ (1998) Interaction of hunting-
Huang Z, Hou Q, Cheung NS, Li QT (2006) Neuronal cell death
tin-associated protein with dynactin P150Glued. J Neurosci 18:
caused by inhibition of intracellular cholesterol trafficking is
1261–1269.
caspase dependent and associated with activation of the mito-
Li SH, Li XJ (2004) Huntingtin-protein interactions and the pathogen-
chondrial apoptosis pathway. J Neurochem 97:280 –291.
Huie RE, Padmaja S (1993) The reaction of no with superoxide. Free esis of Huntington’s disease. Trends Genet 20:146 –154.
Radic Res Commun 18:195–199. Lill R, Muhlenhoff U (2005) Iron-sulfur-protein biogenesis in eu-
Irwin CC, Wexler NS, Young AB, Ozelius LJ, Penney JB, Shoulson I, karyotes. Trends Biochem Sci 30:133–141.
Snodgrass SR, Ramos-Arroyo MA, Sanchez-Ramos J, Pen- Lodi R, Cooper JM, Bradley JL, Manners D, Styles P, Taylor DJ,
chaszadeh GK et al. (1989) The role of mitochondrial DNA in Schapira AH (1999) Deficit of in vivo mitochondrial ATP production
Huntington’s disease. J Mol Neurosci 1:129 –136. in patients with Friedreich ataxia. Proc Natl Acad Sci U S A 96:
Jenkins BG, Rosas HD, Chen YC, Makabe T, Myers R, MacDonald M, 11492–11495.
Rosen BR, Beal MF, Koroshetz WJ (1998) 1H NMR spectroscopy Lodi R, Schapira AH, Manners D, Styles P, Wood NW, Taylor DJ,
studies of Huntington’s disease: correlations with CAG repeat Warner TT (2000) Abnormal in vivo skeletal muscle energy me-
numbers. Neurology 50:1357–1365. tabolism in Huntington’s disease and dentatorubropallidoluysian
Jo E, Darabie AA, Han K, Tandon A, Fraser PE, McLaurin J (2004) atrophy. Ann Neurol 48:72–76.
alpha-Synuclein-synaptosomal membrane interactions: implica- Lodi R, Tonon C, Calabrese V, Schapira AH (2006) Friedreich’s ataxia:
tions for fibrillogenesis. Eur J Biochem 271:3180 –3189. From disease mechanisms to therapeutic interventions. Antioxid
Karthikeyan G, Lewis LK, Resnick MA (2002) The mitochondrial pro- Redox Signal 8:438 – 443.
tein frataxin prevents nuclear damage. Hum Mol Genet 11: MacMillan-Crow LA, Crow JP, Thompson JA (1998) Peroxynitrite-
1351–1362. mediated inactivation of manganese superoxide dismutase in-
Karthikeyan G, Santos JH, Graziewicz MA, Copeland WC, Isaya G, volves nitration and oxidation of critical tyrosine residues. Bio-
Van Houten B, Resnick MA (2003) Reduction in frataxin causes chemistry 37:1613–1622.
progressive accumulation of mitochondrial damage. Hum Mol Majumder P, Chattopadhyay B, Mazumder A, Das P, Bhattacharyya
Genet 12:3331–3342. NP (2006) Induction of apoptosis in cells expressing exogenous
Kegel KB, Kim M, Sapp E, McIntyre C, Castano JG, Aronin N, DiFiglia Hippi, a molecular partner of huntingtin-interacting protein Hip1.
M (2000) Huntingtin expression stimulates endosomal-lysosomal Neurobiol Dis 22:242–256.
activity, endosome tubulation, and autophagy. J Neurosci 20: Mann VM, Cooper JM, Javoy-Agid F, Agid Y, Jenner P, Schapira AH
7268 –7278. (1990) Mitochondrial function and parental sex effect in Hunting-
Kegel KB, Meloni AR, Yi Y, Kim YJ, Doyle E, Cuiffo BG, Sapp E, Wang ton’s disease. Lancet 336:749.
Y, Qin ZH, Chen JD, Nevins JR, Aronin N, DiFiglia M (2002) Manna SK, Zhang HJ, Yan T, Oberley LW, Aggarwal BB (1998)
Huntingtin is present in the nucleus, interacts with the transcrip-
Overexpression of manganese superoxide dismutase suppresses
tional corepressor C-terminal binding protein, and represses tran-
tumor necrosis factor-induced apoptosis and activation of nuclear
scription. J Biol Chem 277:7466 –7476.
transcription factor-kappaB and activated protein-1. J Biol Chem
Kegel KB, Sapp E, Yoder J, Cuiffo B, Sobin L, Kim YJ, Qin ZH, Hayden
273:13245–13254.
MR, Aronin N, Scott DL, Isenberg G, Goldmann WH, DiFiglia M
Mao Z, Choo YS, Lesort M (2006) Cystamine and cysteamine prevent
(2005) Huntingtin associates with acidic phospholipids at the
3-NP-induced mitochondrial depolarization of Huntington’s dis-
plasma membrane. J Biol Chem 280:36464 –36473.
ease knock-in striatal cells. Eur J Neurosci 23:1701–1710.
Keyer K, Imlay JA (1996) Superoxide accelerates DNA damage by
elevating free-iron levels. Proc Natl Acad Sci U S A 93:13635– Mariani E, Polidori MC, Cherubini A, Mecocci P (2005) Oxidative
13640. stress in brain aging, neurodegenerative and vascular diseases:
King MP, Attardi G (1989) Human cells lacking mtDNA: repopulation an overview. J Chromatogr B Analyt Technol Biomed Life Sci
with exogenous mitochondria by complementation. Science 246: 827:65–75.
500 –503. Mattson MP (2004) Metal-catalyzed disruption of membrane protein
Koroshetz WJ, Jenkins BG, Rosen BR, Beal MF (1997) Energy me- and lipid signaling in the pathogenesis of neurodegenerative dis-
tabolism defects in Huntington’s disease and effects of coenzyme orders. Ann N Y Acad Sci 1012:37–50.
Q10. Ann Neurol 41:160 –165. McCormick ML, Buettner GR, Britigan BE (1998) Endogenous super-
Kuo CF, Mashino T, Fridovich I (1987) alpha, beta-Dihydroxyisovaler- oxide dismutase levels regulate iron-dependent hydroxyl radical
ate dehydratase. A superoxide-sensitive enzyme. J Biol Chem formation in Escherichia coli exposed to hydrogen peroxide.
262:4724 – 4727. J Bacteriol 180:622– 625.
Kruman II, Wersto RP, Cardozo-Pelaez F, Smilenov L, Chan SL, McMurray CT (2005) To die or not to die: DNA repair in neurons.
Chrest FJ, Emokpae R Jr, Gorospe M, Mattson MP (2004) Cell Mutant Res 577:260 –274.
1246 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

Michalik A, Van Broeckhoven C (2003) Pathogenesis of polyglutamine Puranam KL, Wu G, Strittmatter WJ, Burke JR (2006) Polyglutamine
disorders: aggregation revisited. Hum Mol Genet 12 (Spec No 2): expansion inhibits respiration by increasing reactive oxygen spe-
R173–R186. cies in isolated mitochondria. Biochem Biophys Res Commun
Michiels C, Raes M, Toussaint O, Remacle J (1994) Importance of 341:607– 613.
Se-glutathione peroxidase, catalase, and Cu/Zn-SOD for cell Radi R, Rodriguez M, Castro L, Telleri R (1994) Inhibition of mitochon-
survival against oxidative stress. Free Radic Biol Med 17: drial electron transport by peroxynitrite. Arch Biochem Biophys
235–248. 308:89 –95.
Milakovic T, Johnson GV (2005) Mitochondrial respiration and ATP Radisky DC, Babcock MC, Kaplan J (1999) The yeast frataxin homo-
production are significantly impaired in striatal cells expressing logue mediates mitochondrial iron efflux. Evidence for a mitochon-
mutant huntingtin. J Biol Chem 280:30773–30782. drial iron cycle. J Biol Chem 274:4497– 4499.
Montermini L, Andermann E, Labuda M, Richter A, Pandolfo M, Cav- Rao VR, Finkbeiner S (2003) Secrets of a secretase: N-cadherin
alcanti F, Pianese L, Iodice L, Farina G, Monticelli A, Turano M, proteolysis regulates CBP function. Cell 114:533–535.
Filla A, De Michele G, Cocozza S (1997a) The Friedreich ataxia Read MA, Whitley MZ, Gupta S, Pierce JW, Best J, Davis RJ, Collins
GAA triplet repeat: premutation and normal alleles. Hum Mol Genet T (1997) Tumor necrosis factor alpha-induced E-selectin expres-
6:1261–1266. sion is activated by the nuclear factor-kappaB and c-JUN N-termi-
Montermini L, Kish SJ, Jiralerspong S, Lamarche JB, Pandolfo M nal kinase/p38 mitogen-activated protein kinase pathways. J Biol
(1997b) Somatic mosaicism for Friedreich’s ataxia GAA triplet Chem 272:2753–2761.
repeat expansions in the central nervous system. Neurology 49: Reddy PH (2006) Amyloid precursor protein-mediated free radicals
606 – 610. and oxidative damage: implications for the development and pro-
Muhlenhoff U, Richhardt N, Ristow M, Kispal G, Lill R (2002) The yeast gression of Alzheimer’s disease. J Neurochem 96:1–13.
frataxin homolog Yfh1p plays a specific role in the maturation of Rigamonti D, Bauer JH, De-Fraja C, Conti L, Sipione S, Sciorati C,
cellular Fe/S proteins. Hum Mol Genet 11:2025–2036. Clementi E, Hackam A, Hayden MR, Li Y, Cooper JK, Ross CA,
Nucifora FC Jr, Sasaki M, Peters MF, Huang H, Cooper JK, Yamada Govoni S, Vincenz C, Cattaneo E (2000) Wild-type huntingtin
M, Takahashi H, Tsuji S, Troncoso J, Dawson VL, Dawson TM, protects from apoptosis upstream of caspase-3. J Neurosci 20:
Ross CA (2001) Interference by huntingtin and atrophin-1 with 3705–3713.
cbp-mediated transcription leading to cellular toxicity. Science Ross CA, Poirier MA (2004) Protein aggregation and neurodegenera-
291:2423–2428.
tive disease. Nat Med 10 (Suppl):S10 –S17.
Ohshima K, Montermini L, Wells RD, Pandolfo M (1998) Inhibitory
Rötig A, de Lonlay P, Chretien D, Foury F, Koenig M, Sidi D,
effects of expanded GAA. TTC triplet repeats from intron I of the
Munnich A, Rustin P (1997) Aconitase and mitochondrial iron-
Friedreich ataxia gene on transcription and replication in vivo.
sulphur protein deficiency in Friedreich ataxia. Nat Genet
J Biol Chem 273:14588 –14595.
17:215–217.
Ono M, Kohda H, Kawaguchi T, Ohhira M, Sekiya C, Namiki M,
Ruan Q, Lesort M, MacDonald ME, Johnson GV (2004) Striatal cells
Takeyasu A, Taniguchi N (1992) Induction of Mn-superoxide dis-
from mutant huntingtin knock-in mice are selectively vulnerable to
mutase by tumor necrosis factor, interleukin-1 and interleukin-6 in
mitochondrial complex II inhibitor-induced cell death through a
human hepatoma cells. Biochem Biophys Res Commun 182:
non-apoptotic pathway. Hum Mol Genet 13:669 – 681.
1100 –1107.
Saft C, Zange J, Andrich J, Muller K, Lindenberg K, Landwehrmeyer B,
Panov AV, Burke JR, Strittmatter WJ, Greenamyre JT (2003) In vitro
Vorgerd M, Kraus PH, Przuntek H, Schols L (2005) Mitochondrial
effects of polyglutamine tracts on Ca2⫹-dependent depolarization
impairment in patients and asymptomatic mutation carriers of Hun-
of rat and human mitochondria: relevance to Huntington’s disease.
tington’s disease. Mov Disord 20:674 – 679.
Arch Biochem Biophys 410:1– 6.
Sakamoto N, Ohshima K, Montermini L, Pandolfo M, Wells RD (2001)
Panov AV, Gutekunst CA, Leavitt BR, Hayden MR, Burke JR, Stritt-
matter WJ, Greenamyre JT (2002) Early mitochondrial calcium Sticky DNA, a self-associated complex formed at long GAA*TTC
defects in Huntington’s disease are a direct effect of polyglu- repeats in intron 1 of the frataxin gene, inhibits transcription. J Biol
tamines. Nat Neurosci 5:731–736. Chem 276:27171–27177.
Park S, Gakh O, O’Neill HA, Mangravita A, Nichol H, Ferreira GC, Sawa A (2001) Mechanisms for neuronal cell death and dysfunction in
Isaya G (2003) Yeast frataxin sequentially chaperones and stores Huntington’s disease: pathological cross-talk between the nucleus
iron by coupling protein assembly with iron oxidation. J Biol Chem and the mitochondria? J Mol Med 79:375–381.
278:31340 –31351. Sawa A, Wiegand GW, Cooper J, Margolis RL, Sharp AH, Lawler JF
Perez-Severiano F, Rios C, Segovia J (2000) Striatal oxidative dam- Jr, Greenamyre JT, Snyder SH, Ross CA (1999) Increased apo-
age parallels the expression of a neurological phenotype in mice ptosis of Huntington disease lymphoblasts associated with repeat
transgenic for the mutation of Huntington’s disease. Brain Res length-dependent mitochondrial depolarization. Nat Med 5:1194 –
862:234 –237. 1198.
Perutz MF (1999) Glutamine repeats and neurodegenerative Seong IS, Ivanova E, Lee JM, Choo YS, Fossale E, Anderson M,
diseases: molecular aspects. Trends Biochem Sci 24:58 – 63. Gusella JF, Laramie JM, Myers RH, Lesort M, MacDonald ME
Piemonte F, Pastore A, Tozzi G, Tagliacozzi D, Santorelli FM, Car- (2005) HD CAG repeat implicates a dominant property of hunting-
rozzo R, Casali C, Damiano M, Federici G, Bertini E (2001) Glu- tin in mitochondrial energy metabolism. Hum Mol Genet 14:
tathione in blood of patients with Friedreich’s ataxia. Eur J Clin 2871–2880.
Invest 31:1007–1011. Schiefer J, Landwehrmeyer GB, Luesse HG, Sprunken A, Puls C,
Polidori MC, Mecocci P, Browne SE, Senin U, Beal MF (1999) Oxida- Milkereit A, Milkereit E, Kosinski CM (2002) Riluzole prolongs
tive damage to mitochondrial DNA in Huntington’s disease parietal survival time and alters nuclear inclusion formation in a trans-
cortex. Neurosci Lett 272:53–56. genic mouse model of Huntington’s disease. Mov Disord
Poulsen HE (2005) Oxidative DNA modifications. Exp Toxicol Pathol 17:748 –757.
57 (Suppl 1):161–169. Schilling G, Coonfield ML, Ross CA, Borchelt DR (2001) Coenzyme
Puccio H, Simon D, Cossee M, Criqui-Filipe P, Tiziano F, Melki J, Q10 and remacemide hydrochloride ameliorate motor deficits in a
Hindelang C, Matyas R, Rustin P, Koenig M (2001) Mouse models Huntington’s disease transgenic mouse model. Neurosci Lett
for Friedreich ataxia exhibit cardiomyopathy, sensory nerve defect 315:149 –153.
and Fe-S enzyme deficiency followed by intramitochondrial iron Schulz JB, Dehmer T, Schols L, Mende H, Hardt C, Vorgerd M, Burk
deposits. Nat Genet 27:181–186. K, Matson W, Dichgans J, Beal MF, Bogdanov MB (2000) Oxida-
E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248 1247

tive stress in patients with Friedreich ataxia. Neurology 55: associated protein 1 influence neuronal calcium signaling
1719 –1721. mediated by inositol-(1,4,5) triphosphate receptor type 1. Neu-
Shi X, Dong Z, Huang C, Ma W, Liu K, Ye J, Chen F, Leonard SS, Ding ron 39:227–239.
M, Castranova V, Vallyathan V (1999) The role of hydroxyl radical Tang TS, Tu H, Orban PC, Chan EY, Hayden MR, Bezprozvanny I
as a messenger in the activation of nuclear transcription factor (2004) HAP1 facilitates effects of mutant huntingtin on inositol
NF-kappaB. Mol Cell Biochem 194:63–70. 1,4,5-trisphosphate-induced Ca release in primary culture of
Shimohata T, Onodera O, Tsuji S (2000) Interaction of expanded striatal medium spiny neurons. Eur J Neurosci 20:1779 –
polyglutamine stretches with nuclear transcription factors leads to 1787.
aberrant transcriptional regulation in polyglutamine diseases. Neu- The Huntington’s Disease Collaborative Research Group (1993) A
ropathology 20:326 –333. novel gene containing a trinucleotide repeat that is expanded
Shoichet SA, Baumer AT, Stamenkovic D, Sauer H, Pfeiffer AF, Kahn and unstable on Huntington’s disease chromosomes. Cell 72:
CR, Muller-Wieland D, Richter C, Ristow M (2002) Frataxin pro- 971–983.
motes antioxidant defense in a thiol-dependent manner resulting in Trushina E, Dyer RB, Badger JD 2nd, Ure D, Eide L, Tran DD,
diminished malignant transformation in vitro. Hum Mol Genet Vrieze BT, Legendre-Guillemin V, McPherson PS, Mandavilli
11:815– 821. BS, Van Houten B, Zeitlin S, McNiven M, Aebersold R, Hayden
Singh KK (2006) Mitochondria damage checkpoint, aging, and cancer. M, Parisi JE, Seeberg E, Dragatsis I, Doyle K, Bender A, Chacko
Ann N Y Acad Sci 1067:182–190. C, McMurray CT (2004) Mutant huntingtin impairs axonal traf-
Sjogren M, Mielke M, Gustafson D, Zandi P, Skoog I (2006) Choles- ficking in mammalian neurons in vivo and in vitro. Mol Cell Biol
terol and Alzheimer’s disease: is there a relation? Mech Ageing 24:8195– 8209.
Dev 127:138 –147. Trushina E, Heldebrant MP, Perez-Terzic CM, Bortolon R, Kovtun IV,
Squitieri F, Cannella M, Sgarbi G, Maglione V, Falleni A, Lenzi P, Badger JD 2nd, Terzic A, Estevez A, Windebank AJ, Dyer RB, Yao
Baracca A, Cislaghi G, Saft C, Ragona G, Russo MA, Thompson J, McMurray CT (2003) Microtubule destabilization and nuclear
LM, Solaini G, Fornai F (2006) Severe ultrastructural mitochondrial entry are sequential steps leading to toxicity in Huntington’s dis-
changes in lymphoblasts homozygous for Huntington disease mu- ease. Proc Natl Acad Sci U S A 100:12171–12176.
tation. Mech Ageing Dev 127:217–220. Tukamoto T, Nukina N, Ide K, Kanazawa I (1997) Huntington’s dis-
Steffan JS, Kazantsev A, Spasic-Boskovic O, Greenwald M, Zhu YZ, ease gene product, huntingtin, associates with microtubules in
Gohler H, Wanker EE, Bates GP, Housman DE, Thompson LM
vitro. Brain Res Mol Brain Res 51:8 –14.
(2000) The Huntington’s disease protein interacts with p53 and
Valenza M, Rigamonti D, Goffredo D, Zuccato C, Fenu S, Jamot L,
CREB-binding protein and represses transcription. Proc Natl Acad
Strand A, Tarditi A, Woodman B, Racchi M, Mariotti C, Di
Sci U S A 97:6763– 6768.
Donato S, Corsini A, Bates G, Pruss R, Olson JM, Sipione S,
Sugars KL, Brown R, Cook LJ, Swartz J, Rubinsztein DC (2004)
Tartari M, Cattaneo E (2005) Dysfunction of the cholesterol
Decreased cAMP response element-mediated transcription: an
biosynthetic pathway in Huntington’s disease. J Neurosci
early event in exon 1 and full-length cell models of Huntington’s
25:9932–9939.
disease that contributes to polyglutamine pathogenesis. J Biol
van Dellen A, Grote HE, Hannan AJ (2005) Gene-environment
Chem 279:4988 – 4999.
interactions, neuronal dysfunction and pathological plasticity in
Suopanki J, Gotz C, Lutsch G, Schiller J, Harjes P, Herrmann A,
Huntington’s disease. Clin Exp Pharmacol Physiol 32:1007–
Wanker EE (2006) Interaction of huntingtin fragments with brain
1019.
membranes: clues to early dysfunction in Huntington’s disease.
van Loon AP, Pesold-Hurt B, Schatz G (1986) A yeast mutant lacking
J Neurochem 96:870 – 884.
mitochondrial manganese-superoxide dismutase is hypersensitive
Swerdlow RH, Parks JK, Cassarino DS, Shilling AT, Bennett JP Jr,
to oxygen. Proc Natl Acad Sci U S A 83:3820 –3824.
Harrison MB, Parker WD Jr (1999) Characterization of cybrid cell
lines containing mtDNA from Huntington’s disease patients. Bio- Visner GA, Dougall WC, Wilson JM, Burr IA, Nick HS (1990) Regula-
chem Biophys Res Commun 261:701–704. tion of manganese superoxide dismutase by lipopolysaccharide,
Szebenyi G, Morfini GA, Babcock A, Gould M, Selkoe K, Stenoien DL, interleukin-1, and tumor necrosis factor. Role in the acute inflam-
Young M, Faber PW, MacDonald ME, McPhaul MJ, Brady ST matory response. J Biol Chem 265:2856 –2864.
(2003) Neuropathogenic forms of huntingtin and androgen recep- Vorgerd M, Schols L, Hardt C, Ristow M, Epplen JT, Zange J (2000)
tor inhibit fast axonal transport. Neuron 40:41–52. Mitochondrial impairment of human muscle in Friedreich ataxia in
Tabrizi SJ, Blamire AM, Manners DN, Rajagopalan B, Styles P, Scha- vivo. Neuromuscul Disord 10:430 – 435.
pira AH, Warner TT (2005) High-dose creatine therapy for Hun- Waelter S, Scherzinger E, Hasenbank R, Nordhoff E, Lurz R, Goehler
tington disease: a 2-year clinical and MRS study. Neurology H, Gauss C, Sathasivam K, Bates GP, Lehrach H, Wanker EE
64:1655–1656. (2001) The huntingtin interacting protein HIP1 is a clathrin and
Tabrizi SJ, Cleeter MW, Xuereb J, Taanman JW, Cooper JM, Schapira alpha-adaptin-binding protein involved in receptor-mediated endo-
AH (1999) Biochemical abnormalities and excitotoxicity in Hunting- cytosis. Hum Mol Genet 10:1807–1817.
ton’s disease brain. Ann Neurol 45:25–32. White JK, Auerbach W, Duyao MP, Vonsattel JP, Gusella JF, Joyner
Takahashi K, Avissar N, Whitin J, Cohen H (1987) Purification and AL, MacDonald ME (1997) Huntingtin is required for neurogenesis
characterization of human plasma glutathione peroxidase: a sel- and is not impaired by the Huntington’s disease CAG expansion.
enoglycoprotein distinct from the known cellular enzyme. Arch Nat Genet 17:404 – 410.
Biochem Biophys 256:677– 686. Wilson RB (2003) Frataxin and frataxin deficiency in Friedreich’s
Tan G, Chen LS, Lonnerdal B, Gellera C, Taroni FA, Cortopassi GA ataxia. J Neurol Sci 207:103–105.
(2001) Frataxin expression rescues mitochondrial dysfunctions in Wong A, Yang J, Cavadini P, Gellera C, Lonnerdal B, Taroni F,
FRDA cells. Hum Mol Genet 10:2099 –2107. Cortopassi G (1999) The Friedreich’s ataxia mutation confers cel-
Tang TS, Slow E, Lupu V, Stavrovskaya IG, Sugimori M, Llinas R, lular sensitivity to oxidant stress which is rescued by chelators of
Kristal BS, Hayden MR, Bezprozvanny I (2005) Disturbed Ca2⫹ iron and calcium and inhibitors of apoptosis. Hum Mol Genet
signaling and apoptosis of medium spiny neurons in Hunting- 8:425– 430.
ton’s disease. Proc Natl Acad Sci U S A 102:2602– Wyttenbach A, Swartz J, Kita H, Thykjaer T, Carmichael J, Bradley J,
2607. Brown R, Maxwell M, Schapira A, Orntoft TF, Kato K, Rubinsztein
Tang TS, Tu H, Chan EY, Maximov A, Wang Z, Wellington CL, DC (2001) Polyglutamine expansions cause decreased CRE-me-
Hayden MR, Bezprozvanny I (2003) Huntingtin and huntingtin- diated transcription and early gene expression changes prior to cell
1248 E. Trushina and C. T. McMurray / Neuroscience 145 (2007) 1233–1248

death in an inducible cell model of Huntington’s disease. Hum Mol Zeron MM, Hansson O, Chen N, Wellington CL, Leavitt BR, Brundin P,
Genet 10:1829 –1845. Hayden MR, Raymond LA (2002) Increased sensitivity to N-meth-
Yu W, Gong JS, Ko M, Garver WS, Yanagisawa K, Michikawa M yl-D-aspartate receptor-mediated excitotoxicity in a mouse model
(2005) Altered cholesterol metabolism in Niemann-Pick type C1 of Huntington’s disease. Neuron 33:849 – 860.
mouse brains affects mitochondrial function. J Biol Chem 280: Zhou Z, Kang YJ (2000) Cellular and subcellular localization of catalase
11731–11739. in the heart of transgenic mice. J Histochem Cytochem 48:585–594.

(Accepted 27 October 2006)


(Available online 14 February 2007)

You might also like