Abstract
By the time a patient first presents with symptoms of Parkinson’s disease at the clinic, a significant proportion (50-70%) of the cells in the substantia nigra (SN) has already been destroyed. This degeneration progresses until, within a few years, most of the cells have died. Except for rare cases of familial PD, the initial trigger for cell loss is unknown. However, we do have some clues as to why the damage, once initiated, progresses unabated. It would represent a major advance in therapy to arrest cell loss at the stage when the patient first presents at the clinic. Current therapies for Parkinson’s disease focus on relieving the motor symptoms of the disease, these unfortunately lose their effectiveness as the neurodegeneration and symptoms progress. Many experimental approaches are currently being investigated attempting to alter the progression of the disease. These range from replacement of the lost neurons to neuroprotective therapies; each of these will be briefly discussed in this review. The main thrust of this review is to explore the interactions between dopamine, alpha synuclein and redox-active metals. There is abundant evidence suggesting that destruction of SN cells occurs as a result of a self-propagating series of reactions involving dopamine, alpha synuclein and redox-active metals. A potent reducing agent, the neurotransmitter dopamine has a central role in this scheme, acting through redox metallo-chemistry to catalyze the formation of toxic oligomers of alpha-synuclein and neurotoxic metabolites including 6-hydroxydopamine. It has been hypothesized that these feed the cycle of neurodegeneration by generating further oxidative stress. The goal of dissecting and understanding the observed pathological changes is to identify therapeutic targets to mitigate the progression of this debilitating disease.
Key Words: Parkinson’s disease, pathology, redox chemistry, metallo- chemistry, review, iron.
1. INTRODUCTION
Classified as the second most common neurodegenerative disorder, Parkinson’s disease (PD) is a debilitating motor related disease and is presently incurable. Resulting from a gradual progressive degeneration of nigrostriatal neurons, PD affects approximately 1% of the population over 50 years of age [333].
The pathological characteristics of PD require careful differential diagnosis of specific features which are only confirmed post mortem. Determined by microscopic examination, the key pathological characteristics of PD are loss of nigral neurons (and loss of pigmentation in this area), and the presence of insoluble proteinaceous cytoplasmic inclusions termed Lewy bodies (LB) and Lewy neurites in the remaining cells. The inclusions are rich in the fibrils of the presynaptic protein alpha synuclein (α-synuclein) and ubiquitin which are thought to arise from the failure of normal degradation in the proteasome pathway (see section 2.3) [197, 280, 381]. Motor symptoms that are associated with the disease are most likely to be caused by a loss of dopamine (DA) producing cells within the substantia nigra pars compacta (SN), in particular, in the interconnections between the SN and the caudate and putamen. The five nuclei which comprise the basal ganglion are; Substantia Nigra (SN), Globus Pallidus (GP), Subthalamic Nucleus (STN), Caudate Nucleus (CN), and Putamen (CPu). The CPu seems to be the most affected structure, losing up to 95% of dopaminergic inputs compared to 80% in the caudate nucleus [184, 214]. Animal models that have shown dynamic rearrangement of the dopaminergic terminals after lesioning suggests that this is likely to be occurring in the preclinical stages of the human disease [114].
In recent times, a new concept in the progression of PD has emerged [45] proposing that a number of nuclei are involved before the SN. The first appearance of disease related symptoms correlates with functional deficits in the lower brainstem and olfactory bulb, then dysfunction progresses up the brainstem to produce classical PD [45]. Changes in other nuclei are observed and are thought to be secondary to the primary disease [175, 176]. The disease progresses until cortical and cognitive changes develop. In this review we will focus on interventions which may prevent the chronic degeneration of the dopaminergic neurons of the SN.
Clinical behavioral symptoms of PD are linked to uncontrolled motor deficits such as akinesia (absence of movement or temporary paralysis), bradykinesia (abnormal slowness of movement), abnormalities in gait, resting tremor and rigidity. Cognitive dysfunction such as speech, executive function and memory loss can also develop as later symptoms.
The study by Braak and colleagues (2003) [45] has also shown that patients that exhibit early stage dysfunction (such as REM sleep disturbance, restless leg syndrome) go on to develop PD within 20 years. In the future, these clinical developments may identify patients that would be suitable candidates for neuroprotective therapies before the SN cells are lost.
PD is a progressive neurodegenerative disease with up to 10% of the remaining dopaminergic cells dying each year [295]. It has been suggested that this progression of the disease explains the loss of responsiveness to drug therapies such as L-dihydroxyphenylalanine (L-DOPA), ultimately failing as the number of DA producing cells falls below a critical level [111]. Most patients benefit from L-DOPA without complications for only approximately five years from the commencement of treatment. Subsequently, the side effects of the treatment become apparent with the percentage of motor fluctuations and dyskinesia ranging from 10% of patients at 5 years, up to 80–90% in later years [265, 266]. Some researchers within the field suggest that pharmacological intervention with neuroprotective therapies in the preclinical stage of PD will have a better chance of prolonging near normal function in patients [200, 264]. New diagnostic tools are currently being developed with several able to provide diagnosis at an earlier stage than is commonly achieved at present [380, 431]. With our expanded clinical perspective on parkinsonism, there is a real chance of utilizing new neuroprotective therapies when they become available. Current studies have hypothesized an interaction of multi-factorial risk factors which can cause the disease. These can be classified as: genetic, environmental, metabolic (oxidative stress; reactive species (RS) production) and biochemical (protein aggregation). New diagnostic techniques and promising neuroprotective pharmacological agents are becoming a reality enabling the next stage in PD therapy, slowing down the progression of SN cell loss in the disease. This review focuses on the possible contributing factors that cause PD, in order to discuss possible strategies to intervene and to slow the progression of PD.
2. PUTATIVE CAUSES AND RISK FACTORS INVOLVED IN PD
Various contributing factors such as drugs of abuse, industrial/ agricultural chemicals and genetic mutations have all been known to cause PD or PD type symptoms in humans. Therefore animal models that utilize these factors are valuable tools to help identify the molecular mechanisms of neurodegeneration that are associated with PD. The exact mechanistic trigger that causes loss of nigral cells found in sporadic PD remains unknown. Many studies have revealed that in combination with a genetic predisposition, contributing factors for oxidative stress (due to the dysregulation in metabolic homeostasis) are involved with the degenerative processes. Although only a minor proportion of these PD patients (<10%) harbor all known genetic deficits, investigation of these mutations may reveal the mechanisms through which a therapeutic target could be identified [400]. With this caveat in mind, the genetic mutations point toward the direction of possible pharmacological targets.
2.1. Genetic Factors
PD is primarily a sporadic disorder that is believed to be ‘multi-factorial’ occurring through the involvement of interactions between genetic and environmental factors. Epidemiological studies have shown that while sporadic PD occurs at a later onset of 60 years of age, familial onset tends to develop at a younger age (<50 years), and occurs in approximately 1% of all PD cases [333]. Thus, genetic mutations alone cannot explain the majority of disease cases. However, mutations in specific genes in familial PD have given emphasis to particular genes involved in the molecular pathogenesis of parkinsonism. Six gene loci have been identified that have a PD pathological phenotype (see Table 1). Mutations in the α-synuclein (α-syn), Parkin, PTEN- Induced putative Kinase 1 (PINK1), DJ-1, Leucine rich repeat kinase 2 (LRRK2), ATP13A2 and UCH-L1 genes have all been shown to be involved in familial PD.
Table 1.
Genes and their Associated Mutations that have been Identified in PD
| Gene | Function | Major Mutations Identified | Mode of Inheritance | Clinical | Pathology (Human) | Pathology In Vivo Model |
|---|---|---|---|---|---|---|
| α-Syn [198, 333](PARK1) | Possibly functions as a neurotransmitter modulator | A53T, A30P, E46K | Autosomal dominant | Similar to sporadic PD, early onset | SN depigmentation, DA loss, gliosis, LB & neuritis [382] Increases in amyloidal fibrillation and tau inclusion [156]LBs | DA loss, LB are present in areas which are not associated with PD [204, 227] |
| Parkin(PARK2) | E3 Ubiquitin protein ligase [361] | EX3-7DEL EX4DEL T240N [169, 170, 215, 256, 273] | Autosomal recessive | Early onset, slow progression parkinsonism [361] | Variations in the presence of LB | Data not available |
| (PARK3) | Unknown | Data not available | Autosomal dominant | Data not available | Data not available | Data not available |
| (PARK4) | See α-Syn above | α-Syn Triplication (up to 4 full copies) | Autosomal dominant | Early Onset PD | Nigral and locus coeruleus degeneration, presence of LB in hippocampus, locus coeruleus and cortices [297, 368] | Data not available |
| UCH L1 (PARK5) | C-terminal ubiquitin hydrolyse and, ubiquitin ligase [241, 250] | V66M, S18Y, I93M [216, 262, 290, 351, 393, 401, 430, 444] | Autosomal dominant | PD | Data not available | Mutant mice display gracile axonal dystrophy [213] |
| PINK1(PARK6) | Mitochondrial serine/threonine kinase [230, 231] | H271Q, L347P, 1573TTAG, 1602CAA, R279H, DEL EX6-8, T313M, A217N L489P, L347P, E240K, A340T [132, 402, 420] | Autosomal recessive | Similar to atypical sporadic PD, early onset | Data not available | PINK1 silencing showed rapid eye degeneration and progressive DA loss in a drosophila model [419, 435] |
| DJ-1(PARK7) | Protection against oxidative stress [196, 286, 288] | 14-KB DEL, L166P, M26I, D149A, G64D, E163K + 18-BP DUP | Autosomal recessive | Early onset PD | Data not available | DJ-1 null mice showed no loss of striatal DA neurons [213] |
| LRRK2(PARK8) | Protein kinase (unknown substrates) | R1441G, Y1699C, R1441C, L1122V, G2019S, I2020T, R1441H, G2385R, P755L, [85] | Autosomal dominant | PD | LB, nigral degeneration without LB, And tau aggregation [452] | Data not available |
α-synuclein
Specifically enriched in the adult brain, the α-synuclein protein is ubiquitously expressed within the central nervous system (CNS) in neurons and glia and is localized in neuronal structures [198]. Belonging to a structurally homologous protein family which includes β- and γ-synuclein, its function has not been well defined. Recent publications have implicated α-synuclein as a neurotransmitter modulator regulating vesicle handling (recycling and docking) [121, 122, 228], synaptic transmission and re-uptake [249]. Structural assays have determined that α-synuclein has the potential to bind to phospholipids particularly on synaptic vesicles [80, 300]. The binding to phospholipids occurs through the N-amphiphatic domain which is a highly conserved region. α-Synuclein knockout animals have deficits in expression of synaptic-associated proteins as well as a reduction in synaptic vesicles [50], but surprisingly only subtle behavioral deficits. Most recently, other novel functions have shown α-synuclein as a protein trafficking chaperone as defects in the gene obstructs ER-Golgi vesicular trafficking [74]. This evidence suggests that α-synuclein plays an essential role in different cellular functions which may be disrupted in PD and other diseases where α-synuclein aggregates are present such as Dementia with Lewy Bodies (DLB) and Multiple System Atrophy (MSA) - (synucleinopathies). The parkinsonian disease phenotype to genotype correlation was firstly linked to a mutation in the α-synuclein gene derived from an Italian kindred [333, 334]. The penetrance in this family alone was 85%, comprising of a single point mutation, a G to A conversion at position 209 which ultimately changed the amino acid code from an Alanine to Threonine at residue 53 (A53T). Since then, two other point mutations (A30P and E46K) have been characterized and shown to segregate with the disease. Whole gene duplication [58, 195] and triplication resulting in over-expression of wild type α-synuclein [368] have been found to cause familial PD, all increase the rate of onset and rate of disease progression [301]. The A53T mutation increases the mean rate of onset of PD (to approximately 46 years of age), muscle rigidity and bradykinesia being the prominent clinical features [14]. Systematic mutational screening of the α-synuclein gene revealed another mutation, A30P, which displayed similar features to those observed in sporadic cases, with classic fundamental behavioural deficits associated with L-DOPA responsive PD [227]. Unlike the other missense mutations identified in this gene, the E46K mutation is linked to LB with dementia [440] which is associated with amyloid fibrilization [156]. The mutations in α-synuclein (A53T, A30P) can form filaments at an increased rate in vitro [72]. Wild type α-synuclein undergoes oligomerization when DA is present (in a dose dependent manner). These oligomers are found to be soluble, but are not amyloidogenic [53]. In the presence of iron and copper, wild type α-synuclein can rapidly form soluble SDS resistant oligomers but the formation of amyloidogenic α-synuclein aggregates can be completely inhibited by the addition of DA [53]. This strongly suggests that DA acts as a dominant modulator of α-synuclein aggregation [53]. The structural morphology of the aggregates of α-synuclein differs across genetic mutation species and when copper or iron is present [28]. Interestingly, the three mutations have been shown to reside in potential metal binding sites (for both copper and iron) which could alter the binding of metal-protein complexes, this may be important in fibril formation [29]. In its native state, α-synuclein is an unfolded protein [143, 409, 423] due to an overall low net hydrophobicity [408]. However, like many amyloidogenic proteins such as the beta amyloid protein (Aβ), the α-synuclein protein has a propensity to aggregate specifically in vitro to form higher order soluble oligomers, an intermediate species, which in turn further polymerizes into fibrils. Insoluble filamentous aggregates of α-synuclein are the major component of LB in PD and other neurodegenerative synucleinopathies [381]. There is no clear evidence that demonstrates if the development of LB occurs as the cause or a result of the disease and it remains to be established whether the toxic form of α-synuclein is a soluble oligomer, as has been postulated for Aβ in Alzheimer’s disease, or the classic insoluble fibrils. The oligomeric species is considered to be the most neurotoxic, possibly by causing vesicle permeability [416]. On the other hand the accelerated pathology of the α-synuclein mutants (particularly A53T), is associated with an increased rate of self-fibrilization [301].
Parkin
Parkin (PARK2) mutants have been linked to autosomal recessive juvenile parkinsonism. An array of mutations including deletions [170, 215, 256], multiplications, rearrangements, missense and several point mutations have now been reported. Parkin mutants present as a wide range of clinical PD phenotypes but differ from idiopathic PD as they tend to be more early onset, with slow progression and neuronal death in the absence of LB formation (see Table 1) [361]. Shimura and colleagues (2000) [361] showed that Parkin is an E3 ubiquitin ligase the loss of functional activity of which leads to protein accumulation. E3 proteins attach polyubiquitin chains to target proteins that are to be degraded by the proteasome. Mutations that have been described result in greatly diminished Parkin production which in turn results in the failure of Parkin associated substrates to be degraded. However, this fact seems to be confounded as the survival of DA neurons in Parkin knockout mice remains unaffected [145].
PINK1
PINK1 was first identified in cancer expression profiles [407] and shown to be localized to mitochondria by both immunofluorescence and western blot analysis [411]. In a study of three PD kindreds, two families harbored a mutation within the kinase domain of the PINK1 gene; a missense mutation and a nonsense mutation. To date, around 40 mutations have been characterized (see Table 1), most of which have not yet been correlated to a neuropathological phenotype in human PD cases. Many mutations have been discovered in late onset cases, equivalent to sporadic PD cases [4] and also to atypical early onset PD. Mutations found in PINK1 in early onset cases maybe the second most common mutation after Parkin to induce parkinsonism [168]. However, pathogenic PINK1 mutants seem to be a rare cause of early onset PD [344]. The potential PINK1 substrates seem to phosphorylate mitochondrial proteins that may act to preserve mitochondrial integrity [411]. However, these target substrates remain unknown. Expression of mutant forms of PINK1 are said to be loss of function mutations, which suggests that the potential kinase activity of PINK1 is required and therefore highlights the relevance of mitochondrial dysfunction as one of the processes that mediate PD.
Expression of mutations in this mitochondrial kinase, demonstrates a heterogeneous spectrum of pathological phenotypes ranging from early onset to late onset PD. Less than 10% of PINK1 protein has been shown to be found in LB which could be due to poor solubility of the protein and a greater propensity for its aggregation in culture [23]. There have been no post mortem studies looking at the neuropathology of PINK1 mutation carriers.
LRRK2
Mutations in the LRRK2 gene have been identified in both the sporadic and familial forms of PD and display an autosomal dominant inheritance pattern of PD [452]. More than 30 mutations in the LRRK2 gene have now been reported and many of these reside within a highly conserved functional domain of the protein [85, 86, 452]. It has been determined that LRRK2 possesses kinase activity but little is known about its phosphorylation substrates and its specific role in PD pathogenesis. Expression of LRRK2 is shown in all tissues and at low levels in the brain [316]. The average age of onset in familial cases is the late 50s and 60s, indicating that of all the genes identified in familial PD cases, this gene bears the closest resemblance to sporadic forms of the disease. Varying pathological phenotypes have been observed in PD patients with mutations in LRRK2 and may reflect multiple roles or target substrates for this protein.
DJ-1
DJ-1 mutants were first reported in a consanguineous Dutch family [413]. The first mutation identified in this family was a homozygous exon 1 to 5 deletion [413] which effectively caused a deletion of the entire protein. Another reported pathogenic homozygous mutation L166P showed co-segregation with PD in an Italian family [44]. Other reports of DJ-1 mutants (see Table 1) have been described. The loss of function from specific mutations has been shown to cause autosomal recessive early onset PD. Mutations in DJ-1 are extremely rare occurring in approximately 1-2% of early onset familial cases [3]. There have so far been no reports of any DJ-1 mutations in late onset sporadic cases [3]. DJ-1 is suggested to be co-localized in the mitochondria [446], and is more predominantly up-regulated under high oxidative stress conditions [38]. DJ-1 related cellular processes include, attenuating oxidation [196, 286, 288], RNA binding [181], cell transformation, and androgen receptor signaling [331]. The role of DJ-1 in neuroprotection against the oxidative stress response is of particular interest to PD. Drosophila models which lack DJ-1 show increased susceptibility to oxidative stress and environmental toxins such as paraquat and rotenone [235, 320]. It has been shown that loss of DJ-1 alone cannot sufficiently induce parkinsonism but increases the susceptibility of DA neurons under an oxidative environment [213].
UCH-L1
The only mutation described in the UCH-L1 gene was the I93M mutation found in a German family [240]. One other reported polymorphism (S18Y) was found in exon 3 of UCH-L1 [247] but is suggested to be non-pathogenic since it is found in about 20% of the Caucasian population [247]. UCH-L1 mutations have yet to be correlated to a cellular morphological phenotype in PD. However, in vivo transgenic mice models with this mutation display gracile axonal dystrophy. This autosomal disease results in axonal degeneration [389], Aβ protein and accumulation of ubiquitinated proteins within the sensory and motor systems [347, 389].
UCH-L1 has a high sequence homology to the gene family that hydrolyzes ubiquitin c-terminals to form monomeric ubiquitin (ubiquitin hydrolase) [241]. It has been suggested that UCH-L1 may have multi-functional capabilities including the recycling of ubiquitin by hydrolyzing ubiquitinated proteins and ubiquitin ligase activity [250].
2.2. Environmental Toxins
While the genetic contribution to the disease seems to be only a small proportion of cases, environmental factors are thought to play a pivotal role in PD etiology. In the early 1980’s it was suspected that exogenous neurotoxins caused PD-like symptoms. These gave rise to correlative studies which suggest that long term exposure to common pesticides cause PD-like symptoms. Mitochondrial dysfunction plays a vital role in DA neuron degeneration. Mitochondrial inefficiency is considered to be a result of oxidative stress and is proposed as a primary mechanism for nigral loss following exposure to a range of substances [399]. These neurotoxic agents potentially elicit their effects on mitochondria.
Large scale cohort studies of both chronic and low dose exposures to pesticides have shown a 70% increase in risk of developing PD compared to people not exposed to these chemicals [13]. As both farmers and non-farmers seem to share the same level of risks associated with PD and pesticide exposure, Ascherio and colleagues (2006) [13] suggests that non-farmers could have been exposed to harmful pesticides while gardening. It is becoming apparent that common chemicals, such as Paraquat and Rotenone, could be implicated in some of the PD cases.
2.2.1. Agricultural Toxins
Paraquat (PQ; N,N'-Dimethyl-4,4'-bipyridinium dichloride) is a potential neurotoxin as it has the ability to cross the blood brain barrier [75, 427]. PQ is thought to participate in redox cycling as it can be easily reduced to form free radicals, resembling a structural analogue of a known neurotoxic metabolite 1-methyl-4-phenylpyridinium ion (MPP+). Like MPP+, PQ selectively destroys dopaminergic neurons. Animal models have shown that systemic [276] and long term infusion of PQ has a toxic effect on rodent dopaminergic neurons [312]. PQ causes a loss of TH positive neurons together with loss of striatal terminal projections, decreases in Dopamine transporter (DAT) and accelerated fibrilization of α-synuclein [260, 410].
The insecticide Rotenone is widely used as an in vivo animal model of inducing PD. It is thought that the rotenone model is of key importance as the specific neuropathology reflects the selective loss of DA neurons in the SN and LB formation [27, 287, 399]. However the specificity of the model has been called into question as neurodegeneration has also been observed in non-dopaminergic systems [182]. Rats chronically infused with rotenone show severe behavioral deficits characteristic of PD including an unsteady gait and bradykinesia [7, 8]. Chronic low doses of rotenone also show an increase in oxidative stress markers as well as α-synuclein positive LB [360]. Similar behavioral observations have been shown in mice treated with rotenone which do not exhibit LB inclusions or other pathological changes [342].
Rotenone induces the cellular death cascade via inhibition of the mitochondrial metallo-protein complex I [27, 359, 360] resulting in oxidative phosphorylation dysfunction. Rotenone causes the inhibition of complex I throughout the brain, however not all investigations have shown loss of SN neurons [27]. The presence of oxidative metabolites in samples from PD tissue is similar to those oxidative metabolites produced by rotenone suggests that oxidative stress could play an important role in the chain of events leading to PD [399].
2.2.2. Drugs of Abuse and Parkinsonism
Drug-induced parkinsonism has been reported in both humans and in animal models [116]. The commonly abused amphetamine derivatives have been reported to damage catecholamine neurons in both humans and in animal models of the disease [193]. Abuse of drugs such as amphetamine, ecstasy and cocaine are known to induce alterations in striatal DA [220] with specific effects on Tyrosine Hydroxylase (TH) positive cells [220, 377, 403] in mice models. Drugs of these sorts have specific modes of action which make them potential catalysts for PD: i) amphetamine inhibits DA uptake causing a marked elevation in extracellular DA. ii) A single dose of cocaine results in elevated levels of hydrogen peroxide and lipid peroxidation products for up to 50 hours [96]. iii) There is increased DA turnover as the result of DA and its metabolites causing an elevation in extracellular DA [319]. iv) The increase in DA production due to amphetamine generates a toxic cascade of free radicals [77]. v) Post mortem brain tissue from human chronic cocaine abusers exhibit over-expression of α-synuclein mRNA with a three fold increase in the α-synuclein protein levels in dopaminergic neurons [269] although no data are available on the histopathology of abusers [93]. vi) Amphetamine and similar drugs have been shown to redistribute DA into the cytoplasm where it can be oxidized into semi-quinones and other oxidative metabolites [117]. vii) Drug induced toxicity has been shown to form inclusions containing α-synuclein specifically within the SN [116]. While the use of recreational drugs has raised the possibility of an associated increased risk of developing PD, this link has not been clearly established using population based studies.
2.2.3. MPTP
In the early 1980’s, drug addicts who tried to manufacture their own drugs, instead synthesized MPTP and developed a syndrome symptomatically very similar to sporadic PD (L-DOPA responsive, progressive, with neuronal inclusions) [233, 234]. This neurotoxin has subsequently been extensively used in non-human primates to replicate the cardinal symptoms of PD [354]. Although rodents are more resistant to MPTP than primates, they have contributed greatly to the understanding of the mechanisms of toxicity. Rodents are commonly employed in developing a new generation of drugs to target the causes rather than the symptoms of PD [287]. To date, most of the work on MPTP using mice demonstrates that cell death following acute exposure of the toxin occurs in two phases: initial cell death commences within hours after the insult [166], peaking at around 5 days [199] with the second phase lasting ≈ 21-28 days [36, 95, 192]. Like most animals models, the acute MPTP model does not reflect all aspects of the human disease. For example, acute intoxication with MPTP does not result in inclusions within the SN [118, 395] which suggests that DA neurons are dying before protein aggregation can occur. More recently, rodent models that employ chronically administered MPTP, exhibit intracellular α-synuclein-positive electron dense inclusions [118]. Chronic MPTP administration is shown to up regulate the expression, the nitration and also the aggregation of α-synuclein within the SN [414]. Conversely, animals that lack α-synuclein are spared and have been shown to be protected against the toxic effects of chronic MPTP treatment [116]. This suggests that the uptake of MPTP is rapid due to high affinity DA re-uptake transporters. The conversion of MPTP into MPP+ by Monoamine Oxidase B (MAO-B) allows the reduced form to affect mitochondrial metabolism eventually leading to the inhibition of complex I and release of cytochrome c from the inner membrane of the mitochondria [237]. This model is one of the keystones of the oxidative stress hypothesis for PD.
2.3. Protein Aggregation
The failure to clear damaged and cytotoxic protein aggregates is often a common feature of most neurodegenerative disorders including PD. Proteasome dysfunction has been observed in dopaminergic neurons in PD patients [278] and cortical neurons in AD cases [208]. Mutations in genes encoding proteins within the ubiquitin proteasome system (parkin and UCH-L1) further link this pathway to PD. Functional and structural deficits within the proteasome pathway prevent protein clearance, leading to proteolytic stress [278]. There has been growing speculation that proteasome dysfunction via proteolytic stress could be the underlying mechanism of LB formation and neurodegeneration in the SN in both familial and sporadic PD [278-282].
As the cellular machinery for protein degradation or repair becomes overwhelmed in the disease, failure of this pathway leads to protein accumulation and ultimately cell death [415]. It is clear that an abundance of damaged proteins which occur through oxidation or nitration processes can lead to proteasome inhibition as seen in sporadic PD cases. Proteasome inhibition seems to initially affect DA neurons by modifying DA re-uptake [281, 299]. In cell culture models, the presence of oxidative products such as hydrogen peroxide (H2O2) and peroxynitrite (ONOO-) inhibits the activity of the 20S proteasome [339]. Excessive levels of oxidative modifications to the proteasome causes a decrease in proteolysis, decreases in solubility, stabilization of proteins via cross-linkages and protein aggregation [142, 314]. However, what remains unclear is whether the precise nature of LB formation is a systemic defense mechanism acting as a neuroprotectant or is a result of the disease.
2.4. Oxidative Stress and Parkinson’s Disease
Neurodegeneration is a multifaceted process and the mechanisms that result in cellular death are linked to events that cause oxidative stress. Although there is much evidence in favor of this hypothesis; there is no definitive study. In the current section, the discussion focuses on: the markers of oxidative stress that have been observed in PD; how the cell becomes stressed; what oxidative damage does to cells; and cellular defense mechanisms to help overcome oxidative stress.
Oxidative stress is a key pathological process that is common to all neurodegenerative diseases. While oxidative stress occurs over the entire brain, the nigral environment appears to be more sensitive to oxidative stress [10, 62, 92, 104, 139, 142, 164, 259, 358, 436]. Although loss of DA producing neurons is seen in both normal aging individuals and PD cases it has been hypothesized that these catecholaminergic neurons are particularly susceptible to oxidative stress [67, 68, 119].
Oxidative stress markers have been observed in SN specimens in many PD studies. Oxidative biomarkers shown to be elevated in PD brain tissue include: 4-hydroxy-2-nonenal (HNE) [437], protein carbonyls [9] and 8-hydroxyguanosine (8-OHG) [445]. Reduction and protection of remaining neurons from oxidative stress is currently the focus of the pharmaceutical industry with the aim of developing new classes of therapeutic agents for PD. In this review, reactive species (RS) will be used as a broad term of all reactive species including nitration species. In normal healthy aerobes, O2 is utilized for cellular respiration processes by the mitochondria [164]. Mitochondria are a major source of RS production as leakage of electrons from the electron transport chain are slowly accepted by O2 producing free radicals including the superoxide radical (O2•). Superoxide is chemically inactive however, if allowed, substantial production of this species can cause biological damage. The covalent bonding of O2• to nitroxide (NO) forms peroxynitrite (ONOO-) which under physiological conditions can be very reactive and can oxidize lipids [338], DNA [391] and proteins [40, 95, 257]. Superoxide can also be converted to other RS products such as H2O2 and hydroxyl radicals (OH•) which may in turn attack other macromolecules such as proteins, lipids, sugars, and polynucleotides which are susceptible to oxidative damage. The abundance of O2• and ONOO- can accelerate the oxidization of iron or copper sulfur clusters found in many proteins that require these clusters for functional activity [209, 210]. Superoxide has been demonstrated to “leach” iron from metalloenzymes, increasing the free iron content [209]. The presence of these free redox active metals can in turn accelerate RS production. Excessive accumulation of H2O2 and O2• in the presence of catalytically reduced transition metals such as iron, copper and manganese can generate a potent hydroxyl radical species OH• via Fenton chemistry (1).
| (1) |
Superoxide as well as ascorbic acid and thiols can rereduce oxidized metals via the Haber-Weiss reaction to produce OH• from H2O2. Purines and pyrmidines in DNA and RNA can be attacked by the hydroxyl radical, producing irreparable breakages and oxidized RNA products such as 8-OHG [107, 309, 356].
Radicals can remove H+ from polyunsaturated side chains found in membrane lipids. The effect of lipid peroxidation is dramatic, causing disruption of membrane fluidity allowing the leakage of molecules that normally cannot cross the membrane. Metal ions can also rapidly induce lipid peroxidation by continually removing H+ in fatty acid side chains resulting in toxic hydrocarbons and aldehydes (malonaldehydes and 4-HNE). Byproducts of lipid peroxides such as HNE have a high affinity for and inactivate integral proteins such as Ca2+ and K+ ion channels and receptors [54] and glutamate transporters leading to a greater excitatory effect and possible excitotoxicity [39, 323].
DA is highly reactive and produces RS through two pathways. In the first instance, DA can undergo oxidation in the presence of molecular oxygen to form H2O2, 6-OHDA, quinone intermediates and O2• (see Fig. 1). The generation of oxidized DA metabolites can further feed into the redox cycle, leading to the amplification of RS products eventuating in neuronal death. In vivo models have shown that DA oxidative metabolites, in particular 6-OHDA, can induce toxicity by generating RS and initiating caspase activation [11]. Secondly, DA can also be broken down by MAO-B through deamination to produce H2O2 [67]. Further redox cycling can be driven by excess O2• which reacts with both DA and metal ions to produce more RS.
Fig. (1).
An Oxidative stress model suggesting the roles of Iron (metals), α-synuclein and Dopamine (DA) in the propagation of neurondegeneration in PD (modified from [41]). The stars indicate the points on the pathways that MPAC compounds can influence the reaction.
It is inevitable that aerobic organisms produce RS. RS can be cleared under normal conditions by the anti-oxidative enzymatic activity of catalase, Copper/Zinc superoxide dismutase (Cu/Zn SOD) or glutathione (GSH) peroxidase. The importance of these enzymes has been demonstrated in transgenic and knockout mice [130, 405]. Cu/ZnSOD, catalase and GSH peroxidase transgenic mice have been tested in both the MPTP and the 6-OHDA lesion models and data demonstrate that there is increased susceptibility to oxidative stress in the absence of these protective proteins [218, 443] while over expression of these proteins result in increased protection [335]. Anti-oxidant systems are essential for defense against cellular endogenous or exogenous oxidants. A decrease in total GSH has been reported in PD post mortem SN tissue [63, 322, 365, 374]. The homeostatic balance of RS production to anti-oxidative mechanisms is shifted as the cell becomes burdened and stressed. Elevation in RS [62, 64, 119, 177, 202, 436, 437] and perturbation of anti-oxidative mechanisms ultimately leads to cellular death of individual cells and it is widely hypothesized that there is a feed forward system that leads to progressive neurodegeneration of the nucleus [12, 353]. A decrease in total GSH has been reported in PD post mortem tissue [63, 365] where the decrease has been observed specifically within the SN [63, 322, 365, 374]. Neurons seem to be more susceptible to oxidative damage as (i) neuronal membranes are rich in polyunsaturated side chains which are freely attacked increasing the fluidity of the membrane (leaky membrane), (ii) these cells express small amounts of anti-oxidant enzymes such as catalase, GSH peroxidase and Cu/ZnSOD compared to other cells in the body. Much effort has thus gone into the therapeutic intervention of these oxidative processes.
2.4.1. Metals in PD
Transition metals have been implicated in many neurological diseases such as Alzheimer’s disease (AD) [49, 274, 426], Multiple Sclerosis (MS) [242, 284] as well as PD [19, 20, 90, 92, 259, 375]. Aberrant brain metal levels have been associated with normal aging and a variety of diseases however this is still debated [19, 258].
Dopaminergic neurons seem to be highly sensitive to oxidative stress providing a potential link to environmental exposures of metals and PD susceptibility. Epidemiological literature has assessed the potential risk of developing PD when exposed to certain metals such as iron, manganese and to a lesser degree, copper. The risks reflect the potential of redox active metals such as iron and copper acting as catalysts to drive oxidative stress.
It is thought that dysregulation in metal ion homeostasis acts as a potential catalyst to further produce RS as previously mentioned. The current section further discusses how metal ions and in particular iron and manganese are associated with PD.
2.4.1.1. Iron
Iron is required for numerous critical biological processes. These processes are involved in cellular respiration pathways, acting as the central core for metallo-proteins, neurotransmission and myelination. The balance of iron content is essential, as excess iron is highly toxic to cells, seen in some neurological (PD, AD and MS) and peripheral diseases (Haemachomatosis and Friedrich’s Ataxia).
2.4.1.2. Iron Distribution in the Brain
Dynamically, the brain has the capacity to adapt to abnormal iron levels and redistribute iron to regions of high requirement. An average adult brain has approximately 60 mg of non-heme iron, with some cerebral regions such as the SN, GP, caudate nucleus and putamen retaining the highest level of iron [19, 267]. Most of the iron in brain is found specifically in neurons as iron is a co-factor for many enzymatic reactions (such as the production of DA). Tyrosine hydroxylase (TH) is a non-heme iron enzyme which uses molecular oxygen to hydroxylate tyrosine to form L-DOPA [153]. The brain acquires the critical level of iron necessary for human adult stores through breast milk [219]. At birth, very little iron is present within the developing brain. However, cerebral iron levels rapidly increase specifically during the early years of life. Approximately 0.3 mg/L of iron is found within human breast milk [118] and uptake of iron occurs within the first 12 to 18 months of human life. In rodents, the uptake of cerebral iron occurs during the 3rd week post partum [79] and during these critical developmental periods; the brain requires iron for normal neurological maturation.
The manner in which iron is transported into the brain is a complex process which is still not well understood. Circulating iron, once oxidized to its ferric state by the serum protein ceruloplasmin cannot readily cross the blood brain barrier [294]. For iron to be transferred across the BBB, the endothelial cells lining the cerebrovasculature require a transfer protein Transferrin (Tf) to which the complex (Tf–iron) binds to transferrin receptors (Tfr) found on the luminal side of the membrane. The complex enters the cells via endocytosis and is transported to various cell types within the brain such as neurons, glia and oligodendrocytes. This process is highly regulated by the abundance or the deficiency in Tfr and the Tf-iron complex. Transcriptional regulation of iron binding proteins transferrin and ferritin is controlled by iron regulatory proteins (IRP) which bind to iron responsive elements (IRE) on RNA to alter the expression levels [56, 212]. IRP2 knockout mice develop motor deficits (tremor and bradykinesia), progressive neurodegeneration and increases in ferritin levels within affected neurons [236].
Ferritin is a common iron storage protein within the brain and is expressed in microglia, oligodendrocytes and neurons [71, 190]. Ferritin possesses ferroxidase activity that catalyzes the conversion of ferrous iron to ferric iron. Ferritin acts to reduce the amount of free cytosolic iron by catalyzing iron to its non reactive or ferric state [154]. This mechanism prevents iron from being available to participate in Fenton chemistry and generating RS.
Neuromelanin (NM) is another storage protein with a high affinity for iron [441, 442] and is localized within regions of high metabolic turnover such as in DA producing neurons in the SN and the noradrenalin neurons in the locus coeruleus [42, 349]. Catecholaminergic neurons found in primates contain NM, as in humans, but are less visible and are not localized within the SN [263]. Rodents possess DA and noradrenalin neurons, but these neurons do not appear to contain NM [17]. NM is a byproduct of catecholamine metabolism and is synthesized from quinone intermediates when cytosolic DA is in excess [390]. It has been suggested that NM is a neuroprotectant, preventing degeneration of nigral neurons by binding transition metals and other DA oxidative products that are abundant in the SN. It has been suggested that in situations of high iron overload, NM retains iron or other heavy metals within dihydroxyindole groups on its chemical backbone. The functional role of NM upon its binding to heavy metals is beneficial, by sequestering and preventing any free metal ion from participating in Fenton chemistry. This role allows the SN neurons to tolerate a higher iron load than other neurons within the brain that do not possess this protein. However, during the progression of PD; the NM molecule reaches saturation. Once the NM iron-chelating capabilities are saturated the iron-saturated molecule could be available to create RS [102]. NM appears to be an important buffering molecule that is involved in DA neuron protection and possibly vulnerability.
2.4.1.3. Dysregulation in Brain Iron and Neurodegeneration
There is an overwhelming consensus that iron accumulation has a pathogenic role and this has been seen in many neurological diseases. This highlights the use of metal altering drugs as a potential therapy for PD (see later section). Iron dys-homeostasis is highly evident in post mortem PD brains [157]. The association of elevated iron found in parkinsonian brains may be linked to age related changes in redox active metals. Post mortem studies have shown that nigral cells are associated with elevated levels of both ferric and ferrous ions within the SN in severe cases of PD [90, 126]. Furthermore, iron has been shown to accumulate in the SN in animals following 6-OHDA and MPTP lesion [112]. Iron is potentially pathogenic as it is a highly redox active metal and can participate in metallo-redox reactions (as discussed in section 2.4). Changes in iron homeostasis which particularly increase the labile iron pool potentially promote neuronal toxicity by catalyzing conversion of a less reactive inert species (H2O2) to the highly reactive hydroxyl radical. Currently, it is unknown if the dysregulation in iron homeostasis may be a primary or a secondary cause of PD, however, many genetic and biochemical studies have suggested that iron accumulation may be a primary event.
Proteins that regulate iron are disrupted in PD patients [21, 90, 112]. Mutations in iron regulating or binding proteins have been reported in other iron overloading diseases with overlapping clinical features to PD such as neuroferritinopathy, Friedrichs ataxia, and haemochromatosis. Increased levels of the iron storage protein ferritin have been shown in PD patients [21, 90]. Transgenic animals that express increased ferritin levels within dopaminergic neurons in the SN have shown age-related progressive neurodegeneration, loss of axonal projections and decreased DA in the striatum together with loss of spontaneous movement [76, 206]. Gene knockout mouse models of haemochromatosis showed severe deficits in motor performance [147] attributable to the CNS.
Point mutations near the iron binding site of the TH molecule have been found in cases of L-DOPA responsive PD and Segawa’s syndrome that results in TH deficiency [152]. Furthermore, iron plays a critical role in electron transport and the metabolism of various neurotransmitters including DA, norepinephrine and GABA as well as in DA D2 receptor function [373].
Direct bolus intranigral injection of iron has shown to increase the levels of iron within the SN [25, 424] and results in severe loss of nigral cells associated with reduced DA levels in the striatum and deficits in locomotor activity. The risks of high dietary iron at a given critical vulnerable stage have been highly correlated to late onset development of PD [124, 125, 205]. During brain maturation, dietary iron induced in rat and mice neonates resulted in patterns of PD like behavior and nigral cell degeneration in adulthood [124, 125]. Excess dietary iron post partum, increases the levels of iron within the whole brain but to the greatest extent in the GP and the SN [330]. The transport of iron to the brain peaks between birth and 21 days post partum in neonatal mice, with little entering the brain after that time. Feeding a high iron diet generates characteristics of PD later in life in these animal models [125, 373]. Feeding of 20000 ppm for 12 weeks causes behavioral dysfunction and severely affects the latency in motor activity [373]. Mice that are exposed to a high iron diet during neonatal development have an increased susceptibility to oxidative stress and a reduction in TH positive cells at 24 months of age [205]. Rodents deficient in neonatal iron are deficient in brain ferritin [167], and have reduced cognitive functioning [84, 148, 330]. This evidence notwithstanding, the direct effects of iron during early human development and its contribution to a potential increase in susceptibility to PD is a controversial topic that has not been fully addressed to date.
2.4.1.4. Copper
The interrelationship with iron and copper has been discussed in brief in papers [90-92]. Few studies have investigated copper and its role is yet to be established in PD.
2.4.1.5. Manganese
Manganese toxicity, also known as manganism has been characterized in miners following long term exposure to manganese ore [187-189] and occupational studies have correlated welders with a high prevalence of parkinsonian like symptoms, such as abnormalities in gait and speech [337] following exposure to high levels of manganese from the welding rods. One clinical difference between PD and manganism is that manganism produces dystonia [16]. This difference is attributed to the GP being primarily affected in manganism [363] with only minor damage to the SN [432]. These commonalities suggest that manganese and iron affect the dopaminergic system where they accumulate within these regions. Following a 6-OHDA lesion to the SN significantly higher levels of manganese were detected in the GP, SN, amygdala, hypothalamus, and hippocampus [394]. It has also been suggested that the increase in manganese content found in the SN after a 6-OHDA lesion directly contributes to the accumulation of iron levels within the SN [394]. Like iron, manganese can also participate in redox chemistry and generate RS causing cell death [318]. Evidence demonstrating that manganese affects dopaminergic neurons via oxidative stress damage to DNA of these neurons [310] include elevation of DA oxidative metabolites [358] and induction of apoptotic pathways in cultured cells [88]. Furthermore, manganese can reduce anti-oxidants such as glutathione (GSH), catalase and thiols [87, 246].
2.4.1.6. Aluminum
The hypothetic role of aluminum and its pathogenic role seen in neurodegenerative diseases, has managed to survive in the neuroscience field in spite of ambivalent support. Aluminum in drinking water was linked to AD, ALS and PD until it was discovered that the epidemiological studies were inadequately designed and that aluminum is poorly absorbed by the digestive system [115]. The presence of aluminum is quite low within living organisms, but is highly abundant within the environment. Aluminum can be detected in foods, pharmaceutical agents (antacids) and can also be found in drinking water. Aluminum has been generally considered biologically inert. In spite of this, the toxicity of aluminum has been well established. Few epidemiological studies have made connections between aluminum and neurodegenerative diseases such as AD [138, 155, 161, 325, 332, 345], ALS and PD [26, 369]. Currently, there is no data that suggests aluminum (a trace element) is essential for bodily function. While no reliable measurement of total body aluminum content is available, the body’s acquisition of aluminum is largely by ingestion [292, 396, 397]. Microdialysis studies have shown that aluminum can cross the blood brain barrier via the aid of a transporter mechanism which included an iron carrier molecule transferrin [293]. Historically there has been concern, concentrations of aluminum in drinking water have been investigated as a potential factor to give rise to AD [115, 138, 345, 379]. Whilst there is evidence which evinced no relationship [268, 425], a considerable number of studies have attempted to defined this link [345]. There is pathological data showing elevated aluminum in the spinal cord and the hippocampus from ALS and PD with dementia [137, 216, 326, 329] and in tissues from PD patients [151, 180]. However the elevation in aluminum seen in these diseases may not be related to dietary intake.
The biological effects of artificially increased aluminum in experimental animals [211, 216, 379] is to cause pathological changes; including neurofibrillary tangles which closely resemble those in AD, and neuronal loss [137, 211, 216, 224, 325, 406]. Interestingly, these aluminum treated animals showed signs of loss in motor functions found in the hind limbs [216] which suggests that aluminum affects other modalities in the CNS. Together with iron, aluminum has been reported to be significantly concentrated in melanized granules within the SN [151, 180]. The presence of aluminum and iron accelerate the formation of lipid peroxides [159] and thus act in a synergistic fashion. Aluminum can participate in chemistry that induces changes in membrane fluidity that can facilitate lipid peroxidation [283]. While aluminum is redox inactive, a 3 week exposure of rats to aluminum salts induced increased levels of RS in cortical regions [43]. The speculation that aluminum can potentiate and generate a pro-oxidative environment has been supported by studies suggesting that aluminum can trigger inflammation responses [51]. Interestingly, Mendez-Alvarez and colleagues (2002) found that in a 6-OHDA animal model, the presence of aluminum reduced the OH• production and attenuated the neurotoxic effects of the 6-OHDA lesion [285] by the prevention of lipid peroxide formation. It was concluded that the effects of aluminum accumulation could be by binding to 6-OHDA preventing the interaction between 6-OHDA and hydrogen peroxide [285], thereby reducing the toxic effects of the 6-OHDA.
Aluminum toxicity has recently been revisited because of its effect in vitro and in animal models but there is little evidence on aluminum has any involvement in PD and it is therefore is not considered as a therapeutic target at this stage.
2.5. Integration of Current Models for Therapeutics
Dopaminergic neurons appear to be generally under high oxidative loads and are thus suspected of being susceptible to neurodegeneration. Iron, DA and α-synuclein, all co-localize to the SN and these factors are implicated in the etiology of PD. However it has been substantiated that none of these elements alone is sufficient to cause the observed chronic neurodegeneration. Elevated iron and DA in the presence of increased or altered metabolism of α-synuclein, may act synergistically to propagate a series of reactions that result in destruction of SN neurons. To address potential new pharmacological intervention therapies for drug development, the observed changes in metabolism that result in PD must be considered.
The highly favorable oxidative stress environment for DA interaction with α-synuclein and iron resulting in RS-mediated toxicity and protein aggregation is one of the most likely mechanistic explanations for PD (refer to Fig. (1)). Singular constituents of this model such as DA, α-synuclein or iron alone are not capable of emulating PD-like neurodegeneration. In this section of the review several therapeutic targets are highlighted and discussed.
DA has been implicated to play a role in the neurodegenerative cascade in PD as it is susceptible to oxidation (discussed in section 2.4). Following a partial lesion of the dopaminergic SN neurons the remaining neurons compensate by increasing the amount of DA produced as well as increasing the rate of DA turnover [383]. In surviving neurons, the DA turnover rate is increased by the deamination of DA by MAO-B thus increasing the production of H2O2 and oxidative stress metabolites [67]. DA is readily auto-oxidized through metallo-redox reactions to produce the neurotoxin 6-OHDA and further oxidized into quinone intermediates to generate O2• [202] (See Fig. 1). As 6-OHDA is analogous to catecholamine neurotransmitters, it may be taken up by the corresponding reuptake transporters [335] resulting in rapid loss of nigral neurons by activation of caspase pathways [99]. Direct injection of 6-OHDA into the SN or the medial forebrain bundle is widely used in animal models to induce parkinsonism [131, 194, 439].
Ferrous ions catalyze the formation of oxidized DA, generating free radicals but also converting oxidative metabolites such as 6-OHDA into melanin [103]. Manganese can also participate in the oxidation of DA leading to the generation of RS [310] which implements further redox (active or inactive) metals that can be substituted in this reaction.
The familial A53T α-synuclein mutant raises cytosolic DA which increases the available substrates for this cascade [203, 254, 296]. Over-expression of DA metabolic genes such as α-synuclein accelerate the rate of DA re-uptake via DAT therefore increasing DA vulnerability [203, 392], further RS production and DA induced apoptosis [392].
α-Synuclein mRNA expression has been seen to be only elevated within the SN in later stages of PD [61, 158, 414]. This effect could be due to the relationship between DA levels and α-synuclein that facilitate the transmission of DA. Synaptic regulation is shown to be severely affected when α-synuclein expression is altered [412]. In the presence of α-synuclein, DA-α-synuclein adducts form through the stabilization of protofibrillar structures [73]. This is exacerbated by the presence of iron which also triggers aggregation of α-synuclein by changing the protein conformation structure from helices into an unfolded beta-sheet structure [313]. The fibrillated protein is the major component in LB [154]. Formation of protein aggregates is likely to be by the direct association between metals and α-synuclein.
It is interesting to note that the familial mutations of α-synuclein are localized within the metal binding site of α-synuclein and each of these mutations affect metal interactions with the protein [29]. A metal responsive element has been demonstrated in the promoter located within the 5’ Untranslated Region (UTR) of the α-synuclein mRNA transcript [129]. This suggests that α-synuclein may be a metallo-protein which can be manipulated to help treat PD.
Metal ions in the presence of DA directly cause α-synuclein protein aggregates [53] and modifications of α-synuclein protein occur in the presence of RS generated by Fenton chemistry. Post translational modifications such as phosphorylation, glycosylation, oxidation and nitration of α-synuclein promote protein aggregation [139]. Oxidatively modified α-synuclein and aggregates stabilized through the dityrosine cross-linkages are observed in LB [378]. Through further oxidization of α-synuclein, sequential oligomerization is enhanced by the presence of copper [315]. Nitration of α-synuclein promotes the formation of high order oligomerization [303] which may perforate vesicular membranes such as DA storage synaptic vesicles, resulting in leakage of DA into the cytoplasm [255]. In addition, aggregated α-synuclein may over-stimulate TH activity causing overproduction of DA, propagating a feed forward degenerative cascade [53, 324]. α-synuclein mutants as well as increased levels of normal α-synuclein generate RS [239], which is accelerated in the presence of DA and increases susceptibility of cells to oxidative stress [203].
Mutated α-synuclein has a greater propensity to polymerize in vitro and α-synuclein transgenic mice develop age-dependent intracellular α-synuclein inclusions within aberrant areas of the CNS. Further, the expression patterns of mutated human α-synuclein and the location of inclusions, in the various transgenic mouse models, appear to be influenced by the different promoters utilized [270]. Wild type α-synuclein has a half life of 48 hours while the A53T mutation has an approximately 50% longer half life [245] suggesting that these mutant proteins are selectively preventing degradation via the lysosome-mediated pathway [78].
Oxidative stress (e.g. from mitochondrial inefficiency, anti-oxidant depletion, or transition metal perturbation) provides conditions under which DA may interact aberrantly with α-synuclein and iron resulting in the generation of H2O2, oxidation of DA to toxic intermediates such as 6-OHDA, and aggregation of α-synuclein. A self-propagating cascade is engendered as oxidative products of these reactions create conditions for further local elevation of DA and generation of metal mediated RS. Therefore subsequent sections discuss how intervening therapies which target these interactions may be capable of modulating the disease.
3. CURRENT TRENDS IN THERAPEUTICS
Alleviation of parkinsonian symptoms and functional disability is the principal goal of PD management in clinical practice. Most patients in early stages of idiopathic PD will improve in response to medications that are directed at correction of the hypo-dopaminergic biochemical deficit and enhancement of dopaminergic neurotransmission. This approach constitutes symptomatic therapy of PD, but the majority of PD patients will gradually deteriorate. It is thought that an ongoing apoptotic death of dopaminergic neurons in SN underpins this relentless natural history of PD. Presynaptic dopaminergic terminals in the basal ganglia release vesicular DA on demand and also carry out DA reuptake via the DAT system. Dopaminergic neurons of the SN receive innervation from the basal ganglia, thus creating a complex feedback loop. This illustrates the role of dopaminergic neurons in biochemical processes and also emphasizes that dopaminergic neurons are intimately incorporated into neural circuits. Neuroprotective therapy sets out to rescue the apoptotic dopaminergic neurons in SN. Neuroprotective therapy still remains mostly an experimental approach, but putative neuroprotective drugs may alter the relentless course of PD. Detailed synopsis of current symptomatic treatment is out of the scope of this review and is dealt with elsewhere [185, 186]. In this section we summarize only the current most common therapeutic pharmacological strategies and focus on some experimental neuroprotective therapies currently undergoing human trial.
3.1. Symptomatic Therapy
Levodopa
At present, Levodopa (L-DOPA or 3, 4-dihydroxy-L-phenylalanine) is the most useful drug for symptomatic treatment of idiopathic PD. Unlike DA, L-DOPA crosses the BBB. After oral administration L-DOPA is taken up by the dopaminergic neurons and converted into DA by the enzyme Aromatic Amino Acid Decarboxylase (AADC).
L-DOPA effectively alleviates PD symptoms in the early stages of disease. The current “storage hypothesis” holds that at this stage of PD the available dopaminergic neurons and pre-synaptic dopaminergic terminals maintain the capacity to process exogenous L-DOPA and carry out physiological handling of synthesized DA [183, 304-306]. It has been suggested that the benefits of L-DOPA wear off with disease progression and ongoing death of dopaminergic neurons [238]. This view may be misleading due to the inability to discriminate against the treatment effects and the natural progression of the disease. According to the “storage hypothesis”, in the absence of dopaminergic neurons L-DOPA is metabolized into DA by neural cells that lack “dopaminergic machinery”. As a result DA release becomes pulsatile rather than continuous and eventually leads to post-synaptic changes and development of motor complications [5, 55, 429].
At present there is some evidence that L-DOPA can be neuroprotective to dopaminergic neurons. The Early versus Late Levodopa study (ELLDOPA) indicates some neuroprotection all be it with diminishes striatal innervation [111]. The DATATOP study also suggested that L-DOPA slowed the rate of disease progression [1, 57, 183]. In contrast, in vitro experiments suggest that L-DOPA accelerates degeneration of residual dopaminergic neurons through enhanced oxidative stress. However, L-DOPA was not toxic to dopaminergic neurons in vivo in experimental rodents. Recent human trials presented unequivocal evidence that L-DOPA treatment did not cause clinical deterioration over a period of 40 weeks compared to the placebo [69, 110, 229, 298, 385, 418]. However, the potential long-term effects of L-DOPA on dopaminergic neurons remain unclear.
Direct Agonists of Dopaminergic Receptors (or Dopamine Agonists)
The rationale for developing this class of drugs was the delivery of continuous stimulation of dopaminergic receptors, thought necessary to prevent development of motor fluctuations in long-term. This approach was put forward as an alternative to L-DOPA treatment, based on the hypothesis that L-DOPA treatment set pulsatile stimulation of postsynaptic dopaminergic receptors and promoted development of motor fluctuations.
Numerous in vitro and in vivo laboratory studies have shown neuroprotective potential of dopaminergic agonists that can be mediated via several mechanisms including free radical scavenging [149, 221], and anti-oxidative properties [352, 453].
Data from human trials are not conclusive as to neuroprotective properties of DA agonists in PD patients, chiefly because it is very difficult to discriminate between symptomatic and putative neuroprotective effects in the settings of clinical trials and requires a sophisticated approach to the design and analysis of the study. However, current experience with PD patients suggests that the impact of direct DA agonists on the natural course of PD may not be of a clinically meaningful magnitude [6].
3.2. Drugs with Dual, Symptomatic and Neuroprotective Effect
MAO-B Inhibitors
There are currently two selective irreversible MAO-B inhibitor drugs approved for clinical use, rasagiline (Azilect) and selegiline (Deprenyl). Two isoforms of MAO have been identified, A and B. In the human brain, MAO-B is the predominant isoform responsible for the breakdown of DA. Selective inhibition of MAO-B results in the elevation of synaptosomal DA concentrations. The primary rationale for MAO-B inhibition in PD is enhancement of striatal DA through inhibition of DA metabolism and the role of MAO-B inhibitors in symptomatic treatment of PD has been well established [317].
Interestingly, both selegiline and rasagiline possess potent neuroprotective and anti-apoptotic properties that are not related to MAO-B inhibition. This effect has been demonstrated in vitro using primary cultures of cortical neurons; both drugs enhanced survival of dopaminergic neurons. Neuroprotection has been demonstrated in vivo in rodent models of Parkinson’s disease. It has been proposed that stabilization of mitochondrial membranes, enhancement of intracellular anti-oxidant systems and induction of pro-survival genes underlies this effect (for review, see www.rasagiline.com).
Recently, the interest in neuroprotective properties of MAO-B inhibitors has been sparked by the study showing that Selegiline slows progression of PD symptoms by about 35% over 5 years [317]. This suggests that there may be neuroprotective effects on the nigro-striatal system. Rasagiline has only recently gained FDA approval, but preliminary results have been promising [366].
3.3. Cell Based Therapies
The neuropathological and neurochemical alterations of the dopaminergic nigro-striatal system are responsible for the major symptoms of PD (see above section). This constitutes the premise of DA cell-replacement therapy, whereby introducing DA-producing cells into the parkinsonian brain might replenish the diminishing levels of DA and alleviate or cure PD.
Over the last 20 years there has been an enormous research effort in this field of neuroscience. Swedish neuroscientists pioneered transplantation experiments in the mid-70’s and early 80’s [33, 36, 108]. In early transplantation experiments the grafts of DA-producing cells were placed into the striatum because this approach yielded best survival of the grafted cells with subsequent dopaminergic reinner-vation of the basal ganglia [31, 32, 34, 35, 106, 133]. In parallel, the demand grew for the sources of DA-producing cells. Traditionally, fetal ventral mesencephalic tissue has been used for grafting because this region of the developing brain contains precursors of dopaminergic cells which differentiate into functional DA-producing cells in vivo. Ethical issues essentially preclude large-scale use of the fetal-derived ventral mesencephalic tissue. An alternative approach has been developed, whereby embryonic stem cells or committed neural precursors can undergo directed in vitro differentiation into DA-producing cells, these are then harvested and used for transplantation [37, 248, 346, 387].
Several groups in Europe reported that PD symptoms improved following grafting of the fetal mesencephalic tissue into the putamen or head of caudate area of PD patients [162, 163]. Based on the promising preliminary results, NIH funded the first prospective, double-blind, placebo-controlled trial in which 40 PD patients received fetal mesencephalic transplants or placebo operations [126]. The functional improvement of participants was assessed 12 months following grafting using the Unified Parkinson’s Disease Rating Scale (UPDRS). Fetal mesencephalic transplants induced statistically significant improvement in a cohort of patients under the age of 60. Long-term follow-up of the participants revealed five patients who underwent transplantation developed dystonia and dyskinesia. Another prospective, 24-month, double-blind, placebo-controlled trial of human fetal nigral transplantation [311] failed to detect significant differences between grafted and placebo groups. The incidence of dyskinesias was high in this study affecting almost half of the patients that had received mesencephalic transplants.
The two human trials are commonly designated as “proof-of-concept” studies and the negative result are perceived as compromising the entire concept of the cell-replacement approach. However, several factors have been identified that confound interpretation of the negative results of human transplantation trials and require further clarification: surgical technique needs improvement; non-dopaminergic cells within fetal tissue transplants also have been implicated in post-surgical complications [30, 127, 128]. The current mainstream of research is directed at producing a reliable and standardized population of DA-producing cells such as neural progenitor cells (NPC) that can be used for further transplantation trials.
3.4. Neurosurgical Therapies
Neurosurgical interventions have developed symptomatic treatments for motor related disorders particularly for advanced PD patients with ensuing dyskinesias.
With the increasing knowledge of the neuroanatomical circuitry, surgical treatments such as precision surgical ablation (pallidotomy and thalamotomy) and Deep Brain Stimulation (DBS) are favorable procedures due to the shortcomings of pharmacological therapies. Surgical ablation therapy has been used in many instances until the late 1990’s to reduce severe behavioral symptoms such as bradykinesia, dyskinesia, and rigidity and to some extent resting tremor. Targets for functional neurosurgery include the ventral intermediate nucleus (ViN), STN or the internal Globus Pallidus (GPi). It is though that the reduction of GPi activity through ablative surgery rebalances the inhibitory effect of the abundance in striatal GABA due to the loss of DA production.
In many models [174] and also human parkinsonism, STN and GPi ablation have shown to improve behavioral outcomes associated with the disease. The procedure itself is irreversible, with serious complications that could lead to permanent disability particularly impairment of speech and visual modalities.
Unilateral pallidotomies are still preformed today without any knowledge of the long term effects of the surgery. While results of bilateral GPi lesions are indicative of a reduction in dyskinesia [81], there is an association with increased risk of inherent adverse side effects [81]. Patients with unilateral subthalamic lesions showed pronounced improvement particularly after surgery [321]. However, efficacy was limited in that tremors would reoccur in about 20 % of cases. A current alternative surgical treatment, high frequency DBS, has replaced stereotaxic lesioning. Chronic high frequency stimulation of the STN (via ViM) in a pilot study in 1987, yielded some promising results by a reduction in extra pyramidal side effects [24]. This new treatment paved the way for a new type of functional motor disorder neurosurgery without subsequent adverse side effects associated with surgical ablation. In PD, the motor deficits are attributed to increased neuronal activity within the STN and the GPi. The surgery involves an insertion of an electrode attached to a neurostimulator. The neurostimulator sends out electrical signals that modulate neuronal circuitry in target areas in the brain to inhibit the impulses that give rise to motor dysfunction. The electrode is placed into a region to address a particular motor symptom accordingly. DBS is proven to be an efficacious treatment in studies that target regions of both the STN and GPi. These studies revealed a reduction in symptoms such as tremor, bradykinesia and rigidity [217, 388]. The stimulation of either the STN [226] or the GPi [2] resulted in significant improvements in UPDRS motor scales.
The mechanisms of STN-DBS and GPi-DBS are paradoxical and still remain unknown. It was hypothesized that electrical stimulation of the STN and GPi would suppress these structural inputs [101] and would therefore act as a counteractive measure of DA replacement therapy. Conversely, Stefani and colleagues (2005) [384] questioned this mechanism suggesting that STN-DBS increases GPi firing rate and synchronizes the STN activity. In a microdialysis model of PD, cGMP was used as a measure of glutamate transmission and was found to be increased by 6 fold in GPi dialysate [384].
Concurrent electrical stimulation is a reversible procedure with fewer surgical complications compared to its lesioning counterpart. Follow-up studies of bilateral STN-DBS patients showed improvement in motor symptoms [109] suggesting that DBS is a beneficial long term treatment. The DBS is a useful procedure because it allows the stimulation of an affected region without further destruction of brain tissue [160]. At the same time, long term stimulation of the STN could aid in slowing the disease progression. However, this is yet to be confirmed since Hilker and colleagues (2005) [179] established that bilateral STN stimulation did not alter the rate of disease progression.
4. NEW THERAPEUTIC STRATEGIES
Current therapeutics for PD is neither curative nor preventative as they only temporarily alleviate some of the symptoms of the disease. Drug intervention needs to aim at halting the progression of PD. Current treatments of PD are successfully improving quality of life but unfortunately largely without the ability to control or reduce the rate of disease progression. An integrative model that combines the putative fundamental aspects of nigral degeneration is needed for appropriate therapeutic targeting to potentially prevent further DA nigral loss.
4.1. Gene Therapy
With the current knowledge of molecular characterization of vital genes involved in the neurodegenerative process, several research groups have embarked on using gene therapy to help protect and also repair neuronal damage. Deliverance of protein products is difficult as the BBB limits the transfer to the intended destinations. Genetic manipulation has many advantageous applications with many vehicles aiding in the delivery of the gene target such as viral (lentivirus, adenovirus and herpes virus) or non viral (polyplexes) that can infect cells without inducing inflammatory responses and has the ability to affect both dividing and non dividing cells [105, 277]. Furthermore, the regulatory control of an element that allows the expression of the gene is the primary mechanism for genetic manipulation. Whilst many genes have been uncovered acting as potential ‘players’ in the degenerative cascade, this has allowed the makings of in vivo gene therapy promising a new future treatment for PD. The conceptual difficulty in this approach comes from the unknown cause of sporadic PD [400]. Once the cause has been identified, gene therapy may then take greater prominence. While this interventional approach is still a new concept, only a few genes have been trialed in animal models of PD. These gene targets include α-synuclein [123, 171, 239, 350, 451] and Parkin [192, 251, 362, 434].
The use of various neurotrophins in support of the nigral neurons has proved effective in various animal models [59, 70, 225]. The Glial cell line-Derived Neurotrophic Factor (GDNF) has shown the propensity to increase the rate of DA neuronal survival under neurotoxic cell culture conditions [48, 65] and in animal models [59, 70]. Reports of GDNF therapy delivered with an adenovirus have been used in animal models and it was found that GDNF can rescue DA cell loss if administered prior to or shortly after delivery of 6-OHDA [225, 372] or MPTP [222]. Significant improvement in motor behavior is a reflection in significant DA cell recovery of function [421, 450] and correlated with a higher level of DA production in the striatum [421]. These higher levels of DA may occur because of increased TH expression [371]. The results from the various clinical trials have not been so definitive. Lang and colleagues (2006) [232], showed that there was no significant clinical benefit in the UPDRS in a phase II trial that investigated the effect of intra-putamen infusion of GDNF, yet similar studies have shown significant progressive improvements in open-label designed trials [141, 370]. It has been debated that Lang and colleagues (2006) did not adequately take into account the placebo effect, the catheter design or the rate of delivery of GDNF [232]. Further, recalculation of the statistical power showed that the power of the study was unable to investigate the effects of GDNF in PD [191]. In light of these difficulties, GDNF properties of promoting cell survival have not been adequately tested in trial for neuroprotection therapy as this would require a longer period of evaluation and sophisticated study design.
Preliminary data using gene therapy to target the STN instead the SN has showed some promise as a therapy in PD. Utilizing a viral expression system, an enzyme (glutamic acid decarboxylase) that synthesizes a neurotransmitter (GABA) was surgically introduced into the STN of patients with PD [113]. The rationale of targeting the STN instead of the SN (which is a primary target seen in many gene therapy trials) was to functionally increase the production of GABA to decrease the aberrant increase in signals to the thalamus [113]. Significant improvement was reported clinically with the functional restoration of circuitry and improvements in motor behavior. Whilst successful in a pilot phase, the question of the placebo effect seen in many surgeries has not been adequately addressed [82]. The clinical presentation of PD symptoms occurs when at least 70% of nigral cells are lost. Goals of therapeutic interventions must therefore address the recovery and prevent the progressive nature of neuronal death in the SN. GDNF partially fulfils these criteria and is therefore an interesting target. However, many factors need to be considered: i) Regulated controlled delivery of gene products [223]. ii) Transfection is notoriously difficult and inefficient in neuronal cultured systems [422] and growing concerns of random integration posing a risk of insertion mutagenesis. iii) Adverse immune reaction [271] and iv) How chronic delivery of these “foreign” genetic products will be restricted to the correct brain region. Gene therapy still has a promising future and remains in an experimental stage. These factors need to be carefully tested before its emergence as an effective therapy for PD.
4.2. Anti-Oxidant Based Therapies
Over the last decade, neuroprotective approaches for PD have been tried in an attempt to slow the rate of disease progression. There have been a number of intervention strategies focusing on decreasing oxidative stress. Anti-oxidants can be naturally found in the diet in the form of vitamins (A, C and E), polyphenols, flavonoids and carotenoids. Interestingly, reports of dietary intake (such as high intakes of saturated fats and cholesterols) could possibly influence the susceptibility of developing PD [172, 173, 201, 253]. Dietary anti-oxidants can be found highly in fruits, vegetables, green/ black teas and red wine [144, 289], it appears that moderate ingestion of these foods results in a reduction in risk of PD [144, 252, 353]. The properties of anti-oxidants possesses is the ability to scavenge for free radicals such as the hydroxyl and the O2• radical [52, 89, 94, 165, 348, 398]. RS damage can be prevented by selected flavonoids and related phenols (polyphenols) by directly inhibiting both the formation of RS [52, 165, 398] and they enzymes that produce them [140]. A lot of research to date has focused on the properties of phenols found in tea extracts [348]. These potent anti-oxidants have shown to attenuate the toxic effects of 6-OHDA both in cultured PC12 cells [244] and an animal model of PD [243].
The protection with the use of carotenoids and both vitamin C (ascorbate) and E (α-tocopherol) has been seen in cellular based models of oxidative stress [367]. However these findings are inconsistent with the data produced by epidemiological studies. The use of vitamin supplements has been assessed in large cohort studies and found that there was no association with reduced risks of developing PD [447]. Among individuals who have a high intake of foods that were rich in vitamin E showed significant reduction in the associated risks [83, 136, 144]. Other studies had not found this association [12, 353].
Clinical based trials have investigated the use of anti-oxidants in PD patients. One of the first of its kind Deprenyl, and α-tocopherol Anti-oxidant Therapy of Parkinsonism (DATATOP) evaluated the use of these agents in a controlled clinical trial setting [1]. The study revealed that α-tocopherol did not benefit in slowing down or reducing the severity of symptoms of PD. This result has been suggested to be a cause of slow absorption and poor penetration into the CNS [1]. Animal models of PD using MPTP evinced conflicting views on the effects of vitamin E [150, 340], vitamin C and carotenoids [327, 328, 417]. Vitamin E deficient mice have an increased susceptibility to MPTP which severely affected the SN [307]. Dietary intake of vitamin E, C and carotenoids in the form of some foods remains consistent over a lifetime, and should be regarded and used in a staple diet from an earlier age. The supplementations of these vitamins are yet to be convincing as a therapy to be used at a clinical level.
A potential new anti-oxidant agent coenzyme Q10 seemingly has some promise as a therapy in mitochondrial disorders and neurodegenerative diseases. Improvement in patients with mitochondrial defects is seen biochemically and clinically with coenzyme Q10 treatment [46, 47, 308]. In neurological diseases that show mitochondrial deficit as a clinical and pathological feature, treatment with coenzyme Q10 could be of benefit. Serving as a potent anti-oxidant, coenzyme Q10 is a lipid soluble molecule which sits in the inner membrane of mitochondria and transfers electrons in the electron transport chain [404]. The anti-oxidative properties involve the ability to scavenge and inhibit the formation of RS [120, 302, 376]. The neuroprotective effects of coenzyme Q10 is seen in many models of neurotoxicity such as rotenone [291] and MPTP [22, 66]. These studies revealed that in animal lesion models, coenzyme Q10 significantly protected against; loss of TH positive cells in the SN; the depletion of striatal DA and the prevention in the formation of α-synuclein aggregates [66].
Phase II PD clinical trial showed a reduction (44%) in motor deficits measured by UPDRS using a maximal dose of 1200mg (per day) [364]. Storch and colleagues (2007) [386] attempted to replicate the earlier study with a withdrawal phase using participants with middle stage PD in a more rigorous study design. The treatment with coenzyme Q10 in this study showed no significant motor improvements at a dose of 300mg a day [386]. It was concluded that dosage is not sufficient enough to have a symptomatic effect at this stage of the disease. Further trials need to explore the protective effects in PD using the anti-oxidant coenzyme Q10 at a high dosage and for an extended period of time.
4.3. Therapeutics that Focus on Metals
There has been substantial research into pharmacological interventions that are involved in the modulation of biometals in neurodegenerative disorders. Iron dysregulation seems to play a vital role in disease pathogenesis in PD patients. These disruptions in the iron homeostatic mechanism observed in PD offer the potential for future therapeutic intervention. Controlling the bioavailability of metals could prevent not only the generation of RS through metallo-redox reactions but also the interaction with other known ‘culprit contenders’ such as α-synuclein (as shown in Fig. 1).
4.3.1. Therapies which Target Metal-Associated Proteins
As discussed previously in section 2.4.1, ferritin is a protein that regulates iron storage and can potentially remove any free redox active iron that is present within the cell. Transgenic mice that express high levels of H-Ferritin have been shown to effectively protect the further loss of nigral cells in MPTP [207] and paraquat [275] animal models of PD. H-Ferritin possesses a modifying oxidase activity, which sequesters the iron and converts it to the less bioreactive form [205, 207]. This increase in ferroxidase activity reduces the free iron pool preventing its further participation in redox chemistry [207].
4.3.2. Metal Chelation Therapies
The primary mechanism of chelators is to chemically bind metal ions to form complexes rendering the ions less reactive and allow removal of these ions via the bloodstream for excretion. Pharmacological chelators such as desferroxamine have shown some promise in modulating metal ions. In vitro studies have shown that Desferal intervenes in mitochondrial inhibition by directly enhancing the activation of NADH dehydrogenase [438]. Unfortunately, desferroxamine has poor penetration through the BBB [448]. New iron chelators such as VK-28 were synthesized in order to overcome potential barrier impermeability. VK-28 has been shown to protect nigral cells against 6-OHDA induced lesions [357].
In an induced proteasome dysfunction animal model, chelation therapy with desferroxamine reduced the inhibitory effects on proteasome inhibitors [448]. Iron potentially acerbates the rapid formation of the α-synuclein structure to promote high molecular weight insoluble aggregates (see section 2.4.1). Proteasome dysfunction in this model of PD is relieved by sequestering iron to prevent this aggregation from occurring and protect the nigral cells [448]. Pyridoxal Isonicotinoyl Hyrdazone (PIH) and 2-Pyridylcarboxaldehyde isonicotinoyl Hydrazone (PCIH) share similar potency to desferroxamine, possess high and potent chelating activity, have the ability to cross the BBB and are highly specific for iron overload diseases [178, 341]. PIH and its analogues seem to act in a dose-dependant manner in the immobilization of iron from ferritin and allowing excretion [433].
The complexity of parkinsonism has been highlighted and discussed in this review. An observation of this complexity has lead biochemists to develop bifunctional compounds which have two modes of action. Only recently, new compounds have emerged based on known MAO-B inhibitors. These have been synthesized to possess both neuroprotective effects and iron chelating properties [449]. M30, which has similar structure to that of VK-28, is a hydroxyquinoline which acts as both a selective inhibitor of MAO-A and MAO-B with chelating capabilities similar to that of desferroxamine [15, 134]. Both in vivo and in vitro models have shown promising effects to both increase levels of DA and prevent further MPTP toxicity [15, 134, 135, 449]. While there is potential to synthesize more ‘multi-functional’ compounds, however there is a need to develop a more clinical based approach and evaluate the effects of these drugs both at a physical and biochemical level.
4.3.3. Metal Protein Attenuating Compounds (MPAC)
Metal protein attenuating compounds (MPAC) may offer future therapies for PD. Clioquinol (5-chloro-7-iodo-8-hydroxyquinoline, CQ) is the prototype MPAC and acts by competing with proteins for metal ions [100]. Clioquinol (CQ) is an orally bioavailable drug with moderate affinity for copper, zinc and iron. Differing from traditional chelators as mentioned above, these compounds do not remove metals from tissues. CQ appears to act as an ionophore to redistribute metals from areas of superabundance to those which may be deficient. Unlike traditional chelators such as EDTA, CQ does not cause bulk excretion of metals but permeates the BBB and potently inhibits metal-mediated hydrogen peroxide production [18]. CQ and analogues are being investigated in a number of conditions in which oxidative stress is a feature. These include; cancer [97, 98] stroke [272], AD [336, 343] and PD [207].
This type of therapeutic approach using CQ type MPACs appears to be encouraging for AD. Animal trials with 21 month old transgenic mice over expressing the amyloid precursor protein (APP) with the Swedish mutation showed a significant reduction in Aβ plaques after treatment with CQ [60]. A phase II double-blinded clinical trial showed that CQ treatment for 36 weeks resulted in a reduction of Aβ1-42 in plasma, with minimal cognitive decline [343]. While this study had a very small subset which reflected within the non significant difference between the groups, these results support the idea that metals play an important role in neurological diseases.
There is also proof of concept that MPACs may be useful for PD therapeutics. A parkinsonian animal model study showed that treatment of animals with CQ for eight weeks prior to induction of lesions resulted in 50% decrease in nigral cell loss compared to animals treated with the parkinsonian toxin MPTP alone [207]. An 8 week pretreatment of CQ resulted in reduction in iron within the SN in MPTP lesioned mice [146]. More recently data showing that CQ treatment commencing only 6 hours after induction of the lesion is equally effective at attenuating SN lesions provoked by intra-nigral injection of 6-OHDA [428] Further the data from a neuronal cell line that expresses the A30P mutant human α-synuclein was rescued by either catalase or CQ [428].
CONCLUSION
Understanding the causes and the pathology of PD is pivotal in identifying specific targets for drug intervention. Surgical interventions have a limited niche in the treatment of symptoms of PD, while other restorative measures (such as gene therapy) are still experimental and pre-clinical. The challenge is to develop novel therapeutic agents which are capable of slowing SN cell loss and reducing disease progression. Recent studies into new classes of drugs (such as MPACs) have shown that they may intervene with interactions between dopamine, alpha synuclein and redox-active metals. This class of therapeutic drug offers a new pharmacological approach that could potentially modify the progression of PD.
ACKNOWLEDGEMENT
This work is funded by The National Health and Medical Research Council of Australia (NHMRC), the Australian Brain Foundation, Supported by Operational Infrastructure Support (OIS) from the Victorian State Government and Prana Biotechnology.
REFERENCES
- 1.Parkinson’s Disease Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson's disease. The Parkinson Study Group. N. Engl. J. Med. 1993;328:176–183. doi: 10.1056/NEJM199301213280305. [DOI] [PubMed] [Google Scholar]
- 2.Parkinson’s Disease Study Group. Deep-brain stimulation of the subthalamic nucleus or the pars interna of the globus pallidus in Parkinson's disease. N. Engl. J. Med. 2001;345:956–963. doi: 10.1056/NEJMoa000827. [DOI] [PubMed] [Google Scholar]
- 3.Abou-Sleiman PM, Healy DG, Quinn N, Lees AJ, Wood NW. The role of pathogenic DJ-1 mutations in Parkinson's disease. Ann. Neurol. 2003;54:283–286. doi: 10.1002/ana.10675. [DOI] [PubMed] [Google Scholar]
- 4.Abou-Sleiman PM, Muqit MM, McDonald NQ, Yang YX, Gandhi S, Healy DG, Harvey K, Harvey RJ, Deas E, Bhatia K, Quinn N, Lees A, Latchman DS, Wood NW. A heterozygous effect for PINK1 mutations in Parkinson's disease? Ann. Neurol. 2006;60:414–419. doi: 10.1002/ana.20960. [DOI] [PubMed] [Google Scholar]
- 5.Agid Y, Bonnet AM, Ruberg M, Javoy-Agid F. Pathophysiology of L-dopa-induced abnormal involuntary movements. Psychopharmacol. Suppl. 1985;2:145–159. doi: 10.1007/978-3-642-70140-5_19. [DOI] [PubMed] [Google Scholar]
- 6.Ahlskog JE. Parkinson's disease: is the initial treatment established? Curr. Neurol. Neurosci. Rep. 2003;3:289–295. doi: 10.1007/s11910-003-0005-1. [DOI] [PubMed] [Google Scholar]
- 7.Alam M, Schmidt WJ. Rotenone destroys dopaminergic neurons and induces parkinsonian symptoms in rats. Behav. Brain Res. 2002;136:317–324. doi: 10.1016/s0166-4328(02)00180-8. [DOI] [PubMed] [Google Scholar]
- 8.Alam M, Schmidt WJ. L-DOPA reverses the hypokinetic behaviour and rigidity in rotenone-treated rats. Behav. Brain Res. 2004;153:439–446. doi: 10.1016/j.bbr.2003.12.021. [DOI] [PubMed] [Google Scholar]
- 9.Alam ZI, Daniel SE, Lees AJ, Marsden DC, Jenner P, Halliwell B. A generalised increase in protein carbonyls in the brain in Parkinson's but not incidental Lewy body disease. J. Neurochem. 1997;69:1326–1329. doi: 10.1046/j.1471-4159.1997.69031326.x. [DOI] [PubMed] [Google Scholar]
- 10.Alam ZI, Jenner A, Daniel SE, Lees AJ, Cairns N, Marsden CD, Jenner P, Halliwell B. Oxidative DNA damage in the parkinsonian brain: an apparent selective increase in 8-hydroxyguanine levels in substantia nigra. J. Neurochem. 1997;69:1196–1203. doi: 10.1046/j.1471-4159.1997.69031196.x. [DOI] [PubMed] [Google Scholar]
- 11.Alves Da Costa C, Paitel E, Vincent B, Checler F. Alpha-synuclein lowers p53-dependent apoptotic response of neuronal cells. Abolishment by 6-hydroxydopamine and implication for Parkinson's disease. J. Biol. Chem. 2002;277:50980–50984. doi: 10.1074/jbc.M207825200. [DOI] [PubMed] [Google Scholar]
- 12.Anderson C, Checkoway H, Franklin GM, Beresford S, Smith-Weller T, Swanson PD. Dietary factors in Parkinson's disease: the role of food groups and specific foods. Mov. Disord. 1999;14:21–27. doi: 10.1002/1531-8257(199901)14:1<21::aid-mds1006>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
- 13.Ascherio A, Chen H, Weisskopf MG, O'Reilly E, McCullough ML, Calle EE, Schwarzschild MA, Thun MJ. Pesticide exposure and risk for Parkinson's disease. Ann. Neurol. 2006;60:197–203. doi: 10.1002/ana.20904. [DOI] [PubMed] [Google Scholar]
- 14.Athanassiadou A, Voutsinas G, Psiouri L, Leroy E, Polymeropoulos MH, Ilias A, Maniatis GM, Papapetropoulos T. Genetic analysis of families with Parkinson disease that carry the Ala53Thr mutation in the gene encoding alpha-synuclein. Am. J. Hum. Genet. 1999;65:555–558. doi: 10.1086/302486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Avramovich-Tirosh Y, Amit T, Bar-Am O, Zheng H, Fridkin M, Youdim MB. Therapeutic targets and potential of the novel brain- permeable multifunctional iron chelator-monoamine oxidase inhibitor drug, M-30, for the treatment of Alzheimer's disease. J. Neurochem. 2007;100:490–502. doi: 10.1111/j.1471-4159.2006.04258.x. [DOI] [PubMed] [Google Scholar]
- 16.Barbeau A, Inoue N, Cloutier T. Role of manganese in dystonia. Adv. Neurol. 1976;14:339–352. [PubMed] [Google Scholar]
- 17.Barden H, Levine S. Histochemical observations on rodent brain melanin. Brain Res. Bull. 1983;10:847–851. doi: 10.1016/0361-9230(83)90218-6. [DOI] [PubMed] [Google Scholar]
- 18.Barnham KJ, Haeffner F, Ciccotosto GD, Curtain CC, Tew D, Mavros C, Beyreuther K, Carrington D, Masters CL, Cherny RA, Cappai R, Bush AI. Tyrosine gated electron transfer is key to the toxic mechanism of Alzheimer's disease beta-amyloid. FASEB J. 2004;18:1427–1429. doi: 10.1096/fj.04-1890fje. [DOI] [PubMed] [Google Scholar]
- 19.Bartzokis G, Beckson M, Hance DB, Marx P, Foster JA, Marder SR. MR evaluation of age-related increase of brain iron in young adult and older normal males. Magn. Reson. Imaging. 1997;15:29–35. doi: 10.1016/s0730-725x(96)00234-2. [DOI] [PubMed] [Google Scholar]
- 20.Bartzokis G, Cummings JL, Markham CH, Marmarelis PZ, Treciokas LJ, Tishler TA, Marder SR, Mintz J. MRI evaluation of brain iron in earlier- and later-onset Parkinson's disease and normal subjects. Magn. Reson. Imaging. 1999;17:213–222. doi: 10.1016/s0730-725x(98)00155-6. [DOI] [PubMed] [Google Scholar]
- 21.Bartzokis G, Tishler TA, Shin IS, Lu PH, Cummings JL. Brain ferritin iron as a risk factor for age at onset in neurodegenerative diseases. Ann. N. Y. Acad. Sci. 2004;1012:224–236. doi: 10.1196/annals.1306.019. [DOI] [PubMed] [Google Scholar]
- 22.Beal MF, Matthews RT, Tieleman A, Shults CW. Coenzyme Q10 attenuates the 1-methyl-4-phenyl-1,2,3,tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice. Brain Res. 1998;783:109–114. doi: 10.1016/s0006-8993(97)01192-x. [DOI] [PubMed] [Google Scholar]
- 23.Beilina A, Van Der Brug M, Ahmad R, Kesavapany S, Miller DW, Petsko GA, Cookson MR. Mutations in PTEN-induced putative kinase 1 associated with recessive parkinsonism have differential effects on protein stability. Proc. Natl. Acad. Sci. USA. 2005;102:5703–5708. doi: 10.1073/pnas.0500617102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Benabid AL, Pollak P, Louveau A, Henry S, de Rougemont J. Combined (thalamotomy and stimulation) stereotactic surgery of the VIM thalamic nucleus for bilateral Parkinson disease. Appl. Neurophysiol. 1987;50:344–346. doi: 10.1159/000100803. [DOI] [PubMed] [Google Scholar]
- 25.Ben-Shachar D, Youdim MB. Intranigral iron injection induces behavioral and biochemical "parkinsonism" in rats. J. Neurochem. 1991;57:2133–2135. doi: 10.1111/j.1471-4159.1991.tb06432.x. [DOI] [PubMed] [Google Scholar]
- 26.Bergomi M, Vinceti M, Nacci G, Pietrini V, Bratter P, Alber D, Ferrari A, Vescovi L, Guidetti D, Sola P, Malagu S, Aramini C, Vivoli G. Environmental exposure to trace elements and risk of amyotrophic lateral sclerosis: a population-based case-control study. Environ. Res. 2002;89:116–123. doi: 10.1006/enrs.2002.4361. [DOI] [PubMed] [Google Scholar]
- 27.Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat. Neurosci. 2000;3:1301–1306. doi: 10.1038/81834. [DOI] [PubMed] [Google Scholar]
- 28.Bharathi Indi SS, Rao KS. Copper- and iron-induced differential fibril formation in alpha-synuclein: TEM study. Neurosci. Lett. 2007;424:78–82. doi: 10.1016/j.neulet.2007.06.052. [DOI] [PubMed] [Google Scholar]
- 29.Bharathi Rao KS. Thermodynamics imprinting reveals differential binding of metals to alpha-synuclein: relevance to Parkinson's disease. Biochem. Biophys. Res. Commun. 2007;359:115–120. doi: 10.1016/j.bbrc.2007.05.060. [DOI] [PubMed] [Google Scholar]
- 30.Bjorklund A, Dunnett SB, Brundin P, Stoessl A, Freed CR, Breeze RE, Levivier M, Peschanski M, Studer L, Barker R. Neural transplantation for the treatment of Parkinson's disease. Lancet Neurol. 2003;2:437–445. doi: 10.1016/s1474-4422(03)00442-3. [DOI] [PubMed] [Google Scholar]
- 31.Bjorklund A, Gage FH, Schmidt RH, Stenevi U, Dunnett SB. Intracerebral grafting of neuronal cell suspensions. VII. Recovery of choline acetyltransferase activity and acetylcholine synthesis in the denervated hippocampus reinnervated by septal suspension implants. Acta Physiol. Scand. Suppl. 1983;522:59–66. [PubMed] [Google Scholar]
- 32.Bjorklund A, Gage FH, Stenevi U, Dunnett SB. Intracerebral grafting of neuronal cell suspensions. VI. Survival and growth of intrahippocampal implants of septal cell suspensions. Acta Physiol. Scand. Suppl. 1983;522:49–58. [PubMed] [Google Scholar]
- 33.Bjorklund A, Stenevi U, Dunnett SB, Iversen SD. Functional reactivation of the deafferented neostriatum by nigral transplants. Nature. 1981;289:497–499. doi: 10.1038/289497a0. [DOI] [PubMed] [Google Scholar]
- 34.Bjorklund A, Stenevi U, Schmidt RH, Dunnett SB, Gage FH. Intracerebral grafting of neuronal cell suspensions. I. Introduction and general methods of preparation. Acta Physiol. Scand. Suppl. 1983;522:1–7. [PubMed] [Google Scholar]
- 35.Bjorklund A, Stenevi U, Schmidt RH, Dunnett SB, Gage FH. Intracerebral grafting of neuronal cell suspensions. II. Survival and growth of nigral cell suspensions implanted in different brain sites. Acta Physiol. Scand. Suppl. 1983;522:9–18. [PubMed] [Google Scholar]
- 36.Bjorklund A, Stenevi U, Svendgaard N. Growth of transplanted monoaminergic neurones into the adult hippocampus along the perforant path. Nature. 1976;262:787–790. doi: 10.1038/262787a0. [DOI] [PubMed] [Google Scholar]
- 37.Bjorklund LM, Sanchez-Pernaute R, Chung S, Andersson T, Chen IY, McNaught KS, Brownell AL, Jenkins BG, Wahlestedt C, Kim KS, Isacson O. Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model. Proc. Natl. Acad. Sci. USA. 2002;99:2344–2349. doi: 10.1073/pnas.022438099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Blackinton J, Ahmad R, Miller DW, van der Brug MP, Canet-Aviles RM, Hague SM, Kaleem M, Cookson MR. Effects of DJ-1 mutations and polymorphisms on protein stability and subcellular localization. Brain Res. Mol. Brain Res. 2005;134:76–83. doi: 10.1016/j.molbrainres.2004.09.004. [DOI] [PubMed] [Google Scholar]
- 39.Blanc EM, Keller JN, Fernandez S, Mattson MP. 4-hydroxynonenal, a lipid peroxidation product, impairs glutamate transport in cortical astrocytes. Glia. 1998;22:149–160. doi: 10.1002/(sici)1098-1136(199802)22:2<149::aid-glia6>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
- 40.Blanchard-Fillion B, Souza JM, Friel T, Jiang GC, Vrana K, Sharov V, Barron L, Schoneich C, Quijano C, Alvarez B, Radi R, Przedborski S, Fernando GS, Horwitz J, Ischiropoulos H. Nitration and inactivation of tyrosine hydroxylase by peroxynitrite. J. Biol. Chem. 2001;276:46017–46023. doi: 10.1074/jbc.M105564200. [DOI] [PubMed] [Google Scholar]
- 41.Blum D, Torch S, Nissou MF, Verna JM. 6-hydroxydopamine-induced nuclear factor-kappa B activation in PC12 cells. Biochem. Pharmacol. 2001;62:473–481. doi: 10.1016/s0006-2952(01)00680-3. [DOI] [PubMed] [Google Scholar]
- 42.Bogerts B. A brainstem atlas of catecholaminergic neurons in man, using melanin as a natural marker. J. Comp. Neurol. 1981;197:63–80. doi: 10.1002/cne.901970106. [DOI] [PubMed] [Google Scholar]
- 43.Bondy SC, Ali SF, Guo-Ross S. Aluminum but not iron treatment induces pro-oxidant events in the rat brain. Mol. Chem. Neuropathol. 1998;34:219–232. doi: 10.1007/BF02815081. [DOI] [PubMed] [Google Scholar]
- 44.Bonifati V, Rizzu P, Squitieri F, Krieger E, Vanacore N, van Swieten JC, Brice A, van Duijn CM, Oostra B, Meco G, Heutink P. DJ-1( PARK7), a novel gene for autosomal recessive, early onset parkinsonism. Neurol. Sci. 2003;24:159–160. doi: 10.1007/s10072-003-0108-0. [DOI] [PubMed] [Google Scholar]
- 45.Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol. Aging. 2003;24:197–211. doi: 10.1016/s0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
- 46.Bresolin N, Bet L, Binda A, Moggio M, Comi G, Nador F, Ferrante C, Carenzi A, Scarlato G. Clinical and biochemical correlations in mitochondrial myopathies treated with coenzyme Q10. Neurology. 1988;38:892–899. doi: 10.1212/wnl.38.6.892. [DOI] [PubMed] [Google Scholar]
- 47.Bresolin N, Bet L, Ferrante C, Binda A, Carenzi A, Moggio M, Comi G, Scarlato G. Immunological and biochemical studies and pilot therapeutic trial with ubidecarenone in Kearns-Sayre patients. Adv. Neurol. 1988;48:239–256. [PubMed] [Google Scholar]
- 48.Burke RE, Antonelli M, Sulzer D. Glial cell line-derived neurotrophic growth factor inhibits apoptotic death of postnatal substantia nigra dopamine neurons in primary culture. J. Neurochem. 1998;71:517–525. doi: 10.1046/j.1471-4159.1998.71020517.x. [DOI] [PubMed] [Google Scholar]
- 49.Bush AI, Masters CL, Tanzi RE. Copper, beta-amyloid, and Alzheimer's disease: tapping a sensitive connection. Proc. Natl. Acad. Sci. USA. 2003;100:11193–11194. doi: 10.1073/pnas.2135061100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cabin DE, Shimazu K, Murphy D, Cole NB, Gottschalk W, McIlwain KL, Orrison B, Chen A, Ellis CE, Paylor R, Lu B, Nussbaum RL. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein. J. Neurosci. 2002;22:8797–8807. doi: 10.1523/JNEUROSCI.22-20-08797.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Campbell A, Yang EY, Tsai-Turton M, Bondy SC. Pro-inflammatory effects of aluminum in human glioblastoma cells. Brain Res. 2002;933:60–65. doi: 10.1016/s0006-8993(02)02305-3. [DOI] [PubMed] [Google Scholar]
- 52.Cao G, Sofic E, Prior RL. Antioxidant and prooxidant behavior of flavonoids: structure-activity relationships. Free Radic. Biol. Med. 1997;22:749–760. doi: 10.1016/s0891-5849(96)00351-6. [DOI] [PubMed] [Google Scholar]
- 53.Cappai R, Leck SL, Tew DJ, Williamson NA, Smith DP, Galatis D, Sharples RA, Curtain CC, Ali FE, Cherny RA, Culvenor JG, Bottomley SP, Masters CL, Barnham KJ, Hill AF. Dopamine promotes alpha-synuclein aggregation into SDS-resistant soluble oligomers via a distinct folding pathway. FASEB J. 2005;19:1377–1379. doi: 10.1096/fj.04-3437fje. [DOI] [PubMed] [Google Scholar]
- 54.Carini R, Bellomo G, Paradisi L, Dianzani MU, Albano E. 4-Hydroxynonenal triggers Ca2+ influx in isolated rat hepatocytes. Biochem. Biophys. Res. Commun. 1996;218:772–776. doi: 10.1006/bbrc.1996.0137. [DOI] [PubMed] [Google Scholar]
- 55.Carta M, Lindgren HS, Lundblad M, Stancampiano R, Fadda F, Cenci MA. Role of striatal L-DOPA in the production of dyskinesia in 6-hydroxydopamine lesioned rats. J. Neurochem. 2006;96:1718–1727. doi: 10.1111/j.1471-4159.2006.03696.x. [DOI] [PubMed] [Google Scholar]
- 56.Caughman SW, Hentze MW, Rouault TA, Harford JB, Klausner RD. The iron-responsive element is the single element responsible for iron-dependent translational regulation of ferritin biosynthesis. Evidence for function as the binding site for a translational repressor. J. Biol. Chem. 1988;263:19048–19052. [PubMed] [Google Scholar]
- 57.Chan PL, Nutt JG, Holford NH. Levodopa slows progression of Parkinson's disease: external validation by clinical trial simulation. Pharm. Res. 2007;24:791–802. doi: 10.1007/s11095-006-9202-3. [DOI] [PubMed] [Google Scholar]
- 58.Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J, Hulihan M, Waucquier N, Defebvre L, Amouyel P, Farrer M, Destee A. Alpha-synuclein locus duplication as a cause of familial Parkinson's disease. Lancet. 2004;364:1167–1169. doi: 10.1016/S0140-6736(04)17103-1. [DOI] [PubMed] [Google Scholar]
- 59.Chen YH, Harvey BK, Hoffman AF, Wang Y, Chiang YH, Lupica CR. MPTP-induced deficits in striatal synaptic plasticity are prevented by glial cell line-derived neurotrophic factor expressed via an adeno-associated viral vector. FASEB J. 2007;22(1):261–275. doi: 10.1096/fj.07-8797com. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cherny RA, Atwood CS, Xilinas ME, Gray DN, Jones WD, McLean CA, Barnham KJ, Volitakis I, Fraser FW, Kim Y, Huang X, Goldstein LE, Moir RD, Lim JT, Beyreuther K, Zheng H, Tanzi RE, Masters CL, Bush AI. Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer's disease transgenic mice. Neuron. 2001;30:665–676. doi: 10.1016/s0896-6273(01)00317-8. [DOI] [PubMed] [Google Scholar]
- 61.Chiba-Falek O, Lopez GJ, Nussbaum RL. Levels of alpha-synuclein mRNA in sporadic Parkinson disease patients. Mov. Disord. 2006;21:1703–1708. doi: 10.1002/mds.21007. [DOI] [PubMed] [Google Scholar]
- 62.Chinta SJ, Andersen JK. Redox imbalance in Parkinson's disease. Biochim. Biophys. Acta. 2008;1780(11):1362–1367. doi: 10.1016/j.bbagen.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chinta SJ, Kumar MJ, Hsu M, Rajagopalan S, Kaur D, Rane A, Nicholls DG, Choi J, Andersen JK. Inducible alterations of glutathione levels in adult dopaminergic midbrain neurons result in nigrostriatal degeneration. J. Neurosci. 2007;27:13997–14006. doi: 10.1523/JNEUROSCI.3885-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Chiueh CC, Wu RM, Mohanakumar KP, Sternberger LM, Krishna G, Obata T, Murphy DL. In vivo generation of hydroxyl radicals and MPTP-induced dopaminergic toxicity in the basal ganglia. Ann. N. Y. Acad. Sci. 1994;738:25–36. doi: 10.1111/j.1749-6632.1994.tb21786.x. [DOI] [PubMed] [Google Scholar]
- 65.Clarkson ED, Edwards-Prasad J, Freed CR, Prasad KN. Immortalized dopamine neurons: A model to study neurotoxicity and neuroprotection. Proc. Soc. Exp. Biol. Med. 1999;222:157–163. doi: 10.1046/j.1525-1373.1999.d01-126.x. [DOI] [PubMed] [Google Scholar]
- 66.Cleren C, Yang L, Lorenzo B, Calingasan NY, Schomer A, Sireci A, Wille EJ, Beal MF. Therapeutic effects of coenzyme Q10 (CoQ10) and reduced CoQ10 in the MPTP model of Parkinsonism. J. Neurochem. 2008;104:1613–1621. doi: 10.1111/j.1471-4159.2007.05097.x. [DOI] [PubMed] [Google Scholar]
- 67.Cohen G. Monoamine oxidase, hydrogen peroxide, and Parkinson's disease. Adv. Neurol. 1987;45:119–125. [PubMed] [Google Scholar]
- 68.Cohen G. Monoamine oxidase and oxidative stress at dopaminergic synapses. J. Neural. Transm. Suppl. 1990;32:229–238. doi: 10.1007/978-3-7091-9113-2_33. [DOI] [PubMed] [Google Scholar]
- 69.Colosimo C, Merello M, Hughes AJ, Sieradzan K, Lees AJ. Motor response to acute dopaminergic challenge with apomorphine and levodopa in Parkinson's disease: implications for the pathogenesis of the on-off phenomenon. J. Neurol. Neurosurg. Psychiatry. 1996;60:634–637. doi: 10.1136/jnnp.60.6.634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Connor B, Kozlowski DA, Unnerstall JR, Elsworth JD, Tillerson JL, Schallert T, Bohn MC. Glial cell line-derived neurotrophic factor (GDNF) gene delivery protects dopaminergic terminals from degeneration. Exp. Neurol. 2001;169:83–95. doi: 10.1006/exnr.2001.7638. [DOI] [PubMed] [Google Scholar]
- 71.Connor JR, Menzies SL, St Martin SM, Mufson EJ. Cellular distribution of transferrin, ferritin, and iron in normal and aged human brains. J. Neurosci. Res. 1990; 27:595–611. doi: 10.1002/jnr.490270421. [DOI] [PubMed] [Google Scholar]
- 72.Conway KA, Harper JD, LansburyP.T Jr. Fibrils formed in vitro from alpha-synuclein and two mutant forms linked to Parkinson's disease are typical amyloid. Biochemistry. 2000;39:2552–2563. doi: 10.1021/bi991447r. [DOI] [PubMed] [Google Scholar]
- 73.Conway KA, Rochet JC, Bieganski RM, Lansbury P.T Jr. Kinetic stabilization of the alpha-synuclein protofibril by a dopamine-alpha-synuclein adduct. Science. 2001;294:1346–1349. doi: 10.1126/science.1063522. [DOI] [PubMed] [Google Scholar]
- 74.Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, Cao S, Caldwell KA, Caldwell GA, Marsischky G, Kolodner RD, Labaer J, Rochet JC, Bonini NM, Lindquist S. Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. Science. 2006;313:324–328. doi: 10.1126/science.1129462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Corasaniti MT, Strongoli MC, Pisanelli A, Bruno P, Rotiroti D, Nappi G, Nistico G. Distribution of paraquat into the brain after its systemic injection in rats. Funct. Neurol. 1992;7:51–56. [PubMed] [Google Scholar]
- 76.Crompton DE, Chinnery PF, Fey C, Curtis AR, Morris CM, Kierstan J, Burt A, Young F, Coulthard A, Curtis A, Ince PG, Bates D, Jackson MJ, Burn J. Neuroferritinopathy: a window on the role of iron in neurodegeneration. Blood Cells Mol. Dis. 2002;29:522–531. doi: 10.1006/bcmd.2002.0589. [DOI] [PubMed] [Google Scholar]
- 77.Cubells JF, Rayport S, Rajendran G, Sulzer D. Methamphetamine neurotoxicity involves vacuolation of endocytic organelles and dopamine-dependent intracellular oxidative stress. J. Neurosci. 1994;14:2260–2271. doi: 10.1523/JNEUROSCI.14-04-02260.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D. Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science. 2004;305:1292–1295. doi: 10.1126/science.1101738. [DOI] [PubMed] [Google Scholar]
- 79.Dallman PR, Spirito RA. Brain iron in the rat: extremely slow turnover in normal rats may explain long-lasting effects of early iron deficiency. J. Nutr. 1977;107:1075–1081. doi: 10.1093/jn/107.6.1075. [DOI] [PubMed] [Google Scholar]
- 80.Davidson WS, Jonas A, Clayton DF, George JM. Stabilization of alpha-synuclein secondary structure upon binding to synthetic membranes. J. Biol. Chem. 1998;273:9443–9449. doi: 10.1074/jbc.273.16.9443. [DOI] [PubMed] [Google Scholar]
- 81.De Bie RM, Schuurman PR, Esselink RA, Bosch DA, Speelman JD. Bilateral pallidotomy in Parkinson's disease: a retrospective study. Mov. Disord. 2002;17:533–538. doi: 10.1002/mds.10090. [DOI] [PubMed] [Google Scholar]
- 82.de la Fuente-Fernandez R, Schulzer M, Stoessl AJ. Placebo mechanisms and reward circuitry: clues from Parkinson's disease. Biol. Psychiatry. 2004;56:67–71. doi: 10.1016/j.biopsych.2003.11.019. [DOI] [PubMed] [Google Scholar]
- 83.de Rijk MC, Breteler MM, den Breeijen JH, Launer LJ, Grobbee DE, van der Meche FG, Hofman A. Dietary antioxidants and Parkinson disease. The Rotterdam Study. Arch. Neurol. 1997;54:762–765. doi: 10.1001/archneur.1997.00550180070015. [DOI] [PubMed] [Google Scholar]
- 84.Deinard AS, List A, Lindgren B, Hunt JV, Chang PN. Cognitive deficits in iron-deficient and iron-deficient anemic children. J. Pediatr. 1986;108:681–689. doi: 10.1016/s0022-3476(86)81041-1. [DOI] [PubMed] [Google Scholar]
- 85.Deng H, Le W, Guo Y, Hunter CB, Xie W, Huang M, Jankovic J. Genetic analysis of LRRK2 mutations in patients with Parkinson disease. J. Neurol. Sci. 2006;251:102–106. doi: 10.1016/j.jns.2006.09.017. [DOI] [PubMed] [Google Scholar]
- 86.Deng J, Lewis PA, Greggio E, Sluch E, Beilina A, Cookson MR. Structure of the ROC domain from the Parkinson's disease-associated leucine-rich repeat kinase 2 reveals a dimeric GTPase. Proc. Natl. Acad. Sci. USA. 2008;105:1499–1504. doi: 10.1073/pnas.0709098105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Desole MS, Esposito G, Migheli R, Sircana S, Delogu MR, Fresu L, Miele M, de Natale G, Miele E. Glutathione deficiency potentiates manganese toxicity in rat striatum and brainstem and in PC12 cells. Pharmacol. Res. 1997;36:285–292. doi: 10.1006/phrs.1997.0197. [DOI] [PubMed] [Google Scholar]
- 88.Desole MS, Sciola L, Delogu MR, Sircana S, Migheli R, Miele E. Role of oxidative stress in the manganese and 1-methyl-4-(2'-ethylphenyl)-1,2,3,6-tetrahydropyridine-induced apoptosis in PC12 cells. Neurochem. Int. 1997;31:169–176. doi: 10.1016/s0197-0186(96)00146-5. [DOI] [PubMed] [Google Scholar]
- 89.Devasagayam TP, Werner T, Ippendorf H, Martin HD, Sies H. Synthetic carotenoids, novel polyene polyketones and new capsorubin isomers as efficient quenchers of singlet molecular oxygen. Photochem. Photobiol. 1992;55:511–514. doi: 10.1111/j.1751-1097.1992.tb04271.x. [DOI] [PubMed] [Google Scholar]
- 90.Dexter DT, Carayon A, Javoy-Agid F, Agid Y, Wells FR, Daniel SE, Lees AJ, Jenner P, Marsden CD. Alterations in the levels of iron, ferritin and other trace metals in Parkinson's disease and other neurodegenerative diseases affecting the basal ganglia. Brain. 1991;114 (Pt 4):1953–1975. doi: 10.1093/brain/114.4.1953. [DOI] [PubMed] [Google Scholar]
- 91.Dexter DT, Carayon A, Vidailhet M, Ruberg M, Agid F, Agid Y, Lees AJ, Wells FR, Jenner P, Marsden CD. Decreased ferritin levels in brain in Parkinson's disease. J. Neurochem. 1990;55:16–20. doi: 10.1111/j.1471-4159.1990.tb08814.x. [DOI] [PubMed] [Google Scholar]
- 92.Dexter DT, Wells FR, Lees AJ, Agid F, Agid Y, Jenner P, Marsden CD. Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson's disease. J. Neurochem. 1989;52:1830–1836. doi: 10.1111/j.1471-4159.1989.tb07264.x. [DOI] [PubMed] [Google Scholar]
- 93.Dhopesh VP, Yagnik PM, Weddington WW. Can cocaine abuse cause parkinsonism? Am. J. Addict. 1997;6:177–179. [PubMed] [Google Scholar]
- 94.Di Mascio P, Devasagayam TP, Kaiser S, Sies H. Carotenoids, tocopherols and thiols as biological singlet molecular oxygen quenchers. Biochem. Soc. Trans. 1990;18:1054–1056. doi: 10.1042/bst0181054. [DOI] [PubMed] [Google Scholar]
- 95.Di Stasi AM, Mallozzi C, Macchia G, Petrucci TC, Minetti M. Peroxynitrite induces tryosine nitration and modulates tyrosine phosphorylation of synaptic proteins. J. Neurochem. 1999;73:727–735. doi: 10.1046/j.1471-4159.1999.0730727.x. [DOI] [PubMed] [Google Scholar]
- 96.Dietrich JB, Mangeol A, Revel MO, Burgun C, Aunis D, Zwiller J. Acute or repeated cocaine administration generates reactive oxygen species and induces antioxidant enzyme activity in dopaminergic rat brain structures. Neuropharmacology. 2005;48:965–974. doi: 10.1016/j.neuropharm.2005.01.018. [DOI] [PubMed] [Google Scholar]
- 97.Ding WQ, Liu B, Vaught JL, Palmiter RD, Lind SE. Clioquinol and docosahexaenoic acid act synergistically to kill tumor cells. Mol. Cancer Ther. 2006;5:1864–1872. doi: 10.1158/1535-7163.MCT-06-0067. [DOI] [PubMed] [Google Scholar]
- 98.Ding WQ, Liu B, Vaught JL, Yamauchi H, Lind SE. Anticancer activity of the antibiotic clioquinol. Cancer Res. 2005;65:3389–3395. doi: 10.1158/0008-5472.CAN-04-3577. [DOI] [PubMed] [Google Scholar]
- 99.Ding YM, Jaumotte JD, Signore AP, Zigmond MJ. Effects of 6-hydroxydopamine on primary cultures of substantia nigra: specific damage to dopamine neurons and the impact of glial cell line-derived neurotrophic factor. J. Neurochem. 2004;89:776–787. doi: 10.1111/j.1471-4159.2004.02415.x. [DOI] [PubMed] [Google Scholar]
- 100.Doraiswamy PM, Finefrock AE. Metals in our minds: therapeutic implications for neurodegenerative disorders. Lancet Neurol. 2004;3:431–434. doi: 10.1016/S1474-4422(04)00809-9. [DOI] [PubMed] [Google Scholar]
- 101.Dostrovsky JO, Levy R, Wu JP, Hutchison WD, Tasker RR, Lozano AM. Microstimulation-induced inhibition of neuronal firing in human globus pallidus. J. Neurophysiol. 2000;84:570–574. doi: 10.1152/jn.2000.84.1.570. [DOI] [PubMed] [Google Scholar]
- 102.Double KL. Functional effects of neuromelanin and synthetic melanin in model systems. J. Neural. Transm. 2006;113:751–756. doi: 10.1007/s00702-006-0450-5. [DOI] [PubMed] [Google Scholar]
- 103.Double KL, Ben-Shachar D, Youdim MB, Zecca L, Riederer P, Gerlach M. Influence of neuromelanin on oxidative pathways within the human substantia nigra. Neurotoxicol. Teratol. 2002;24:621–628. doi: 10.1016/s0892-0362(02)00218-0. [DOI] [PubMed] [Google Scholar]
- 104.Double KL, Gerlach M, Youdim MB, Riederer P. Impaired iron homeostasis in Parkinson's disease. J. Neural. Transm. Suppl. 2000:37–58. doi: 10.1007/978-3-7091-6301-6_3. [DOI] [PubMed] [Google Scholar]
- 105.Du B, Wu P, Boldt-Houle DM, Terwilliger EF. Efficient transduction of human neurons with an adeno-associated virus vector. Gene Ther. 1996;3:254–261. [PubMed] [Google Scholar]
- 106.Dunnett SB, Bjorklund A, Schmidt RH, Stenevi U, Iversen SD. Intracerebral grafting of neuronal cell suspensions. V. Behavioural recovery in rats with bilateral 6-OHDA lesions following implantation of nigral cell suspensions. Acta Physiol. Scand. Suppl. 1983;522:39–47. [PubMed] [Google Scholar]
- 107.Emerit I, Keck M, Levy A, Feingold J, Michelson AM. Activated oxygen species at the origin of chromosome breakage and sister-chromatid exchanges. Mutat. Res. 1982;103:165–172. doi: 10.1016/0165-7992(82)90024-0. [DOI] [PubMed] [Google Scholar]
- 108.Emson PC, Bjorklund A, Stenevi U. Possible regeneration of gamma-aminobutyric acid-containing fibres into irides transplanted into the central nervous system. Nature. 1976;259:567–570. doi: 10.1038/259567a0. [DOI] [PubMed] [Google Scholar]
- 109.Erola T, Heikkinen ER, Haapaniemi T, Tuominen J, Juolasmaa A, Myllyla VV. Efficacy of bilateral subthalamic nucleus (STN) stimulation in Parkinson's disease. Acta Neurochir. (Wien) 2006;148:389–394. doi: 10.1007/s00701-005-0662-8. [DOI] [PubMed] [Google Scholar]
- 110.Fahn S. A new look at levodopa based on the ELLDOPA study. J. Neural. Transm. Suppl. 2006:419–426. doi: 10.1007/978-3-211-45295-0_63. [DOI] [PubMed] [Google Scholar]
- 111.Fahn S, Oakes D, Shoulson I, Kieburtz K, Rudolph A, Lang A, Olanow CW, Tanner C, Marek K. Levodopa and the progression of Parkinson's disease. N. Engl. J. Med. 2004;351:2498–2508. doi: 10.1056/NEJMoa033447. [DOI] [PubMed] [Google Scholar]
- 112.Faucheux BA, Martin ME, Beaumont C, Hunot S, Hauw JJ, Agid Y, Hirsch EC. Lack of up-regulation of ferritin is associated with sustained iron regulatory protein-1 binding activity in the substantia nigra of patients with Parkinson's disease. J. Neurochem. 2002;83:320–330. doi: 10.1046/j.1471-4159.2002.01118.x. [DOI] [PubMed] [Google Scholar]
- 113.Feigin A, Kaplitt MG, Tang C, Lin T, Mattis P, Dhawan V, During MJ, Eidelberg D. Modulation of metabolic brain networks after subthalamic gene therapy for Parkinson's disease. Proc. Natl. Acad. Sci. USA. 2007;104:19559–19564. doi: 10.1073/pnas.0706006104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Finkelstein DI, Stanic D, Parish CL, Tomas D, Dickson K, Horne MK. Axonal sprouting following lesions of the rat substantia nigra. Neuroscience. 2000;97:99–112. doi: 10.1016/s0306-4522(00)00009-9. [DOI] [PubMed] [Google Scholar]
- 115.Flaten TP. Aluminium as a risk factor in Alzheimer's disease, with emphasis on drinking water. Brain Res. Bull. 2001;55:187–196. doi: 10.1016/s0361-9230(01)00459-2. [DOI] [PubMed] [Google Scholar]
- 116.Fornai F, Lenzi P, Ferrucci M, Lazzeri G, di Poggio AB, Natale G, Busceti CL, Biagioni F, Giusiani M, Ruggieri S, Paparelli A. Occurrence of neuronal inclusions combined with increased nigral expression of alpha-synuclein within dopaminergic neurons following treatment with amphetamine derivatives in mice. Brain Res. Bull. 2005;65:405–413. doi: 10.1016/j.brainresbull.2005.02.022. [DOI] [PubMed] [Google Scholar]
- 117.Fornai F, Lenzi P, Gesi M, Ferrucci M, Lazzeri G, Capobianco L, de Blasi A, Battaglia G, Nicoletti F, Ruggieri S, Paparelli A. Similarities between methamphetamine toxicity and proteasome inhibition. Ann. N. Y. Acad. Sci. 2004;1025:162–170. doi: 10.1196/annals.1316.021. [DOI] [PubMed] [Google Scholar]
- 118.Fornai F, Schluter OM, Lenzi P, Gesi M, Ruffoli R, Ferrucci M, Lazzeri G, Busceti CL, Pontarelli F, Battaglia G, Pellegrini A, Nicoletti F, Ruggieri S, Paparelli A, Sudhof TC. Parkinson-like syndrome induced by continuous MPTP infusion: convergent roles of the ubiquitin-proteasome system and alpha-synuclein. Proc. Natl. Acad. Sci. USA. 2005;102:3413–3418. doi: 10.1073/pnas.0409713102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Fornstedt B. Role of catechol autooxidation in the degeneration of dopamine neurons. Acta Neurol. Scand. Suppl. 1990;129:12–14. doi: 10.1111/j.1600-0404.1990.tb02595.x. [DOI] [PubMed] [Google Scholar]
- 120.Forsmark-Andree P, Lee CP, Dallner G, Ernster L. Lipid peroxidation and changes in the ubiquinone content and the respiratory chain enzymes of submitochondrial particles. Free Radic. Biol. Med. 1997;22:391–400. doi: 10.1016/s0891-5849(96)00330-9. [DOI] [PubMed] [Google Scholar]
- 121.Fortin DL, Nemani VM, Voglmaier SM, Anthony MD, Ryan TA, Edwards RH. Neural activity controls the synaptic accumulation of alpha-synuclein. J. Neurosci. 2005;25:10913–10921. doi: 10.1523/JNEUROSCI.2922-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Fortin DL, Troyer MD, Nakamura K, Kubo S, Anthony MD, Edwards RH. Lipid rafts mediate the synaptic localization of alpha-synuclein. J. Neurosci. 2004;24:6715–6723. doi: 10.1523/JNEUROSCI.1594-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Fountaine TM, Wade-Martins R. RNA interference-mediated knockdown of alpha-synuclein protects human dopaminergic neuroblastoma cells from MPP(+) toxicity and reduces dopamine transport. J. Neurosci. Res. 2007;85:351–363. doi: 10.1002/jnr.21125. [DOI] [PubMed] [Google Scholar]
- 124.Fredriksson A, Archer T. Postnatal iron overload destroys NA-DA functional interactions. J. Neural. Transm. 2007;114:195–203. doi: 10.1007/s00702-006-0522-6. [DOI] [PubMed] [Google Scholar]
- 125.Fredriksson A, Schroder N, Eriksson P, Izquierdo I, Archer T. Neonatal iron potentiates adult MPTP-induced neurodegenerative and functional deficits. Parkinsonism Relat. Disord. 2001;7:97–105. doi: 10.1016/s1353-8020(00)00028-6. [DOI] [PubMed] [Google Scholar]
- 126.Freed CR, Greene PE, Breeze RE, Tsai WY, DuMouchel W, Kao R, Dillon S, Winfield H, Culver S, Trojanowski JQ, Eidelberg D, Fahn S. Transplantation of embryonic dopamine neurons for severe Parkinson's disease. N. Engl. J. Med. 2001;344:710–719. doi: 10.1056/NEJM200103083441002. [DOI] [PubMed] [Google Scholar]
- 127.Freed CR, Leehey MA, Zawada M, Bjugstad K, Thompson L, Breeze RE. Do patients with Parkinson's disease benefit from embryonic dopamine cell transplantation? J. Neurol. 2003;250(3):III44–46. doi: 10.1007/s00415-003-1308-5. [DOI] [PubMed] [Google Scholar]
- 128.Freed WJ. A perspective on transplantation therapy and stem cells for Parkinson's disease. Cell Transplant. 2004;13:319–327. [PubMed] [Google Scholar]
- 129.Friedlich AL, Tanzi RE, Rogers JT. The 5'-untranslated region of Parkinson's disease alpha-synuclein messengerRNA contains a predicted iron responsive element. Mol. Psychiatry. 2007;12:222–223. doi: 10.1038/sj.mp.4001937. [DOI] [PubMed] [Google Scholar]
- 130.Fujimura M, Morita-Fujimura Y, Kawase M, Copin JC, Calagui B, Epstein CJ, Chan PH. Manganese superoxide dismutase mediates the early release of mitochondrial cytochrome C and subsequent DNA fragmentation after permanent focal cerebral ischemia in mice. J. Neurosci. 1999;19:3414–3422. doi: 10.1523/JNEUROSCI.19-09-03414.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Fulceri F, Biagioni F, Lenzi P, Falleni A, Gesi M, Ruggieri S, Fornai F. Nigrostriatal damage with 6-OHDA: validation of routinely applied procedures. Ann. N. Y. Acad. Sci. 2006;1074:344–348. doi: 10.1196/annals.1369.032. [DOI] [PubMed] [Google Scholar]
- 132.Fung HC, Scholz S, Matarin M, Simon-Sanchez J, Hernandez D, Britton A, Gibbs JR, Langefeld C, Stiegert ML, Schymick J, Okun MS, Mandel RJ, Fernandez HH, Foote KD, Rodriguez RL, Peckham E, De Vrieze FW, Gwinn-Hardy K, Hardy JA, Singleton A. Genome-wide genotyping in Parkinson's disease and neurologically normal controls: first stage analysis and public release of data. Lancet Neurol. 2006;5:911–916. doi: 10.1016/S1474-4422(06)70578-6. [DOI] [PubMed] [Google Scholar]
- 133.Gage FH, Bjorklund A, Stenevi U, Dunnett SB. Intracerebral grafting of neuronal cell suspensions. VIII. Survival and growth of implants of nigral and septal cell suspensions in intact brains of aged rats. Acta Physiol. Scand. Suppl. 1983;522:67–75. [PubMed] [Google Scholar]
- 134.Gal S, Fridkin M, Amit T, Zheng H, Youdim MB. M30, a novel multifunctional neuroprotective drug with potent iron chelating and brain selective monoamine oxidase-ab inhibitory activity for Parkinson's disease. J. Neural. Transm. Suppl. 2006;00:447–456. doi: 10.1007/978-3-211-45295-0_68. [DOI] [PubMed] [Google Scholar]
- 135.Gal S, Zheng H, Fridkin M, Youdim MB. Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selective brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine depletion. J. Neurochem. 2005;95:79–88. doi: 10.1111/j.1471-4159.2005.03341.x. [DOI] [PubMed] [Google Scholar]
- 136.Gao X, Chen H, Fung TT, Logroscino G, Schwarzschild MA, Hu FB, Ascherio A. Prospective study of dietary pattern and risk of Parkinson disease. Am. J. Clin. Nutr. 2007;86:1486–1494. doi: 10.1093/ajcn/86.5.1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Garruto RM, Fukatsu R, Yanagihara R, Gajdusek DC, Hook G, Fiori CE. Imaging of calcium and aluminum in neurofibrillary tangle-bearing neurons in parkinsonism-dementia of Guam. Proc. Natl. Acad. Sci. USA. 1984;81:1875–1879. doi: 10.1073/pnas.81.6.1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Gauthier E, Fortier I, Courchesne F, Pepin P, Mortimer J, Gauvreau D. Aluminum forms in drinking water and risk of Alzheimer's disease. Environ. Res. 2000;84:234–246. doi: 10.1006/enrs.2000.4101. [DOI] [PubMed] [Google Scholar]
- 139.Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, Hurtig HI, Ischiropoulos H, Trojanowski JQ, Lee VM. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science. 2000;290:985–989. doi: 10.1126/science.290.5493.985. [DOI] [PubMed] [Google Scholar]
- 140.Gil B, Sanz MJ, Terencio MC, Ferrandiz ML, Bustos G, Paya M, Gunasegaran R, Alcaraz MJ. Effects of flavonoids on Naja naja and human recombinant synovial phospholipases A2 and inflammatory responses in mice. Life Sci. 1994;54:PL333–338. doi: 10.1016/0024-3205(94)90021-3. [DOI] [PubMed] [Google Scholar]
- 141.Gill SS, Patel NK, Hotton GR, O'Sullivan K, McCarter R, Bunnage M, Brooks DJ, Svendsen CN, Heywood P. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat. Med. 2003;9:589–595. doi: 10.1038/nm850. [DOI] [PubMed] [Google Scholar]
- 142.Giulivi C, Davies KJ. Dityrosine and tyrosine oxidation products are endogenous markers for the selective proteolysis of oxidatively modified red blood cell hemoglobin by (the 19 S) proteasome. J. Biol. Chem. 1993;268:8752–8759. [PubMed] [Google Scholar]
- 143.Goedert M. Familial Parkinson's disease. The awakening of alpha-synuclein. Nature. 1997;388:232–233. doi: 10.1038/40767. [DOI] [PubMed] [Google Scholar]
- 144.Golbe LI, Farrell TM, Davis PH. Case-control study of early life dietary factors in Parkinson's disease. Arch. Neurol. 1988;45:1350–1353. doi: 10.1001/archneur.1988.00520360068014. [DOI] [PubMed] [Google Scholar]
- 145.Goldberg MS, Fleming SM, Palacino JJ, Cepeda C, Lam HA, Bhatnagar A, Meloni EG, Wu N, Ackerson LC, Klapstein GJ, Gajendiran M, Roth BL, Chesselet MF, Maidment NT, Levine MS, Shen J. Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J. Biol. Chem. 2003;278:43628–43635. doi: 10.1074/jbc.M308947200. [DOI] [PubMed] [Google Scholar]
- 146.Goldstein LE, Leopold MC, Huang X, Atwood CS, Saunders AJ, Hartshorn M, Lim JT, Faget KY, Muffat JA, Scarpa RC, Chylack LT, Jr., Bowden EF, Tanzi RE, Bush AI. 3-Hydroxykynurenine and 3-hydroxyanthranilic acid generate hydrogen peroxide and promote alpha-crystallin cross-linking by metal ion reduction. Biochemistry. 2000;39:7266–7275. doi: 10.1021/bi992997s. [DOI] [PubMed] [Google Scholar]
- 147.Golub MS, Germann SL, Araiza RS, Reader JR, Griffey SM, Lloyd KC. Movement disorders in the Hfe knockout mouse. Nutr. Neurosci. 2005;8:239–244. doi: 10.1080/10284150500277685. [DOI] [PubMed] [Google Scholar]
- 148.Golub MS, Hogrefe CE, Germann SL. Iron deprivation during fetal development changes the behavior of juvenile rhesus monkeys. J. Nutr. 2007;137:979–984. doi: 10.1093/jn/137.4.979. [DOI] [PubMed] [Google Scholar]
- 149.Gomez-Vargas M, Nishibayashi-Asanuma S, Asanuma M, Kondo Y, Iwata E, Ogawa N. Pergolide scavenges both hydroxyl and nitric oxide free radicals in vitro and inhibits lipid peroxidation in different regions of the rat brain. Brain Res. 1998;790:202–208. doi: 10.1016/s0006-8993(97)01521-7. [DOI] [PubMed] [Google Scholar]
- 150.Gong L, Daigneault EA, Acuff RV, Kostrzewa RM. Vitamin E supplements fail to protect mice from acute MPTP neurotoxicity. Neuroreport. 1991;2:544–546. doi: 10.1097/00001756-199109000-00012. [DOI] [PubMed] [Google Scholar]
- 151.Good PF, Olanow CW, Perl DP. Neuromelanin-containing neurons of the substantia nigra accumulate iron and aluminum in Parkinson's disease: a LAMMA study. Brain Res. 1992;593:343–346. doi: 10.1016/0006-8993(92)91334-b. [DOI] [PubMed] [Google Scholar]
- 152.Goodwill KE, Sabatier C, Marks C, Raag R, Fitzpatrick PF, Stevens RC. Crystal structure of tyrosine hydroxylase at 2.3 A and its implications for inherited neurodegenerative diseases. Nat. Struct. Biol. 1997;4:578–585. doi: 10.1038/nsb0797-578. [DOI] [PubMed] [Google Scholar]
- 153.Goodwill KE, Sabatier C, Stevens RC. Crystal structure of tyrosine hydroxylase with bound cofactor analogue and iron at 2.3 A resolution: self-hydroxylation of Phe300 and the pterin-binding site. Biochemistry. 1998;37:13437–13445. doi: 10.1021/bi981462g. [DOI] [PubMed] [Google Scholar]
- 154.Gotz ME, Double K, Gerlach M, Youdim MB, Riederer P. The relevance of iron in the pathogenesis of Parkinson's disease. Ann. N. Y. Acad. Sci. 2004;1012:193–208. doi: 10.1196/annals.1306.017. [DOI] [PubMed] [Google Scholar]
- 155.Graves AB, Rosner D, Echeverria D, Mortimer JA, Larson EB. Occupational exposures to solvents and aluminium and estimated risk of Alzheimer's disease. Occup. Environ. Med. 1998;55:627–633. doi: 10.1136/oem.55.9.627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Greenbaum EA, Graves CL, Mishizen-Eberz AJ, Lupoli MA, Lynch DR, Englander SW, Axelsen PH, Giasson BI. The E46K mutation in alpha-synuclein increases amyloid fibril formation. J. Biol. Chem. 2005;280:7800–7807. doi: 10.1074/jbc.M411638200. [DOI] [PubMed] [Google Scholar]
- 157.Griffiths PD, Dobson BR, Jones GR, Clarke DT. Iron in the basal ganglia in Parkinson's disease. An in vitro study using extended X-ray absorption fine structure and cryo-electron microscopy. Brain. 1999;122(Pt 4):667–673. doi: 10.1093/brain/122.4.667. [DOI] [PubMed] [Google Scholar]
- 158.Grundemann J, Schlaudraff F, Haeckel O, Liss B. Elevated alpha-synuclein mRNA levels in individual UV-laser-microdissected dopaminergic substantia nigra neurons in idiopathic Parkinson's disease. Nucleic Acids Res. 2008;36:e38. doi: 10.1093/nar/gkn084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gutteridge JM, Quinlan GJ, Clark I, Halliwell B. Aluminium salts accelerate peroxidation of membrane lipids stimulated by iron salts. Biochim. Biophys. Acta. 1985;835:441–447. doi: 10.1016/0005-2760(85)90113-4. [DOI] [PubMed] [Google Scholar]
- 160.Haberler C, Alesch F, Mazal PR, Pilz P, Jellinger K, Pinter MM, Hainfellner JA, Budka H. No tissue damage by chronic deep brain stimulation in Parkinson's disease. Ann. Neurol. 2000;48:372–376. [PubMed] [Google Scholar]
- 161.Hachinski V. Aluminum exposure and risk of Alzheimer disease. Arch. Neurol. 1998;55:742. doi: 10.1001/archneur.55.5.742. [DOI] [PubMed] [Google Scholar]
- 162.Hallett M, Litvan I. Evaluation of surgery for Parkinson's disease: a report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. The Task Force on Surgery for Parkinson's Disease. Neurology. 1999;53:1910–1921. doi: 10.1212/wnl.53.9.1910. [DOI] [PubMed] [Google Scholar]
- 163.Hallett M, Litvan I. Scientific position paper of the Movement Disorder Society evaluation of surgery for Parkinson's disease. Task Force on Surgery for Parkinson's Disease of the American Academy of Neurology Therapeutic and Technology Assessment Committee. Mov. Disord. 2000;15:436–438. [PubMed] [Google Scholar]
- 164.Halliwell B. Oxidative stress and neurodegeneration: where are we now? J. Neurochem. 2006;97:1634–1658. doi: 10.1111/j.1471-4159.2006.03907.x. [DOI] [PubMed] [Google Scholar]
- 165.Hanasaki Y, Ogawa S, Fukui S. The correlation between active oxygens scavenging and antioxidative effects of flavonoids. Free Radic. Biol. Med. 1994;16:845–850. doi: 10.1016/0891-5849(94)90202-x. [DOI] [PubMed] [Google Scholar]
- 166.Hanrott K, Gudmunsen L, O'Neill MJ, Wonnacott S. 6-hydroxydopamine-induced apoptosis is mediated via extracellular auto-oxidation and caspase 3-dependent activation of protein kinase Cdelta. J. Biol. Chem. 2006;281:5373–5382. doi: 10.1074/jbc.M511560200. [DOI] [PubMed] [Google Scholar]
- 167.Hansen TM, Nielsen H, Bernth N, Moos T. Expression of ferritin protein and subunit mRNAs in normal and iron deficient rat brain. Brain Res. Mol. Brain Res. 1999;65:186–197. doi: 10.1016/s0169-328x(99)00011-x. [DOI] [PubMed] [Google Scholar]
- 168.Hatano Y, Li Y, Sato K, Asakawa S, Yamamura Y, Tomiyama H, Yoshino H, Asahina M, Kobayashi S, Hassin-Baer S, Lu CS, Ng AR, Rosales RL, Shimizu N, Toda T, Mizuno Y, Hattori N. Novel PINK1 mutations in early-onset parkinsonism. Ann. Neurol. 2004;56:424–427. doi: 10.1002/ana.20251. [DOI] [PubMed] [Google Scholar]
- 169.Hattori N, Kitada T, Matsumine H, Asakawa S, Yamamura Y, Yoshino H, Kobayashi T, Yokochi M, Wang M, Yoritaka A, Kondo T, Kuzuhara S, Nakamura S, Shimizu N, Mizuno Y. Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann. Neurol. 1998;44:935–941. doi: 10.1002/ana.410440612. [DOI] [PubMed] [Google Scholar]
- 170.Hayashi S, Wakabayashi K, Ishikawa A, Nagai H, Saito M, Maruyama M, Takahashi T, Ozawa T, Tsuji S, Takahashi H. An autopsy case of autosomal-recessive juvenile parkinsonism with a homozygous exon 4 deletion in the parkin gene. Mov. Disord. 2000;15:884–888. doi: 10.1002/1531-8257(200009)15:5<884::aid-mds1019>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- 171.Hayashita-Kinoh H, Yamada M, Yokota T, Mizuno Y, Mochizuki H. Down-regulation of alpha-synuclein expression can rescue dopaminergic cells from cell death in the substantia nigra of Parkinson's disease rat model. Biochem. Biophys. Res. Commun. 2006;341:1088–1095. doi: 10.1016/j.bbrc.2006.01.057. [DOI] [PubMed] [Google Scholar]
- 172.Hellenbrand W, Boeing H, Robra BP, Seidler A, Vieregge P, Nischan P, Joerg J, Oertel WH, Schneider E, Ulm G. Diet and Parkinson's disease. II: A possible role for the past intake of specific nutrients. Results from a self-administered food-frequency questionnaire in a case-control study. Neurology. 1996;47:644–650. doi: 10.1212/wnl.47.3.644. [DOI] [PubMed] [Google Scholar]
- 173.Hellenbrand W, Seidler A, Boeing H, Robra BP, Vieregge P, Nischan P, Joerg J, Oertel WH, Schneider E, Ulm G. Diet and Parkinson's disease. I: A possible role for the past intake of specific foods and food groups. Results from a self-administered food-frequency questionnaire in a case-control study. Neurology. 1996;47:636–643. doi: 10.1212/wnl.47.3.636. [DOI] [PubMed] [Google Scholar]
- 174.Henderson J, Doherty K, Allbutt H, Billing R. Effects of pallidotomy on motor symptoms in an animal model of Parkinson's disease. Behav. Brain Res. 2006;169:29–38. doi: 10.1016/j.bbr.2005.11.028. [DOI] [PubMed] [Google Scholar]
- 175.Henderson JM, Carpenter K, Cartwright H, Halliday GM. Degeneration of the centre median-parafascicular complex in Parkinson's disease. Ann. Neurol. 2000;47:345–352. [PubMed] [Google Scholar]
- 176.Henderson JM, Stanic D, Tomas D, Patch J, Horne MK, Bourke D, Finkelstein DI. Postural changes after lesions of the substantia nigra pars reticulata in hemiparkinsonian monkeys. Behav. Brain Res. 2005;160:267–276. doi: 10.1016/j.bbr.2004.12.008. [DOI] [PubMed] [Google Scholar]
- 177.Hermida-Ameijeiras A, Mendez-Alvarez E, Sanchez-Iglesias S, Sanmartin-Suarez C, Soto-Otero R. Autoxidation and MAO-mediated metabolism of dopamine as a potential cause of oxidative stress: role of ferrous and ferric ions. Neurochem. Int. 2004;45:103–116. doi: 10.1016/j.neuint.2003.11.018. [DOI] [PubMed] [Google Scholar]
- 178.Hershko C, Avramovici-Grisaru S, Link G, Gelfand L, Sarel S. Mechanism of in vivo iron chelation by pyridoxal isonicotinoyl hydrazone and other imino derivatives of pyridoxal. J. Lab. Clin. Med. 1981;98:99–108. [PubMed] [Google Scholar]
- 179.Hilker R, Portman AT, Voges J, Staal MJ, Burghaus L, van Laar T, Koulousakis A, Maguire RP, Pruim J, de Jong BM, Herholz K, Sturm V, Heiss WD, Leenders KL. Disease progression continues in patients with advanced Parkinson's disease and effective subthalamic nucleus stimulation. J. Neurol. Neurosurg. Psychiatry. 2005;76:1217–1221. doi: 10.1136/jnnp.2004.057893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Hirsch EC, Brandel JP, Galle P, Javoy-Agid F, Agid Y. Iron and aluminum increase in the substantia nigra of patients with Parkinson's disease: an X-ray microanalysis. J. Neurochem. 1991;56:446–451. doi: 10.1111/j.1471-4159.1991.tb08170.x. [DOI] [PubMed] [Google Scholar]
- 181.Hod Y, Pentyala SN, Whyard TC, El-Maghrabi MR. Identification and characterization of a novel protein that regulates RNA-protein interaction. J. Cell Biochem. 1999;72:435–444. [PubMed] [Google Scholar]
- 182.Hoglinger GU, Feger J, Prigent A, Michel PP, Parain K, Champy P, Ruberg M, Oertel WH, Hirsch EC. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. J. Neurochem. 2003;84:491–502. doi: 10.1046/j.1471-4159.2003.01533.x. [DOI] [PubMed] [Google Scholar]
- 183.Holford NH, Chan PL, Nutt JG, Kieburtz K, Shoulson I. Disease progression and pharmacodynamics in Parkinson disease - evidence for functional protection with levodopa and other treatments. J. Pharmacokinet. Pharmacodyn. 2006;33:281–311. doi: 10.1007/s10928-006-9012-6. [DOI] [PubMed] [Google Scholar]
- 184.Hornykiewicz O, Kish SJ. Biochemical pathophysiology of Parkinson's disease. Adv. Neurol. 1987;45:19–34. [PubMed] [Google Scholar]
- 185.Horstink M, Tolosa E, Bonuccelli U, Deuschl G, Friedman A, Kanovsky P, Larsen JP, Lees A, Oertel W, Poewe W, Rascol O, Sampaio C. Review of the therapeutic management of Parkinson's disease. Report of a joint task force of the European Federation of Neurological Societies (EFNS) and the Movement Disorder Society-European Section (MDS-ES). Part II: late (complicated) Parkinson's disease. Eur. J. Neurol. 2006;13:1186–1202. doi: 10.1111/j.1468-1331.2006.01548.x. [DOI] [PubMed] [Google Scholar]
- 186.Horstink M, Tolosa E, Bonuccelli U, Deuschl G, Friedman A, Kanovsky P, Larsen JP, Lees A, Oertel W, Poewe W, Rascol O, Sampaio C. Review of the therapeutic management of Parkinson's disease. Report of a joint task force of the European Federation of Neurological Societies and the Movement Disorder Society-European Section. Part I: early (uncomplicated) Parkinson's disease. Eur. J. Neurol. 2006;13:1170–1185. doi: 10.1111/j.1468-1331.2006.01547.x. [DOI] [PubMed] [Google Scholar]
- 187.Huang CC, Chu NS, Lu CS, Chen RS, Calne DB. Long-term progression in chronic manganism: ten years of follow-up. Neurology. 1998;50:698–700. doi: 10.1212/wnl.50.3.698. [DOI] [PubMed] [Google Scholar]
- 188.Huang CC, Chu NS, Lu CS, Chen RS, Schulzer M, Calne DB. The natural history of neurological manganism over 18 years. Parkinsonism Relat. Disord. 2007;13:143–145. doi: 10.1016/j.parkreldis.2006.09.002. [DOI] [PubMed] [Google Scholar]
- 189.Huang CC, Lu CS, Chu NS, Hochberg F, Lilienfeld D, Olanow W, Calne DB. Progression after chronic manganese exposure. Neurology. 1993;43:1479–1483. doi: 10.1212/wnl.43.8.1479. [DOI] [PubMed] [Google Scholar]
- 190.Hulet SW, Heyliger SO, Powers S, Connor JR. Oligodendrocyte progenitor cells internalize ferritin via clathrin-dependent receptor mediated endocytosis. J. Neurosci. Res. 2000;61:52–60. doi: 10.1002/1097-4547(20000701)61:1<52::AID-JNR6>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
- 191.Hutchinson M, Gurney S, Newson R. GDNF in Parkinson disease: an object lesson in the tyranny of type II. J. Neurosci. Methods. 2007;163:190–192. doi: 10.1016/j.jneumeth.2006.06.015. [DOI] [PubMed] [Google Scholar]
- 192.Hyun DH, Lee M, Halliwell B, Jenner P. Effect of overexpression of wild-type or mutant parkin on the cellular response induced by toxic insults. J. Neurosci. Res. 2005;82:232–244. doi: 10.1002/jnr.20638. [DOI] [PubMed] [Google Scholar]
- 193.Iacovelli L, Fulceri F, De Blasi A, Nicoletti F, Ruggieri S, Fornai F. The neurotoxicity of amphetamines: bridging drugs of abuse and neurodegenerative disorders. Exp. Neurol. 2006;201:24–31. doi: 10.1016/j.expneurol.2006.02.130. [DOI] [PubMed] [Google Scholar]
- 194.Iancu R, Mohapel P, Brundin P, Paul G. Behavioral characterization of a unilateral 6-OHDA-lesion model of Parkinson's disease in mice. Behav. Brain Res. 2005;162:1–10. doi: 10.1016/j.bbr.2005.02.023. [DOI] [PubMed] [Google Scholar]
- 195.Ibanez P, Bonnet AM, Debarges B, Lohmann E, Tison F, Pollak P, Agid Y, Durr A, Brice A. Causal relation between alpha-synuclein gene duplication and familial Parkinson's disease. Lancet. 2004;364:1169–1171. doi: 10.1016/S0140-6736(04)17104-3. [DOI] [PubMed] [Google Scholar]
- 196.Inden M, Taira T, Kitamura Y, Yanagida T, Tsuchiya D, Takata K, Yanagisawa D, Nishimura K, Taniguchi T, Kiso Y, Yoshimoto K, Agatsuma T, Koide-Yoshida S, Iguchi-Ariga SM, Shimohama S, Ariga H. PARK7 DJ-1 protects against degeneration of nigral dopaminergic neurons in Parkinson's disease rat model. Neurobiol. Dis. 2006;24:144–158. doi: 10.1016/j.nbd.2006.06.004. [DOI] [PubMed] [Google Scholar]
- 197.Irizarry MC, Growdon W, Gomez-Isla T, Newell K, George JM, Clayton DF, Hyman BT. Nigral and cortical Lewy bodies and dystrophic nigral neurites in Parkinson's disease and cortical Lewy body disease contain alpha-synuclein immunoreactivity. J. Neuropathol. Exp. Neurol. 1998;57:334–337. doi: 10.1097/00005072-199804000-00005. [DOI] [PubMed] [Google Scholar]
- 198.Jakes R, Spillantini MG, Goedert M. Identification of two distinct synucleins from human brain. FEBS Lett. 1994;345:27–32. doi: 10.1016/0014-5793(94)00395-5. [DOI] [PubMed] [Google Scholar]
- 199.Jakowec MW, Petzinger GM. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned model of parkinson's disease, with emphasis on mice and nonhuman primates. Comp. Med. 2004;54:497–513. [PubMed] [Google Scholar]
- 200.Jenner P, Olanow CW. The pathogenesis of cell death in Parkinson's disease. Neurology. 2006;66:S24–36. doi: 10.1212/wnl.66.10_suppl_4.s24. [DOI] [PubMed] [Google Scholar]
- 201.Johnson CC, Gorell JM, Rybicki BA, Sanders K, Peterson EL. Adult nutrient intake as a risk factor for Parkinson's disease. Int. J. Epidemiol. 1999;28:1102–1109. doi: 10.1093/ije/28.6.1102. [DOI] [PubMed] [Google Scholar]
- 202.Jonsson G, Sachs C. Actions of 6-hydroxydopamine quinones on catecholamine neurons. J. Neurochem. 1975;25:509–516. doi: 10.1111/j.1471-4159.1975.tb04357.x. [DOI] [PubMed] [Google Scholar]
- 203.Junn E, Mouradian MM. Human alpha-synuclein over-expression increases intracellular reactive oxygen species levels and susceptibility to dopamine. Neurosci. Lett. 2002;320:146–150. doi: 10.1016/s0304-3940(02)00016-2. [DOI] [PubMed] [Google Scholar]
- 204.Kahle PJ, Neumann M, Ozmen L, Muller V, Odoy S, Okamoto N, Jacobsen H, Iwatsubo T, Trojanowski JQ, Takahashi H, Wakabayashi K, Bogdanovic N, Riederer P, Kretzschmar HA, Haass C. Selective insolubility of alpha-synuclein in human Lewy body diseases is recapitulated in a transgenic mouse model. Am. J. Pathol. 2001;159:2215–2225. doi: 10.1016/s0002-9440(10)63072-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Kaur D, Peng J, Chinta SJ, Rajagopalan S, Di Monte DA, Cherny RA, Andersen JK. Increased murine neonatal iron intake results in Parkinson-like neurodegeneration with age. Neurobiol. Aging. 2006;28(6):907–913. doi: 10.1016/j.neurobiolaging.2006.04.003. [DOI] [PubMed] [Google Scholar]
- 206.Kaur D, Rajagopalan S, Chinta S, Kumar J, Di Monte D, Cherny RA, Andersen JK. Chronic ferritin expression within murine dopaminergic midbrain neurons results in a progressive age-related neurodegeneration. Brain Res. 2007;1140:188–194. doi: 10.1016/j.brainres.2006.03.006. [DOI] [PubMed] [Google Scholar]
- 207.Kaur D, Yantiri F, Rajagopalan S, Kumar J, Mo JQ, Boonplueang R, Viswanath V, Jacobs R, Yang L, Beal MF, DiMonte D, Volitaskis I, Ellerby L, Cherny RA, Bush AI, Andersen JK. Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: a novel therapy for Parkinson's disease. Neuron. 2003;37:899–909. doi: 10.1016/s0896-6273(03)00126-0. [DOI] [PubMed] [Google Scholar]
- 208.Keller JN, Hanni KB, Markesbery WR. Impaired proteasome function in Alzheimer's disease. J. Neurochem. 2000;75:436–439. doi: 10.1046/j.1471-4159.2000.0750436.x. [DOI] [PubMed] [Google Scholar]
- 209.Keyer K, Imlay JA. Superoxide accelerates DNA damage by elevating free-iron levels. Proc. Natl. Acad. Sci. USA. 1996;93:13635–13640. doi: 10.1073/pnas.93.24.13635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Keyer K, Imlay JA. Inactivation of dehydratase [4Fe-4S] clusters and disruption of iron homeostasis upon cell exposure to peroxynitrite. J. Biol. Chem. 1997;272:27652–27659. doi: 10.1074/jbc.272.44.27652. [DOI] [PubMed] [Google Scholar]
- 211.Kihira T, Yoshida S, Yase Y, Ono S, Kondo T. Chronic low-Ca/Mg high-Al diet induces neuronal loss. Neuropathology. 2002;22:171–179. doi: 10.1046/j.1440-1789.2002.00441.x. [DOI] [PubMed] [Google Scholar]
- 212.Kikinis Z, Eisenstein RS, Bettany AJ, Munro HN. Role of RNA secondary structure of the iron-responsive element in translational regulation of ferritin synthesis. Nucleic Acids Res. 1995;23:4190–4195. doi: 10.1093/nar/23.20.4190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, Wakeham A, You-Ten AJ, Kalia SK, Horne P, Westaway D, Lozano AM, Anisman H, Park DS, Mak TW. Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc. Natl. Acad. Sci. USA. 2005;102:5215–5220. doi: 10.1073/pnas.0501282102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Kish SJ, Shannak K, Hornykiewicz O. Uneven pattern of dopamine loss in the striatum of patients with idiopathic Parkinson's disease. Pathophysiologic and clinical implications. N. Engl. J. Med. 1988;318:876–880. doi: 10.1056/NEJM198804073181402. [DOI] [PubMed] [Google Scholar]
- 215.Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998;392:605–608. doi: 10.1038/33416. [DOI] [PubMed] [Google Scholar]
- 216.Klatzo I, Wisniewski H, Streicher E. Experimental production of neurofibrillary degeneration. I. Light microscopic observations. J. Neuropathol. Exp. Neurol. 1965;24:187–199. doi: 10.1097/00005072-196504000-00002. [DOI] [PubMed] [Google Scholar]
- 217.Kleiner-Fisman G, Fisman DN, Sime E, Saint-Cyr JA, Lozano AM, Lang AE. Long-term follow up of bilateral deep brain stimulation of the subthalamic nucleus in patients with advanced Parkinson disease. J. Neurosurg. 2003;99:489–495. doi: 10.3171/jns.2003.99.3.0489. [DOI] [PubMed] [Google Scholar]
- 218.Klivenyi P, Andreassen OA, Ferrante RJ, Dedeoglu A, Mueller G, Lancelot E, Bogdanov M, Andersen JK, Jiang D, Beal MF. Mice deficient in cellular glutathione peroxidase show increased vulnerability to malonate, 3-nitropropionic acid, and 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. J. Neurosci. 2000;20:1–7. doi: 10.1523/JNEUROSCI.20-01-00001.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Koeppen AH. The history of iron in the brain. J. Neurol. Sci. 1995;134(Suppl):1–9. doi: 10.1016/0022-510x(95)00202-d. [DOI] [PubMed] [Google Scholar]
- 220.Kogan FJ, Nichols WK, Gibb JW. Influence of methamphetamine on nigral and striatal tyrosine hydroxylase activity and on striatal dopamine levels. Eur. J. Pharmacol. 1976;36:363–371. doi: 10.1016/0014-2999(76)90090-x. [DOI] [PubMed] [Google Scholar]
- 221.Kondo T, Ito T, Sugita Y. Bromocriptine scavenges methamphetamine-induced hydroxyl radicals and attenuates dopamine depletion in mouse striatum. Ann. N. Y. Acad. Sci. 1994;738:222–229. doi: 10.1111/j.1749-6632.1994.tb21807.x. [DOI] [PubMed] [Google Scholar]
- 222.Kordower JH, Emborg ME, Bloch J, Ma SY, Chu Y, Leventhal L, McBride J, Chen EY, Palfi S, Roitberg BZ, Brown WD, Holden JE, Pyzalski R, Taylor MD, Carvey P, Ling Z, Trono D, Hantraye P, Deglon N, Aebischer P. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease. Science. 2000;290:767–773. doi: 10.1126/science.290.5492.767. [DOI] [PubMed] [Google Scholar]
- 223.Kordower JH, Palfi S, Chen EY, Ma SY, Sendera T, Cochran EJ, Mufson EJ, Penn R, Goetz CG, Comella CD. Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson's disease. Ann. Neurol. 1999;46:419–424. doi: 10.1002/1531-8249(199909)46:3<419::aid-ana21>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
- 224.Kowall NW, Pendlebury WW, Kessler JB, Perl DP, Beal MF. Aluminum-induced neurofibrillary degeneration affects a subset of neurons in rabbit cerebral cortex, basal forebrain and upper brainstem. Neuroscience. 1989;29:329–337. doi: 10.1016/0306-4522(89)90060-2. [DOI] [PubMed] [Google Scholar]
- 225.Kozlowski DA, Connor B, Tillerson JL, Schallert T, Bohn MC. Delivery of a GDNF gene into the substantia nigra after a progressive 6-OHDA lesion maintains functional nigrostriatal connections. Exp. Neurol. 2000;166:1–15. doi: 10.1006/exnr.2000.7463. [DOI] [PubMed] [Google Scholar]
- 226.Krack P, Batir A, Van Blercom N, Chabardes S, Fraix V, Ardouin C, Koudsie A, Limousin PD, Benazzouz A, LeBas JF, Benabid AL, Pollak P. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson's disease. N. Engl. J. Med. 2003;349:1925–1934. doi: 10.1056/NEJMoa035275. [DOI] [PubMed] [Google Scholar]
- 227.Kruger R, Kuhn W, Leenders KL, Sprengelmeyer R, Muller T, Woitalla D, Portman AT, Maguire RP, Veenma L, Schroder U, Schols L, Epplen JT, Riess O, Przuntek H. Familial parkinsonism with synuclein pathology: clinical and PET studies of A30P mutation carriers. Neurology. 2001;56:1355–1362. doi: 10.1212/wnl.56.10.1355. [DOI] [PubMed] [Google Scholar]
- 228.Kubo S, Nemani VM, Chalkley RJ, Anthony MD, Hattori N, Mizuno Y, Edwards RH, Fortin DL. A combinatorial code for the interaction of alpha-synuclein with membranes. J. Biol. Chem. 2005;280:31664–31672. doi: 10.1074/jbc.M504894200. [DOI] [PubMed] [Google Scholar]
- 229.Kurlan R. "Levodopa phobia": a new iatrogenic cause of disability in Parkinson disease. Neurology. 2005;64:923–924. doi: 10.1212/01.WNL.0000152880.77812.5B. [DOI] [PubMed] [Google Scholar]
- 230.Kuroda Y, Mitsui T, Kunishige M, Matsumoto T. Parkin affects mitochondrial function and apoptosis in neuronal and myogenic cells. Biochem. Biophys. Res. Commun. 2006;348:787–793. doi: 10.1016/j.bbrc.2006.06.201. [DOI] [PubMed] [Google Scholar]
- 231.Kuroda Y, Mitsui T, Kunishige M, Shono M, Akaike M, Azuma H, Matsumoto T. Parkin enhances mitochondrial biogenesis in proliferating cells. Hum. Mol. Genet. 2006;15:883–895. doi: 10.1093/hmg/ddl006. [DOI] [PubMed] [Google Scholar]
- 232.Lang AE, Gill S, Patel NK, Lozano A, Nutt JG, Penn R, Brooks DJ, Hotton G, Moro E, Heywood P, Brodsky MA, Burchiel K, Kelly P, Dalvi A, Scott B, Stacy M, Turner D, Wooten VG, Elias WJ, Laws ER, Dhawan V, Stoessl AJ, Matcham J, Coffey RJ, Traub M. Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann. Neurol. 2006;59:459–466. doi: 10.1002/ana.20737. [DOI] [PubMed] [Google Scholar]
- 233.Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science. 1983;219:979–980. doi: 10.1126/science.6823561. [DOI] [PubMed] [Google Scholar]
- 234.Langston JW, BallardP.A Jr. Parkinson's disease in a chemist working with 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. N. Engl. J. Med. 1983;309:310. doi: 10.1056/nejm198308043090511. [DOI] [PubMed] [Google Scholar]
- 235.Lavara-Culebras E, Paricio N. Drosophila DJ-1 mutants are sensitive to oxidative stress and show reduced lifespan and motor deficits. Gene. 2007;400:158–165. doi: 10.1016/j.gene.2007.06.013. [DOI] [PubMed] [Google Scholar]
- 236.LaVaute T, Smith S, Cooperman S, Iwai K, Land W, Meyron-Holtz E, Drake SK, Miller G, Abu-Asab M, Tsokos M, Switzer R 3rd, Grinberg A, Love P, Tresser N, Rouault TA. Targeted deletion of the gene encoding iron regulatory protein-2 causes misregulation of iron metabolism and neurodegenerative disease in mice. Nat. Genet. 2001;27:209–214. doi: 10.1038/84859. [DOI] [PubMed] [Google Scholar]
- 237.Lee CS, Park WJ, Ko HH, Han ES. Differential involvement of mitochondrial permeability transition in cytotoxicity of 1-methyl-4-phenylpyridinium and 6-hydroxydopamine. Mol. Cell Biochem. 2006;289:193–200. doi: 10.1007/s11010-006-9164-0. [DOI] [PubMed] [Google Scholar]
- 238.Lee J, Zhu WM, Stanic D, Finkelstein DI, Horne MH, Henderson J, Lawrence AJ, O'Connor L, Tomas D, Drago J, Horne MK. Sprouting of dopamine terminals and altered dopamine release and uptake in Parkinsonian dyskinaesia. Brain. 2008;131:1574–1587. doi: 10.1093/brain/awn085. [DOI] [PubMed] [Google Scholar]
- 239.Lee M, Hyun D, Halliwell B, Jenner P. Effect of the overexpression of wild-type or mutant alpha-synuclein on cell susceptibility to insult. J. Neurochem. 2001;76:998–1009. doi: 10.1046/j.1471-4159.2001.00149.x. [DOI] [PubMed] [Google Scholar]
- 240.Leroy E, Boyer R, Auburger G, Leube B, Ulm G, Mezey E, Harta G, Brownstein MJ, Jonnalagada S, Chernova T, Dehejia A, Lavedan C, Gasser T, Steinbach PJ, Wilkinson KD, Polymeropoulos MH. The ubiquitin pathway in Parkinson's disease. Nature. 1998;395:451–452. doi: 10.1038/26652. [DOI] [PubMed] [Google Scholar]
- 241.Leroy E, Boyer R, Polymeropoulos MH. Intron-exon structure of ubiquitin c-terminal hydrolase-L1. DNA Res. 1998;5:397–400. doi: 10.1093/dnares/5.6.397. [DOI] [PubMed] [Google Scholar]
- 242.LeVine SM. Iron deposits in multiple sclerosis and Alzheimer's disease brains. Brain Res. 1997;760:298–303. doi: 10.1016/s0006-8993(97)00470-8. [DOI] [PubMed] [Google Scholar]
- 243.Levites Y, Weinreb O, Maor G, Youdim MB, Mandel S. Green tea polyphenol (-)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J. Neurochem. 2001;78:1073–1082. doi: 10.1046/j.1471-4159.2001.00490.x. [DOI] [PubMed] [Google Scholar]
- 244.Levites Y, Youdim MB, Maor G, Mandel S. Attenuation of 6-hydroxydopamine (6-OHDA)-induced nuclear factor-kappaB (NF-kappaB) activation and cell death by tea extracts in neuronal cultures. Biochem. Pharmacol. 2002;63:21–29. doi: 10.1016/s0006-2952(01)00813-9. [DOI] [PubMed] [Google Scholar]
- 245.Li W, Lesuisse C, Xu Y, Troncoso JC, Price DL, Lee MK. Stabilization of alpha-synuclein protein with aging and familial parkinson's disease-linked A53T mutation. J. Neurosci. 2004;24:7400–7409. doi: 10.1523/JNEUROSCI.1370-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Liccione JJ, Maines MD. Selective vulnerability of glutathione metabolism and cellular defense mechanisms in rat striatum to manganese. J. Pharmacol. Exp. Ther. 1988;247:156–161. [PubMed] [Google Scholar]
- 247.Lincoln S, Vaughan J, Wood N, Baker M, Adamson J, Gwinn-Hardy K, Lynch T, Hardy J, Farrer M. Low frequency of pathogenic mutations in the ubiquitin carboxy-terminal hydrolase gene in familial Parkinson's disease. Neuroreport. 1999;10:427–429. doi: 10.1097/00001756-199902050-00040. [DOI] [PubMed] [Google Scholar]
- 248.Ling ZD, Potter ED, Lipton JW, Carvey PM. Differentiation of mesencephalic progenitor cells into dopaminergic neurons by cytokines. Exp. Neurol. 1998;149:411–423. doi: 10.1006/exnr.1998.6715. [DOI] [PubMed] [Google Scholar]
- 249.Liu S, Ninan I, Antonova I, Battaglia F, Trinchese F, Narasanna A, Kolodilov N, Dauer W, Hawkins RD, Arancio O. alpha-Synuclein produces a long-lasting increase in neurotransmitter release. EMBO J. 2004;23:4506–4516. doi: 10.1038/sj.emboj.7600451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Liu Y, Fallon L, Lashuel HA, Liu Z, Lansbury P.T Jr. The UCH-L1 gene encodes two opposing enzymatic activities that affect alpha-synuclein degradation and Parkinson's disease susceptibility. Cell. 2002;111:209–218. doi: 10.1016/s0092-8674(02)01012-7. [DOI] [PubMed] [Google Scholar]
- 251.Lo Bianco C, Schneider BL, Bauer M, Sajadi A, Brice A, Iwatsubo T, Aebischer P. Lentiviral vector delivery of parkin prevents dopaminergic degeneration in an alpha-synuclein rat model of Parkinson's disease. Proc. Natl. Acad. Sci. USA. 2004 ;101:17510–17515. doi: 10.1073/pnas.0405313101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Logroscino G, Marder K, Cote L, Tang MX, Shea S, Mayeux R. Dietary lipids and antioxidants in Parkinson's disease: a population-based, case-control study. Ann. Neurol. 1996 ;39:89–94. doi: 10.1002/ana.410390113. [DOI] [PubMed] [Google Scholar]
- 253.Logroscino G, Marder K, Graziano J, Freyer G, Slavkovich V, Lojacono N, Cote L, Mayeux R. Dietary iron, animal fats, and risk of Parkinson's disease. Mov. Disord. 1998;13(1):13–16. [PubMed] [Google Scholar]
- 254.Lotharius J, Barg S, Wiekop P, Lundberg C, Raymon HK, Brundin P. Effect of mutant alpha-synuclein on dopamine homeostasis in a new human mesencephalic cell line. J. Biol. Chem. 2002;277:38884–38894. doi: 10.1074/jbc.M205518200. [DOI] [PubMed] [Google Scholar]
- 255.Lotharius J, Brundin P. Pathogenesis of Parkinson's disease: dopamine, vesicles and alpha-synuclein. Nat. Rev. Neurosci. 2002;3:932–942. doi: 10.1038/nrn983. [DOI] [PubMed] [Google Scholar]
- 256.Lucking CB, Abbas N, Durr A, Bonifati V, Bonnet AM, de Broucker T, De Michele G, Wood NW, Agid Y, Brice A. Homozygous deletions in parkin gene in European and North African families with autosomal recessive juvenile parkinsonism. The European Consortium on Genetic Susceptibility in Parkinson's Disease and the French Parkinson's Disease Genetics Study Group. Lancet. 1998;352:1355–1356. doi: 10.1016/s0140-6736(05)60746-5. [DOI] [PubMed] [Google Scholar]
- 257.MacMillan-Crow LA, Crow JP, Thompson JA. Peroxynitrite-mediated inactivation of manganese superoxide dismutase involves nitration and oxidation of critical tyrosine residues. Biochemistry. 1998;37:1613–1622. doi: 10.1021/bi971894b. [DOI] [PubMed] [Google Scholar]
- 258.Magaki S, Mueller C, Yellon SM, Fox J, Kim J, Snissarenko E, Chin V, Ghosh MC, Kirsch WM. Regional dissection and determination of loosely bound and non-heme iron in the developing mouse brain. Brain Res. 2007;1158:144–150. doi: 10.1016/j.brainres.2007.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Mann VM, Cooper JM, Daniel SE, Srai K, Jenner P, Marsden CD, Schapira AH. Complex I, iron, and ferritin in Parkinson's disease substantia nigra. Ann. Neurol. 1994;36:876–881. doi: 10.1002/ana.410360612. [DOI] [PubMed] [Google Scholar]
- 260.Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Di Monte DA. The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein. J. Biol. Chem. 2002;277:1641–1644. doi: 10.1074/jbc.C100560200. [DOI] [PubMed] [Google Scholar]
- 261.Maraganore DM, Farrer MJ, Hardy JA, Lincoln SJ, McDonnell SK, Rocca WA. Case-control study of the ubiquitin carboxy-terminal hydrolase L1 gene in Parkinson's disease. Neurology. 1999;53:1858–1860. doi: 10.1212/wnl.53.8.1858. [DOI] [PubMed] [Google Scholar]
- 262.Maraganore DM, Lesnick TG, Elbaz A, Chartier-Harlin MC, Gasser T, Kruger R, Hattori N, Mellick GD, Quattrone A, Satoh J, Toda T, Wang J, Ioannidis JP, de Andrade M, Rocca WA. UCHL1 is a Parkinson's disease susceptibility gene. Ann. Neurol. 2004;55:512–521. doi: 10.1002/ana.20017. [DOI] [PubMed] [Google Scholar]
- 263.Marsden CD. Pigmentation in the nucleus substantiae nigrae of mammals. J. Anat. 1961;95:256–261. [PMC free article] [PubMed] [Google Scholar]
- 264.Marsden CD, Olanow CW. The causes of Parkinson's disease are being unraveled and rational neuroprotective therapy is close to reality. Ann. Neurol. 1998;44:S189–196. doi: 10.1002/ana.410440727. [DOI] [PubMed] [Google Scholar]
- 265.Marsden CD, Parkes JD. "On-off" effects in patients with Parkinson's disease on chronic levodopa therapy. Lancet. 1976;1:292–296. doi: 10.1016/s0140-6736(76)91416-1. [DOI] [PubMed] [Google Scholar]
- 266.Marsden CD, Parkes JD. Success and problems of long-term levodopa therapy in Parkinson's disease. Lancet. 1977;1:345–349. doi: 10.1016/s0140-6736(77)91146-1. [DOI] [PubMed] [Google Scholar]
- 267.Martin WR, Ye FQ, Allen PS. Increasing striatal iron content associated with normal aging. Mov. Disord. 1998;13:281–286. doi: 10.1002/mds.870130214. [DOI] [PubMed] [Google Scholar]
- 268.Martyn CN, Coggon DN, Inskip H, Lacey RF, Young WF. Aluminum concentrations in drinking water and risk of Alzheimer's disease. Epidemiology. 1997;8:281–286. doi: 10.1097/00001648-199705000-00009. [DOI] [PubMed] [Google Scholar]
- 269.Mash DC, Ouyang Q, Pablo J, Basile M, Izenwasser S, Lieberman A, Perrin RJ. Cocaine abusers have an overexpression of alpha-synuclein in dopamine neurons. J. Neurosci. 2003;23:2564–2571. doi: 10.1523/JNEUROSCI.23-07-02564.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Maskri L, Zhu X, Fritzen S, Kuhn K, Ullmer C, Engels P, Andriske M, Stichel CC, Lubbert H. Influence of different promoters on the expression pattern of mutated human alpha-synuclein in transgenic mice. Neurodegener. Dis. 2004;1:255–265. doi: 10.1159/000085064. [DOI] [PubMed] [Google Scholar]
- 271.Mastakov MY, Baer K, Symes CW, Leichtlein CB, Kotin RM, During MJ. Immunological aspects of recombinant adeno-associated virus delivery to the mammalian brain. J. Virol. 2002;76:8446–8454. doi: 10.1128/JVI.76.16.8446-8454.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Masuda T, Hida H, Kanda Y, Aihara N, Ohta K, Yamada K, Nishino H. Oral administration of metal chelator ameliorates motor dysfunction after a small hemorrhage near the internal capsule in rat. J. Neurosci. Res. 2007;85:213–222. doi: 10.1002/jnr.21089. [DOI] [PubMed] [Google Scholar]
- 273.Matsumine H, Saito M, Shimoda-Matsubayashi S, Tanaka H, Ishikawa A, Nakagawa-Hattori Y, Yokochi M, Kobayashi T, Igarashi S, Takano H, Sanpei K, Koike R, Mori H, Kondo T, Mizutani Y, Schaffer AA, Yamamura Y, Nakamura S, Kuzuhara S, Tsuji S, Mizuno Y. Localization of a gene for an autosomal recessive form of juvenile Parkinsonism to chromosome 6q25.2-27. Am. J. Hum. Genet. 1997;60:588–596. [PMC free article] [PubMed] [Google Scholar]
- 274.Maynard CJ, Cappai R, Volitakis I, Cherny RA, White AR, Beyreuther K, Masters CL, Bush AI, Li QX. Overexpression of Alzheimer's disease amyloid-beta opposes the age-dependent elevations of brain copper and iron. J. Biol. Chem. 2002;277:44670–44676. doi: 10.1074/jbc.M204379200. [DOI] [PubMed] [Google Scholar]
- 275.McCormack AL, Atienza JG, Johnston LC, Andersen JK, Vu S, Di Monte DA. Role of oxidative stress in paraquat-induced dopaminergic cell degeneration. J. Neurochem. 2005;93:1030–1037. doi: 10.1111/j.1471-4159.2005.03088.x. [DOI] [PubMed] [Google Scholar]
- 276.McCormack AL, Thiruchelvam M, Manning-Bog AB, Thiffault C, Langston JW, Cory-Slechta DA, Di Monte DA. Environmental risk factors and Parkinson's disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol. Dis. 2002;10:119–127. doi: 10.1006/nbdi.2002.0507. [DOI] [PubMed] [Google Scholar]
- 277.McCown TJ, Xiao X, Li J, Breese GR, Samulski RJ. Differential and persistent expression patterns of CNS gene transfer by an adeno-associated virus (AAV) vector. Brain Res. 1996;713:99–107. doi: 10.1016/0006-8993(95)01488-8. [DOI] [PubMed] [Google Scholar]
- 278.McNaught KS, Belizaire R, Isacson O, Jenner P, Olanow CW. Altered proteasomal function in sporadic Parkinson's disease. Exp. Neurol. 2003;179:38–46. doi: 10.1006/exnr.2002.8050. [DOI] [PubMed] [Google Scholar]
- 279.McNaught KS, Bjorklund LM, Belizaire R, Isacson O, Jenner P, Olanow CW. Proteasome inhibition causes nigral degeneration with inclusion bodies in rats. Neuroreport. 2002;13:1437–1441. doi: 10.1097/00001756-200208070-00018. [DOI] [PubMed] [Google Scholar]
- 280.McNaught KS, Mytilineou C, Jnobaptiste R, Yabut J, Shashidharan P, Jennert P, Olanow CW. Impairment of the ubiquitin-proteasome system causes dopaminergic cell death and inclusion body formation in ventral mesencephalic cultures. J. Neurochem. 2002;81:301–306. doi: 10.1046/j.1471-4159.2002.00821.x. [DOI] [PubMed] [Google Scholar]
- 281.McNaught KS, Perl DP, Brownell AL, Olanow CW. Systemic exposure to proteasome inhibitors causes a progressive model of Parkinson's disease. Ann. Neurol. 2004;56:149–162. doi: 10.1002/ana.20186. [DOI] [PubMed] [Google Scholar]
- 282.McNaught KS, Shashidharan P, Perl DP, Jenner P, Olanow CW. Aggresome-related biogenesis of Lewy bodies. Eur. J. Neurosci. 2002;16:2136–2148. doi: 10.1046/j.1460-9568.2002.02301.x. [DOI] [PubMed] [Google Scholar]
- 283.Meglio L, Oteiza PI. Aluminum enhances melanin-induced lipid peroxidation. Neurochem. Res. 1999;24:1001–1008. doi: 10.1023/a:1021000709082. [DOI] [PubMed] [Google Scholar]
- 284.Melo TM, Larsen C, White LR, Aasly J, Sjobakk TE, Flaten TP, Sonnewald U, Syversen T. Manganese, copper, and zinc in cerebrospinal fluid from patients with multiple sclerosis. Biol. Trace Elem. Res. 2003;93:1–8. doi: 10.1385/BTER:93:1-3:1. [DOI] [PubMed] [Google Scholar]
- 285.Mendez-Alvarez E, Soto-Otero R, Hermida-Ameijeiras A, Lopez-Real AM, Labandeira-Garcia JL. Effects of aluminum and zinc on the oxidative stress caused by 6-hydroxy-dopamine autoxidation: relevance for the pathogenesis of Parkinson's disease. Biochim. Biophys. Acta. 2002;1586:155–168. doi: 10.1016/s0925-4439(01)00077-1. [DOI] [PubMed] [Google Scholar]
- 286.Menzies FM, Yenisetti SC, Min KT. Roles of Drosophila DJ-1 in survival of dopaminergic neurons and oxidative stress. Curr. Biol. 2005;15:1578–1582. doi: 10.1016/j.cub.2005.07.036. [DOI] [PubMed] [Google Scholar]
- 287.Meredith GE, Halliday GM, Totterdell S. A critical review of the development and importance of proteinaceous aggregates in animal models of Parkinson's disease: new insights into Lewy body formation. Parkinsonism Relat. Disord. 2004;10:191–202. doi: 10.1016/j.parkreldis.2004.01.001. [DOI] [PubMed] [Google Scholar]
- 288.Meulener MC, Xu K, Thomson L, Ischiropoulos H, Bonini NM. Mutational analysis of DJ-1 in Drosophila implicates functional inactivation by oxidative damage and aging. Proc. Natl. Acad. Sci. USA. 2006;103:12517–12522. doi: 10.1073/pnas.0601891103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Middleton E Jr. Effect of plant flavonoids on immune and inflammatory cell function. Adv. Exp. Med. Biol. 1998;439:175–182. doi: 10.1007/978-1-4615-5335-9_13. [DOI] [PubMed] [Google Scholar]
- 290.Momose Y, Murata M, Kobayashi K, Tachikawa M, Nakabayashi Y, Kanazawa I, Toda T. Association studies of multiple candidate genes for Parkinson's disease using single nucleotide polymorphisms. Ann. Neurol. 2002;51:133–136. doi: 10.1002/ana.10079. [DOI] [PubMed] [Google Scholar]
- 291.Moon Y, Lee KH, Park JH, Geum D, Kim K. Mitochondrial membrane depolarization and the selective death of dopaminergic neurons by rotenone: protective effect of coenzyme Q10. J. Neurochem. 2005;93:1199–1208. doi: 10.1111/j.1471-4159.2005.03112.x. [DOI] [PubMed] [Google Scholar]
- 292.Moore PB, Day JP, Taylor GA, Ferrier IN, Fifield LK, Edwardson JA. Absorption of aluminium-26 in Alzheimer's disease, measured using accelerator mass spectrometry. Dement. Geriatr. Cogn. Disord. 2000;11:66–69. doi: 10.1159/000017216. [DOI] [PubMed] [Google Scholar]
- 293.Morris CM, Candy JM, Oakley AE, Taylor GA, Mountfort S, Bishop H, Ward MK, Bloxham CA, Edwardson JA. Comparison of the regional distribution of transferrin receptors and aluminium in the forebrain of chronic renal dialysis patients. J. Neurol. Sci. 1989;94:295–306. doi: 10.1016/0022-510x(89)90238-4. [DOI] [PubMed] [Google Scholar]
- 294.Morris CM, Keith AB, Edwardson JA, Pullen RG. Uptake and distribution of iron and transferrin in the adult rat brain. J. Neurochem. 1992;59:300–306. doi: 10.1111/j.1471-4159.1992.tb08904.x. [DOI] [PubMed] [Google Scholar]
- 295.Morrish PK, Rakshi JS, Bailey DL, Sawle GV, Brooks DJ. Measuring the rate of progression and estimating the preclinical period of Parkinson's disease with [18F]dopa PET. J. Neurol. Neurosurg. Psychiatry. 1998;64:314–319. doi: 10.1136/jnnp.64.3.314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Mosharov EV, Staal RG, Bove J, Prou D, Hananiya A, Markov D, Poulsen N, Larsen KE, Moore CM, Troyer MD, Edwards RH, Przedborski S, Sulzer D. Alpha-synuclein overexpression increases cytosolic catecholamine concentration. J. Neurosci. 2006;26:9304–9311. doi: 10.1523/JNEUROSCI.0519-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Muenter MD, Forno LS, Hornykiewicz O, Kish SJ, Maraganore DM, Caselli RJ, Okazaki H, Howard FM, Jr., Snow BJ, Calne DB. Hereditary form of parkinsonism--dementia. Ann. Neurol. 1998;43:768–781. doi: 10.1002/ana.410430612. [DOI] [PubMed] [Google Scholar]
- 298.Muller T, Hefter H, Hueber R, Jost WH, Leenders KL, Odin P, Schwarz J. Is levodopa toxic? J. Neurol. 2004;251(6):VI/44–46. doi: 10.1007/s00415-004-1610-x. [DOI] [PubMed] [Google Scholar]
- 299.Mytilineou C, McNaught KS, Shashidharan P, Yabut J, Baptiste RJ, Parnandi A, Olanow CW. Inhibition of proteasome activity sensitizes dopamine neurons to protein alterations and oxidative stress. J. Neural. Transm. 2004;111:1237–1251. doi: 10.1007/s00702-004-0167-2. [DOI] [PubMed] [Google Scholar]
- 300.Narayanan V, Scarlata S. Membrane binding and self-association of alpha-synucleins. Biochemistry. 2001;40:9927–9934. doi: 10.1021/bi002952n. [DOI] [PubMed] [Google Scholar]
- 301.Narhi L, Wood SJ, Steavenson S, Jiang Y, Wu GM, Anafi D, Kaufman SA, Martin F, Sitney K, Denis P, Louis JC, Wypych J, Biere AL, Citron M. Both familial Parkinson's disease mutations accelerate alpha-synuclein aggregation. J. Biol. Chem. 1999;274:9843–9846. doi: 10.1074/jbc.274.14.9843. [DOI] [PubMed] [Google Scholar]
- 302.Noack H, Kube U, Augustin W. Relations between tocopherol depletion and coenzyme Q during lipid peroxidation in rat liver mitochondria. Free Radic. Res. 1994;20:375–386. doi: 10.3109/10715769409145637. [DOI] [PubMed] [Google Scholar]
- 303.Norris EH, Giasson BI, Ischiropoulos H, Lee VM. Effects of oxidative and nitrative challenges on alpha-synuclein fibrillogenesis involve distinct mechanisms of protein modifications. J. Biol. Chem. 2003;278:27230–27240. doi: 10.1074/jbc.M212436200. [DOI] [PubMed] [Google Scholar]
- 304.Obeso JA, Rodriguez-Oroz M, Marin C, Alonso F, Zamarbide I, Lanciego JL, Rodriguez-Diaz M. The origin of motor fluctuations in Parkinson's disease: importance of dopaminergic innervation and basal ganglia circuits. Neurology. 2004;62:S17–30. doi: 10.1212/wnl.62.1_suppl_1.s17. [DOI] [PubMed] [Google Scholar]
- 305.Obeso JA, Rodriguez-Oroz MC, Lanciego JL, Rodriguez Diaz M. How does Parkinson's disease begin? The role of compensatory mechanisms. Trends Neurosci. 2004;27:125–127. doi: 10.1016/j.tins.2003.12.006. [DOI] [PubMed] [Google Scholar]
- 306.Obeso JA, Rodriguez-Oroz MC, Rodriguez M, Lanciego JL, Artieda J, Gonzalo N, Olanow CW. Pathophysiology of the basal ganglia in Parkinson's disease. Trends Neurosci. 2000;23:S8–19. doi: 10.1016/s1471-1931(00)00028-8. [DOI] [PubMed] [Google Scholar]
- 307.Odunze IN, Klaidman LK, Adams J.D Jr. MPTP toxicity in the mouse brain and vitamin E. Neurosci. Lett. 1990;108:346–349. doi: 10.1016/0304-3940(90)90665-v. [DOI] [PubMed] [Google Scholar]
- 308.Ogasahara S, Yorifuji S, Nishikawa Y, Takahashi M, Wada K, Hazama T, Nakamura Y, Hashimoto S, Kono N, Tarui S. Improvement of abnormal pyruvate metabolism and cardiac conduction defect with coenzyme Q10 in Kearns-Sayre syndrome. Neurology. 1985;35:372–377. doi: 10.1212/wnl.35.3.372. [DOI] [PubMed] [Google Scholar]
- 309.Ohshima H, Gilibert I, Bianchini F. Induction of DNA strand breakage and base oxidation by nitroxyl anion through hydroxyl radical production. Free Radic. Biol. Med. 1999;26:1305–1313. doi: 10.1016/s0891-5849(98)00327-x. [DOI] [PubMed] [Google Scholar]
- 310.Oikawa S, Hirosawa I, Tada-Oikawa S, Furukawa A, Nishiura K, Kawanishi S. Mechanism for manganese enhancement of dopamine-induced oxidative DNA damage and neuronal cell death. Free Radic. Biol. Med. 2006;41:748–756. doi: 10.1016/j.freeradbiomed.2006.05.018. [DOI] [PubMed] [Google Scholar]
- 311.Olanow CW, Goetz CG, Kordower JH, Stoessl AJ, Sossi V, Brin MF, Shannon KM, Nauert GM, Perl DP, Godbold J, Freeman TB. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease. Ann. Neurol. 2003;54:403–414. doi: 10.1002/ana.10720. [DOI] [PubMed] [Google Scholar]
- 312.Ossowska K, Wardas J, Smialowska M, Kuter K, Lenda T, Wieronska JM, Zieba B, Nowak P, Dabrowska J, Bortel A, Kwiecinski A, Wolfarth S. A slowly developing dysfunction of dopaminergic nigrostriatal neurons induced by long-term paraquat administration in rats: an animal model of preclinical stages of Parkinson's disease? Eur. J. Neurosci. 2005;22:1294–1304. doi: 10.1111/j.1460-9568.2005.04301.x. [DOI] [PubMed] [Google Scholar]
- 313.Ostrerova-Golts N, Petrucelli L, Hardy J, Lee JM, Farer M, Wolozin B. The A53T alpha-synuclein mutation increases iron-dependent aggregation and toxicity. J. Neurosci. 2000;20:6048–6054. doi: 10.1523/JNEUROSCI.20-16-06048.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Pacifici RE, Kono Y, Davies KJ. Hydrophobicity as the signal for selective degradation of hydroxyl radical-modified hemoglobin by the multicatalytic proteinase complex, proteasome. J. Biol. Chem. 1993;268:15405–15411. [PubMed] [Google Scholar]
- 315.Paik SR, Shin HJ, Lee JH, Chang CS, Kim J. Copper(II)-induced self-oligomerization of alpha-synuclein. Biochem. J. 1999;340(Pt 3):821–828. [PMC free article] [PubMed] [Google Scholar]
- 316.Paisan-Ruiz C, Jain S, Evans EW, Gilks WP, Simon J, van der Brug M, Lopez de Munain A, Aparicio S, Gil AM, Khan N, Johnson J, Martinez JR, Nicholl D, Carrera IM, Pena AS, de Silva R, Lees A, Marti-Masso JF, Perez-Tur J, Wood NW, Singleton AB. Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron. 2004;44:595–600. doi: 10.1016/j.neuron.2004.10.023. [DOI] [PubMed] [Google Scholar]
- 317.Palhagen S, Heinonen E, Hagglund J, Kaugesaar T, Maki-Ikola O, Palm R. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology. 2006;66:1200–1206. doi: 10.1212/01.wnl.0000204007.46190.54. [DOI] [PubMed] [Google Scholar]
- 318.Parenti M, Rusconi L, Cappabianca V, Parati EA, Groppetti A. Role of dopamine in manganese neurotoxicity. Brain Res. 1988;473:236–240. doi: 10.1016/0006-8993(88)90852-9. [DOI] [PubMed] [Google Scholar]
- 319.Parish CL, Drago J, Stanic D, Borrelli E, Finkelstein DI, Horne MK. Haloperidol treatment reverses behavioural and anatomical changes in cocaine-dependent mice. Neurobiol. Dis. 2005;19:301–311. doi: 10.1016/j.nbd.2005.01.009. [DOI] [PubMed] [Google Scholar]
- 320.Park J, Kim SY, Cha GH, Lee SB, Kim S, Chung J. Drosophila DJ-1 mutants show oxidative stress-sensitive locomotive dysfunction. Gene. 2005;361:133–139. doi: 10.1016/j.gene.2005.06.040. [DOI] [PubMed] [Google Scholar]
- 321.Patel NK, Heywood P, O'Sullivan K, McCarter R, Love S, Gill SS. Unilateral subthalamotomy in the treatment of Parkinson's disease. Brain. 2003;126:1136–1145. doi: 10.1093/brain/awg111. [DOI] [PubMed] [Google Scholar]
- 322.Pearce RK, Owen A, Daniel S, Jenner P, Marsden CD. Alterations in the distribution of glutathione in the substantia nigra in Parkinson's disease. J. Neural. Transm. 1997;104:661–677. doi: 10.1007/BF01291884. [DOI] [PubMed] [Google Scholar]
- 323.Pedersen WA, Cashman NR, Mattson MP. The lipid peroxidation product 4-hydroxynonenal impairs glutamate and glucose transport and choline acetyltransferase activity in NSC-19 motor neuron cells. Exp. Neurol. 1999;155:1–10. doi: 10.1006/exnr.1998.6890. [DOI] [PubMed] [Google Scholar]
- 324.Perez RG, Waymire JC, Lin E, Liu JJ, Guo F, Zigmond MJ. A role for alpha-synuclein in the regulation of dopamine biosynthesis. J. Neurosci. 2002;22:3090–3099. doi: 10.1523/JNEUROSCI.22-08-03090.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Perl DP. Exposure to aluminium and the subsequent development of a disorder with features of Alzheimer's disease. J. Neurol. Neurosurg. Psychiatry. 2006;77:811. doi: 10.1136/jnnp.2006.090613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Perl DP, Gajdusek DC, Garruto RM, Yanagihara RT, Gibbs CJ. Intraneuronal aluminum accumulation in amyotrophic lateral sclerosis and Parkinsonism-dementia of Guam. Science. 1982;217:1053–1055. doi: 10.1126/science.7112111. [DOI] [PubMed] [Google Scholar]
- 327.Perry TL, Yong VW, Clavier RM, Jones K, Wright JM, Foulks JG, Wall RA. Partial protection from the dopaminergic neurotoxin N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine by four different antioxidants in the mouse. Neurosci. Lett. 1985;60:109–114. doi: 10.1016/0304-3940(85)90229-0. [DOI] [PubMed] [Google Scholar]
- 328.Perry TL, Yong VW, Hansen S, Jones K, Bergeron C, Foulks JG, Wright JM. Alpha-tocopherol and beta-carotene do not protect marmosets against the dopaminergic neurotoxicity of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. J. Neurol. Sci. 1987;81:321–331. doi: 10.1016/0022-510x(87)90106-7. [DOI] [PubMed] [Google Scholar]
- 329.Piccardo P, Yanagihara R, Garruto RM, Gibbs C.J Jr, Gajdusek DC. Histochemical and X-ray microanalytical localization of aluminum in amyotrophic lateral sclerosis and parkinsonism-dementia of Guam. Acta Neuropathol. 1988;77:1–4. doi: 10.1007/BF00688235. [DOI] [PubMed] [Google Scholar]
- 330.Pinero D, Jones B, Beard J. Variations in dietary iron alter behavior in developing rats. J. Nut. 2001;131:311–318. doi: 10.1093/jn/131.2.311. [DOI] [PubMed] [Google Scholar]
- 331.Pitkanen-Arsiola T, Tillman JE, Gu G, Yuan J, Roberts RL, Wantroba M, Coetzee GA, Cookson MS, Kasper S. Androgen and anti-androgen treatment modulates androgen receptor activity and DJ-1 stability. Prostate. 2006;66:1177–1193. doi: 10.1002/pros.20450. [DOI] [PubMed] [Google Scholar]
- 332.Polizzi S, Pira E, Ferrara M, Bugiani M, Papaleo A, Albera R, Palmi S. Neurotoxic effects of aluminium among foundry workers and Alzheimer's disease. Neurotoxicology. 2002;23:761–774. doi: 10.1016/S0161-813X(02)00097-9. [DOI] [PubMed] [Google Scholar]
- 333.Polymeropoulos MH, Higgins JJ, Golbe LI, Johnson WG, Ide SE, Di Iorio G, Sanges G, Stenroos ES, Pho LT, Schaffer AA, Lazzarini AM, Nussbaum RL, Duvoisin RC. Mapping of a gene for Parkinson's disease to chromosome 4q21-q23. Science. 1996;274:1197–1199. doi: 10.1126/science.274.5290.1197. [DOI] [PubMed] [Google Scholar]
- 334.Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio G, Golbe LI, Nussbaum RL. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. 1997;276:2045–2047. doi: 10.1126/science.276.5321.2045. [DOI] [PubMed] [Google Scholar]
- 335.Przedborski S, Kostic V, Jackson-Lewis V, Naini AB, Simonetti S, Fahn S, Carlson E, Epstein CJ, Cadet JL. Transgenic mice with increased Cu/Zn-superoxide dismutase activity are resistant to N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity. J. Neurosci. 1992;12:1658–1667. doi: 10.1523/JNEUROSCI.12-05-01658.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336.Puglielli L, Friedlich AL, Setchell KD, Nagano S, Opazo C, Cherny RA, Barnham KJ, Wade JD, Melov S, Kovacs DM, Bush AI. Alzheimer disease beta-amyloid activity mimics cholesterol oxidase. J. Clin. Invest. 2005;115:2556–2563. doi: 10.1172/JCI23610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337.Racette BA, Tabbal SD, Jennings D, Good L, Perlmutter JS, Evanoff B. Prevalence of parkinsonism and relationship to exposure in a large sample of Alabama welders. Neurology. 2005;64:230–235. doi: 10.1212/01.WNL.0000149511.19487.44. [DOI] [PubMed] [Google Scholar]
- 338.Radi R, Beckman JS, Bush KM, Freeman BA. Peroxynitrite-induced membrane lipid peroxidation: the cytotoxic potential of superoxide and nitric oxide. Arch. Biochem. Biophys. 1991;288:481–487. doi: 10.1016/0003-9861(91)90224-7. [DOI] [PubMed] [Google Scholar]
- 339.Reinheckel T, Sitte N, Ullrich O, Kuckelkorn U, Davies KJ, Grune T. Comparative resistance of the 20S and 26S proteasome to oxidative stress. Biochem. J. 1998;335(Pt 3):637–642. doi: 10.1042/bj3350637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340.Ren YR, Nishida Y, Yoshimi K, Yasuda T, Jishage K, Uchihara T, Yokota T, Mizuno Y, Mochizuki H. Genetic vitamin E deficiency does not affect MPTP susceptibility in the mouse brain. J. Neurochem. 2006;98:1810–1816. doi: 10.1111/j.1471-4159.2006.03994.x. [DOI] [PubMed] [Google Scholar]
- 341.Richardson DR, Mouralian C, Ponka P, Becker E. Development of potential iron chelators for the treatment of Friedreich's ataxia: ligands that mobilize mitochondrial iron. Biochim. Biophys. Acta. 2001;1536:133–140. doi: 10.1016/s0925-4439(01)00041-2. [DOI] [PubMed] [Google Scholar]
- 342.Richter F, Hamann M, Richter A. Chronic rotenone treatment induces behavioral effects but no pathological signs of parkinsonism in mice. J. Neurosci. Res. 2007;85:681–691. doi: 10.1002/jnr.21159. [DOI] [PubMed] [Google Scholar]
- 343.Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, MacGregor L, Kiers L, Cherny R, Li QX, Tammer A, Carrington D, Mavros C, Volitakis I, Xilinas M, Ames D, Davis S, Beyreuther K, Tanzi RE, Masters CL. Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting Abeta amyloid deposition and toxicity in Alzheimer disease: a pilot phase 2 clinical trial. Arch. Neurol. 2003;60:1685–1691. doi: 10.1001/archneur.60.12.1685. [DOI] [PubMed] [Google Scholar]
- 344.Rogaeva E, Johnson J, Lang AE, Gulick C, Gwinn-Hardy K, Kawarai T, Sato C, Morgan A, Werner J, Nussbaum R, Petit A, Okun MS, McInerney A, Mandel R, Groen JL, Fernandez HH, Postuma R, Foote KD, Salehi-Rad S, Liang Y, Reimsnider S, Tandon A, Hardy J, St George-Hyslop P, Singleton AB. Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease. Arch. Neurol. 2004;61:1898–1904. doi: 10.1001/archneur.61.12.1898. [DOI] [PubMed] [Google Scholar]
- 345.Rondeau V, Commenges D, Jacqmin-Gadda H, Dartigues JF. Relation between aluminum concentrations in drinking water and Alzheimer's disease: an 8-year follow-up study. Am. J. Epidemiol. 2000;152:59–66. doi: 10.1093/aje/152.1.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346.Roy NS, Cleren C, Singh SK, Yang L, Beal MF, Goldman SA. Functional engraftment of human ES cell-derived dopaminergic neurons enriched by coculture with telomerase-immortalized midbrain astrocytes. Nat. Med. 2006;12:1259–1268. doi: 10.1038/nm1495. [DOI] [PubMed] [Google Scholar]
- 347.Saigoh K, Wang YL, Suh JG, Yamanishi T, Sakai Y, Kiyosawa H, Harada T, Ichihara N, Wakana S, Kikuchi T, Wada K. Intragenic deletion in the gene encoding ubiquitin carboxy-terminal hydrolase in gad mice. Nat. Genet. 1999;23:47–51. doi: 10.1038/12647. [DOI] [PubMed] [Google Scholar]
- 348.Salah N, Miller NJ, Paganga G, Tijburg L, Bolwell GP, Rice-Evans C. Polyphenolic flavanols as scavengers of aqueous phase radicals and as chain-breaking antioxidants. Arch. Biochem. Biophys. 1995;322:339–346. doi: 10.1006/abbi.1995.1473. [DOI] [PubMed] [Google Scholar]
- 349.Saper CB, Petito CK. Correspondence of melanin-pigmented neurons in human brain with A1-A14 catecholamine cell groups. Brain. 1982;105:87–101. doi: 10.1093/brain/105.1.87. [DOI] [PubMed] [Google Scholar]
- 350.Sapru MK, Yates JW, Hogan S, Jiang L, Halter J, Bohn MC. Silencing of human alpha-synuclein in vitro and in rat brain using lentiviral-mediated RNAi. Exp. Neurol. 2006;198:382–390. doi: 10.1016/j.expneurol.2005.12.024. [DOI] [PubMed] [Google Scholar]
- 351.Satoh J, Kuroda Y. A polymorphic variation of serine to tyrosine at codon 18 in the ubiquitin C-terminal hydrolase-L1 gene is associated with a reduced risk of sporadic Parkinson's disease in a Japanese population. J. Neurol. Sci. 2001;189:113–117. doi: 10.1016/s0022-510x(01)00555-x. [DOI] [PubMed] [Google Scholar]
- 352.Sawada H, Ibi M, Kihara T, Urushitani M, Akaike A, Kimura J, Shimohama S. Dopamine D2-type agonists protect mesencephalic neurons from glutamate neurotoxicity: mechanisms of neuroprotective treatment against oxidative stress. Ann. Neurol. 1998;44:110–119. doi: 10.1002/ana.410440117. [DOI] [PubMed] [Google Scholar]
- 353.Scheider WL, Hershey LA, Vena JE, Holmlund T, Marshall JR Freudenheim. Dietary antioxidants and other dietary factors in the etiology of Parkinson's disease. Mov. Disord. 1997;12:190–196. doi: 10.1002/mds.870120209. [DOI] [PubMed] [Google Scholar]
- 354.Schober A. Classic toxin-induced animal models of Parkinson's disease: 6-OHDA and MPTP. Cell Tissue Res. 2004;318:215–224. doi: 10.1007/s00441-004-0938-y. [DOI] [PubMed] [Google Scholar]
- 355.Schwarting RK, Huston JP. Unilateral 6-hydroxydopamine lesions of meso-striatal dopamine neurons and their physiological sequelae. Prog. Neurobiol. 1996;49:215–266. doi: 10.1016/s0301-0082(96)00015-9. [DOI] [PubMed] [Google Scholar]
- 356.Sestili P, Piedimonte G, Cattabeni F, Cantoni O. Induction of DNA breakage and suppression of DNA synthesis by the OH radical generated in a Fenton-like reaction. Biochem. Int. 1986;12:493–501. [PubMed] [Google Scholar]
- 357.Shachar DB, Kahana N, Kampel V, Warshawsky A, Youdim MB. Neuroprotection by a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats. Neuropharmacology. 2004;46:254–263. doi: 10.1016/j.neuropharm.2003.09.005. [DOI] [PubMed] [Google Scholar]
- 358.Shen XM, Dryhurst G. Iron- and manganese-catalyzed autoxidation of dopamine in the presence of L-cysteine: possible insights into iron- and manganese-mediated dopaminergic neurotoxicity. Chem. Res. Toxicol. 1998;11:824–837. doi: 10.1021/tx980036t. [DOI] [PubMed] [Google Scholar]
- 359.Sherer TB, Betarbet R, Stout AK, Lund S, Baptista M, Panov AV, Cookson MR, Greenamyre JT. An in vitro model of Parkinson's disease: linking mitochondrial impairment to altered alpha-synuclein metabolism and oxidative damage. J. Neurosci. 2002;22:7006–7015. doi: 10.1523/JNEUROSCI.22-16-07006.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360.Sherer TB, Kim JH, Betarbet R, Greenamyre JT. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp. Neurol. 2003;179:9–16. doi: 10.1006/exnr.2002.8072. [DOI] [PubMed] [Google Scholar]
- 361.Shimura H, Hattori N, Kubo S, Mizuno Y, Asakawa S, Minoshima S, Shimizu N, Iwai K, Chiba T, Tanaka K, Suzuki T. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat. Genet. 2000;25:302–305. doi: 10.1038/77060. [DOI] [PubMed] [Google Scholar]
- 362.Shimura H, Schlossmacher MG, Hattori N, Frosch MP, Trockenbacher A, Schneider R, Mizuno Y, Kosik KS, Selkoe DJ. Ubiquitination of a new form of alpha-synuclein by parkin from human brain: implications for Parkinson's disease. Science. 2001;293:263–269. doi: 10.1126/science.1060627. [DOI] [PubMed] [Google Scholar]
- 363.Shinotoh H, Snow BJ, Chu NS, Huang CC, Lu CS, Lee C, Takahashi H, Calne DB. Presynaptic and postsynaptic striatal dopaminergic function in patients with manganese intoxication: a positron emission tomography study. Neurology. 1997;48:1053–1056. doi: 10.1212/wnl.48.4.1053. [DOI] [PubMed] [Google Scholar]
- 364.Shults CW, Oakes D, Kieburtz K, Beal MF, Haas R, Plumb S, Juncos JL, Nutt J, Shoulson I, Carter J, Kompoliti K, Perlmutter JS, Reich S, Stern M, Watts RL, Kurlan R, Molho E, Harrison M, Lew M. Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch. Neurol. 2002;59:1541–1550. doi: 10.1001/archneur.59.10.1541. [DOI] [PubMed] [Google Scholar]
- 365.Sian J, Dexter DT, Lees AJ, Daniel S, Agid Y, Javoy-Agid F, Jenner P, Marsden CD. Alterations in glutathione levels in Parkinson's disease and other neurodegenerative disorders affecting basal ganglia. Ann. Neurol. 1994;36:348–355. doi: 10.1002/ana.410360305. [DOI] [PubMed] [Google Scholar]
- 366.Siderowf A, Stern M. Clinical trials with rasagiline: evidence for short-term and long-term effects. Neurology. 2006;66:S80–88. doi: 10.1212/wnl.66.10_suppl_4.s80. [DOI] [PubMed] [Google Scholar]
- 367.Sies H, Stahl W, Sundquist AR. Antioxidant functions of vitamins. Vitamins E and C, beta-carotene, and other carotenoids. Ann. N. Y. Acad. Sci. 1992;669:7–20. doi: 10.1111/j.1749-6632.1992.tb17085.x. [DOI] [PubMed] [Google Scholar]
- 368.Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, Lincoln S, Crawley A, Hanson M, Maraganore D, Adler C, Cookson MR, Muenter M, Baptista M, Miller D, Blancato J, Hardy J, Gwinn-Hardy K. alpha-Synuclein locus triplication causes Parkinson's disease. Science. 2003;302:841. doi: 10.1126/science.1090278. [DOI] [PubMed] [Google Scholar]
- 369.Sjogren B, Iregren A, Frech W, Hagman M, Johansson L, Tesarz M, Wennberg A. Effects on the nervous system among welders exposed to aluminium and manganese. Occup. Environ. Med. 1996;53:32–40. doi: 10.1136/oem.53.1.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 370.Slevin JT, Gerhardt GA, Smith CD, Gash DM, Kryscio R, Young B. Improvement of bilateral motor functions in patients with Parkinson disease through the unilateral intraputaminal infusion of glial cell line-derived neurotrophic factor. J. Neurosurg. 2005;102:216–222. doi: 10.3171/jns.2005.102.2.0216. [DOI] [PubMed] [Google Scholar]
- 371.Smith AD, Kozlowski DA, Bohn MC, Zigmond MJ. Effect of AdGDNF on dopaminergic neurotransmission in the striatum of 6-OHDA-treated rats. Exp. Neurol. 2005;193:420–426. doi: 10.1016/j.expneurol.2005.01.010. [DOI] [PubMed] [Google Scholar]
- 372.Smith MP, Cass WA. GDNF reduces oxidative stress in a 6-hydroxydopamine model of Parkinson's disease. Neurosci. Lett. 2006;412(3):259–263. doi: 10.1016/j.neulet.2006.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373.Sobotka TJ, Whittaker P, Sobotka JM, Brodie RE, Quander DY, Robl M, Bryant M, Barton CN. Neurobehavioral dysfunctions associated with dietary iron overload. Physiol. Behav. 1996;59:213–219. doi: 10.1016/0031-9384(95)02030-6. [DOI] [PubMed] [Google Scholar]
- 374.Sofic E, Lange KW, Jellinger K, Riederer P. Reduced and oxidized glutathione in the substantia nigra of patients with Parkinson's disease. Neurosci. Lett. 1992;142:128–130. doi: 10.1016/0304-3940(92)90355-b. [DOI] [PubMed] [Google Scholar]
- 375.Sofic E, Riederer P, Heinsen H, Beckmann H, Reynolds GP, Hebenstreit G, Youdim MB. Increased iron (III) and total iron content in post mortem substantia nigra of parkinsonian brain. J. Neural. Transm. 1988;74:199–205. doi: 10.1007/BF01244786. [DOI] [PubMed] [Google Scholar]
- 376.Somayajulu M, McCarthy S, Hung M, Sikorska M, Borowy-Borowski H, Pandey S. Role of mitochondria in neuronal cell death induced by oxidative stress; neuroprotection by Coenzyme Q10. Neurobiol. Dis. 2005;18:618–627. doi: 10.1016/j.nbd.2004.10.021. [DOI] [PubMed] [Google Scholar]
- 377.Sonsalla PK, Jochnowitz ND, Zeevalk GD, Oostveen JA, Hall ED. Treatment of mice with methamphetamine produces cell loss in the substantia nigra. Brain Res. 1996;738:172–175. doi: 10.1016/0006-8993(96)00995-x. [DOI] [PubMed] [Google Scholar]
- 378.Souza JM, Giasson BI, Chen Q, Lee VM, Ischiropoulos H. Dityrosine cross-linking promotes formation of stable alpha-synuclein polymers. Implication of nitrative and oxidative stress in the pathogenesis of neurodegenerative synucleinopathies. J. Biol. Chem. 2000;275:18344–18349. doi: 10.1074/jbc.M000206200. [DOI] [PubMed] [Google Scholar]
- 379.Sparks DL, Lochhead J, Horstman D, Wagoner T, Martin T. Water quality has a pronounced effect on cholesterol-induced accumulation of Alzheimer amyloid beta (Abeta) in rabbit brain. J. Alzheimers Dis. 2002;4:523–529. doi: 10.3233/jad-2002-4609. [DOI] [PubMed] [Google Scholar]
- 380.Spiegel J, Hellwig D, Samnick S, Jost W, Mollers MO, Fassbender K, Kirsch CM, Dillmann U. Striatal FP-CIT uptake differs in the subtypes of early Parkinson's disease. J. Neural. Transm. 2007;114:331–335. doi: 10.1007/s00702-006-0518-2. [DOI] [PubMed] [Google Scholar]
- 381.Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–840. doi: 10.1038/42166. [DOI] [PubMed] [Google Scholar]
- 382.Spira PJ, Sharpe DM, Halliday G, Cavanagh J, Nicholson GA. Clinical and pathological features of a Parkinsonian syndrome in a family with an Ala53Thr alpha-synuclein mutation. Ann. Neurol. 2001;49:313–319. [PubMed] [Google Scholar]
- 383.Stanic D, Finkelstein DI, Bourke DW, Drago J, Horne MK. Timecourse of striatal re-innervation following lesions of dopaminergic SNpc neurons of the rat. Eur. J. Neurosci. 2003;18:1175–1188. doi: 10.1046/j.1460-9568.2003.02800.x. [DOI] [PubMed] [Google Scholar]
- 384.Stefani A, Fedele E, Galati S, Pepicelli O, Frasca S, Pierantozzi M, Peppe A, Brusa L, Orlacchio A, Hainsworth AH, Gattoni G, Stanzione P, Bernardi G, Raiteri M, Mazzone P. Subthalamic stimulation activates internal pallidus: evidence from cGMP microdialysis in PD patients. Ann. Neurol. 2005;57:448–452. doi: 10.1002/ana.20402. [DOI] [PubMed] [Google Scholar]
- 385.Stefanova N, Kollensperger M, Hainzer M, Cenci A, Poewe W, Wenning GK. High dose levodopa therapy is not toxic in multiple system atrophy: experimental evidence. Mov. Disord. 2007;22:969–973. doi: 10.1002/mds.21468. [DOI] [PubMed] [Google Scholar]
- 386.Storch A, Jost WH, Vieregge P, Spiegel J, Greulich W, Durner J, Muller T, Kupsch A, Henningsen H, Oertel WH, Fuchs G, Kuhn W, Niklowitz P, Koch R, Herting B, Reichmann H. Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme Q(10) in Parkinson disease. Arch. Neurol. 2007;64:938–944. doi: 10.1001/archneur.64.7.nct60005. [DOI] [PubMed] [Google Scholar]
- 387.Storch A, Paul G, Csete M, Boehm BO, Carvey PM, Kupsch A, Schwarz J. Long-term proliferation and dopaminergic differentiation of human mesencephalic neural precursor cells. Exp. Neurol. 2001;170:317–325. doi: 10.1006/exnr.2001.7706. [DOI] [PubMed] [Google Scholar]
- 388.Stover NP, Okun MS, Evatt ML, Raju DV, Bakay RA, Vitek JL. Stimulation of the subthalamic nucleus in a patient with Parkinson disease and essential tremor. Arch. Neurol. 2005;62:141–143. doi: 10.1001/archneur.62.1.141. [DOI] [PubMed] [Google Scholar]
- 389.Suh JG, Yamanishi T, Matsui K, Tanaka K, Wada K. Mapping of the gracile axonal dystrophy (gad gene to a region between D5Mit197 and D5Mit113 on proximal mouse chromosome 5. Genomics. 1995;27:549–551. doi: 10.1006/geno.1995.1091. [DOI] [PubMed] [Google Scholar]
- 390.Sulzer D, Bogulavsky J, Larsen KE, Behr G, Karatekin E, Kleinman MH, Turro N, Krantz D, Edwards RH, Greene LA, Zecca L. Neuromelanin biosynthesis is driven by excess cytosolic catecholamines not accumulated by synaptic vesicles. Proc. Natl. Acad. Sci. USA. 2000;97:11869–11874. doi: 10.1073/pnas.97.22.11869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 391.Szabo C, Zingarelli B, O'Connor M, Salzman AL. DNA strand breakage, activation of poly (ADP-ribose synthetase, and cellular energy depletion are involved in the cytotoxicity of macrophages and smooth muscle cells exposed to peroxynitrite. Proc. Natl. Acad. Sci. USA. 1996;93:1753–1758. doi: 10.1073/pnas.93.5.1753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392.Tabrizi SJ, Orth M, Wilkinson JM, Taanman JW, Warner TT, Cooper JM, Schapira AH. Expression of mutant alpha-synuclein causes increased susceptibility to dopamine toxicity. Hum. Mol. Genet. 2000;9:2683–2689. doi: 10.1093/hmg/9.18.2683. [DOI] [PubMed] [Google Scholar]
- 393.Tan EK, Puong KY, Fook-Chong S, Chua E, Shen H, Yuen Y, Pavanni R, Wong MC, Puvan K, Zhao Y. Case-control study of UCHL1 S18Y variant in Parkinson's disease. Mov. Disord. 2006;21:1765–1768. doi: 10.1002/mds.21064. [DOI] [PubMed] [Google Scholar]
- 394.Tarohda T, Ishida Y, Kawai K, Yamamoto M, Amano R. Regional distributions of manganese, iron, copper, and zinc in the brains of 6-hydroxydopamine-induced parkinsonian rats. Anal. Bioanal. Chem. 2005;383:224–234. doi: 10.1007/s00216-005-3423-x. [DOI] [PubMed] [Google Scholar]
- 395.Tatton NA, Kish SJ. In situ detection of apoptotic nuclei in the substantia nigra compacta of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice using terminal deoxynucleotidyl transferase labelling and acridine orange staining. Neuroscience. 1997;77:1037–1048. doi: 10.1016/s0306-4522(96)00545-3. [DOI] [PubMed] [Google Scholar]
- 396.Taylor GA, Ferrier IN, McLoughlin IJ, Fairbairn AF, McKeith IG, Lett D, Edwardson JA. Gastrointestinal absorption of aluminium in Alzheimer's disease: response to aluminium citrate. Age Ageing. 1992;21:81–90. doi: 10.1093/ageing/21.2.81. [DOI] [PubMed] [Google Scholar]
- 397.Taylor GA, Moore PB, Ferrier IN, Tyrer SP, Edwardson JA. Gastrointestinal absorption of aluminium and citrate in man. J. Inorg. Biochem. 1998;69:165–169. doi: 10.1016/s0162-0134(97)10014-9. [DOI] [PubMed] [Google Scholar]
- 398.Teixeira S, Siquet C, Alves C, Boal I, Marques MP, Borges F, Lima JL, Reis S. Structure-property studies on the antioxidant activity of flavonoids present in diet. Free. Radic. Biol. Med. 2005;39:1099–1108. doi: 10.1016/j.freeradbiomed.2005.05.028. [DOI] [PubMed] [Google Scholar]
- 399.Testa CM, Sherer TB, Greenamyre JT. Rotenone induces oxidative stress and dopaminergic neuron damage in organotypic substantia nigra cultures. Brain Res. Mol. Brain Res. 2005;134:109–118. doi: 10.1016/j.molbrainres.2004.11.007. [DOI] [PubMed] [Google Scholar]
- 400.Thomas B, Beal MF. Parkinson's disease. Hum. Mol. Genet. 2007;16(Spec No. 2):R183–194. doi: 10.1093/hmg/ddm159. [DOI] [PubMed] [Google Scholar]
- 401.Toda T, Momose Y, Murata M, Tamiya G, Yamamoto M, Hattori N, Inoko H. Toward identification of susceptibility genes for sporadic Parkinson's disease. J. Neurol. 2003;250(3):III40–43. doi: 10.1007/s00415-003-1307-6. [DOI] [PubMed] [Google Scholar]
- 402.Toft M, Myhre R, Pielsticker L, White LR, Aasly JO, Farrer MJ. PINK1 mutation heterozygosity and the risk of Parkinson's disease. J. Neurol. Neurosurg. Psychiatry. 2007;78:82–84. doi: 10.1136/jnnp.2006.097840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 403.Trulson ME, Cannon MS, Faegg TS, Raese JD. Effects of chronic methamphetamine on the nigral-striatal dopamine system in rat brain: tyrosine hydroxylase immunochemistry and quantitative light microscopic studies. Brain Res. Bull. 1985;15:569–577. doi: 10.1016/0361-9230(85)90206-0. [DOI] [PubMed] [Google Scholar]
- 404.Turunen M, Olsson J, Dallner G. Metabolism and function of coenzyme Q. Biochim. Biophys. Acta. 2004;1660:171–199. doi: 10.1016/j.bbamem.2003.11.012. [DOI] [PubMed] [Google Scholar]
- 405.Uchiyama S, Shimizu T, Shirasawa T. CuZn-SOD deficiency causes ApoB degradation and induces hepatic lipid accumulation by impaired lipoprotein secretion in mice. J. Biol. Chem. 2006;281:31713–31719. doi: 10.1074/jbc.M603422200. [DOI] [PubMed] [Google Scholar]
- 406.Uemura E. Intranuclear aluminum accumulation in chronic animals with experimental neurofibrillary changes. Exp. Neurol. 1984;85:10–18. doi: 10.1016/0014-4886(84)90155-9. [DOI] [PubMed] [Google Scholar]
- 407.Unoki M, Nakamura Y. Growth-suppressive effects of BPOZ and EGR2, two genes involved in the PTEN signaling pathway. Oncogene. 2001;20:4457–4465. doi: 10.1038/sj.onc.1204608. [DOI] [PubMed] [Google Scholar]
- 408.Uversky VN, Gillespie JR, Fink AL. Why are "natively unfolded" proteins unstructured under physiologic conditions? Proteins. 2000;41:415–427. doi: 10.1002/1097-0134(20001115)41:3<415::aid-prot130>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
- 409.Uversky VN, Li J, Fink AL. Evidence for a partially folded intermediate in alpha-synuclein fibril formation. J. Biol. Chem. 2001;276:10737–10744. doi: 10.1074/jbc.M010907200. [DOI] [PubMed] [Google Scholar]
- 410.Uversky VN, Li J, Fink AL. Pesticides directly accelerate the rate of alpha-synuclein fibril formation: a possible factor in Parkinson's disease. FEBS Lett. 2001;500:105–108. doi: 10.1016/s0014-5793(01)02597-2. [DOI] [PubMed] [Google Scholar]
- 411.Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, Gonzalez-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science. 2004;304:1158–1160. doi: 10.1126/science.1096284. [DOI] [PubMed] [Google Scholar]
- 412.van der Putten H, Wiederhold KH, Probst A, Barbieri S, Mistl C, Danner S, Kauffmann S, Hofele K, Spooren WP, Ruegg MA, Lin S, Caroni P, Sommer B, Tolnay M, Bilbe G. Neuropathology in mice expressing human alpha-synuclein. J. Neurosci. 2000;20:6021–6029. doi: 10.1523/JNEUROSCI.20-16-06021.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 413.van Duijn CM, Dekker MC, Bonifati V, Galjaard RJ, Houwing-Duistermaat JJ, Snijders PJ, Testers L, Breedveld GJ, Horstink M, Sandkuijl LA, van Swieten JC, Oostra BA, Heutink P. Park7, a novel locus for autosomal recessive early-onset parkinsonism, on chromosome 1p36. Am. J. Hum. Genet. 2001;69:629–634. doi: 10.1086/322996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 414.Vila M, Vukosavic S, Jackson-Lewis V, Neystat M, Jakowec M, Przedborski S. Alpha-synuclein up-regulation in substantia nigra dopaminergic neurons following administration of the parkinsonian toxin MPTP. J. Neurochem. 2000;74:721–729. doi: 10.1046/j.1471-4159.2000.740721.x. [DOI] [PubMed] [Google Scholar]
- 415.Voges D, Zwickl P, Baumeister W. The 26S proteasome: a molecular machine designed for controlled proteolysis. Ann. Rev. Biochem. 1999;68:1015–1068. doi: 10.1146/annurev.biochem.68.1.1015. [DOI] [PubMed] [Google Scholar]
- 416.Volles MJ, Lee SJ, Rochet JC, Shtilerman MD, Ding TT, Kessler JC, Lansbury P.T Jr. Vesicle permeabilization by protofibrillar alpha-synuclein: implications for the pathogenesis and treatment of Parkinson's disease. Biochemistry. 2001;40:7812–7819. doi: 10.1021/bi0102398. [DOI] [PubMed] [Google Scholar]
- 417.Wagner GC, Jarvis MF, Carelli RM. Ascorbic acid reduces the dopamine depletion induced by MPTP. Neuropharmacology. 1985;24:1261–1262. doi: 10.1016/0028-3908(85)90164-9. [DOI] [PubMed] [Google Scholar]
- 418.Walton-Hadlock JL. Levodopa and the progression of Parkinson's disease. N. Engl. J. Med. 2005;352:1386. doi: 10.1056/NEJM200503313521324. [DOI] [PubMed] [Google Scholar]
- 419.Wang D, Qian L, Xiong H, Liu J, Neckameyer WS, Oldham S, Xia K, Wang J, Bodmer R, Zhang Z. Antioxidants protect PINK1-dependent dopaminergic neurons in Drosophila. Proc. Natl. Acad. Sci. USA. 2006;103:13520–13525. doi: 10.1073/pnas.0604661103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420.Wang F, Feng X, Ma J, Zou H, Chan P. A common A340T variant in PINK1 gene associated with late-onset Parkinson's disease in Chinese. Neurosci. Lett. 2006;410:121–125. doi: 10.1016/j.neulet.2006.09.080. [DOI] [PubMed] [Google Scholar]
- 421.Wang L, Muramatsu S, Lu Y, Ikeguchi K, Fujimoto K, Okada T, Mizukami H, Hanazono Y, Kume A, Urano F, Ichinose H, Nagatsu T, Nakano I, Ozawa K. Delayed delivery of AAV-GDNF prevents nigral neurodegeneration and promotes functional recovery in a rat model of Parkinson's disease. Gene Ther. 2002;9:381–389. doi: 10.1038/sj.gt.3301682. [DOI] [PubMed] [Google Scholar]
- 422.Washbourne P, McAllister AK. Techniques for gene transfer into neurons. Curr. Opin. Neurobiol. 2002;12:566–573. doi: 10.1016/s0959-4388(02)00365-3. [DOI] [PubMed] [Google Scholar]
- 423.Weinreb PH, Zhen W, Poon AW, Conway KA, Lansbury P.T Jr. NACP, a protein implicated in Alzheimer's disease and learning, is natively unfolded. Biochemistry. 1996;35:13709–13715. doi: 10.1021/bi961799n. [DOI] [PubMed] [Google Scholar]
- 424.Wesemann W, Blaschke S, Solbach M, Grote C, Clement HW, Riederer P. Intranigral injected iron progressively reduces striatal dopamine metabolism. J. Neural. Transm. Park. Dis. Dement. Sect. 1994;8:209–214. doi: 10.1007/BF02260941. [DOI] [PubMed] [Google Scholar]
- 425.Wettstein A, Aeppli J, Gautschi K, Peters M. Failure to find a relationship between mnestic skills of octogenarians and aluminum in drinking water. Int. Arch. Occup. Environ. Health. 1991;63:97–103. doi: 10.1007/BF00379071. [DOI] [PubMed] [Google Scholar]
- 426.White AR, Reyes R, Mercer JF, Camakaris J, Zheng H, Bush AI, Multhaup G, Beyreuther K, Masters CL, Cappai R. Copper levels are increased in the cerebral cortex and liver of APP and APLP2 knockout mice. Brain Res. 1999;842:439–444. doi: 10.1016/s0006-8993(99)01861-2. [DOI] [PubMed] [Google Scholar]
- 427.Widdowson PS, Farnworth MJ, Simpson MG, Lock EA. Influence of age on the passage of paraquat through the blood-brain barrier in rats: a distribution and pathological examination. Hum. Exp. Toxicol. 1996;15:231–236. doi: 10.1177/096032719601500308. [DOI] [PubMed] [Google Scholar]
- 428.Groenewegen H, Wilkins S, Masters CL, Bush AI, Cherny RA, Finkelstein DI. Cliquinol protects against cell death in Parkinson’s disease models in vivo and in vitro. In: Groenewegen H, editor. Advances in Behavioral Biology. IBAGS IX. New York: Springer; 2008. [Google Scholar]
- 429.Winkler C, Kirik D, Bjorklund A, Cenci MA. L-DOPA-induced dyskinesia in the intrastriatal 6-hydroxydopamine model of parkinson's disease: relation to motor and cellular parameters of nigrostriatal function. Neurobiol. Dis. 2002;10:165–186. doi: 10.1006/nbdi.2002.0499. [DOI] [PubMed] [Google Scholar]
- 430.Wintermeyer P, Kruger R, Kuhn W, Muller T, Woitalla D, Berg D, Becker G, Leroy E, Polymeropoulos M, Berger K, Przuntek H, Schols L, Epplen JT, Riess O. Mutation analysis and association studies of the UCHL1 gene in German Parkinson's disease patients. Neuroreport. 2000;11:2079–2082. doi: 10.1097/00001756-200007140-00004. [DOI] [PubMed] [Google Scholar]
- 431.Wolters EC, Francot C, Bergmans P, Winogrodzka A, Booij J, Berendse HW, Stoof JC. Preclinical (premotor) Parkinson's disease. J. Neurol. 2000;247(2):II103–109. [PubMed] [Google Scholar]
- 432.Wolters EC, Huang CC, Clark C, Peppard RF, Okada J, Chu NS, Adam MJ, Ruth TJ, Li D, Calne DB. Positron emission tomography in manganese intoxication. Ann. Neurol. 1989;26:647–651. doi: 10.1002/ana.410260510. [DOI] [PubMed] [Google Scholar]
- 433.Wong CS, Kwok JC, Richardson DR. PCTH: a novel orally active chelator of the aroylhydrazone class that induces iron excretion from mice. Biochim. Biophys. Acta. 2004;1739:70–80. doi: 10.1016/j.bbadis.2004.09.001. [DOI] [PubMed] [Google Scholar]
- 434.Yamada M, Mizuno Y, Mochizuki H. Parkin gene therapy for alpha-synucleinopathy: a rat model of Parkinson's disease. Hum. Gene Ther. 2005;16:262–270. doi: 10.1089/hum.2005.16.262. [DOI] [PubMed] [Google Scholar]
- 435.Yang Y, Gehrke S, Imai Y, Huang Z, Ouyang Y, Wang JW, Yang L, Beal MF, Vogel H, Lu B. Mitochondrial pathology and muscle and dopaminergic neuron degeneration caused by inactivation of Drosophila Pink1 is rescued by Parkin. Proc. Natl. Acad. Sci. USA. 2006;103:10793–10798. doi: 10.1073/pnas.0602493103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 436.Yoo MS, Chun HS, Son JJ, DeGiorgio LA, Kim DJ, Peng C, Son JH. Oxidative stress regulated genes in nigral dopaminergic neuronal cells: correlation with the known pathology in Parkinson's disease. Brain Res. Mol. Brain Res. 2003;110:76–84. doi: 10.1016/s0169-328x(02)00586-7. [DOI] [PubMed] [Google Scholar]
- 437.Yoritaka A, Hattori N, Uchida K, Tanaka M, Stadtman ER, Mizuno Y. Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease. Proc. Natl. Acad. Sci. USA. 1996;93:2696–2701. doi: 10.1073/pnas.93.7.2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 438.Youdim MB, Stephenson G, Ben Shachar D. Ironing iron out in Parkinson's disease and other neurodegenerative diseases with iron chelators: a lesson from 6-hydroxydopamine and iron chelators, desferal and VK-28. Ann. N. Y. Acad. Sci. 2004;1012:306–325. doi: 10.1196/annals.1306.025. [DOI] [PubMed] [Google Scholar]
- 439.Yuan H, Sarre S, Ebinger G, Michotte Y. Histological, behavioural and neurochemical evaluation of medial forebrain bundle and striatal 6-OHDA lesions as rat models of Parkinson's disease. J. Neurosci. Methods. 2005;144:35–45. doi: 10.1016/j.jneumeth.2004.10.004. [DOI] [PubMed] [Google Scholar]
- 440.Zarranz JJ, Alegre J, Gomez-Esteban JC, Lezcano E, Ros R, Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atares B, Llorens V, Gomez Tortosa E, del Ser T, Munoz DG, de Yebenes JG. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 2004;55:164–173. doi: 10.1002/ana.10795. [DOI] [PubMed] [Google Scholar]
- 441.Zecca L, Pietra R, Goj C, Mecacci C, Radice D, Sabbioni E. Iron and other metals in neuromelanin, substantia nigra, and putamen of human brain. J. Neurochem. 1994;62:1097–1101. doi: 10.1046/j.1471-4159.1994.62031097.x. [DOI] [PubMed] [Google Scholar]
- 442.Zecca L, Swartz HM. Total and paramagnetic metals in human substantia nigra and its neuromelanin. J. Neural. Transm. Park. Dis. Dement. Sect. 1993;5:203–213. doi: 10.1007/BF02257675. [DOI] [PubMed] [Google Scholar]
- 443.Zhang J, Graham DG, Montine TJ, Ho YS. Enhanced N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity in mice deficient in CuZn-superoxide dismutase or glutathione peroxidase. J. Neuropathol. Exp. Neurol. 2000;59:53–61. doi: 10.1093/jnen/59.1.53. [DOI] [PubMed] [Google Scholar]
- 444.Zhang J, Hattori N, Leroy E, Morris HR, Kubo S, Kobayashi T, Wood NW, Polymeropoulos MH, Mizuno Y. Association between a polymorphism of ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) gene and sporadic Parkinson's disease. Parkinsonism Relat. Disord. 2000;6:195–197. doi: 10.1016/s1353-8020(00)00015-8. [DOI] [PubMed] [Google Scholar]
- 445.Zhang J, Perry G, Smith MA, Robertson D, Olson SJ, Graham DG, Montine TJ. Parkinson's disease is associated with oxidative damage to cytoplasmic DNA and RNA in substantia nigra neurons. Am. J. Pathol. 1999;154:1423–1429. doi: 10.1016/S0002-9440(10)65396-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 446.Zhang L, Shimoji M, Thomas B, Moore DJ, Yu SW, Marupudi NI, Torp R, Torgner IA, Ottersen OP, Dawson TM, Dawson VL. Mitochondrial localization of the Parkinson's disease related protein DJ-1: implications for pathogenesis. Hum. Mol Genet. 2005;14:2063–2073. doi: 10.1093/hmg/ddi211. [DOI] [PubMed] [Google Scholar]
- 447.Zhang SM, Hernan MA, Chen H, Spiegelman D, Willett WC, Ascherio A. Intakes of vitamins E and C, carotenoids, vitamin supplements, and PD risk. Neurology. 2002;59:1161–1169. doi: 10.1212/01.wnl.0000028688.75881.12. [DOI] [PubMed] [Google Scholar]
- 448.Zhang X, Xie W, Qu S, Pan T, Wang X, Le W. Neuroprotection by iron chelator against proteasome inhibitor-induced nigral degeneration. Biochem. Biophys. Res. Commun. 2005;333:544–549. doi: 10.1016/j.bbrc.2005.05.150. [DOI] [PubMed] [Google Scholar]
- 449.Zheng H, Gal S, Weiner LM, Bar-Am O, Warshawsky A, Fridkin M, Youdim MB. Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases: in vitro studies on antioxidant activity, prevention of lipid peroxide formation and monoamine oxidase inhibition. J. Neurochem. 2005;95:68–78. doi: 10.1111/j.1471-4159.2005.03340.x. [DOI] [PubMed] [Google Scholar]
- 450.Zheng JS, Tang LL, Zheng SS, Zhan RY, Zhou YQ, Goudreau J, Kaufman D, Chen AF. Delayed gene therapy of glial cell line-derived neurotrophic factor is efficacious in a rat model of Parkinson's disease. Brain Res. Mol. Brain Res. 2005;134:155–161. doi: 10.1016/j.molbrainres.2004.06.029. [DOI] [PubMed] [Google Scholar]
- 451.Zhou W, Schaack J, Zawada WM, Freed CR. Overexpression of human alpha-synuclein causes dopamine neuron death in primary human mesencephalic culture. Brain Res. 2002;926:42–50. doi: 10.1016/s0006-8993(01)03292-9. [DOI] [PubMed] [Google Scholar]
- 452.Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Muller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–607. doi: 10.1016/j.neuron.2004.11.005. [DOI] [PubMed] [Google Scholar]
- 453.Zou L, Jankovic J, Rowe DB, Xie W, Appel SH, Le W. Neuroprotection by pramipexole against dopamine- and levodopa-induced cytotoxicity. Life Sci. 1999;64:1275–1285. doi: 10.1016/s0024-3205(99)00062-4. [DOI] [PubMed] [Google Scholar]

