Anda di halaman 1dari 13

Free Radical Biology and Medicine 62 (2013) 132144

Contents lists available at SciVerse ScienceDirect

Free Radical Biology and Medicine


journal homepage: www.elsevier.com/locate/freeradbiomed

Review Article

Parkinson disease: from pathology to molecular disease mechanisms


David T. Dexter a, Peter Jenner b,n
a
Parkinsons Disease Research Group, Centre for Neuroinammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College
London, Hammersmith Hospital Campus, London, UK
b
Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, Kings College London, London SE1 9NH, UK

a r t i c l e i n f o a b s t r a c t

Available online 4 February 2013 Parkinson disease (PD) is a complex neurodegenerative disorder with both motor and nonmotor
Keywords: symptoms owing to a spreading process of neuronal loss in the brain. At present, only symptomatic
Parkinson disease treatment exists and nothing can be done to halt the degenerative process, as its cause remains unclear.
Genes Risk factors such as aging, genetic susceptibility, and environmental factors all play a role in the onset
Oxidative stress of the pathogenic process but how these interlink to cause neuronal loss is not known. There have been
Mitochondrial dysfunction major advances in the understanding of mechanisms that contribute to nigral dopaminergic cell death,
Protein handling including mitochondrial dysfunction, oxidative stress, altered protein handling, and inammation.
Inammation However, it is not known if the same processes are responsible for neuronal loss in nondopaminergic
Neuroprotection
brain regions. Many of the known mechanisms of cell death are mirrored in toxin-based models of PD,
Free radicals
but neuronal loss is rapid and not progressive and limited to dopaminergic cells, and drugs that protect
against toxin-induced cell death have not translated into neuroprotective therapies in humans. Gene
mutations identied in rare familial forms of PD encode proteins whose functions overlap widely with
the known molecular pathways in sporadic disease and these have again expanded our knowledge of
the neurodegenerative process but again have so far failed to yield effective models of sporadic disease
when translated into animals. We seem to be missing some key parts of the jigsaw, the trigger event
starting many years earlier in the disease process, and what we are looking at now is merely part of a
downstream process that is the end stage of neuronal death.
& 2013 Elsevier Inc. All rights reserved.

Contents

Dening Parkinson disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133


Motor and nonmotor symptoms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133
Spreading pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133
Current approaches to treatmentno cure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Risk factors for developing PD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Predisposing factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Protective factors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Genetics of PDgene mutations and genome-wide association studies (GWAS). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Mechanisms of neurodegeneration. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Oxidative stress in Parkinson disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Altered mitochondrial function in PD. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Altered proteolysis in PDproteasomal and lysosomal. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
Inammatory change . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
Excitotoxic mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138

Abbreviations: SNc, substantia nigra pars compacta; PD, Parkinson disease; NMS, nonmotor symptoms; MAO-B, monoamine oxidase-B; MPP , 1-methyl-4-phenylpyridinium;
MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; 6-OHDA, 6-hydroxy dopamine; GBA, glucocerebrosidase; STN, subthalamic nucleus; DBS, deep brain stimulation; LPS,
lipopolysaccharide; UCH-L1, ubiquitin carboxyl-terminal hydrolase L1; PINK1, phosphatase and tensin homolog-inducible kinase 1; LRRK2, leucine-rich repeat kinase 2; GWAS,
genome-wide association studies; mtDNA, mitochondrial DNA; PGC-1a, peroxisome proliferator-activated receptor g coactivator-1a; UPS, ubiquitinproteasome system; LAMP2A,
lysosome-associated membrane protein type 2A; HLA, human leukocyte antigen; IL, interleukin; TNF-a, tumor necrosis factor-a
n
Corresponding author. Fax: 44 20 7848 6034.
E-mail address: peter.jenner@kcl.ac.uk (P. Jenner).

0891-5849/$ - see front matter & 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.freeradbiomed.2013.01.018
D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144 133

Bringing it all together? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138


Translation into animal models of PD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
Toxin relevance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
Gene relevance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
From pathogenesis to neuroprotection? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141

Dening Parkinson disease


From the perspective of this review, the most important
Parkinson disease (PD)1 is the second most common neurode- feature of NMS is that some, for example, olfactory decits,
generative disorder after Alzheimer disease, with prevalence in constipation, rapid-eye-movement sleep behavior disorder, and
industrialized countries of approximately 0.3% of the population. depression, may precede the onset of motor symptoms by many
This rises with age from 1% in those over 60 years of age to 4% of years (although they can occur at the same time as motor
the population over 80, illustrating the effect of aging. The mean symptoms or follow the onset of motor abnormalities) [105].
age of onset is approximately 60 years; however, 10% of cases are NMS may in the future be used for the early diagnosis of PD,
classied as young onset, occurring between 20 and 50 years of enabling neuroprotective strategies to be introduced at an early
age, which may represent a distinct disease group. PD is more stage, and studies of large populations of apparently normal older
prevalent in men than in women, with reports of ratios of 1.1:1 to individuals are ongoing at this time to enable such early detection
almost 3:1 being quoted [135], which may be attributable to the to occur [6,143]. However, they provide another and perhaps vital
protective effects of estrogen in women [121]. The socioeconomic clue to the search for the pathogenic processes that underlie PD,
cost of PD is high and in the United Kingdom is estimated to be as they suggest that it is a disease of both the peripheral and the
approximately 3.3 billion. In the United States, the cost per central nervous systems and that it is a multisystem disorder that
patient per year is around $10,000, with a total economic burden spreads with time, affecting movement only at a relatively late
of $23 billion. The greatest proportion of cost comes in the later stage in its course, and may thus be a target for early diagnosis
stages of the illness for inpatient care and nursing homes and far and identication of at-risk populations. Current dopamine repla-
less for medication [27]. cement strategies for treating PD are effective against the motor
This review aims to provide a current overview of the clinical, features of PD but are largely ineffective at addressing NMS.
neuropathological, and biochemical features of PD, the genetic Hence a greater effort needs to be made not only in under-
components of the disease, and risk factors for its development in standing the molecular mechanisms that cause NMS but also in
relation to the pathogenic mechanisms thought to be involved in how to treat them.
neuronal death. Currently drug treatment provides only sympto-
matic relief and we explore how knowledge of the pathogenic
processes and the use of experimental models of PD interlink Spreading pathology
to assist in the search for neuroprotective/neurorestorative
treatments. Neuronal loss in the substantia nigra pars compacta (SNc) and
the subsequent loss of striatal dopamine content are accepted as
being responsible for the classical motor features of PD. The
neuropathological diagnosis of PD requires the detection of
Motor and nonmotor symptoms marked dopaminergic neuronal loss in the SNc and the presence
of Lewy bodies, eosinophilic inclusions consisting of a dense core
Impaired motor function is classically used to make a clinical surrounded by a pale-staining halo of radiating laments. The role
diagnosis of PD. The main features are bradykinesia, rigidity, of the Lewy body in pathogenesis remains unknown, but the
tremor, and postural instability with an asymmetric onset spread- discovery that misfolded a-synuclein is a major component of the
ing to become bilateral with time. Other motor features include radiating laments and is also present in neuronal processes as
gait and posture changes that manifest as festination (rapid Lewy neurites has altered views on their formation and role in
shufing steps with a forward-exed posture when walking), neuronal loss and has led to a major shift in thinking about the
speech and swallowing difculties, and a masklike facial expres- onset and progression of the disease process from a pathological
sion and micrographia [58]. A good response to dopaminergic perspective and to the classication of PD as a synucleinopathy
medication is conrmatory of the diagnosis. Although this has [21]. However, the neuropathological picture of PD has been
been the classical textbook description of PD, more recently it has known to be more widespread for many decades, with many
become recognized as a more complex illness encompassing both nondopaminergic nuclei affected, including the locus coeruleus,
motor and nonmotor symptoms (NMS), such as depression, sleep reticular formation of the brain stem, raphe nucleus, dorsal motor
disturbance, sensory abnormalities, autonomic dysfunction, and nucleus of the vagus, basal nucleus of the Meynert, amygdala, and
cognitive decline [74]. NMS affect all patients with PD, the hippocampus [59]. Importantly, all of these nuclei degenerate
frequency of which increases with disease severity, with late- with Lewy body pathology, suggesting a pathogenic process in
stage patients exhibiting 610 NMS. NMS create the biggest common with that occurring in the SNc. It is the Lewy body
demand on clinical resources, they are poorly diagnosed and pathology in these nondopaminergic areas that then results in
treated and they are the major determinant of disease outcome, some NMS. For example, hyposmia is associated with the pre-
increasing disability, poor quality of life, and entry into long-term sence of Lewy bodies and Lewy neurites in the olfactory bulb and
care [13]. Whereas the causes of motor dysfunction in PD are brain centers such as the amygdala and perirhinal nucleus
reasonably well understood (see later), the cause of NMS in [78,162]. But it is the presence of Lewy bodies in peripheral
PD remains poorly researched and they may largely relate to tissues that takes the pathology associated with PD to another
pathology outside of the basal ganglia. level. Orthostatic hypotension in PD is associated with
134 D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144

sympathetic autonomic denervation of the heart with the pre- rate of nigral cell degeneration compared to that seen in patients
sence of Lewy bodies [28,112]. Lewy bodies are found in the treated with L-dopa [117,161]. However, the drugs themselves
myenteric plexus, reecting the alterations in gastrointestinal turned out to affect the imaging markers employed [104]. Even in
motility occurring in PD. However, there may be a connection mild PD, the early use of pramipexole showed no advantage on
between changes originating in the periphery. For example, disease progression over the later introduction of the drug
constipation as a common NMS is associated with neuronal loss (Pramipexole on Underlying DiseasePROUD study) [131].
and the presence of Lewy bodies in the dorsal motor nucleus of L-dopa has had a checkered career as a potential reason for
the vagus, which provides parasympathetic innervation to the both acceleration of the cell death in PD and its slowing. For many
stomach and intestine [10,20,159], and changes such as these can years, the ability of L-dopa to generate free radicals and to kill
be initiated by injection of proteasomal inhibitors into the dopaminergic cells in culture tainted the drug with a neurotoxic
stomach wall and retrograde degeneration of the dorsal motor label, but overall, based on a range of in vivo studies in animals,
nucleus, at least in rats [102]. postmortem studies in humans, and clinical experience, this does
Indeed, what has become clear is that pathology in PD does not not seem to be true, although the debate continues [1,108,120].
start in the SNc, but rather Lewy body pathology and the deposi- From the Earlier versus Late Levodopa (ELLDOPA) clinical trial,
tion of a-synuclein are proposed to originate in the olfactory bulb among others, early use of L-dopa seems to slow disease progres-
and lower brain stem, from where they spread in stages to involve sion based on clinical rating scales, but not when using imaging
the midbrain, eventually spreading to cortical regions. A staging endpoints [26].
of the caudorostral spread of Lewy body and a-synuclein patho- It has been the MAO inhibitors selegiline and rasagiline,
logy in PD was rst proposed by Braak and colleagues in 2003 however, that have attracted most attention as potential disease
[11], when they proposed that a-synuclein deposition begins in modiers. It was the ability of these drugs to prevent 1-methyl-
the dorsal motor nucleus of the vagus (stage 1), from where it is 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity to nigral
thought to proceed in an upward direction via the pons (stage 2) dopaminergic neurons through inhibition of 1-methyl-4-phenyl-
to the midbrain (stage 3) and from there to the basal prosenca- pyridinium (MPP ) by blocking MAO-B activity [60]. However,
phalon and mesocortex (stage 4), nally reaching the neocortex both drugs also are associated with a range of actions that
(stages 5 and 6). It is only at stage 3 after extensive loss of encompass antiapoptotic actions, antioxidant effects, antigluta-
dopaminergic neurons in the SNc that motor features of PD matergic effects, and neurotrophic actions, among others, that
become apparent. Although it is recognized that not all PD cases underlie a belief in their ability to alter the rate of loss of
exactly follow this formal pattern of spread of PD pathology [68], dopaminergic neurons [62,84]. Both selegiline and rasagiline have
it is now widely accepted that spreading pathology does occur in been examined in detail in clinical trials to assess whether disease
PD. This raises the important concept that PD pathology may be modication occurs in humans. The Deprenyl and Tocopherol
propagated from one neuron to another [34]. Exactly how this Antioxidative Therapy of Parkinsonism (DATATOP) study initially
occurs is not known, but altered a-synuclein released by an suggested that selegiline treatment could delay the need for the
affected neuron that is taken up by an adjacent unaffected introduction of L-dopa in early PD but this now seems largely due
neuron, or direct transfer between neurons, could act as a seed to a symptomatic effect of the drug, although open label exten-
in a prion-like mechanism to perpetuate the cycle of a-synuclein sion studies in other clinical trials still suggest that disease
misfolding and the spread of a-synuclein pathology [23]. Although progression is slower in those patients with PD that receive early
this hypothesis is controversial some groups have demonstrated selegiline treatment [115,119]. Similarly, for rasagiline, the
such a seeding mechanism in cell culture, animal models, and results of the TEMPO (TVP-1012 in Early Monotherapy for
fetal mesencephalic cells transplanted into the PD striatum [23]. Parkinsons Disease Outpatients) and ADAGIO (the Attenuation
If such a hypothesis is correct, it will have a major impact on the of Disease Progression with Azilect Given Once Daily) studies
molecular understanding of pathogenesis in PD. suggested that early use had a positive effect on clinical scores
compared to later introduction of the drug, most notably in those
patients with the highest degree of motor disability [109,118].
Current approaches to treatmentno cure However, so far none of the studies undertaken have convinced
the regulatory authorities to label either selegiline or rasagiline as
The treatment of PD has not changed substantially in the past 30 disease modifying.
years, with dopamine replacement therapy employing L-dopa and One other approach to current therapy deserves mention,
dopamine agonists as the mainstay, supported by the use of a series namely, suppression of glutamatergic activity in PD. As will be
of enzyme inhibitors, namely peripheral decarboxylase inhibitors, explained later, excitotoxicity may contribute to the pathogenic
catechol-O-methyl transferase inhibitors, and monoamine oxidase-B process and as a consequence inhibition of glutamatergic function
(MAO-B) inhibitors. These dopaminergic medications now come in a may inuence disease progression. This has stimulated new
variety of oral, intraduodenal, intravenous, subcutaneous, and trans- interest in the actions of amantadine, which is currently used
dermal forms, providing differing degrees of drug delivery. Outside of largely to suppress dyskinesia in PD. However, there is anecdotal
the dopaminergic arena only anticholinergics and the weak N- evidence that treatment with amantadine from the beginning of
methyl-D-aspartate receptor antagonist amantadine have had any therapy may lead to a better outcome than would otherwise be
use. But these are all symptomatic approaches to treating the motor expected, and new clinical studies are being started with the drug
decits of PD with little effect on nonmotor symptoms and no proven to provide evidence-based medicine for its effects.
effect on disease progression.
There have been several attempts to determine whether
current therapy has any effect on the disease process. The Risk factors for developing PD
dopamine agonists ropinirole and pramipexole show neuropro-
tective actions in preclinical models of PD of dopaminergic cell Predisposing factors
loss, but in clinical trial (REAL-PET; the Comparison of the Agonist
Pramipexole versus Levodopa on Motor Complications of Parkin- Age represents the biggest predisposing factor for PD (young-
sons DiseaseCALM-PD) there was no slowing of progression of onset cases may form a different patient population) for the
motor symptoms, although imaging studies suggested a lower majority of individuals developing the illness. However, it
D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144 135

remains unknown whether it is chronological age or the aging by negative reports on these compounds [5,146,148,169]. Perhaps
process that is responsible [69]. Loss of estrogen production in surprisingly, but of enormous potential interest, is a role for
women with age may also remove a protective effect and there is exercise and metabolic factors associated with the risk of developing
some evidence that early menopause, hysterectomy, or removal PD [169].
of the ovaries increases risk in women to that seen in men
[121,123]. Genetic causes are becoming more common as gene
hunting continues (see later) and may account for up to 40% of Genetics of PDgene mutations and genome-wide association
those at risk in the population from autosomal dominant, reces- studies (GWAS)
sive, and susceptibility genes [41]. However, there are also
environmental factors that contribute to the risk of developing Investigation of familial PD has so far revealed at least 17
PD [148]. autosomal dominant and autosomal recessive gene mutations
The discovery of the toxicity of MPTP to nigral dopaminergic responsible for variants of the disease [47]. These include
cells generated interest in environmental causes of the disease, a-synuclein mutations and triplication, parkin, ubiquitin carboxyl-
although no other MPTP-like toxins have been discovered [53]. terminal hydrolase L1 (UCH-L1), DJ-1, phosphatase and tensin
The closest has been the ability of the structurally related homolog-inducible kinase 1 (PINK1), leucine-rich repeat kinase 2
paraquat, a commercial weed killer, to destroy dopaminergic cells (LRRK2), and glucocerebrosidase (GBA) (see Table 1). Of these
in rodents and this links nicely to the increased risk of developing parkin and LRRK2 are probably the most common genetic link to
PD that repeatedly emerges from epidemiological studies, young-onset and late-onset PD, respectively, whereas GBA muta-
although there is no association with any specic compound tions may be the most common risk factor. Autosomal dominant
[90,149,150]. The same might apply to the complex I inhibitor mutations are typied by mutations in a-synuclein and LRRK2.
rotenone as a constituent of derris, which is commonly used by Although a-synuclein mutations are only rarely encountered,
gardeners as a pesticide, but no cases have specically been their discovery led directly to a-synuclein being identied as a
reported [7]. In contrast, exposure to annonacin derived from major component of Lewy bodies and Lewy neurites and the
fruits in Guadeloupe was shown to produce parkinsonism [75]. labeling of PD as a synucleinopathy, underpinning much of the
Other environmental causes include solvent exposure (n-hexane, consensus on the nal stages of neuronal loss in PD being related
methanol), carbon monoxide poisoning, hydrogen sulde intox- to altered protein aggregation [33,35]. Mutations in LRRK2 seem
ication, and perhaps manganese, although the resulting syndrome to alter kinase/GTPase of this mixed lineage-like kinase found in
involves striatal cell degeneration and dystonic features and cytoplasm and outer membrane of mitochondria [17,39]. LRRK2
there now seems to be no increased risk in welders, who can be interacts with another PD-related protein, parkin, and mutant
exposed to manganese vapor [70]. LRRK2 induces apoptotic cell death in cultured neurons. Auto-
One last factor worthy of note is head trauma, as it has somal loss of function mutations include those in the ubiquitin
repeatedly emerged as borderline signicant for increased risk E3 ligase parkin, which in combination with the ubiquitin-
of PD in population studies. A recent study in ex-National Football conjugating enzyme causes the attachment of ubiquitin as a
League players in the United States suggests that head trauma marker on proteins destined for destruction by the proteasome.
does increase the risk of developing PD [81] and so this may be a Additionally mutations in the mitochondrial PINK1 protect cells
more relevant factor than previously thought, although contro- from mitochondrial stress/dysfunction, and the rarest mutation,
versial [128]. in the redox-sensitive chaperone DJ-1, protects cells against
oxidative stress [17,39]. Familial forms of PD and the associated
Protective factors gene mutations currently account for approximately 10% of cases
and they have distinct clinical and pathological phenotypes.
Factors that protect against the development of PD may reveal However, there is sufcient overlap with sporadic PD that
a lot about underlying pathogenic mechanisms. Perhaps the investigations into such gene mutations have revealed important
protective effect of cigarette smoking is the best documented clues as to the molecular mechanisms that underlie the disease
and most reproducible of observations [72]. Nicotine is the process in PD (see Fig. 1). Indeed, many of these familial PD
immediate candidate for a neuroprotective action and there is neurodegeneration mechanisms overlap with the pathogenic
currently interest in developing nicotinic agonists, for example, a- mechanisms discovered in sporadic PD, such as mitochondrial
7 nicotinic agonists, for both symptomatic treatment and neuro- dysfunction, oxidative stress, and altered protein handling (see
protection [122]. However, cigarette smoke contains more than below and Fig. 1) [47]. Furthermore a combination of advances
4000 components, so other agents may contribute to the protec- in genetic analysis techniques, the ability to genotype cost
tive effects, for example, smoking decreases MAO-B activity in
brain [165]. Additionally, it may be that a genetic disposition Table 1
against reward-seeking habits, such as cigarette smoking, may Common gene mutations causing familial Parkinson disease.
prevail in those who go on to have a clinical diagnosis of PD.
Caffeine intake also appears protective against the development Gene Locus/disease Mode of inheritance Age of onset (years)

of PDmore so in men than in women and more so in those not SNCA PARK1/4 Autosomal dominant 2085
using hormone replacement therapy [114]. Caffeine has been LRRK2 PARK8 Autosomal dominant 3279
demonstrated to exert neuroprotective effects in a variety of GRN FTDP-17 Autosomal dominant 4583
experimental models of PD, possibly via the involvement of the MAPT FTDP-17 Autosomal dominant 2576
DCTN1 Perry syndrome Autosomal dominant 3561
adenosine A2a receptor [14]. This has highlighted the potential
PRKN PARK2 Autosomal recessive 1672
neuroprotective effect of A2a antagonists, such as istradephylline PINK1 PARK6 Autosomal recessive 2040
and preladenant, which are currently under development as DJ1 PARK7 Autosomal recessive 2040
symptomatic treatments for PD. FBXO2 PARK15/PPS Autosomal recessive 1019
Other potentially protective factors are not so well dened NR4A2/NURR1 Unknown 4567
POLG Unknown 2026
but there have been suggestions of an effect of antihypertensives
(notably calcium antagonists), nonsteroidal anti-inammatory FTDP-17, frontotemporal dementia with parkinsonism linked to chromosome 17;
drugs, and antilipidemics, among others, although all are balanced PPS, palidopyramidal syndrome.
136 D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144

Parkin UCH-L1

Glutamate
-synuclein
ExcitotoxicityParkin
Ubiquitin Proteasome

Misfolded
protein
Proteasome Ca2+
Dysfunction
Dopaminergic neuronal
environment PINK1
Aggregated
Protein

Iron Mitochondria DJ-1


Oxidative
stress

Energy
Cytokines Apoptosis
depletion
ROS
Lack of
growth
Activated factor
Microglia support

Neurodegeneration
Innate
Inflammation

Fig. 1. Key molecular mechanisms that are widely accepted to contribute to the neurodegenerative process in dopaminergic neurons in the substantia nigra in Parkinson
disease. Blue double-headed arrows indicate molecular mechanisms that may not only be toxic in their own right but importantly may also inuence other molecular
mechanisms known to be features in Parkinson disease. Double helix structures identify some of the common gene mutations found in familial PD and brown arrows
indicate where the altered protein may interfere with cell function and overlap with known mechanisms of cell death in sporadic Parkinson disease. ROS, reactive oxygen
species. (For interpretation of the references to color in this gure legend, the reader is referred to the web version of this article.)

effectively, and the formation of large patient sample consortia, plasma urate levels and disease progression in PD has raised the
for example, the International PD Genomics Consortium, has spectrum of altered antioxidant activity that reects the decrease
facilitated highly powered GWAS in idiopathic PD, which have previously reported in reduced glutathione levels (along with changes
revealed some 14 risk gene loci for PD, including a-synuclein, in catalase and glutathione peroxidase) [3,4,15,36,138].
LRRK2, human leukocyte antigen (HLA), and tau [55,106] (see The source of the oxidative stress remains hotly contested, with
Table 2). Although the presence of such risk gene loci is not a both neuronal and glial sources being implicated, but there seems
predictor for the development of PD, they highlight the fact that little doubt that the most likely contributor is increased radical
PD is a complex disease involving both genetic risk and environ- formation originating from mitochondria (see later) and, perhaps,
mental factors in its etiology. endoplasmic reticulum. Altered accumulation of iron in SNc, changes
in calcium channel activity, altered proteolysis (proteasomal and
lysosomal), changes in a-synuclein aggregation, and the presence of
Mechanisms of neurodegeneration mutant proteins (for example DJ-1) are all examples of how oxidative
stress might be induced in PD [126,133,146,152]. This emphasizes the
Oxidative stress in Parkinson disease diverse nature of the causes of cell death in this illness but also the
common nal pathways through which neuronal loss occurs. There is
Oxidative stress remains a cornerstone of the concepts under- nothing specic about a role for oxidative stress in PD, as it also
lying the loss of dopaminergic neurons in PD. Since the 1980s contributes to cell degeneration in a variety of other disease states.
there has been an exponential growth in publications that However, it may offer the opportunity to inuence disease progres-
implicate the formation of reactive oxygen species as a nal step sion although attempts to date have been unsuccessful in clinical
in neuronal death of whatever origin. Starting from the idea of trials despite a multitude of encouraging reports from both in vitro
free radical production resulting from increased chemical and and in vivo models of PD. However, ongoing studies with centrally
enzymatic oxidation of dopamine, through to the mechanism of active iron-chelating drugs and the elevation of urate levels through
action of toxins such 6-hydroxydopamine (6-OHDA) and para- administration of uric acid or inosine administration offer new hope.
quat, to evidence from postmortem and clinical investigations,
oxidative stress and the resulting oxidative damage have emerged Altered mitochondrial function in PD
largely unscathed [36,61,63,64,141]. Whereas direct evidence for
increased free radical generation is almost impossible to detect, Mitochondrial involvement in cell death in PD has returned to
the potential inducers of oxidative stress and markers of its effects center stage and provides part of a unifying concept of how
make a substantial case for its occurrence. Enhanced lipid peroxida- neuronal loss occurs in both sporadic and inherited disease.
tion (malondialdehyde, lipid hydroperoxides, 4-hydroxynonenal, and The discovery of the neurotoxicity of MPTP through its metabolite
advanced glycation end products), protein oxidation (protein carbo- MPP identied a role for the inhibition of complex I in pathogenesis
nyls), and DNA oxidation (8-hydroxyguanosine) in the SNc in PD and, that is shared by other substances toxic to dopaminergic neurons,
perhaps, more generally throughout the body, all point in the same including rotenone and annonacin [32,76,124]. Very quickly, an
direction. More recently, the nding of a negative correlation between inhibition of complex I was shown to be present in the SNc that
D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144 137

Table 2
GWAS risk genes for Parkinson disease.

Locus SNP Protein

MAPT rs2942168 Microtubule-associated protein tau


SNCA rs356221 a-Synuclein
BST1 rs4698412 Bone marrow stromal cell antigen 1
LRRK2 rs1491942 Leucine-rich repeat kinase-2
GAK rs11248051 Cyclin G-associated kinase
HLA-DRB5 rs3129882 Major histocompatibility complex, class II, DR b5
ACMSD rs10928513 Aminocarboxymuconate semialdehyde decarboxylase
STK39 rs2102808 Serine threonine kinase 39
HIP1R rs10847864 Huntingtin-interacting protein 1-related
MCCC1/LAMP3 rs11711441 Lysosomal-associated membrane protein 3
SYT11 rs34372695 Synaptotagmin-11
GPNMB rs156429 Glycoprotein (transmembrane) NMB
STX1B rs4889603 Syntaxin 1B
FGF20 rs591323 Fibroblast growth factor 20

was tissue and disease specic, although also detectable in platelets supported by the discovery of mutations in parkin and UCH-L1, which
and muscle in PD [85,129,130,132]. A key Krebs cycle enzyme, a- function as a ubiquitinprotein ligase and in ubiquitin recycling.
ketoglutarate dehydrogenase, was also shown to be impaired [103]. Examination of the 26S proteasome in PD revealed selective changes
The construction of cybrids using mitochondrial DNA (mtDNA) from in its catalytic activity and composition in the SNc that were
patients with PD has clearly demonstrated the encoding of the associated, perhaps wrongly, with impaired degradation of a-synu-
complex I defect [38], but no consistent alterations in mtDNA have clein [97,100,151]. In cell culture and after direct intracerebral
been demonstrated, although deletions associated with complex IV injection, proteasomal inhibitors, such as lactacystin and proteasome
and oxidative damage occur in PD. Perhaps importantly, only about inhibitor 1 (PSI), were found to selectively destroy dopaminergic
30% of patients with PD have a clear complex I defect, suggesting, neurons [88,94,98,163]. In cell culture, the initiation of cell death
perhaps, that they form an important subgroup within the disease using proteasomal inhibitors led to a cascade of events involving
that offers a specic target for interference with the disease process. increases in oxidative and nitrative stress and damage and alterations
More recently, interest in the role of mitochondria has stemmed to mitochondrial function [50]. Indeed, there is a close association
from genetic investigations in familial PD. Notably mutations in a- between mitochondrial electron transport activity and the regulatory
synuclein, parkin, PINK1, and DJ-1 and perhaps LRRK2 have been caps of the 26S proteasome, which are ATPases. The potential role of
associated with altered mitochondrial function [129,130,133] (see the UPS has been conrmed many times over and supported by
Fig. 1). These mutations can lead to altered protein localization in microarray and transcriptional studies on laser-captured microdis-
mitochondria in PD, abnormalities in mitochondrial structure and sected nigral dopaminergic neurons from PD [37]. However, the
function, and a decrease in complex I assembly and activity. Loss of concept lost credibility when a slow, progressive degeneration of
function of, notably, DJ-1, but also parkin and PINK1, decreases neurons in the brain that resembled that occurring in PD was claimed
mitochondrial protection against oxidative stress, which in turn after systemic administration of proteasomal inhibitors [99], but this
increases mitochondrial dysfunction. Another important role for was never substantively reproduced (compare [73,86] with [12,167]).
parkin and PINK1 is in the turnover of mitochondria by autophagy, As a consequence attention turned to lysosomes and to
specically mitophagy; they act in tandem to regulate this autophagy (macro-autophagy, micro-autophagy, and chaperone-
process. This may be critically important in PD, in which autophagy mediated autophagy) (see [133]). Because autophagy-related
seems impaired, reducing the cells ability to remove damaged proteins and autophagosomes are present in Lewy bodies, this
mitochondria. seemed a viable target for disruption related to dopaminergic cell
GWAS, laser-captured human dopaminergic neuron studies, and death. Indeed, the expression of the chaperone-mediated autop-
SNc transcriptome studies have added to the evidence pointing to the hagy proteins lysosome-associated membrane protein type 2A
key role of decits in the mitochondrial electron transport chain in PD (LAMP2A) and heat shock protein 70 is reduced in SNc in PD [2].
[25]. Genes controlling cellular bioenergetics that are expressed in In addition, silencing of LAMP2A activity in a dopaminergic cell
response to peroxisome proliferator-activated receptor g coactivator- line reduced chaperone-mediated autophagy and increased the
1a (PGC-1a) are underexpressed in PD, whereas PGC-1a expression half-life of a-synuclein. Altered handling of a-synuclein can also
is upregulated in broblasts from PD patients [113,137,168]. This has be induced by disruption/mutation of the lysosomal membrane
important functional consequences, as the loss of dopaminergic protein ATPase (ATP13A2), which is affected in one form of
neurons in primary culture induced by mutant a-synuclein or familial PD [154]. The potential involvement of altered lysosomal
rotenone was prevented by activation of PGC-1a and associated function in PD was supported by studies in MPTP-treated mice
with increased expression of nuclear-encoded subunits of the mito- [18,19]. Toxin treatment caused a depletion of lysosomes in
chondrial respiratory chain. dopaminergic cells secondary to the onset of oxidative stress that
led to the accumulation of autophagosomes and cellular degen-
Altered proteolysis in PDproteasomal and lysosomal eration. The toxicity of MPTP was attenuated by pharmacological
and genetic treatments that enhanced lysosomal formation.
The presence of multiple proteins in Lewy bodies, most notably Alterations in autophagy and mitophagy also occur in response
a-synuclein, led to the idea that the catabolism of unwanted, to nigral neuronal degeneration induced by 6-OHDA [87,139].
damaged, or mutated proteins might be disrupted in PD, leading Another strand supporting lysosomal involvement in neuronal
to cellular aggregation and neuronal death [31,136,140]. This led loss in PD comes from Gaucher disease, a lysosomal storage
to investigation of the roles of the ubiquitinproteasome system disorder due to mutations in the GBA gene. This leads to a
(UPS) and lysosomes in pathogenesis in PD. UPS involvement was deciency of the lysosomal enzyme GCase, which catalyzes the
138 D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144

metabolism of the sphingolipid glucosylceramide to ceramide. consensus seems to be that a mild to moderate peripheral
Gaucher mutations lead to a 20- to 30-fold increase in the risk of inammation can exacerbate the loss of dopaminergic neurons
developing PD and 510% of patients with PD have a GBA initiated by another toxic insult.
mutation (see for example [156]). Recently, a GCase deciency All of these data suggest that one way of attempting to reduce
was reported in the substantia nigra of patients with PD and GBA cell loss in PD may be by reducing inammatory change, and
mutations but, importantly, also in those with sporadic PD [30]. certainly this seems to work in experimental systems (but see
Because not all individuals with GBA mutations develop PD, the later). This may be relevant to both the initiation and the
probability is that these lead to increased susceptibility to other progression of neuronal destruction irrespective of whether this
factors involved in the pathogenic process, such as a-synuclein starts at the level of neurons or glia. The importance of the glial
accumulation and oxidative stress occurring as a result of activation in PD might lie in its long duration. In both humans and
altered mitochondrial function. Indeed, GCase deciency leads to nonhuman primates, activated microglia can be detected years
a-synuclein accumulation both in vitro and in vivo [89,164]. after exposure to MPTP, suggesting a propagating role in ongoing
pathogenic change [93]. Conversely, this long-term activation
Inammatory change may prevent microglia from performing their normal support
role for neuronal viability, for example, through the release of
The concept of inammatory change in the brain in PD started trophic factors. Although the emphasis on the role of glia has
with the description of activated HLA-positive microglia in SNc [92]. centered on activated microglia, it should be borne in mind that
Subsequently, alterations in the cytokines interleukin-1a (IL-1a), astrocytosis also occurs in PD and it too may play a signicant
IL-1b, and tumor necrosis factor-a (TNF-a) were found in the brain role in the sequence of events that lead to cell death.
and cerebrospinal uid, and postmortem studies showed inducible
nitric oxide (NO) synthase to be present in activated microglia [45]. Excitotoxic mechanisms
Inducible NO synthase is a source of NO, which in turn can react with
superoxide from glial or neuronal sources to form the highly reactive Excitotoxicity is always included in the list of pathogenic mechan-
peroxynitrite. This can nitrate proteins and other biomolecules, and 3- isms that are thought to contribute to cell death in PD. However, the
nitrotyrosine adducts are found in the SNc in PD. In addition, there is degree of direct evidence for this is small. The major contributor is the
evidence for the presence of nitrated a-synuclein in Lewy bodies [31]. overactivity of the subthalamic nucleus (STN), which releases gluta-
Microglia activation and inammatory change were thought to be a mate and which innervates the substantia nigra pars compacta and
consequence of neuronal destruction but there is evidence for a more the internal segment of the globus pallidus [8,142]. In addition, there
general systemic inammatory reaction in PD, suggesting it to be a is evidence of altered glutamatergic input from the corticostriatal
primary cause of neuronal loss in some cases. In addition, peripheral pathway. Excitotoxicity might also arise from the dysfunction of
inammation may enhance the adverse effects of inammatory mitochondria that occurs in PD. However, experimental studies show
change occurring in the substantia nigra [43]. This point was brought that excitotoxins can cause destruction of nigral dopaminergic
into focus by recent GWAS that showed HLA to be a risk factor for the neurons and that this process can be blocked by various classes of
occurrence of PD [40]. glutamate antagonists [48,67,142,153,157,158]. Similarly, cell loss
Additionally, there seems to be some cross-talk between the induced by toxins acting through other mechanisms, such as 6-OHDA,
central nervous system innate inammatory response and the is diminished by drugs that either modulate glutamate release or
peripheral immune system. Microarray gene expression studies diminish its action on target cells through actions on either inotropic
have identied modied neuroimmune signaling in peripheral blood or metabotropic glutamate receptors. The scenario would be that as
mononucleated cells from PD patients, and changes in neuroin- PD develops with the onset of neuronal loss in the SNc, the increased
ammatory markers in PD have more recently been detected by activity of the glutamatergic input from the STN acts to amplify cell
studies demonstrating increased concentrations of IL-2 [145], TNF-a, death. Indeed, lesions of the STN have been shown to reduce the
IL-6 [22], osteopontin, and RANTES/chemokine (CC motif) ligand 5 loss of dopaminergic neurons normally induced by the injection of
[125] in serum in PD. In particular, serum levels of RANTES, a 6-OHDA into the SNc [12,99,135]. Overactivity of the glutamatergic
chemokine produced by activated microglia, were not only higher in output pathways from the STN to the globus pallidus also contributes
PD compared to normal subjects but signicantly correlated with to the evolution of some of the clinical symptoms of PD. Indeed, deep
Unied Parkinsons Disease Rating Scale scores [125]. Such ndings brain stimulation (DBS) can correct the overactivity of the STN,
have supported the concept that a selective blood biomarker could producing a clinical benet in PD. Perhaps importantly, recent
be developed and used to rene the clinical diagnosis of PD. investigations utilizing DBS in PD have associated it with a lack of
Multiple experimental studies have been utilized to investi- progression of motor symptoms [147]. Assuming that DBS would also
gate the role of inammatory change in PD in greater detail. decrease the overactivity of the STN pathways innervating the SNc,
Initially toxins that directly destroy dopaminergic neurons, such this does add some support to the concept that excitotoxicity plays a
as 6-OHDA, MPTP, and rotenone, were shown to activate micro- role in cell death in PD.
glia and induce inammatory change. But it has been the use of
lipopolysaccharide (LPS) in both in vitro cellular models and
in vivo studies that focused on glial activation [44,56,57,95,96]. Bringing it all together?
These studies have shown that dopaminergic neuronal loss can
occur as a direct consequence of microglial activation that is In the past 20 years, there have been signicant advances in the
accompanied by increased cytokine formation, increased produc- understanding from a mechanistic perspective of the events that lead
tion of reactive oxygen and nitrogen species, and decreased to the death of nigral dopaminergic neurons in PD. It is worth
secretion of trophic factors responsible for the normal mainte- remembering, however, that the pathology of PD is widespread and
nance of neuronal viability. A potentially important nding is that that the same rigor has not been applied to looking at the underlying
damage after toxin application to SNc is accompanied by the causes of cell death in nondopaminergic nuclei, such as the locus
presence of monocytes, suggesting bloodbrain barrier damage coeruleus and raphe nuclei. Indeed, there has been a presumption
and permeability. Indeed, altered gene and protein expression for that because these neuronal groups also degenerate with the appear-
the adhesion molecule intercellular adhesion molecule-1 occurs ance of Lewy bodies, the same cell death sequences are operative,
in PD and in experimental models of the illness [101]. The current but in reality, this is not known. Cartoons of the sequence of events
D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144 139

that underlie nigral cell loss have been published regularly (and to can be controlled, although the time course of such studies is
which we have added in this review) but these have not changed seldom more than a few days. In this manner, it is possible to
much over the years in terms of the overall concepts espoused, as study the role of specic pathogenic pathways in the induction
there has been a trend to make new information on pathogenic and progression of cell death, although the environment is
processes in PD t into the existing framework of conceptual belief. entirely articial and the cells may not be representative of the
This may or may not be correct. susceptibility of dopaminergic neurons in the aging brain in
Certainly the events that are commonly related to pathogen- humans that are affected in PD. For example, cell lines may not
esis in PD t nicely togetherfor example, mitochondrial dys- have a full dopaminergic phenotype, they may not be derived
function with oxidative stress and altered proteolysis. The from a neuronal source, and they will not be accompanied by glial
evidence from gene mutations identied in inherited PD has cells. Primary cultures are derived from fetal sources, they are
added to this accepted interaction and has again highlighted immature and may be from species in which susceptibility to
mitochondrial abnormalities and altered protein handling as key toxin action is different from that in humans. Even the cell culture
events. The general acceptance of PD as a synucleinopathy has medium may not be reective of the normal environment for cells
added to the weight of the arguments for a common thread in that it may be high in iron and low in antioxidants and so
underlying nigral cell loss originating in both sporadic and provide a highly pro-oxidant environment without the natural
inherited disease [33,35]. There is also a degree of comfort antioxidant defense mechanisms being operative [16]. Never-
associated with containing novel information within an accepted theless, as a primary means of assessing cell death processes,
arena. But is this the right thing to do? these are highly useful techniques and capable of being manipu-
A skeptical view would be that it is not surprising that all the lated, for example, by gene transfection, to study both sporadic
major organelles of a cell are somehow involved in the demise of and familial PD.
the neuron. This is emphasized by a range of studies in which cell The use of toxins in in vivo models of PD is also a highly useful
death is initiated at a point in the accepted cycle of pathogenic way of investigating dopaminergic cell death and drug action, but
events in PD. These illustrate very nicely that it is difcult to has limitations that have proved difcult to overcome (see
distinguish what is horse and what is cart. For example, if cell [9,24,91] for recent reviews; see Table 3). Most toxins require
death is initiated by inhibiting complex I, then this entrains direct intracerebral injection into the SNc, medial forebrain
changes in oxidative and nitrative stress and proteasomal activity. bundle, or striatum. Most need to be injected in concentrations
Inhibiting proteasomal activity leads to altered mitochondrial that rapidly kill the majority of dopaminergic neurons to allow
function and increased oxidative and nitrative stress and so on, motor decits to be observed. It is much more difcult to control
with the same applying to the introduction of mutant parkin or the toxin concentration, the degree of neuronal exposure, and the
a-synuclein into cell lines [5052,79,80]. time course of toxin effect than when using in vitro culture
A more difcult conclusionand one that would decrease an systems. Certainly, specic pathogenic mechanisms, for example,
authors chances of publicationwould be that many of the key oxidative stress and complex I inhibition and inammatory
events that are identied as being involved in cell death in PD and change, can be modeled using 6-OHDA, MPP , and LPS, respec-
that result from gene mutations represent separate pathogenic tively, but toxin exposure is usually acute, in young animals, and
pathways that dene different forms of what is currently deemed in a species that differs in susceptibility to humans. The biggest
to be PD. Indeed, the current view is that PD is not a single disease drawback is that most approaches are one off and do not in any
but rather a syndrome of multiple causes and manifestations. way reect the progressive nature of cell loss in PD. One or two
A simple example is the general absence of Lewy bodies in the exceptions are the injection of 6-OHDA in to the striatum, which
SNc in cases with parkin mutations [42]. Another example from causes retrograde degeneration of dopaminergic neurons, and the
sporadic PD is that only 30% of individuals have a complex I defect use of infusion pumps to provide longer periods of toxin expo-
that is outside of the normal range. Our own limited investiga- sure. But even here, we are talking about days rather than weeks
tions failed to show any correlation in postmortem nigral tissue or months or years. In addition, most pathogenesis-based toxin
between iron accumulation, the extent of oxidative stress, and studies are focused entirely on the SNc and there have been only
mitochondrial dysfunction [65]. limited attempts at destroying other neuronal systems affected in
Perhaps in what is termed sporadic PD, we are missing key PD, which do not mimic one of the key features of PD, that being
pieces of the jigsaw and what we are observing are secondary altered protein deposition.
changes that are the result of one or more primary events that There are, however, exceptions to most of the above criticisms.
remain unknown. It becomes increasingly possible that patho- The peripheral administration of 6-OHDA to neonatal rats
genic events in SNc do not originate in the brain but are the result destroys dopaminergic and noradrenergic neurons as it passes
of some peripheral event that is then transmitted into brain, the incomplete bloodbrain barrier. MPTP can be used systemi-
initiating cell death that propagates itself in a stoichiometric cally in both mice and monkeys to destroy dopaminergic neurons
fashion through synucleinopathy to eventually reach the SNc and in the SNc although, again, the time course of these studies is
that explains the long prodromal period and the occurrence of short and the effects specic to dopaminergic systems. Peripheral
nonmotor symptoms that are now an accepted part of what used administration of LPS has been reported to induce nigral
to be considered a movement disorder (see earlier).

Table 3
Translation into animal models of PD Toxins used in animal models of PD that reect the pathogenic process.

Pathology modeled Toxin


Toxin relevance
Oxidative stress 6-OHDA and paraquat
Numerous toxins are in use in experimental models of PD Nitrative stress LPS
largely based on their ability to destroy dopaminergic neurons Mitochondrial dysfunction Rotenone, MPTP, and MPP
through a variety of relevant pathogenic mechanisms. In cell Excitotoxicity Quinolinic acid, ibotinic acid
Proteasomal dysfunction PSI, epoximycin, lactacystin
culture and in primary neuronal cultures, the concentration of Glial cell activation LPS
toxin to which cells are exposed and the duration of that exposure
140 D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144

dopaminergic cell loss but not consistently, although it may susceptibility to subsequent toxic insult may be enhanced and it
increase susceptibility to the central actions of other toxins. may be that combinations of genetic models and toxin-based
Systemic administration of rotenone provides an interesting approaches will yield important information on pathogenesis
possibility despite its inherent toxicity [7]. There seems to be related to nigral cell death. However, one recent study suggests
agreement that with careful administration, rotenone kills nigral that models more closely related to PD can be generated under
dopaminergic cells as well as affecting neurons in other nuclei appropriate conditions. A subset of DJ-1 null mice backcrossed
and perhaps with sequential pathology [54,116]. The biggest into a normal background was reported to show early onset of
question is whether these are relevant to PD or more reective unilateral nigral dopaminergic cell loss that became bilateral with
of the widespread pathology occurring in multiple-system atro- time and later involved the locus coeruleus and the onset of
phy, another Parkinson-like disorder [46]. Most intriguing is the motor decits [127].
use of proteasomal inhibitors such as epoximycin and PSI. There is It should also be remembered that genetic changes not directly
no doubt that these are toxic to dopaminergic neurons in vitro linked to PD have yielded important animal models in which
and on stereotaxic injection into the SNc (see above). But reports nigral dopaminergic cell death does occur and which again add to
of the slow progressive onset of neuronal loss in a pattern that the knowledge of pathogenic mechanisms. These include the
resembles that occurring in PD have proved difcult to reproduce aphakia mouse lacking the homeobox transcription factor Pitx3,
in some laboratories but not others. This would be a marked the mitopark mouse lacking mitochondrial transcription factor A,
advance in toxin-based models of PD if viable, as cell death and Nurr-1 decient mice [29,49,66].
progresses through the brain, as suggested by Braak and is
accompanied by a-synuclein aggregate formation. In fact, recent
data suggest that PSI does not enter the brain but rather such From pathogenesis to neuroprotection?
proteasomal inhibitors can induce Lewy body-like pathology in
the gut and other organs (unpublished observations) and this Based on the extent of knowledge of pathogenic mechanisms
then invades the brain through the dorsal motor nucleus of the leading to dopaminergic cell death in PD, it would seem likely
vagus [102]. If this is a reality, it would explain much that has that treatment strategies aimed at stopping or slowing the
consumed the debate over the progression of PD in recent years. progression of the disease process would be available but this is
not the case. Neuroprotection has proved a more difcult nut to
Gene relevance crack than most had envisaged and the current situation is
starting to look like that in stroke. Based on experimental studies,
Understanding the function of wild-type and mutated proteins a range of compounds have been advanced into clinical develop-
associated with familial PD has been possible through their ment, but so far, none have proved to have a convincing effect
transfection into cell lines and this has led to advances in on the worsening of motor symptoms [107,110,111,134,144].
discerning their role in the pathogenic process through altera- This has involved antioxidants, dopamine agonists, monoamine
tions in oxidative stress, mitochondrial function, and altered oxidase inhibitors, agents affecting mitochondrial function, cal-
proteasomal and lysosomal activity. But it has been the transla- cium antagonists, antiapoptotic agents, and neurotrophic factors,
tion into in vivo experimental models that has proved more among others. So far, only the monoamine oxidase B inhibitor
daunting. Despite promising ndings in Drosophila and Caenor- rasagiline has been shown to have some effect on motor scores,
habditis elegans, there are no transgenic, overexpression, knock- but this was not sufcient for it to be labeled as disease-
out, or knock-in models of wild-type or mutant a-synuclein that modifying [109]. Although the pace has lessened, there are a
have fully recapitulated the pathological picture of PD (see number of ongoing studies looking to see whether centrally active
[9,83,166] for recent reviews). Viral vector-induced expression iron chelators, precursors of urate, or stimulators of glial cell-
of wild-type and mutant a-synuclein in rats and primates has derived neurotrophic factor production in brain are able to do
been associated with nigral dopaminergic cell degeneration but what has eluded previous attempts. But perhaps the worrying
this has not been a consistent nding. The picture has been issue is whether the current concepts of the pathogenic processes
similar when the same approaches have been attempted using in PD and the nature of PD itself are correct.
parkin, DJ-1, PINK1, and LRRK2. Changes in dopaminergic func- We base the animal models of PD on the processes that we
tion in striatum, altered morphology of dopaminergic neurons, believe to be operative in cell death in humans. Classically, there
changes in other monoaminergic systems, degeneration of motor is great reliance on using MPTP-treated mice and the 6-OHDA-
neurons, gliosis, and inammatory changes occur, among others, lesioned rats to model oxidative stress and mitochondrial dys-
along with mild motor abnormalities, but not the expression of function, respectively. But these may have deceived us as so far
PD that was hoped for. However, there is still value in this there has been no translation into clinical effect. The surprising
approach, as the generation of these animals allows an in vivo thing is that we continue to use these models to nd further
approach to explaining the roles of mutated proteins in the potential neuroprotective strategies despite their history, perhaps
pathogenic process dened using other techniques. It also pro- because there is nowhere else to turn in terms of toxin-related
vides a test bed in which to examine potential neuroprotective effect. Interestingly, there has been relatively little work in
therapies, for example, kinase/GTPase inhibitors in LRRK2 trans- rotenone- or PSI-treated rodents, which may have more to offer.
genic animals [77,82] and the role of passive vaccination in What is particularly worrying is the large numbers of compounds
a-synuclein transgenic mice [155]. There also seems to be a reported as positive in MPTP-treated miceif it were that easy,
signicant role for studying nonmotor symptoms of PD in genetic we would have neuroprotection in PD by now. There is clearly a
models of PD, with reports of everything from cognitive impair- need to reassess the models employed and to ensure that we are
ment to altered gastrointestinal motility and constipation working from a rm knowledge base from postmortem studies
[91,160]. It could be questioned whether mice provide the right and the living PD patient with respect to the events that occur
background to test the pathogenic potential of mutated proteins during neuronal loss and their time course. The last may be very
in PD, as total knockout of single wild-type proteins does important as it may be too late to expect a neuroprotective
not seem to result in alterations in dopaminergic function in treatment to have much effect by the time a diagnosis of PD is
the brain of mice that are null for parkin, DJ-1, and PINK1; made on classical grounds, as neuronal loss will be too advanced.
nothing untoward seems to occur as they age [71]. Increased Perhaps ongoing studies to identify premotor signs of PD will
D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144 141

identify an earlier patient population in which to study neuro- several fundamental gaps in our knowledge, which hamper our
protection and maybe the animal models will turn out to be ability to develop effective neuroprotective strategies for PD.
correct in their prediction of effect. Importantly, we still do not understand the molecular mechan-
The gene defects associated with familial PD may lead to new isms that account for the spreading pathology of the disease.
potential targets for neuroprotection and, at present, mitochon- Additionally, the failure to generate an accurate model of sporadic
drial dysfunction and lysosomal defects seem attractive. The Parkinson disease and the clinical failure of neuroprotective
problem has been, and to a large extent remains, that most strategies developed from such models highlight that we are
aberrant proteins identied do not offer an obvious target for missing a key section of the cell death mechanism jigsaw. Indeed,
therapeutic intervention and, in many cases, their physiological the mechanisms currently used to explain pathogenesis in PD
function remains unknown. This applies to a-synuclein, parkin, may just be the downstream consequence of a so far unknown
DJ-1, and PINK1, among others. LRRK2 mutations are linked to trigger. Certainly, it is clear that PD is not a single disease with a
altered kinase/GTPase activity and this could be an attractive single cause but a syndrome in which the variants have in
target, but to nd a centrally active kinase inhibitor that focuses common nigral dopaminergic cell death and motor dysfunction,
on the areas of the brain affected in PD, that does not impair the but around which multiple other neuronal systems degenerate
physiological function of the protein, and that does not affect and which have multiple primary causes involving a range of
other kinases, is challenging. Indeed, the pharmaceutical industry pathogenic processes.
may nd it difcult to identify viable targets at the current time
on which to unleash the medicinal chemists and preclinical
teams. The other danger is that the defects in familial disease References
have nothing to do with the processes that underlie neuronal loss
in the majority of PD patients with sporadic disease. The means of [1] Agid, Y. Levodopa: is toxicity a myth? Neurology 50:858863; 1998.
assessing molecules that might potentially rectify the effects of [2] Alvarez-Erviti, L.; Rodriguez-Oroz, M. C.; Cooper, J. M.; Caballero, C.;
Ferrer, I.; Obeso, J. A.; Schapira, A. H. Chaperone-mediated autophagy
gene defects also needs assessment. For example, it is perhaps not markers in Parkinson disease brains. Arch. Neurol. 67:14641472; 2010.
too surprising that kinase inhibitors prevent the neuronal loss in [3] Ambani, L. M.; Van Woert, M. H.; Murphy, S. Brain peroxidase and catalase
mice induced by the introduction of a LRRK2 mutation that in Parkinson disease. Arch. Neurol. 32:114118; 1975.
[4] Ascherio, A.; LeWitt, P. A.; Xu, K.; Eberly, S.; Watts, A.; Matson, W. R.;
increases kinase activity. The acceptance that PD is a synucleino-
Marras, C.; Kieburtz, K.; Rudolph, A.; Bogdanov, M. B.; Schwid, S. R.;
pathy suggests that removal of the protein could slow disease Tennis, M.; Tanner, C. M.; Beal, M. F.; Lang, A. E.; Oakes, D.; Fahn, S.;
progression whatever the initial cause. But what type of Shoulson, I.; Schwarzschild, M. A. Urate as a predictor of the rate of clinical
decline in Parkinson disease. Arch. Neurol. 66:14601468; 2009.
a-synuclein should be removed and how much of it? Whether this [5] Ascherio, A.; Tanner, C. M. Use of antihypertensives and the risk of
should be aggregates, brils, or protobrils or inhibition of the Parkinson disease. Neurology 72:578579; 2009.
formation of the protein, all of it, some of it, or none of it remains [6] Berg, D.; Marek, K.; Ross, G. W.; Poewe, W. Dening at-risk populations for
to be determined. Certainly approaches such as passive vaccina- Parkinsons disease: lessons from ongoing studies. Mov. Disord 27:656665;
2012.
tion can remove a-synuclein and prevent the consequences of its [7] Betarbet, R.; Sherer, T. B.; MacKenzie, G.; Garcia-Osuna, M.; Panov, A. V.;
aggregation in a-synuclein overexpressing mice, but will this Greenamyre, J. T. Chronic systemic pesticide exposure reproduces features
work in PD? The worry is that several similar approaches using of Parkinsons disease. Nat. Neurosci. 3:13011306; 2000.
[8] Bevan, M. D.; Magill, P. J.; Terman, D.; Bolam, J. P.; Wilson, C. J. Move to the
human monoclonal antibodies against amyloid have recently rhythm: oscillations in the subthalamic nucleusexternal globus pallidus
failed in Alzheimer disease despite encouraging preclinical data. network. Trends Neurosci. 25:525531; 2002.
Finally, it may be the nature of PD that has so far prevented the [9] Blesa, J.; Phani, S.; Jackson-Lewis, V.; Przedborski, S. Classic and new animal
models of Parkinsons disease. J. Biomed. Biotechnol. 2012:845618; 2012.
translation of pathogenic mechanisms into a neuroprotective [10] Braak, H.; de Vos, R. A.; Bohl, J.; Del, T. K. Gastric alpha-synuclein
treatment. Clinical trials are still undertaken in the classical immunoreactive inclusions in Meissners and Auerbachs plexuses in cases
manner of comparing the effect of an agent between large groups staged for Parkinsons disease-related brain pathology. Neurosci. Lett
396:6772; 2006.
of patients in a placebo arm and an active treatment arm, looking [11] Braak, H.; Del, T. K.; Rub, U.; de Vos, R. A.; Jansen Steur, E. N.; Braak, E.
for a statistically relevant outcome. But this presumes that PD is a Staging of brain pathology related to sporadic Parkinsons disease. Neurobiol.
single illness resulting from the same pathogenic processes and Aging 24:197211; 2003.
[12] Bukhatwa, S.; Zeng, B. Y.; Rose, S.; Jenner, P. The effects of dose and route of
this is increasingly being challenged. Rather there should be
administration of PSI on behavioural and biochemical indices of neuronal
attempts to look at drug effect in individuals with common degeneration in the rat brain. Brain Res 1354:236242; 2010.
features, for example, a mitochondrial complex I defect. At [13] Chaudhuri, K. R.; Schapira, A. H. Non-motor symptoms of Parkinsons
present signicant effects of a specic drug targeted at one disease: dopaminergic pathophysiology and treatment. Lancet Neurol.
8:464474; 2009.
component of the cell death pathway may be missed in a small [14] Chen, J. F.; Xu, K.; Petzer, J. P.; Staal, R.; Xu, Y. H.; Beilstein, M.; Sonsalla, P. K.;
proportion of patients in a clinical trial because of dilution of the Castagnoli, K.; Castagnoli Jr N.; Schwarzschild, M. A. Neuroprotection by
outcome by the majority who have PD associated with other caffeine and A(2A) adenosine receptor inactivation in a model of Parkinsons
disease. J. Neurosci. 21:RC143; 2001.
pathogenic mechanisms and who did not respond. Indeed, it may [15] Cipriani, S.; Chen, X.; Schwarzschild, M. A. Urate: a novel biomarker of
be that we have to think about neuroprotection for small portions Parkinsons disease risk, diagnosis and prognosis. Biomark. Med. 4:701712;
of the PD population who have the disease for specic reasons 2010.
[16] Clement, M. V.; Long, L. H.; Ramalingam, J.; Halliwell, B. The cytotoxicity of
and, notably, this may be those with inherited disease, for dopamine may be an artefact of cell culture. J. Neurochem. 81:414421;
example, a LRRK2 mutation, as a druggable target. The downside 2002.
is that such an approach may not be economically viable for the [17] Dawson, T. M. Unraveling the role of defective genes in Parkinsons disease.
Parkinsonism Relat. Disord 13(Suppl. 3):S248S249; 2007.
pharmaceutical industry. The alternative is to adopt the oncology [18] Dehay, B.; Bove, J.; Rodriguez-Muela, N.; Perier, C.; Recasens, A.; Boya, P.;
approach and to use cocktails of drugs that exert multiple effects Vila, M. Pathogenic lysosomal depletion in Parkinsons disease. J. Neurosci.
on the pathogenic cascade. 30:1253512544; 2010.
[19] Dehay, B.; Ramirez, A.; Martinez-Vicente, M.; Perier, C.; Canron, M. H.;
Doudnikoff, E.; Vital, A.; Vila, M.; Klein, C.; Bezard, E. Loss of P-type ATPase
ATP13A2/PARK9 function induces general lysosomal deciency and leads to
Concluding remarks Parkinson disease neurodegeneration. Proc. Natl. Acad. Sci. USA
109:96119616; 2012.
[20] Del, T. K.; Rub, U.; de Vos, R. A.; Bohl, J. R.; Braak, H. Where does Parkinson
Clearly we have made great strides in improving our under- disease pathology begin in the brain? J. Neuropathol. Exp. Neurol.
standing of the nature and causes of PD. However, there are 61:413426; 2002.
142 D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144

[21] Dickson, D. W.; Braak, H.; Duda, J. E.; Duyckaerts, C.; Gasser, T.; Halliday, G. M.; [47] Houlden, H.; Singleton, A. B. The genetics and neuropathology of Parkin-
Hardy, J.; Leverenz, J. B.; Del, T. K.; Wszolek, Z. K.; Litvan, I. Neuropathological sons disease. Acta Neuropathol. 124:325338; 2012.
assessment of Parkinsons disease: rening the diagnostic criteria. Lancet [48] Hovelso, N.; Sotty, F.; Montezinho, L. P.; Pinheiro, P. S.; Herrik, K. F.; Mork, A.
Neurol. 8:11501157; 2009. Therapeutic potential of metabotropic glutamate receptor modulators. Curr.
[22] Dobbs, R. J.; Charlett, A.; Purkiss, A. G.; Dobbs, S. M.; Weller, C.; Peterson, D. W. Neuropharmacol. 10:1248; 2012.
Association of circulating TNF-alpha and IL-6 with ageing and parkinsonism. [49] Hwang, D. Y.; Ardayo, P.; Kang, U. J.; Semina, E. V.; Kim, K. S. Selective loss
Acta Neurol. Scand. 100:3441; 1999. of dopaminergic neurons in the substantia nigra of Pitx3-decient aphakia
[23] Dunning, C. J.; Reyes, J. F.; Steiner, J. A.; Brundin, P. Can Parkinsons disease mice. Brain Res. Mol. Brain Res. 114:123131; 2003.
pathology be propagated from one neuron to another? Prog. Neurobiol. [50] Hyun, D. H.; Lee, M.; Halliwell, B.; Jenner, P. Proteasomal inhibition causes
97:205219; 2012. the formation of protein aggregates containing a wide range of proteins,
[24] Duty, S.; Jenner, P. Animal models of Parkinsons disease: a source of including nitrated proteins. J. Neurochem. 86:363373; 2003.
novel treatments and clues to the cause of the disease. Br. J. Pharmacol [51] Hyun, D. H.; Lee, M.; Halliwell, B.; Jenner, P. Effect of overexpression of
164:13571391; 2011. wild-type or mutant parkin on the cellular response induced by toxic
[25] Elstner, M.; Morris, C. M.; Heim, K.; Bender, A.; Mehta, D.; Jaros, E.; insults. J. Neurosci. Res. 82:232244; 2005.
Klopstock, T.; Meitinger, T.; Turnbull, D. M.; Prokisch, H. Expression analysis [52] Hyun, D. H.; Lee, M.; Hattori, N.; Kubo, S.; Mizuno, Y.; Halliwell, B.; Jenner, P.
of dopaminergic neurons in Parkinsons disease and aging links transcrip- Effect of wild-type or mutant Parkin on oxidative damage, nitric oxide,
tional dysregulation of energy metabolism to cell death. Acta Neuropathol. antioxidant defenses, and the proteasome. J. Biol. Chem. 277:2857228577;
122:7586; 2011. 2002.
[26] Fahn, S. Does levodopa slow or hasten the rate of progression of Parkinsons [53] Ikeda, H.; Markey, C. J.; Markey, S. P. Search for neurotoxins structurally
disease? J. Neurol. 252(Suppl. 4):IV37IV42; 2005. related to 1-methyl-4-phenylpyridine (MPP ) in the pathogenesis of
[27] Findley, L. J. The economic impact of Parkinsons disease. Parkinsonism Relat. Parkinsons disease. Brain Res 575:285298; 1992.
Disord. 13(Suppl):S8S12; 2007. [54] Inden, M.; Kitamura, Y.; Abe, M.; Tamaki, A.; Takata, K.; Taniguchi, T.
[28] Fujishiro, H.; Frigerio, R.; Burnett, M.; Klos, K. J.; Josephs, K. A.; DelleDonne, A.; Parkinsonian rotenone mouse model: reevaluation of long-term adminis-
Parisi, J. E.; Ahlskog, J. E.; Dickson, D. W. Cardiac sympathetic denervation tration of rotenone in C57BL/6 mice. Biol. Pharm. Bull. 34:9296; 2011.
correlates with clinical and pathologic stages of Parkinsons disease. Mov. [55] International Parkinsons Disease Genomic Consortium. A two-stage meta-
Disord 23:10851092; 2008. analysis identies several new loci for Parkinsons disease. PLoS Genet.
[29] Galter, D.; Pernold, K.; Yoshitake, T.; Lindqvist, E.; Hoffer, B.; Kehr, J.; 7:e1002142; 2011.
Larsson, N. G.; Olson, L. MitoPark mice mirror the slow progression of key [56] Iravani, M. M.; Leung, C. C.; Sadeghian, M.; Haddon, C. O.; Rose, S.; Jenner, P.
symptoms and L-DOPA response in Parkinsons disease. Genes Brain Behav. The acute and the long-term effects of nigral lipopolysaccharide adminis-
9:173181; 2010. tration on dopaminergic dysfunction and glial cell activation. Eur. J.
[30] Gegg, M. E.; Burke, D.; Heales, S. J.; Cooper, J. M.; Hardy, J.; Wood, N. W.; Neurosci. 22:317330; 2005.
Schapira, A. H. Glucocerebrosidase deciency in substantia nigra of Parkin- [57] Iravani, M. M.; Sadeghian, M.; Leung, C. C.; Jenner, P.; Rose, S.
son disease brains. Ann. Neurol. 72:455463; 2012. Lipopolysaccharide-induced nigral inammation leads to increased IL-
[31] Giasson, B. I.; Duda, J. E.; Murray, I. V.; Chen, Q.; Souza, J. M.; Hurtig, H. I.; 1beta tissue content and expression of astrocytic glial cell line-derived
Ischiropoulos, H.; Trojanowski, J. Q.; Lee, V. M. Oxidative damage linked to neurotrophic factor. Neurosci. Lett. 510:138142; 2012.
neurodegeneration by selective alpha-synuclein nitration in synucleinopathy [58] Jankovic, J. Parkinsons disease: clinical features and diagnosis. J. Neurol.
lesions. Science 290:985989; 2000. Neurosurg. Psychiatry 79:368376; 2008.
[32] Glinka, Y.; Gassen, M.; Youdim, M. B. Mechanism of 6-hydroxydopamine [59] Jellinger, K. A. Neuropathology of sporadic Parkinsons disease: evaluation
neurotoxicity. J. Neural Transm. Suppl. 50:5566; 1997. and changes of concepts. Mov. Disord 27:830; 2012.
[33] Goedert, M. Parkinsons disease and other alpha-synucleinopathies. Clin. [60] Jenner, P. Preclinical evidence for neuroprotection with monoamine
Chem. Lab. Med. 39:308312; 2001. oxidase-B inhibitors in Parkinsons disease. Neurology 63:S13S22; 2004.
[34] Goedert, M.; Clavaguera, F.; Tolnay, M. The propagation of prion-like [61] Jenner, P. Oxidative stress in Parkinsons disease. Ann. Neurol. 53(Suppl. 3):
protein inclusions in neurodegenerative diseases. Trends Neurosci. S26S36; 2003.
33:317325; 2010. [62] Jenner, P.; Langston, J. W. Explaining ADAGIO: a critical review of the
[35] Goedert, M.; Spillantini, M. G. Lewy body diseases and multiple system biological basis for the clinical effects of rasagiline. Mov. Disord
atrophy as alpha-synucleinopathies. Mol. Psychiatry 3:462465; 1998. 26:23162323; 2011.
[36] Graham, D. G. Oxidative pathways for catecholamines in the genesis of [63] Jenner, P.; Olanow, C. W. The pathogenesis of cell death in Parkinsons
neuromelanin and cytotoxic quinones. Mol. Pharmacol 14:633643; 1978. disease. Neurology 66:S24S36; 2006.
[37] Grunblatt, E.; Mandel, S.; Jacob-Hirsch, J.; Zeligson, S.; Amariglo, N.; [64] Jenner, P.; Olanow, C. W. Understanding cell death in Parkinsons disease.
Rechavi, G.; Li, J.; Ravid, R.; Roggendorf, W.; Riederer, P.; Youdim, M. B. Ann. Neurol. 44:S72S84; 1998.
Gene expression proling of parkinsonian substantia nigra pars compacta: [65] Jenner, P.; Schapira, A. H.; Marsden, C. D. New insights into the cause of
alterations in ubiquitinproteasome, heat shock protein, iron and oxidative Parkinsons disease. Neurology 42:22412250; 1992.
stress regulated proteins, cell adhesion/cellular matrix and vesicle trafck- [66] Jiang, C.; Wan, X.; He, Y.; Pan, T.; Jankovic, J.; Le, W. Age-dependent
ing genes. J. Neural Transm. 111:15431573; 2004. dopaminergic dysfunction in Nurr1 knockout mice. Exp. Neurol.
[38] Gu, M.; Cooper, J. M.; Taanman, J. W.; Schapira, A. H.; Mitochondrial, DNA 191:154162; 2005.
transmission of the mitochondrial defect in Parkinsons disease. Ann. Neurol. [67] Johnson, K. A.; Conn, P. J.; Niswender, C. M. Glutamate receptors as
44:177186; 1998. therapeutic targets for Parkinsons disease. CNS Neurol. Disord. Drug Targets
[39] Gupta, A.; Dawson, V. L.; Dawson, T. M. What causes cell death in 8:475491; 2009.
Parkinsons disease? Ann. Neurol. 64(Suppl. 2):S315; 2008. [68] Kalaitzakis, M. E.; Graeber, M. B.; Gentleman, S. M.; Pearce, R. K. The dorsal
[40] Hamza, T. H.; Zabetian, C. P.; Tenesa, A.; Laederach, A.; Montimurro, J.; motor nucleus of the vagus is not an obligatory trigger site of Parkinsons
Yearout, D.; Kay, D. M.; Doheny, K. F.; Paschall, J.; Pugh, E.; Kusel, V. I.; disease: a critical analysis of alpha-synuclein staging. Neuropathol. Appl.
Collura, R.; Roberts, J.; Grifth, A.; Samii, A.; Scott, W. K.; Nutt, J.; Factor, S. A.; Neurobiol. 34:284295; 2008.
Payami, H. Common genetic variation in the HLA region is associated with [69] Kempster, P. A.; OSullivan, S. S.; Holton, J. L.; Revesz, T.; Lees, A. J.
late-onset sporadic Parkinsons disease. Nat. Genet. 42:781785; 2010. Relationships between age and late progression of Parkinsons disease:
[41] Hardy, J. Genetic analysis of pathways to Parkinson disease. Neuron a clinico-pathological study. Brain 133:17551762; 2010.
68:201206; 2010. [70] Kenborg, L.; Lassen, C. F.; Hansen, J.; Olsen, J. H. Parkinsons disease and
[42] Hattori, N.; Shimura, H.; Kubo, S.; Wang, M.; Shimizu, N.; Tanaka, K.; other neurodegenerative disorders among welders: a Danish cohort study.
Mizuno, Y. Importance of familial Parkinsons disease and parkinsonism to Mov. Disord 27:12831289; 2012.
the understanding of nigral degeneration in sporadic Parkinsons disease. J. [71] Kitada, T.; Tong, Y.; Gautier, C. A.; Shen, J. Absence of nigral degeneration in aged
Neural Transm. Suppl. 60:101116; 2000. parkin/DJ-1/PINK1 triple knockout mice. J. Neurochem. 111:696702; 2009.
[43] Hernandez-Romero, M. C.; Delgado-Cortes, M. J.; Sarmiento, M.; de Pablos; [72] Kiyohara, C.; Kusuhara, S. Cigarette smoking and Parkinsons disease:
Espinosa-Oliva, A. M.; Arguelles, S.; Bandez, M. J.; Villaran, R. F.; Maurino, R.; a meta-analysis. Fukuoka Igaku Zasshi 102:254265; 2011.
Santiago, M.; Venero, J. L.; Herrera, A. J.; Cano, J.; Machado, A. Peripheral [73] Kordower, J. H.; Kanaan, N. M.; Chu, Y.; Suresh, B. R.; Stansell III J.; Terpstra,
inammation increases the deleterious effect of CNS inammation on the B. T.; Sortwell, C. E.; Steece-Collier, K.; Collier, T. J. Failure of proteasome
nigrostriatal dopaminergic system. Neurotoxicology 33:347360; 2012. inhibitor administration to provide a model of Parkinsons disease in rats
[44] Herrera, A. J.; Castano, A.; Venero, J. L.; Cano, J.; Machado, A. The single and monkeys. Ann. Neurol. 60:264268; 2006.
intranigral injection of LPS as a new model for studying the selective effects [74] Langston, J. W. The Parkinsons complex: parkinsonism is just the tip of the
of inammatory reactions on dopaminergic system. Neurobiol. Dis. iceberg. Ann. Neurol. 59:591596; 2006.
7:429447; 2000. [75] Lannuzel, A.; Hoglinger, G. U.; Champy, P.; Michel, P. P.; Hirsch, E. C.;
[45] Hirsch, E. C.; Breidert, T.; Rousselet, E.; Hunot, S.; Hartmann, A.; Michel, P. P. Ruberg, M. Is atypical parkinsonism in the Caribbean caused by the
The role of glial reaction and inammation in Parkinsons disease. Ann. N. Y. consumption of Annonacae? J. Neural Transm. Suppl. 70:153157; 2006.
Acad. Sci. 991:214228; 2003. [76] Lannuzel, A.; Michel, P. P.; Hoglinger, G. U.; Champy, P.; Jousset, A.;
[46] Hoglinger, G. U.; Feger, J.; Prigent, A.; Michel, P. P.; Parain, K.; Champy, P.; Medja, F.; Lombes, A.; Darios, F.; Gleye, C.; Laurens, A.; Hocquemiller, R.;
Ruberg, M.; Oertel, W. H.; Hirsch, E. C. Chronic systemic complex I Hirsch, E. C.; Ruberg, M. The mitochondrial complex I inhibitor annonacin is
inhibition induces a hypokinetic multisystem degeneration in rats. J. toxic to mesencephalic dopaminergic neurons by impairment of energy
Neurochem. 84:491502; 2003. metabolism. Neuroscience 121:287296; 2003.
D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144 143

[77] Lee, B. D.; Shin, J. H.; VanKampen, J.; Petrucelli, L.; West, A. B.; Ko, H. S.; [104] Morrish, P. K. How valid is dopamine transporter imaging as a surrogate
Lee, Y. I.; Maguire-Zeiss, K. A.; Bowers, W. J.; Federoff, H. J.; Dawson, V. L.; marker in research trials in Parkinsons disease? Mov. Disord 18(Suppl. 7):
Dawson, T. M. Inhibitors of leucine-rich repeat kinase-2 protect against S63S70; 2003.
models of Parkinsons disease. Nat. Med 16:9981000; 2010. [105] Muzerengi, S.; Contrafatto, D.; Chaudhuri, K. R. Non-motor symptoms:
[78] Lee, J. H.; Goedert, M.; Hill, W. D.; Lee, V. M.; Trojanowski, J. Q. Tau proteins identication and management. Parkinsonism Relat. Disord. 13(Suppl. 3):
are abnormally expressed in olfactory epithelium of Alzheimer patients and S450S456; 2007.
developmentally regulated in human fetal spinal cord. Exp. Neurol. [106] Nalls, M. A.; Plagnol, V.; Hernandez, D. G.; Sharma, M.; Sheerin, U. M.;
121:93105; 1993. Saad, M.; Simon-Sanchez, J.; Schulte, C.; Lesage, S.; Sveinbjornsdottir, S.;
[79] Lee, M. H.; Hyun, D. H.; Jenner, P.; Halliwell, B. Effect of proteasome Stefansson, K.; Martinez, M.; Hardy, J.; Heutink, P.; Brice, A.; Gasser, T.;
inhibition on cellular oxidative damage, antioxidant defences and nitric Singleton, A. B.; Wood, N. W. Imputation of sequence variants for identica-
oxide production. J. Neurochem. 78:3241; 2001. tion of genetic risks for Parkinsons disease: a meta-analysis of genome-
[80] Lee, M. H.; Hyun, D. H.; Jenner, P.; Halliwell, B. Effect of proteasome wide association studies. Lancet 377:641649; 2011.
inhibition on cellular oxidative damage, antioxidant defences and nitric [107] Olanow, C. W. Can we achieve neuroprotection with currently available
oxide production. J. Neurochem. 78:3241; 2001. anti-parkinsonian interventions? Neurology 72:S59S64; 2009.
[81] Lehman, E. J.; Hein, M. J.; Baron, S. L.; Gersic, C. M. Neurodegenerative [108] Olanow, C. W.; Agid, Y.; Mizuno, Y.; Albanese, A.; Bonuccelli, U.; Damier, P.;
causes of death among retired National Football League players. Neurology De, Y. J.; Gershanik, O.; Guttman, M.; Grandas, F.; Hallett, M.; Hornykiewicz,
79:19701974; 2012. O.; Jenner, P.; Katzenschlager, R.; Langston, W. J.; LeWitt, P.; Melamed, E.;
[82] Liu, Z.; Hamamichi, S.; Lee, B. D.; Yang, D.; Ray, A.; Caldwell, G. A.; Mena, M. A.; Michel, P. P.; Mytilineou, C.; Obeso, J. A.; Poewe, W.; Quinn, N.;
Caldwell, K. A.; Dawson, T. M.; Smith, W. W.; Dawson, V. L. Inhibitors of Raisman-Vozari, R.; Rajput, A. H.; Rascol, O.; Sampaio, C.; Stocchi, F.
LRRK2 kinase attenuate neurodegeneration and Parkinson-like phenotypes Levodopa in the treatment of Parkinsons disease: current controversies.
in Caenorhabditis elegans and Drosophila Parkinsons disease models. Hum. Mov. Disord 19:9971005; 2004.
Mol. Genet 20:39333942; 2011. [109] Olanow, C. W.; Hauser, R. A.; Jankovic, J.; Langston, W.; Lang, A.; Poewe, W.;
[83] Magen, I.; Chesselet, M. F. Genetic mouse models of Parkinsons disease: the Tolosa, E.; Stocchi, F.; Melamed, E.; Eyal, E.; Rascol, O. A randomized,
state of the art. Prog. Brain Res. 184:5387; 2010. double-blind, placebo-controlled, delayed start study to assess rasagiline
[84] Mandel, S.; Weinreb, O.; Amit, T.; Youdim, M. B. Mechanism of neuropro- as a disease modifying therapy in Parkinsons disease (the ADAGIO study):
tective action of the anti-Parkinson drug rasagiline and its derivatives. Brain rationale, design, and baseline characteristics. Mov. Disord 23:21942201;
Res. Brain Res. Rev. 48:379387; 2005. 2008.
[85] Mann, V. M.; Cooper, J. M.; Krige, D.; Daniel, S. E.; Schapira, A. H.; Marsden, [110] Olanow, C. W.; Kieburtz, K.; Schapira, A. H. Why have we failed to
C. D. Brain, skeletal muscle and platelet homogenate mitochondrial func- achieve neuroprotection in Parkinsons disease? Ann. Neurol. 64(Suppl. 2):
tion in Parkinsons disease. Brain 115(Pt 2):333342; 1992. S101S110; 2008.
[86] Manning-Bog, A. B.; Reaney, S. H.; Chou, V. P.; Johnston, L. C.; McCormack, A. L.; [111] Olanow, C. W.; Schapira, A. H.; Agid, Y. Neuroprotection for Parkinsons
Johnston, J.; Langston, J. W.; Di Monte, D. A. Lack of nigrostriatal pathology in a disease: prospects and promises. Ann. Neurol. 53(Suppl. 3):S1S2; 2003.
rat model of proteasome inhibition. Ann. Neurol. 60:256260; 2006. [112] Orimo, S.; Amino, T.; Itoh, Y.; Takahashi, A.; Kojo, T.; Uchihara, T.; Tsuchiya, K.;
[87] Marin, C.; Aguilar, E. In vivo 6-OHDA-induced neurodegeneration and nigral Mori, F.; Wakabayashi, K.; Takahashi, H. Cardiac sympathetic denervation
autophagic markers expression. Neurochem. Int. 58:521526; 2011. precedes neuronal loss in the sympathetic ganglia in Lewy body disease. Acta
[88] Matsui, H.; Ito, H.; Taniguchi, Y.; Inoue, H.; Takeda, S.; Takahashi, R. Neuropathol. 109:583588; 2005.
Proteasome inhibition in medaka brain induces the features of Parkinsons [113] Pacelli, C.; De, R. D.; Signorile, A.; Grattagliano, I.; di, T. G.; DOrazio, A.;
disease. J. Neurochem. 115:178187; 2010. Nico, B.; Comi, G. P.; Ronchi, D.; Ferranini, E.; Pirolo, D.; Seibel, P.; Schubert, S.;
[89] Mazzulli, J. R.; Xu, Y. H.; Sun, Y.; Knight, A. L.; McLean, P. J.; Caldwell, G. A.; Gaballo, A.; Villani, G.; Cocco, T. Mitochondrial defect and PGC-1alpha
Sidransky, E.; Grabowski, G. A.; Krainc, D. Gaucher disease glucocerebrosi- dysfunction in parkin-associated familial Parkinsons disease. Biochim. Biophys.
dase and alpha-synuclein form a bidirectional pathogenic loop in synuclei- Acta 1041-1053:2011; 1812.
nopathies. Cell 146:3752; 2011. [114] Palacios, N.; Gao, X.; McCullough, M. L.; Schwarzschild, M. A.; Shah, R.;
[90] McCormack, A. L.; Thiruchelvam, M.; Manning-Bog, A. B.; Thiffault, C.; Gapstur, S.; Ascherio, A. Caffeine and risk of Parkinsons disease in a large
Langston, J. W.; Cory-Slechta, D. A.; Di Monte, D. A. Environmental risk cohort of men and women. Mov. Disord 27:12761282; 2012.
factors and Parkinsons disease: selective degeneration of nigral dopami- [115] Palhagen, S.; Heinonen, E.; Hagglund, J.; Kaugesaar, T.; Maki-Ikola, O.;
nergic neurons caused by the herbicide paraquat. Neurobiol. Dis. Palm, R. Selegiline slows the progression of the symptoms of Parkinson
10:119127; 2002. disease. Neurology 66:12001206; 2006.
[91] McDowell, K.; Chesselet, M. F. Animal models of the non-motor features of [116] Pan-Montojo, F.; Anichtchik, O.; Dening, Y.; Knels, L.; Pursche, S.; Jung, R.;
Parkinsons disease. Neurobiol. Dis. 46:597606; 2012. Jackson, S.; Gille, G.; Spillantini, M. G.; Reichmann, H.; Funk, R. H. Progres-
[92] McGeer, P. L.; Itagaki, S.; Boyes, B. E.; McGeer, E. G. Reactive microglia are sion of Parkinsons disease pathology is reproduced by intragastric admin-
positive for HLA-DR in the substantia nigra of Parkinsons and Alzheimers istration of rotenone in mice. PLoS One 5:e8762; 2010.
disease brains. Neurology 38:12851291; 1988. [117] Parkinson Study Group A randomized controlled trial comparing pramipex-
[93] McGeer, P. L.; Schwab, C.; Parent, A.; Doudet, D. Presence of reactive ole with levodopa in early Parkinsons disease: design and methods of the
microglia in monkey substantia nigra years after 1-methyl-4-phenyl- CALM-PD Study. Clin. Neuropharmacol 23:3444; 2000.
1,2,3,6-tetrahydropyridine administration. Ann. Neurol. 54:599604; 2003. [118] Parkinson Study Group A controlled trial of rasagiline in early Parkinson
[94] McNaught, K. S.; Bjorklund, L. M.; Belizaire, R.; Isacson, O.; Jenner, P.; disease: the TEMPO Study. Arch. Neurol 59:19371943; 2002.
Olanow, C. W. Proteasome inhibition causes nigral degeneration with [119] Parkinson Study Group Impact of deprenyl and tocopherol treatment on
inclusion bodies in rats. Neuroreport 13:14371441; 2002. Parkinsons disease in DATATOP subjects not requiring levodopa. Ann.
[95] McNaught, K. S.; Jenner, P. Altered glial function causes neuronal death and Neurol 39:2936; 1996.
increases neuronal susceptibility to 1-methyl-4-phenylpyridinium- and 6- [120] Parkkinen, L.; OSullivan, S. S.; Kuoppamaki, M.; Collins, C.; Kallis, C.;
hydroxydopamine-induced toxicity in astrocytic/ventral mesencephalic Holton, J. L.; Williams, D. R.; Revesz, T.; Lees, A. J. Does levodopa accelerate
co-cultures. J. Neurochem 73:24692476; 1999. the pathologic process in Parkinson disease brain? Neurology 77:14201426;
[96] McNaught, K. S.; Jenner, P. Dysfunction of rat forebrain astrocytes in culture 2011.
alters cytokine and neurotrophic factor release. Neurosci. Lett. 285:6165; [121] Popat, R. A.; Van Den Eeden, S. K.; Tanner, C. M.; McGuire, V.; Bernstein, A. L.;
2000. Bloch, D. A.; Leimpeter, A.; Nelson, L. M. Effect of reproductive factors and
[97] McNaught, K. S.; Jenner, P. Proteasomal function is impaired in substantia postmenopausal hormone use on the risk of Parkinson disease. Neurology
nigra in Parkinsons disease. Neurosci. Lett. 297:191194; 2001. 65:383390; 2005.
[98] McNaught, K. S.; Mytilineou, C.; Jnobaptiste, R.; Yabut, J.; Shashidharan, P.; [122] Quik, M.; Huang, L. Z.; Parameswaran, N.; Bordia, T.; Campos, C.; Perez, X. A.
Jennert, P.; Olanow, C. W. Impairment of the ubiquitin-proteasome system Multiple roles for nicotine in Parkinsons disease. Biochem. Pharmacol.
causes dopaminergic cell death and inclusion body formation in ventral 78:677685; 2009.
mesencephalic cultures. J. Neurochem. 81:301306; 2002. [123] Ragonese, P.; DAmelio, M.; Salemi, G.; Aridon, P.; Gammino, M.; Epifanio, A.;
[99] McNaught, K. S.; Perl, D. P.; Brownell, A. L.; Olanow, C. W. Systemic Morgante, L.; Savettieri, G. Risk of Parkinson disease in women: effect of
exposure to proteasome inhibitors causes a progressive model of Parkin- reproductive characteristics. Neurology 62:20102014; 2004.
sons disease. Ann. Neurol. 56:149162; 2004. [124] Ramsay, R. R.; Salach, J. I.; Singer, T. P. Uptake of the neurotoxin 1-methyl-4-
[100] McNaught, K. S.; Shashidharan, P.; Perl, D. P.; Jenner, P.; Olanow, C. W. phenylpyridine (MPP ) by mitochondria and its relation to the inhibition
Aggresome-related biogenesis of Lewy bodies. Eur. J. Neurosci. of the mitochondrial oxidation of NAD -linked substrates by MPP .
16:21362148; 2002. Biochem. Biophys. Res. Commun 134:743748; 1986.
[101] Miklossy, J.; Doudet, D. D.; Schwab, C.; Yu, S.; McGeer, E. G.; McGeer, P. L. [125] Rentzos, M.; Nikolaou, C.; Andreadou, E.; Paraskevas, G. P.; Rombos, A.;
Role of ICAM-1 in persisting inammation in Parkinson disease and MPTP Zoga, M.; Tsoutsou, A.; Boudou, F.; Kapaki, E.; Vassilopoulos, D. Circulating
monkeys. Exp. Neurol. 197:275283; 2006. interleukin-15 and RANTES chemokine in Parkinsons disease. Acta Neurol.
[102] Miwa, H.; Kubo, T.; Suzuki, A.; Kondo, T. Intragastric proteasome inhibition Scand. 116:374379; 2007.
induces alpha-synuclein-immunopositive aggregations in neurons in the [126] Riederer, P.; Soc, E.; Rausch, W. D.; Schmidt, B.; Reynolds, G. P.; Jellinger, K.;
dorsal motor nucleus of the vagus in rats. Neurosci. Lett 401:146149; 2006. Youdim, M. B. Transition metals, ferritin, glutathione, and ascorbic acid in
[103] Mizuno, Y.; Matuda, S.; Yoshino, H.; Mori, H.; Hattori, N.; Ikebe, S. parkinsonian brains. J. Neurochem. 52:515520; 1989.
An immunohistochemical study on alpha-ketoglutarate dehydrogenase [127] Rousseaux, M. W.; Marcogliese, P. C.; Qu, D.; Hewitt, S. J.; Seang, S.; Kim, R. H.;
complex in Parkinsons disease. Ann. Neurol. 35:204210; 1994. Slack, R. S.; Schlossmacher, M. G.; Lagace, D. C.; Mak, T. W.; Park, D. S.
144 D.T. Dexter, P. Jenner / Free Radical Biology and Medicine 62 (2013) 132144

Progressive dopaminergic cell loss with unilateral-to-bilateral progression in a [150] Tanner, C. M.; Ross, G. W.; Jewell, S. A.; Hauser, R. A.; Jankovic, J.; Factor, S. A.;
genetic model of Parkinson disease. Proc. Natl. Acad. Sci. USA Bressman, S.; Deligtisch, A.; Marras, C.; Lyons, K. E.; Bhudhikanok, G. S.;
109:1591815923; 2012. Roucoux, D. F.; Meng, C.; Abbott, R. D.; Langston, J. W. Occupation and risk
[128] Savica, R.; Parisi, J. E.; Wold, L. E.; Josephs, K. A.; Ahlskog, J. E. High school of parkinsonism: a multicenter casecontrol study. Arch. Neurol.
football and risk of neurodegeneration: a community-based study. Mayo 66:11061113; 2009.
Clin. Proc. 87:335340; 2012. [151] Tofaris, G. K.; Layeld, R.; Spillantini, M. G. alpha-Synuclein metabolism and
[129] Schapira, A. H. Mitochondria in the aetiology and pathogenesis of Parkin- aggregation is linked to ubiquitin-independent degradation by the protea-
sons disease. Lancet Neurol 7:97109; 2008. some. FEBS Lett. 509:2226; 2001.
[130] Schapira, A. H. Mitochondrial pathology in Parkinsons disease. Mt. Sinai J. [152] Tofaris, G. K.; Spillantini, M. G. Alpha-synuclein dysfunction in Lewy body
Med. 78:872881; 2011. diseases. Mov. Disord 20(Suppl. 12):S37S44; 2005.
[131] Schapira, A. H.; Albrecht, S.; Barone, P.; Comella, C. L.; McDermott, M. P.; [153] Turski, L.; Bressler, K.; Rettig, K. J.; Loschmann, P. A.; Wachtel, H. Protection
Mizuno, Y.; Poewe, W.; Rascol, O.; Marek, K. Rationale for delayed-start of substantia nigra from MPP neurotoxicity by N-methyl-D-aspartate
study of pramipexole in Parkinsons disease: the PROUD study. Mov. Disord antagonists. Nature 349:414418; 1991.
25:16271632; 2010. [154] Usenovic, M.; Tresse, E.; Mazzulli, J. R.; Taylor, J. P.; Krainc, D. Deciency of
[132] Schapira, A. H.; Cooper, J. M.; Dexter, D.; Clark, J. B.; Jenner, P.; Marsden, C. D. ATP13A2 leads to lysosomal dysfunction, alpha-synuclein accumulation,
Mitochondrial complex I deciency in Parkinsons disease. J. Neurochem. and neurotoxicity. J. Neurosci. 32:42404246; 2012.
54:823827; 1990. [155] Vekrellis, K.; Stefanis, L. Targeting intracellular and extracellular alpha-
[133] Schapira, A. H.; Jenner, P. Etiology and pathogenesis of Parkinsons disease. synuclein as a therapeutic strategy in Parkinsons disease and other
Mov. Disord 26:10491055; 2011. synucleinopathies. Expert Opin. Ther. Targets 16:421432; 2012.
[134] Schapira, A. H.; Olanow, C. W. Neuroprotection in Parkinson disease: [156] Velayati, A.; Yu, W. H.; Sidransky, E. The role of glucocerebrosidase
mysteries, myths, and misconceptions. JAMA 291:358364; 2004. mutations in Parkinson disease and Lewy body disorders. Curr. Neurol.
[135] Schrag, A.; Ben-Shlomo, Y.; Quinn, N. P. Cross sectional prevalence survey Neurosci. Rep. 10:190198; 2010.
of idiopathic Parkinsons disease and parkinsonism in London. Br. Med. J [157] Vernon, A. C.; Zbarsky, V.; Datla, K. P.; Croucher, M. J.; Dexter, D. T. Subtype
321:2122; 2000. selective antagonism of substantia nigra pars compacta Group I metabo-
[136] Shashidharan, P.; Good, P. F.; Hsu, A.; Perl, D. P.; Brin, M. F.; Olanow, C. W. tropic glutamate receptors protects the nigrostriatal system against
TorsinA accumulation in Lewy bodies in sporadic Parkinsons disease. Brain 6-hydroxydopamine toxicity in vivo. J. Neurochem. 103:10751091; 2007.
Res. 877:379381; 2000. [158] Vernon, A. C.; Zbarsky, V.; Datla, K. P.; Dexter, D. T.; Croucher, M. J. Selective
[137] Shin, J. H.; Ko, H. S.; Kang, H.; Lee, Y.; Lee, Y. I.; Pletinkova, O.; Troconso, J. C.; activation of group III metabotropic glutamate receptors by L-( )-2-amino-4-
Dawson, V. L.; Dawson, T. M. PARIS (ZNF746) repression of PGC-1alpha phosphonobutryic acid protects the nigrostriatal system against
contributes to neurodegeneration in Parkinsons disease. Cell 144:689702; 6-hydroxydopamine toxicity in vivo. J. Pharmacol. Exp. Ther. 320:397409; 2007.
2011. [159] Wakabayashi, K.; Takahashi, H.; Takeda, S.; Ohama, E.; Ikuta, F. Parkinsons
[138] Sian, J.; Dexter, D. T.; Lees, A. J.; Daniel, S.; Agid, Y.; Javoy-Agid, F.; Jenner, P.; disease: the presence of Lewy bodies in Auerbachs and Meissners plexuses.
Marsden, C. D. Alterations in glutathione levels in Parkinsons disease and Acta Neuropathol. 76:217221; 1988.
other neurodegenerative disorders affecting basal ganglia. Ann. Neurol. [160] Wang, L.; Magen, I.; Yuan, P. Q.; Subramaniam, S. R.; Richter, F.; Chesselet,
36:348355; 1994. M. F.; Tache, Y. Mice overexpressing wild-type human alpha-synuclein
[139] Solesio, M. E.; Saez-Atienzar, S.; Jordan, J.; Galindo, M. F. Characterization of display alterations in colonic myenteric ganglia and defecation. Neurogas-
mitophagy in the 6-hydoxydopamine Parkinsons disease model. Toxicol. Sci. troenterol. Motil. 24:e425e436; 2012.
129:411420; 2012. [161] Whone, A. L.; Watts, R. L.; Stoessl, A. J.; Davis, M.; Reske, S.; Nahmias, C.;
[140] Spillantini, M. G.; Crowther, R. A.; Jakes, R.; Hasegawa, M.; Goedert, M. Lang, A. E.; Rascol, O.; Ribeiro, M. J.; Remy, P.; Poewe, W. H.; Hauser, R. A.;
alpha-Synuclein in lamentous inclusions of Lewy bodies from Parkinsons Brooks, D. J. Slower progression of Parkinsons disease with ropinirole
disease and dementia with Lewy bodies. Proc. Natl. Acad. Sci. USA versus levodopa: the REAL-PET study. Ann. Neurol. 54:93101; 2003.
95:64696473; 1998. [162] Witt, M.; Bormann, K.; Gudziol, V.; Pehlke, K.; Barth, K.; Minovi, A.;
[141] Spina, M. B.; Cohen, G. Dopamine turnover and glutathione oxidation: Hahner, A.; Reichmann, H.; Hummel, T. Biopsies of olfactory epithelium in
implications for Parkinson disease. Proc. Natl. Acad. Sci. USA 86:13981400; patients with Parkinsons disease. Mov. Disord 24:906914; 2009.
1989. [163] Xie, W.; Li, X.; Li, C.; Zhu, W.; Jankovic, J.; Le, W. Proteasome inhibition
[142] Steigerwald, F.; Potter, M.; Herzog, J.; Pinsker, M.; Kopper, F.; Mehdorn, H.; modeling nigral neuron degeneration in Parkinsons disease. J. Neurochem.
Deuschl, G.; Volkmann, J. Neuronal activity of the human subthalamic 115:188199; 2010.
nucleus in the parkinsonian and nonparkinsonian state. J. Neurophysiol. [164] Xu, Y. H.; Sun, Y.; Ran, H.; Quinn, B.; Witte, D.; Grabowski, G. A. Accumula-
100:25152524; 2008. tion and distribution of alpha-synuclein and ubiquitin in the CNS of
[143] Stern, M. B.; Lang, A.; Poewe, W. Toward a redenition of Parkinsons Gaucher disease mouse models. Mol. Genet. Metab. 102:436447; 2011.
disease. Mov. Disord 27:5460; 2012. [165] Yong, V. W.; Perry, T. L. Monoamine oxidase B, smoking, and Parkinsons
[144] Stocchi, F.; Olanow, C. W. Neuroprotection in Parkinsons disease: clinical disease. J. Neurol. Sci. 72:265272; 1986.
trials. Ann. Neurol. 53(Suppl. 3):S87S97; 2003. [166] Yue, Z.; Lachenmayer, M. L. Genetic LRRK2 models of Parkinsons disease:
[145] Stypula, G.; Kunert-Radek, J.; Stepien, H.; Zylinska, K.; Pawlikowski, M. dissecting the pathogenic pathway and exploring clinical applications. Mov.
Evaluation of interleukins, ACTH, cortisol and prolactin concentrations in Disord 26:13861397; 2011.
the blood of patients with Parkinsons disease. Neuroimmunomodulation [167] Zeng, B. Y.; Medhurst, A. D.; Jackson, M.; Rose, S.; Jenner, P. Proteasomal
3:131134; 1996. activity in brain differs between species and brain regions and changes with
[146] Surmeier, D. J. Calcium, ageing, and neuronal vulnerability in Parkinsons age. Mech. Ageing Dev. 126:760766; 2005.
disease. Lancet Neurol. 6:933938; 2007. [168] Zheng, B.; Liao, Z.; Locascio, J. J.; Lesniak, K. A.; Roderick, S. S.; Watt, M. L.;
[147] Tagliati, M.; Martin, C.; Alterman, R. Lack of motor symptoms progression in Eklund, A. C.; Zhang-James, Y.; Kim, P. D.; Hauser, M. A.; Grunblatt, E.;
Parkinsons disease patients with long-term bilateral subthalamic deep Moran, L. B.; Mandel, S. A.; Riederer, P.; Miller, R. M.; Federoff, H. J.; Wullner, U.;
brain stimulation. Int. J. Neurosci 120:717723; 2010. Papapetropoulos, S.; Youdim, M. B.; Cantuti-Castelvetri, I.; Young, A. B.;
[148] Tanner, C. M. Advances in environmental epidemiology. Mov. Disord Vance, J. M.; Davis, R. L.; Hedreen, J. C.; Adler, C. H.; Beach, T. G.; Graeber, M.
25(Suppl. 1):S58S62; 2010. B.; Middleton, F. A.; Rochet, J. C.; Scherzer, C. R. PGC-1alpha, a potential
[149] Tanner, C. M.; Kamel, F.; Ross, G. W.; Hoppin, J. A.; Goldman, S. M.; therapeutic target for early intervention in Parkinsons disease. Sci. Transl. Med.
Korell, M.; Marras, C.; Bhudhikanok, G. S.; Kasten, M.; Chade, A. R.; 2; 2010. 52ra73.
Comyns, K.; Richards, M. B.; Meng, C.; Priestley, B.; Fernandez, H. H.; [169] Zigmond, M. J.; Cameron, J. L.; Leak, R. K.; Mirnics, K.; Russell, V. A.;
Cambi, F.; Umbach, D. M.; Blair, A.; Sandler, D. P.; Langston, J. W. Rotenone, Smeyne, R. J.; Smith, A. D. Triggering endogenous neuroprotective processes
paraquat, and Parkinsons disease. Environ. Health Perspect. 119:866872; through exercise in models of dopamine deciency. Parkinsonism Relat.
2011. Disord. 15(Suppl. 3):S42S45; 2009.

Anda mungkin juga menyukai