Skip to main content
CNS Neuroscience & Therapeutics logoLink to CNS Neuroscience & Therapeutics
. 2010 Apr 23;17(5):345–367. doi: 10.1111/j.1755-5949.2010.00150.x

Pharmacological Therapy in Parkinson's Disease: Focus on Neuroprotection

Zsigmond Tamas Kincses 1, Laszlo Vecsei 1
PMCID: PMC6493808  PMID: 20438581

Abstract

Although the number of available therapeutic approaches in Parkinson's disease (PD) is steadily increasing the search for effective neuroprotective agent is continuing. Such research is directed at influencing the key steps in the pathomechanism: the mitochondrial dysfunction, the oxidative stress, the neuroinflammatory processes and the final common apoptotic pathway. Earlier‐developed symptomatic therapies were implicated to be neuroprotective, and promising novel disease modifying approaches were brought into the focus of interest. The current review presents a survey of our current knowledge relating to the pathomechanism of PD and discusses the putative neuroprotective therapy.

Keywords: Dopamine agonists, Genetics, Kynurenine, L‐dopa, Mitochondrion, Parkinson's disease

Introduction

After the original description of the clinical picture by James Parkinson in 1817 [1], the first breakthrough in the therapy of Parkinson's diseases’ (PD) was the introduction of L‐dopa in the 1960s [2]. L‐dopa led to significant improvements in the standard of life of individuals with PD, as measured in the activity of daily living, employability and survival, and is still the most potent mode of treatment, but a number of issues remain unresolved. (1) L‐dopa treatment was associated with adverse motor and nonmotor complications. (2) Whereas various drugs have been suggested as potential neuroprotectants, none of the currently applied forms of therapy has been proven to have a clear disease‐modifying, curative benefit [3]. Rather than focusing on the evidence‐based therapeutic interventions, the present review will offer an overview of the recent developments that might become future therapeutic avenues. We have chosen to base our review to reflect current knowledge relating to the pathomechanism of PD.

Parkinson's Disease

Clinical Features

The incidence of PD has been calculated to be 326.3 per 100,000 person‐years [4]. The incidence rate increases with age in both men and women from 0.3 per 1000 person‐years in subjects aged 55 to 65 years to 4.4 per 1000 person‐years in those aged 85 years or above [5]. The age‐adjusted relative risk in men as compared with women has been reported 2.13 in one study [4] in contrast with no significant difference in another cohort [5].

The disease is defined by distinct clinical features, such as bradykinesia, resting tremor, rigidity, and postural instability. Besides the motor symptoms a cognitive impairment develops [6, 7, 8, 9, 10, 11]. In the early stages of the disease, the symptoms are mild and often missed by the patient and family members, becoming evident only retrospectively. The early symptoms start asymmetrically, usually with deterioration in dexterity of fine movements. Relatives and colleagues may notice general slowing‐down, changes in facial expression, and an impaired arm swing. The disease becomes bilateral and then progresses slowly during a period of years. Eventually, the patients become chair‐ and bed‐bound.

Pathological Findings

Pathologically, the disease is characterized by degeneration of the dopaminergic cells of the substantia nigra pars compacta and the appearance of cytoplasmatic inclusion bodies, the Lewy neurites, and Lewy bodies [12, 13]. The major components of the Lewy bodies are alpha‐synuclein, ubiquitin, phosphorylated neurofilaments, parkin, components of the ubiquitin proteasomal system, molecular chaperones, and lipids [14]. Various nondopaminergic structures are affected by the disease, including the cholinergic neurons of the nucleus basalis Meynert, the noradrenergic neurons of the locus coeruleus, the serotoninergic neurons of the raphe nucleus, the cortical, brainstem, and spinal cord neurons and the peripheral autonomic nervous system. It has recently been suggested that there is a gradual caudo‐rostral progression of the pathological changes, starting from the neurons of the nuclei of the autonomic nervous system, progressing through the lower brainstem, reaching the basal ganglia and finally involving the cortical neurons too [15]. Nonetheless, reviews of earlier studies [16] and more recent pathological studies [17] have demonstrated that a significant proportion of patients do not follow the proposed pathway, and the validity of the hypothesis has been questioned [14, 16].

Pathogenesis

It is currently considered that the pathogenesis of PD involves several major interacting pathways (Figure 1). A mitochondrial dysfunction of various origins was first shown to lead to the development of oxidative stress and a cell energy insufficiency. Similarly, immune mechanisms identified in PD (see below) cause oxidative damage, but were also postulated to give rise to apoptosis through more direct mechanisms. Another important pathway is related to the abnormal aggregation of proteins. Glutamate excitotoxicity has additionally been implicated in striatal cell death in PD. The various pathological processes subsequently coincide into a common pathway of apoptosis.

Figure 1.

Figure 1

Interaction of pathomechanical pathways in PD.

Oxidative Stress

Oxidative stress, a key concept of PD pathogenesis, reflects a disturbed balance between the generation of reactive oxygen species (ROS) and antioxidative mechanisms. The major source of ROS is in the mitochondria, as by‐product of oxidative phosphorylation [18]. The free oxygen radicals generated by the mitochondria are converted to H2O2, spontaneously, or by superoxide dismutase, and in turn this is either reduced to water by glutathione peroxidase or converted to hydroxyl radicals in the Fenton reaction, catalyzed by iron.

The ROS bring about oxidative damage of proteins, lipids, and DNA [19]. An imbalance between ROS generation and antioxidative mechanisms in PD has been demonstrated by a decreased nigral level of the antioxidant glutathione [20], reduced mRNA expressions of Cu/Zn‐superoxide dismutase and glutathione peroxidase [21], and an increased concentration of iron (Fe3+) [22]. Increased iron concentrations were reported in the substantia nigra of severe cases of PD [23, 24] but not in milder cases. The iron deposition was localized to the neuromelanin granules of the substatia nigra pars compacta [25] that is a strong iron chelator. In vivo MRI studies indicated reduced T2 relaxation times that was related also to the increased iron content of the substantia nigra [26, 27]. Dopaminergic cells are prone to oxidative damage as the metabolism of dopamine by monoamine oxidase produces H2O2[28]. Dopamine can also be metabolized to reactive dopamine quinol which has been shown to be neurotoxic through the covalent modification of proteins [29].

Mitochondrial Dysfunction

The primary role of mitochondria is the energy supply of the cell through oxidative phosphorylation. In the double‐membrane organization of the mitochondrion, four distinct compartments can be defined, each having unique functional role: the outer membrane, the intermembrane space, the inner membrane, and the matrix. The elements of the mitochondrial electron transport system are all located on the inner membrane: complex I (nicotinamide adenine dinucleotide ubiquinone oxi‐reductase), complex II (succinate ubiquinone oxi‐reductase), complex III (ubiquinone/cytochrome c reductase), complex IV (cytochrome oxidase), and complex V (ATP synthase). Two mobile electron carriers are located in the intermembrane space and in the inner membrane are the cytochrome c and the coenzyme Q (CoQ; ubiquinone). As the electrons are transported down the respiratory chain (Compex I‐V), the proton concentration in the intermembrane space increases and a strong electrochemical gradient is established across the inner membrane (mitochondrial transmembrane potential). The energy produced by equalization of this gradient through complex V is used for ATP synthesis (Figure 2—inset). Although this process is efficient a small percentage of the electrons may reduce oxygen prematurely and produce ROS.

Figure 2.

Figure 2

Apoptotic processes in PD. Extrinsic pathway initiated from the cell surface receptors (e.g., Fas) and proceeding through a cascade of events, in which caspases are involved. The activation of the intrinsic pathway begins with the permeabilization of the mitochondrial outer membrane, which leads to the release of key mitochondrial proteins (such as cytochrome c, Smacs, AIF, etc.). Members of the Bcl2 family of proteins are important modulators of pro‐ and antiapoptotic mechanisms. The inset shows the structure of the mitochondrion and the respiratory chain (JNK, c‐Jun N‐terminal kinase; Smacs, secondary mitochondria‐derived activator caspases; AIF, apoptosis inducing factor; Apaf‐1, apoptotic peptidase activating factor 1).

The first evidence of the key role of a mitochondrial dysfunction emerged from the observation that the 1‐methyl‐4‐phenylpyridinium ion (MPP+), an active metabolite of 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP; a contaminant of opiate drugs), which is an inhibitor of mitochondrial complex I, causes dopaminergic cell death and rapid‐onset parkinsonism [30, 31]. Consecutively, a deficient function of the complexes of the electron transport system was demonstrated in the platelets and skeletal muscle of PD patients [32, 33, 34]. The background of such a complex I deficiency seems to be linked to the mitochondrial DNA, as indicated by neuroblastoma cells with mitochondria derived from the platelets of PD patients [35, 36]. Knowledge of the mitochondrial electron transport system deficiency led to the development of promising, novel therapeutic interventions, such as near‐infrared light therapy, which was suggested to modulate the activity of complex IV [37], and mitochondrial gene therapy was also postulated as a possible future therapeutic avenue [38].

Whatever the reason for and the specific mechanism of the mitochondrial dysfunction, it was hypothesized to cause neurodegeneration via one of the following pathways: (1) oxidative stress (see above), (2) energy depletion (that itself may cause cell death by activating K‐ATP channels [39], by compromising the proteasomal activity [40] or altering the neural amino acid levels [41]), and (3) the promotion of apoptosis (see below).

Neuroinflammation

The role of neuroinflammation in the pathogenesis of PD was suggested by several early findings and also by recent studies. Microglial activation and astrocytic reactions were described in the substantia nigra of PD patients postmortem [42, 43, 44], though it is not yet clear if this is the cause or a result of the progressing disease. It has also been reported that CD4+ and CD8+ T cells are present in the mouse model of PD close to vessels and dopaminergic neurons [45]. In a series of papers, Mogi and coworkers reported increased levels of tumor necrosis factor‐alpha (TNF‐alpha), interleukin‐1 beta (IL‐1 beta), IL‐6, epidermal growth factor, transforming growth factor‐alpha, transforming growth factor‐beta 1, beta 2‐microglobulin, and IL‐2 in the brain of PD patients [46, 47, 48, 49, 50, 51, 52]. In the serum of human patients increased levels of IL‐2, TNF‐alpha, IL‐6, RANTES, and antibodies against dopamine‐oxidized proteins were found [53, 54, 55, 56]. Similar changes were observed in the cerebrospinal fluid [51, 57]. The contribution of the microglial response to the PD pathogenesis was demonstrated in in vivo imaging studies [58, 59]. Parallel changes in microglial activation and the corresponding dopaminergic terminal loss in the affected nigrostriatal pathway in early PD were revealed by means of PET. An association between polymorphisms and mutations of various neuroinflammation related genes and PD was suggested [60, 61, 62, 63, 64, 65, 66].

The exact mechanism of the neuroinflamation‐related neurotoxicity is not yet fully understood, but several studies have suggested that, besides the direct apoptosis‐inducing effect of cytokines, it is related to the oxidative burst arising from the activated microglias [67, 68, 69, 70, 71].

Protein Aggregation

Alpha‐synuclein is the major component of the Lewy bodies in sporadic PD. Overproduction and reduced clearing of the protein by lysosomes and proteasomes may play a key role in PD pathogenesis. Elevated levels of alpha‐synuclein may result in aggregation and the formation of toxic oligomers that may interfere with the lysosome and proteasome functions [72] creating a vicious circle [73]. Under physiological conditions alpha‐synuclein exists as α‐helical conformation, whereas under pathological conditions it can refold into a β‐sheet‐rich conformation that can furnish toxic oligomers and amyloid fibrils. These protein aggregates can in turn promote the misfolding of wild‐type proteins. This hypothetical mechanism, suggested to be similar to a prion‐like mechanism [74], was confirmed by the demonstration that, some 10 years after implantation, embryonic dopaminergic cells displayed PD pathology [75, 76]. Also Desplats and colleagues found that overexpressed synuclein could be transferred to healthy cells that will express pathological changes [77]. This mechanism would also be in accordance with the observation by Braak that the appearance of alpha‐synuclein follows a predictable sequential spread throughout the nervous system [15].

Exitotoxicity and Calcium Homeostasis

A further key process implicated in PD pathogenesis is glutamate exitotoxicity. Glutamate is the major excitatory neurotransmitter of the nervous system that acts on NMDA (N‐methyl‐D‐aspartate), AMPA (α‐amino‐3‐hydroxy‐5‐methylisoxazole proprionic acid) and metabotropic receptors (mGlu1‐mGlu8) [78, 79]. Besides its key role in neurotransmission by activating NMDA and AMPA receptors and voltage‐gated ion channels, glutamate causes a massive influx of extracellular calcium and also its release from the endoplasmatic reticulum, initiating neurotoxic processes. The increased intracellular calcium concentration may influence apoptosis‐related factors such as the Bcl‐2 family proteins [80], and endoplasmatic reticulum‐related protein processing and folding [81]. Moreover, calcium enters the mitochondria and under normal circumstances potentiates oxidative phosphorylation to meet the increased metabolic demands of restoring the calcium homeostasis [82]. In the event of an enhanced calcium influx, it may ultimately compromise the mitochondrial energy production, further increasing the mitochondrial calcium concentration and leading to depolarization and ultimately to opening of the mitochondrial permeability transition pore and commencing apoptosis [83].

Dopaminergic cells of the substantia nigra pars compacta are critically sensitive to glutamate exitotoxicity as a reduced dopaminergic activity leads to an enhanced compensatory recursive glutamatergic input [84]. Further, in order to maintain the constant striatal dopamine concentration, dopaminergic cells have a pace‐maker activity generated through ion channels [85, 86] that increase the intracellular calcium level [83], thereby increasing the susceptibility of these cells.

Apoptosis

Apoptosis, programmed cell death, is a cascade of biochemical events leading to characteristic morphological changes and consequently to cell death [87] (Figure 2). The apoptotic cascade is initiated by two pathways (extrinsic and intrinsic) that converge to the same final effector mechanisms.

The extrinsic pathway is activated through cell surface death‐receptors belonging in the TNF receptor superfamily; activation of the caspase cascade that in turn will activate the effectors, the death substrates [88]. It has been proposed that inflammatory cytokines released from the activated immune cells and microglia found in the brain of PD patients [42, 43, 44, 45] may induce apoptosis directly through TNF cell surface receptors [89]. However, there are conflicting results about the importance of the TNF receptor‐mediated processes in animal models [90, 91, 92, 93, 94]. Another receptor involved in the initiation of the extrinsic pathway is Fas (also belonging in the TNF receptor superfamily), activated by the Fas‐ligand (Fas‐L: expressed mainly by activated T cells); this has been shown to be overexpressed in PD patients and also in MPTP‐treated mice [95, 96]. However, contradictory results have also been published regarding the role of the Fas‐L/Fas/caspase‐8 pathway in PD [97, 98]. The c‐Jun N‐terminal kinase (JNK) have been identified a key transcriptional modulator of the Fas/Fas‐L system and is a crucial component of the neurodegeneration [99, 100, 101, 102]. Nonetheless, inhibition of the JNK signal transduction pathway upstream at the level of mixed lineage kinases by GEP‐1347 in a prospective clinical trial did not prove to be effective [103].

The pivotal event in the intrinsic or mitochondrial pathway is the increased permeabilization of the mitochondrial outer membrane. This either occurs by opening of the mitochondrial permeability transition pore or is mediated by the direct action of proapoptotic proteins, the Bcl‐2 family members Bax and Bak [104]. Once the outer membrane is permeable, key mitochondrial proapoptotic proteins are released to the cytosol, such as cytochrome c, AIF, and Smacs. Cytochrome c binds to Apaf‐1 and forms a protein complex known as apoptosome that in turn activates procaspase‐9 to initiate an enzymatic reaction cascade leading to the execution of apoptosis in cells [105]. The key regulator of the mitochondrial permeability and the release of the proapoptogenic proteins are the proteins belonging in the Bcl‐2 family [106].

Etiology of PD

Although epidemiology studies have suggested several environmental factors as risk factors for PD, no single etiological factor has yet been conclusively established. MPTP, a contaminant of synthetic opiates, is the first link between environmental factors and the disease. Rural living [107, 108], well‐water [108, 109], pesticides, and herbicides [109, 110] (not necessarily independently) have been implicated as risk factors for PD [111, 112].

Genetic alterations have been identified as the cause of PD in familial cases (Table 1), which account for ∼5–10% of all PD cases.

Table 1.

Genetic background of Parkinson's disease

Protein/Gene Locus Putative physiological and pathogenetic role Key references
Synuclein
PARK‐1 Chr 4
4q21–23 Pathological oligomer formation [113, 114, 115, 116, 117, 118]
Parkin
PARK‐2 Chr 6
6q23–27 Ubiquitin‐protein ligase
By ubiquitation catalyze protein degradation [119, 120, 121, 122, 123, 124]
UCH‐L1
PARK‐5 Chr 4
4q14–15 Ubiquitin C terminal hydrolase
Generates monomeric ubiquitin by cleaving polymeric ubiquitin to monomers and from the ubiquitylated peptides [125, 126, 127, 128, 129]
PINK‐1
PARK‐6 Chr 1
1p35–36 A serine‐threonine kinase
Regulator of mitochondrial functions [130, 131, 132, 133, 134]
DJ1
PARK‐7 Chr 1
1p36 Redox‐sensitive modulator of antiapoptotic pathways [135, 136]
LRRK2
PARK‐8 Chr 12
12p11 GTP‐ase and protein kinase
Modulator of apoptotic processes [137, 138, 139, 140, 141, 142]

The first mutation identified as linked to PD was in the gene of alpha‐synuclein, the major component of Lewy bodies [113]. It was observed that the overproduction and reduced clearing of the protein may play a key role in PD. Increased levels of alpha synuclein result in aggregation and the formation of toxic oligomers that may interfere with the lysosome and proteasome functions, creating a vicious circle [114]. Hence, interventions against the processing, misfolding and aggregation of alpha‐synuclein and those augmenting the clearing processes are promising for the development of novel therapeutic methods [143, 144, 145, 146, 147, 148, 149, 150].

The most frequent mutation among PD patients with recessive inheritance [119] is PARK‐2. Parkin is a ubiquitin‐protein ligase located at the outer mitochondrial membrane [120]. By catalyzing the ubiquitination of many proteins, it facilitates their degradation by the proteasome [151, 152, 153, 154] and this effect is linked to the attenuation of JNK and p38 mitogen‐activated protein kinase and the subsequent cleavage of caspase‐3. The above results certainly justify the delivery of parkin gene to the midbrain dopaminergic cells of patients carrying this mutation. Preclinical animal studies are already under way [155, 156], but several issues must be resolved before this new treatment modality can be applied in clinical trials [155].

Ubiquitin C‐terminal hydrolase (UCH‐L1) was first linked to PD by an autosomal dominant point mutation (I93M) in two siblings with a strong family history, but incomplete penentrance [125]. The possibility was also raised that a single nucleotide polymorphism (S18Y) is linked to a reduced susceptibility to PD [126, 127, 128], though contradictory results were also published recently [157]. UCH‐L1 is a neuronal enzyme that catalyzes the hydrolysis of C‐terminal ubiquitin esters and amides [129]. The major function of the enzyme is the generation of monomeric ubiquitin by cleaving polymeric ubiquitin to monomers and from the ubiquitylated peptides that are the main products of proteasomal degradation of polyubiquinated proteins [129]. It has been proposed that there is a fine balance between the concentration‐dependent alpha‐synuclein aggregation, the ubiquitination of alpha‐synuclein by parkin for proteasomal degradation and the subsequent recycling of ubiquitin by UCH‐L1 [158]. A disturbance of this balance at any point may give rise to a shift to pathological changes, and hence the suggested key roles of mutations in these genes in the pathogenesis of PD.

Ubiquinated protein, PTEN‐induced putative kinase 1 (PINK1) [130] is a nuclearly encoded protein localized to the intermembrane space of the mitochondrion [131, 132]. The protein exerts serine‐threonine kinase activity and is also capable of autophosphorylation, which may have an important regulatory role. The role of PINK1 in the pathogenesis of PD may be related to defective oxidative phosporylation in consequence of changes in the phosphorylation of key mitochondrial proteins of the respiratory complexes [159]. Alternatively, failing physiological phosphorylation triggers a signaling cascade that opens the mitochondrial transition pores and apoptosis commences [160, 161].

Although the physiological role of the highly conserved protein DJ‐1 is not yet clear, it is very likely that it is a transcriptional factor and redox‐sensitive modulator of the antioxidative and antiapoptotic pathways [135, 162, 163]. As a potential therapeutic alternative, DJ‐1 transferred either by the gene or by the recombinant protein itself reduces MPTP or 6‐hydroxy‐dopamine‐induced (6‐OHDA) neurotoxicity [164, 165]. Compounds that prevent oxidation of DJ‐1 also successfully prevent oxidative stress‐induced dopaminergic cell death in the rat [166].

LRKK2/dardarin, a member of the ROCO protein family is characterized by GTP‐ase and protein kinase activity [137]. Mutations markedly augment the kinase activity [138, 139] or change the GTP‐ase activity, which in turn influences the kinase activity [139]. The kinase activity seems to be crucial in the development of LRRK2‐related apoptotic processes mediated by cytochrome c and caspase‐3 [167, 168, 169], but domains without kinase activity have also been demonstrated to be important for the initiation of apoptosis [170]. From a possible therapeutic aspect, heat shock protein 90 has been shown to connect to LRRK2 in vivo[171] and inhibition of the chaperone activity of this protein disrupts this association and leads to the proteasomal degradation of LRRK2 [172].

These observations furnish important clues concerning the pathomechanism of PD and have been utilized as starting points for the development of several drugs. However, a recent large‐scale study did not find significant difference in the pairwise concordance of parkinsonism in mono‐ and diziygotic twin brothers older than 50 years. The authors concluded that genetic factors do not play an important role in sporadic PD when the disease begins after 50 years of age, which is the situation in the majority of cases [173]. However, genetic factors appear to be important when disease begins before age 50 years.

Neuroprotection in PD

L‐Dopa

L‐Dopa was introduced into PD therapy in the late 1960s, and is still the most effective symptomatic therapy. Essentially, all PD patients, but especially those with early PD benefit from L‐Dopa therapy [174]. L‐Dopa is usually coadministered with a peripheral decarboxylase inhibitor (carbidopa or benserasid) to prevent peripheral conversion to dopamine, thereby stabilizing the bioavailability of dopamine and reducing the peripheral side effects. To reduce the elimination of L‐Dopa further, it may be coadministered with catechol‐O‐methyl transferase (COMT) inhibitor entacapone and tolcapone [175, 176, 177, 178].

L‐Dopa is the direct biosynthetic precursor of dopamine, is actively transported across the blood–brain barrier and is transformed to dopamine by aromatic‐L‐aminoacid decarboxylase. One site of the decarboxylation is in the remaining dopaminergic neurons, but serotoninergic, noradrenergic, and “D” neurons and glial cells have also been shown to take up L‐Dopa and transform it to dopamine [179, 180, 181]. The importance of decarboxylation in the nondopaminergic neurons and glia is not yet clear. Dopamine produced in nondopaminergic cells may be taken up by dopamine transporters in dopaminergic neuron terminals and packed into dopamine‐containing synaptic vesicles by vesicular monoamine transporter 2 (VMAT2). A reduced level of VMAT2 has been demonstrated to lead to dopamine‐mediated neurodegeneration [182], and gain‐of‐function haplotypes that display increased transcriptional activity have been found to be protective against PD in women [183]. Cytosolic dopamine accumulation itself has been suggested to be neurotoxic through the generation of ROS [184]. The Earlier versus Later Levodopa (ELLDOPA) study, which examined the question of whether the L‐Dopa therapy hastens the disease course [185], did not reveal detrimental effect of L‐dopa therapy, and after 2 weeks of wash‐out patients on L‐Dopa therapy still exhibited beneficial effects. However, the wash‐out time is much longer, and interpretation of the results of the imaging substudy of ELLDOPA led to the possible neuroprotective effect of L‐Dopa remaining in question.

Dopamine Agonists

Dopamine agonists, which were introduced into the therapy of PD in the 1970s have several advantages over L‐Dopa. As they do not need metabolic modification, their effect is not dependent on the surviving dopaminergic cell pool. Their effect does not depend on the dopamine metabolism, and consequently is not related to the possible toxic effect of dopamine. The derivation drugs selectively acting only on certain types of dopamine receptor might afford a possibility to reduce the dopaminergic adverse effects.

Dopamine agonists such as bromocriptine (a potent D2 agonist and mild D1 receptor antagonist), carbergolide (a long‐acting D2 agonist), pergolide (a D1, D2, and somewhat selective D3 receptor agonist), and ropinirole (a nonergot dopamine agonist) have been shown to exert free radical‐scavenging action and to protect against MPTP, dopamine, 6‐OHDA, and nitric oxide toxicity both in vitro and in vivo animal models [186, 187, 188, 189, 190, 191, 192]. The antioxidant effects of these drugs are related to their molecular structure (a hydroxylated benzyl ring), downregulation of dopamine turnover (partly a D2 receptor‐dependent effect) [193, 194], and also the upregulation of protective scavenging enzymes (e.g., they upregulate the expressions of glutathione‐related enzymes) [190, 194].

It has been suggested that apoptogenic cytochrome c released from the mitochondria induces an apoptotic process through Apaf‐1, caspase‐9, and caspase‐3 that is later involved in the pathogenesis of PD [195]. Cytochrome c, a heme protein that is an essential component of the mitochondrial electron transport system, has also been denoted as a stimulator of alpha‐synuclein aggregation [196]. The dopamine agonists talipexole and pramipexole have been shown to possess neuroprotective effects in MPTP‐treated mice [197], 6‐OHDA‐treated rats [198], invertebrate flatworms [199], the human neuroblastoma cell line SH‐SY5Y [200], and nonhuman primates [201], the effect being related to inhibition of the release of cytochrome c and aggregation of alpha‐synuclein [202]. It was also demonstrated that this neuroprotective effect of pramipexole is not dependent on dopamine receptors or either antioxidative effect of the drug [203], but may be related to protection against glutamate excitotoxicity by reduction of the intracellular dopamine content [204]. However, evidence has been provided for at least a partial dopaminergic effect [205].

Recent results have indicated that dopamine agonists may exert their neuroprotective effect through growth factor‐like properties. Pramipexole and ropinirole acted on dopaminergic neurons by modulating the production of endogenous brain‐derived (BDNF) and glial cell line‐derived neurotrophic factors [206] that were shown to mediate neuroprotection [207]. Several lines of evidence suggest that BDNF plays a substantial role in preventing the neuronal degeneration seen in PD. Reduced BDNF mRNA expression was found in the substantia nigra of PD patients [208]. In alpha‐synuclein‐transfected glioma cells, the expression of BDNF was induced, but not by the A30P and A53T‐mutated alpha‐synuclein gene [209]. It was also demonstrated that inhibition of BDNF expression by antisense oligonucleotide infusion causes the loss of nigral dopaminergic neurons [210]. The Val66Met polymorphism of BDNF gene was proposed to be a susceptibility factor for PD [211, 212], but the experimental findings are not clear‐cut [213, 214].

It is important that besides evidence from in vitro and animal studies, dopamine agonists have also been evaluated and their disease‐modifying effects confirmed in clinical trials. The imaging surrogate marker of beta‐CIT SPECT was utilized in the CALM‐PD study [215] and fluoro‐dopa PET the REAL‐PET study [216]. The conclusion of both investigations was that the dopamine agonists pramipexole and ropinirole may have a neuroprotective effect. Whereas the use of imaging biomarkers is certainly justified, the exact relation to clinical measures must be further evaluated before use of such surrogate markers in clinical trials.

MAO‐B Inhibitors

Monoamine oxidase (MAO) embedded in the mitochondrial outer membrane [217] is responsible for the degradation of dopamine. Within the central nervous system, the bulk of the enzyme is present as the B isoenzyme. As one of the key components of the oxidative degradation of dopamine and in the consequent oxidative stress, it has a central role in the pathogenesis of PD.

Selegiline, a selective and irreversible inhibitor of MAO‐B, first synthetized by the Hungarian pharmacologist Ecseri [218] and characterized by Knoll and Magyar [219], improves parkinsonian symptoms and is approved worldwide for use as adjunctive therapy to L‐Dopa. Rasagiline, an indane derivative, is a novel irreversible inhibitor of MAO‐B. In contrast with selegiline, the metabolites of rasagiline do not possess amphetamine‐like effects and are not vasoactive either [220]. A metabolite of selegiline inhibits the neuroprotective effect of the drug, but no similar inhibition is observed in the case of rasagiline [221].

It is well known that MPTP toxicity can be avoided by the use of MAO‐B inhibitors [222, 223]. The metabolism of MPTP by MAO‐B to MPP+ that generates free radicals is linked to the pathogenesis of PD [223, 224]. Nevertheless, the neuroprotective effect of MAO‐B inhibitors does not depend only on the enzyme‐blocker properties of the drugs. The less potent enantiomers of selegiline and rasagiline still have a significant neuroprotective effect and the effect is also observed in the human dopaminergic cell line SH‐SY5Y, which does not express MAO‐B [225, 226]. The antiapoptotic effect is related to inhibition of decrease in the mitochondrial membrane potential. The mitochondrial permeability transition pore that controls the initiation step of the apoptotic death process by reducing the mitochondrial membrane potential has a tertiary structure similar to that of MAO‐B, which may be responsible for the neuroprotective effect of rasagiline. Alternatively, rasagiline has been observed to increase the expression of Bcl‐2, a direct regulator of the mitochondrial permeability transition pore, and hence to have an antiapoptotic property [227, 228]. Importantly, rasagiline exerts neurorestorative action when administered posttreatment after MPTP neurotoxicity. This effect is linked to activation of the pathway involving the neurotrophic tyrosine kinase receptor [229] a ligand of BDNF, the expression of which is also modulated by rasagiline [230, 231].

A major metabolite of rasagiline, the 1‐(R)‐aminoindane was also shown to have neuroprotective effect in several in vitro and in vivo animal studies [232, 233]. The mechanism of neuroprotectant effect of 1‐(R)‐aminoindane was related to modulation of apoptosis related proteins, such as decreasing the level of H2A.X, decreasing the cleavage and increasing the expression of caspase 9 and 3, increasing the level of antiapoptotic proteins Bcl‐2 and Bcl‐xl [232].

Furthermore, novel bifunctional MAO‐B inhibitors possess also an iron chelating property that might be a key factor of the neuroprotectant feature of these drugs (HLA‐20, M30) [234, 235, 236, 237]. In these molecules, the propargylamid and MAO‐B inhibitor moiety of rasagiline is combined with and iron chelating properties, consequently these drugs are able to block the iron‐mediated production of free radicals in the Fenton reaction.

The neuroprotective effect of the MAO‐B inhibitors selegiline and rasagiline has been the subject of several clinical trials. The Deprenyl and Tocopherol Antioxidative Therapy of Parkinsonism (DATATOP) study [238] revealed a beneficial effect of selegiline, but not of tocopherol, when the introduction of L‐Dopa therapy was delayed. The Sinemet‐Deprenyl‐Parlodar (Sindepar) study indicated the superiority of selegiline as compared with placebo, as measured via the primary endpoint of the study: the change in UPDRS between the first and the final visit, after a 2‐month of wash‐out period following a 12‐month treatment [239]. The recently reported initial results of the delayed start design Attenuation of Disease progression with Azilect GIven Once‐daily (ADAGIO) study also supported the neuroprotective effect of rasagiline [240, 241].

Antiinflammatory Therapies

Nonsteroid antiinflammatory drugs (NSAID) and especially nonaspirin NSAIDs such as ibuprophen, have also been suggested to have a beneficial effect in PD [242, 243], though the studies have not proved so far conclusive [244, 245]. The mechanism by which NSAIDs exert their neuroprotective effect is still elusive. One obvious possibility is that they act by inhibiting the proinflammatory cyclooxygenase, but their ROS‐scavenging effect may also be important [246].

Minocycline a semisynthetic second‐generation tetracycline has both antimicrobial and antiinflammatory effects. Minocycline has been found to be neuroprotective in a preclinical MPTP‐induced PD model [247, 248]. The neuroprotective effect is related to inhibition of the MPTP‐induced glial inducible nitrogen monoxide synthase expression and inhibition of the phosphorylation of p38 mitogen‐activated protein kinase [247]. A phase II clinical investigation (the Neuroprotective Exploratory Trials in Parkinson Disease, Futility Study 1 – NET‐PD FS‐1) did not rule out the potential value of the drug, and suggested further phase III trials [249, 250].

There is also substantial interest in glatiramer acetate (Copolymer 1, Copaxone), which induces regulator T cells that secrete high amounts of antiinflammatory T‐helper 2/3 cytokines [251, 252]. It is also known that glatiramer acetate‐specific T‐helper cells can be stimulated to produce BDNF [251, 253, 254]. The neuroprotective effect has been demonstrated in preclinical animal studies [255, 256, 257]. Further studies are warranted to evaluate the effectiveness of Copaxone in PD.

Creatine

Creatine is a natural guanidine compound that plays a pivotal role in the cell energy metabolism. The creatine system serves as a spatial energy buffer between the mitochondria, the energetic centers of the cell, and the cytosolic energy‐consuming processes. Mitochondrial creatine kinase catalyzes the reversible transfer of a phosphate group from ATP, the high‐energy product of the mitochondrial respiratory chain, to creatine, yielding high‐energy phosphocreatinine.

In vitro and in vivo animal studies have indicated the neuroprotective effect of creatine [258]. One possible mechanism of creatine‐induced neuroprotection is stabilization of the energy balance of the cells. Creatine treatment has been shown to increase the level of phosphocreatine [259, 260], which is an important energy supplementation when the mitochondrial respiratory chain is deficient (e.g., complex I inhibition by MPTP). Another possible mechanism of neuroprotection might be inhibition of the mitochondrial permeability transition pore, the initial step of apoptosis. Creatine has been reported to stabilize the mitochondrial isoform of creatine kinase in an octomeric form that in turn can inhibit the mitochondrial permeability transition pore [261, 262], but this was questioned by the report that creatine has a neuroprotective effect in MPTP‐treated mice deficient in ubiquitous mitochondrial creatine kinase [263]. The neuroprotective effect of creatine has also been associated with direct antioxidant properties [264]. The phase II clinical trial NET‐PD FS‐1 did not exclude the potential of creatine in the treatment of PD [249, 250].

Alpha‐tocopherol

Tocopherol, one of the compounds in vitamin E is the main fat‐soluble antioxidant in the organism. It is incorporated into cellular membranes and effectively inhibits the peroxidation of lipids by scavenging the chain‐propagating peroxyl radicals [265]. Tocopherol does not act in isolation, but as part of the antioxidant network, which also involves vitamin C, thiol antioxidants such as glutathione and lipoic acid.

Given that oxidative damage plays a central role in the pathogenesis of PD, antioxidants such as tocopherol are presumed to have a neuroprotective effect. MPTP toxicity was more pronounced in vitamin E‐deficient mice and this effect was related to lipid peroxidation [266]. A dietary vitamin E deficiency itself causes a loss of tyrosine hydroxylase‐immunopositive neurons in the substantia nigra of rats [267], presumably because of a defective antioxidative network. A number of studies have demonstrated neuroprotective effects of vitamin E [268, 269]. On the other hand, vitamin E pretreatment does not attenuate MPTP striatal neurotoxicity [270, 271] and a genetic vitamin E deficiency does not affect MPTP susceptibility [272].

Clinical studies have been carried out on the possible disease‐modifying effect of alpha‐tocopherol. An open‐label study concluded that alpha‐tocopherol and ascorbic acid administered together deferred the initiation of L‐Dopa by 2.5 years [273]. However, the multicenter, placebo‐controlled clinical trial Deprenyl and Tocopherol Antioxidative Therapy of Parkinsonism (DATATOP) [238] did not reveal a positive disease‐modifying effect of tocopherol.

Coenzyme Q

Coenzyme Q10 (CoQ10), also known as ubiquinone, is present in eukaryotic cells mainly in the mitochondria. It contains 1,4‐benzoquinone and 10 isoprenal subunits. It exists in three different redox states (the fully oxidized ubiquinone, the free radical ubisemiquinone, and a fully reduced ubiquinol) and is an important component of the mitochondrial electron transport system. It transports electrons from complexes I and II to complex III of the electron transport chain. CoQ10 behaves as a free radical scavenger [274], this effect depending, at least partly, on the recycling of alpha‐tocopherol [275]. It has been suggested that it acts in concert with dihydrolipoic acid, an organic antioxidant compound that has been also implicated as a potent neuroprotective agent [276, 277].

CoQ10 exerts a neuroprotective effect in various in vivo models, for example, ischemia [278], malonate (an inhibitor of succinate dehydrogenase)‐induced striatal degeneration [279], MPTP toxicity [280], and paraquat‐induced oxidative stress [281]. This latter study is of especial importance because it involved use of the water‐soluble formulation of CoQ10, which is thought to have significantly higher bioavailability [282].

A clinical trial evaluating the efficacy through measurement of the change in UPDRS score also highlighted a possible beneficial effect of CoQ10 in early PD [283], but no effect on the time to initiate symptomatic treatment. The NINDS NET‐PD futility study did not reject CoQ10 as a potential agent and suggested phase III trials; however, the chosen futility threshold was questioned [284].

Tryptophan Metabolism in Neuroprotection

The essential amino acid tryptophan is metabolized in several alternative pathways, one of the most important of which is the kynurenine pathway [285], yielding the final product nicotinamide adenosine dinucleotide (NAD) (Figure 3).

Figure 3.

Figure 3

The kynurenine pathway of the tryptophan metabolism.

The first stable metabolite of the pathway is kynurenine, which is transformed by kynurenine aminotransferase either to kynurenic acid or by kynurenine hydroxylase to 3‐hydroxy kynurenine, which is further metabolized to quinolic acid, the precursor of NAD. These metabolites are usually referred to as neuroactive kynurenines [286, 287]. Kynurenic acid is an antagonist of the strychnine‐insensitive glycine‐binding site of the NMDA receptor [288, 289], a weak antagonist of AMPA and kainite receptors [290] and also an inhibitor of the alpha7 nicotinic receptor [291], which is involved in the presynaptic regulation of glutamate release. Importantly, the neuroinhibitory effect of kynurenic acid is concentration‐dependent: in nanomolar concentrations, it facilitates field excitatory postsynaptic potentials [292]. 3‐hyrdoxykynurenine and quinolic acid are both neurotoxic [293], and were shown to be the direct activator of NMDA receptors [294], modulate the release or reuptake inhibition of glutamate [295], be involved in lipid peroxidation [296] and the production of ROS [297].

The relation of the kynurenine pathway to PD has been demonstrated in several investigations. The level of kynurenine aminotransferase I (the enzyme that converts kynurenine to kynurenic acid) was found to be reduced after MPTP and 6‐OHDA toxicity in the striatum of the rat [298, 299]. MPP+ also reduces the level of kynurenic acid by inhibiting kynurenine aminotransferase II. In a human postmortem study, the concentration of both kynurenine and kynurenic acid proved to be reduced, in contrast with that of 3‐hydroxykynurenine, which concentration was increased in several brain regions. Reduced level of kynurenine has been measured in serum of PD patients, which has been paralleled with reduced kynurenine aminotransferases activity [300]. The importance of these studies was speculated to lie in reduced NMDA antagonism, as a consequence of the reduced level of kynurenic acid or a shift in the tryptophan metabolism in the direction of 3‐hydroxykynurenine and quinolone, both propagating neurotoxicity. In accordance with the former hypothesis, glutamate antagonists were shown to protect against MPP+ toxicity [301], signifying the importance of the glutamatergic exitotoxicity in the pathogenesis of PD. Importantly, the role of the non‐NMDA, AMPA glutamate receptor too was demonstrated in kynurenic acid‐related neuroprotection [302]. Another possible link between the kynurenine pathway and PD is the immune system. Interferon‐gamma modulates indoleamine 2,3‐dioxygenase, the enzyme that catalyzes the tryptophan‐kynurenin transformation [303]. In PD patients, an amplified cellular immune activation was fond to be paralleled by an increased kynurenine/tryptophan ratio [304].

From the above results, it is intuitive that a shift of the balance of the kynurenine pathway in the direction of kynurenic acid may have a potential neuroprotective effect [305]. The increase of endogenous kynurenic acid by nicotinylalanine, inhibiting kynureninase and kynurenine hydroxylase, protects against NMDA and quinolic acid toxicity [306]. Furthermore, Ro 61 8048, a kynurenine hydroxylase inhibitor, reduces the striatal glutamate level besides increasing the kynurenic acid concentration in the striatum, the cortex and the hippocampus [307]. A similar effect has been demonstrated in nonhuman primates [308].

Another option via which to modulate the kynurenine pathway in order to achieve neuroprotection is the use of kynurenine analogues. 7‐Chlorokynurenate, a synthetic derivative of kynurenic acid and an antagonist of the glycine‐binding site of the NMDA receptor, effectively protects against NMDA receptor‐mediated toxicity [309, 310]. The poor penentrance of the drug can be overcome through the use of 4‐chlorokynurenine a precursor of 7‐chlorokynurenate, that readily crosses the blood–brain barrier and is converted enzymatically to the active metabolite [311, 312].

Conclusions

The central mechanism of neurodegeneration shares common pathways in several neurodegenerative diseases, for example, PD, Huntington's disease, amyotrophic lateral sclerosis and Alzheimer's disease. Investigations into these processes have led to development of novel therapeutic approaches shown to be effective in in vitro studies and animal models. A small number of human investigations have also been carried out and positive results have been published, but no medication has yet proved unanimously neuroprotective. Further clinical trials are needed to unravel the potential neuroprotective effects of the above‐cited drugs and approaches. On the other hand, the pathomechanism of PD is far from clear and additional investigations are needed to identify the possible pathways that could be targeted for therapeutic interventions.

Author Contributions

Dr. Kincses and Dr. Vécsei both collaborated on the concept of the article. Drafting was carried out by Dr. Kincses.

Conflict of Interest

Both authors are employed by the Albert Szent‐Györgyi Clinical Center, University of Szeged. Neither of the authors reports any conflict of interest.

Acknowledgment

Funding of the studies reported in the article includes Teller Ede (NAP‐BIO‐06‐BAYBIOSZ), ETT 026‐04, TÁMOP‐4.2.2.‐08/1/2008‐2002, and cNEUPRO (LSHM‐CT‐2007‐037950).

References

  • 1. Parkinson J. An essay on the shaking palsy. London: Sherwood, Neely and Jones; 1817. Reprinted in: Neuropsychiatric classics. J Neuropsychiatry Clin Neurosci 2002;14:223–236. [DOI] [PubMed] [Google Scholar]
  • 2. Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino acids and modification of parkinsonism. N Engl J Med 1967;276:374–379. [DOI] [PubMed] [Google Scholar]
  • 3. Rakoczi K, Klivenyi P, Vecsei L. Neuroprotection in Parkinson's disease and other neurodegenerative disorders: Preclinical and clinical findings. Ideggyogy Sz 2009;62:25–34. [PubMed] [Google Scholar]
  • 4. Baldereschi M, Di Carlo A, Rocca WA, et al Parkinson's disease and parkinsonism in a longitudinal study: Two‐fold higher incidence in men. ILSA Working Group. Italian Longitudinal Study on Aging. Neurology 2000;55:1358–1363. [DOI] [PubMed] [Google Scholar]
  • 5. de Lau LM, Giesbergen PC, de Rijk MC, Hofman A, Koudstaal PJ, Breteler MM. Incidence of parkinsonism and Parkinson disease in a general population: The Rotterdam Study. Neurology 2004;63:1240–1244. [DOI] [PubMed] [Google Scholar]
  • 6. Galvin JE. Cognitive change in Parkinson disease. Alzheimer Dis Assoc Disord 2006;20:302–310. [DOI] [PubMed] [Google Scholar]
  • 7. Antal A, Keri S, Kincses ZT, et al Dopaminergic contributions to the visual categorization of natural scenes: Evidence from Parkinson's disease. J Neural Transm 2003;110:757–770. [DOI] [PubMed] [Google Scholar]
  • 8. Antal A, Keri S, Dibo G, et al Electrophysiological correlates of visual categorization: Evidence for cognitive dysfunctions in early Parkinson's disease. Brain Res Cogn Brain Res 2002;13:153–158. [DOI] [PubMed] [Google Scholar]
  • 9. Antal A, Keri S, Kincses T, et al Corticostriatal circuitry mediates fast‐track visual categorization. Brain Res Cogn Brain Res 2002;13:53–59. [DOI] [PubMed] [Google Scholar]
  • 10. Antal A, Dibo G, Keri S, et al P300 component of visual event‐related potentials distinguishes patients with idiopathic parkinson's disease from patients with essential tremor. J Neural Transm 2000;107:787–797. [DOI] [PubMed] [Google Scholar]
  • 11. Antal A, Bandini F, Keri S, Bodis‐Wollner I. Visuo‐cognitive dysfunctions in Parkinson's disease. Clin Neurosci 1998;5:147–152. [PubMed] [Google Scholar]
  • 12. Forno LS. Concentric hyalin intraneuronal inclusions of Lewy type in the brains of elderly persons (50 incidental cases): Relationship to parkinsonism. J Am Geriatr Soc 1969;17:557–575. [DOI] [PubMed] [Google Scholar]
  • 13. Ishiyama M, Yagishita S, Hasegawa K, Yokoyama T. Ultrastructural study (transmission and scanning EM) of cortical Lewy bodies. Neuropathology 2006;26:A58. [Google Scholar]
  • 14. Jellinger KA. Formation and development of Lewy pathology: A critical update. J Neurol 2009;256(Suppl 3):270–279. [DOI] [PubMed] [Google Scholar]
  • 15. 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] [PubMed] [Google Scholar]
  • 16. Kalaitzakis ME, Graeber MB, Gentleman SM, Pearce RK. Controversies over the staging of alpha‐synuclein pathology in Parkinson's disease. Acta Neuropathol 2008;116:125–128; author reply 9–31. [DOI] [PubMed] [Google Scholar]
  • 17. Parkkinen L, Pirttila T, Alafuzoff I. Applicability of current staging/categorization of alpha‐synuclein pathology and their clinical relevance. Acta Neuropathol 2008;115:399–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Beal MF. Bioenergetic approaches for neuroprotection in Parkinson's disease. Ann Neurol 2003;53(Suppl 3):S39–S47; discussion S‐8. [DOI] [PubMed] [Google Scholar]
  • 19. Bogaerts V, Theuns J, van Broeckhoven C. Genetic findings in Parkinson's disease and translation into treatment: A leading role for mitochondria? Genes Brain Behav 2008;7:129–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. 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] [PubMed] [Google Scholar]
  • 21. Kunikowska G, Jenner P. Alterations in m‐RNA expression for Cu,Zn‐superoxide dismutase and glutathione peroxidase in the basal ganglia of MPTP‐treated marmosets and patients with Parkinson's disease. Brain Res 2003;968:206–218. [DOI] [PubMed] [Google Scholar]
  • 22. Kienzl E, Puchinger L, Jellinger K, Linert W, Stachelberger H, Jameson RF. The role of transition metals in the pathogenesis of Parkinson's disease. J Neurol Sci 1995;134(Suppl):69–78. [DOI] [PubMed] [Google Scholar]
  • 23. Riederer P, Sofic E, Rausch WD, et al Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J Neurochem 1989;52:515–520. [DOI] [PubMed] [Google Scholar]
  • 24. Dexter DT, Wells FR, Lees AJ, et al Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson's disease. J Neurochem 1989;52:1830–1836. [DOI] [PubMed] [Google Scholar]
  • 25. Jellinger K, Kienzl E, Rumpelmair G, et al Iron‐melanin complex in substantia nigra of parkinsonian brains: An x‐ray microanalysis. J Neurochem 1992;59:1168–1171. [DOI] [PubMed] [Google Scholar]
  • 26. Behnke S, Schroeder U, Dillmann U, et al Hyperechogenicity of the substantia nigra in healthy controls is related to MRI changes and to neuronal loss as determined by F‐Dopa PET. Neuroimage 2009;47:1237–1243. [DOI] [PubMed] [Google Scholar]
  • 27. Antonini A, Leenders KL, Meier D, Oertel WH, Boesiger P, Anliker M. T2 relaxation time in patients with Parkinson's disease. Neurology 1993;43:697–700. [DOI] [PubMed] [Google Scholar]
  • 28. Maker HS, Weiss C, Silides DJ, Cohen G. Coupling of dopamine oxidation (monoamine oxidase activity) to glutathione oxidation via the generation of hydrogen peroxide in rat brain homogenates. J Neurochem 1981;36:589–593. [DOI] [PubMed] [Google Scholar]
  • 29. Stokes AH, Hastings TG, Vrana KE. Cytotoxic and genotoxic potential of dopamine. J Neurosci Res 1999;55:659–665. [DOI] [PubMed] [Google Scholar]
  • 30. Gerlach M, Riederer P, Przuntek H, Youdim MB. MPTP mechanisms of neurotoxicity and their implications for Parkinson's disease. Eur J Pharmacol 1991;208:273–286. [DOI] [PubMed] [Google Scholar]
  • 31. 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] [PubMed] [Google Scholar]
  • 32. Blin O, Desnuelle C, Rascol O, et al Mitochondrial respiratory failure in skeletal muscle from patients with Parkinson's disease and multiple system atrophy. J Neurol Sci 1994;125:95–101. [DOI] [PubMed] [Google Scholar]
  • 33. Cardellach F, Marti MJ, Fernandez‐Sola J, et al Mitochondrial respiratory chain activity in skeletal muscle from patients with Parkinson's disease. Neurology 1993;43:2258–2262. [DOI] [PubMed] [Google Scholar]
  • 34. Parker WD Jr., Boyson SJ, Parks JK. Abnormalities of the electron transport chain in idiopathic Parkinson's disease. Ann Neurol 1989;26:719–723. [DOI] [PubMed] [Google Scholar]
  • 35. Swerdlow RH, Parks JK, Miller SW, et al Origin and functional consequences of the complex I defect in Parkinson's disease. Ann Neuro 1996;40:663–671. [DOI] [PubMed] [Google Scholar]
  • 36. Trimmer PA, Bennett JP Jr. The cybrid model of sporadic Parkinson's disease. Exp Neurol 2009;218:320–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Karu TI, Pyatibrat LV, Afanasyeva NI. A novel mitochondrial signaling pathway activated by visible‐to‐near infrared radiation. Photochem Photobiol 2004;80:366–372. [DOI] [PubMed] [Google Scholar]
  • 38. Keeney PM, Quigley CK, Dunham LD, et al Mitochondrial gene therapy augments mitochondrial physiology in a Parkinson's disease cell model. Hum Gene The 2009;20:897–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Liss B, Haeckel O, Wildmann J, Miki T, Seino S, Roeper J. K‐ATP channels promote the differential degeneration of dopaminergic midbrain neurons. Nat Neurosci 2005;8:1742–1751. [DOI] [PubMed] [Google Scholar]
  • 40. Hoglinger GU, Carrard G, Michel PP, et al Dysfunction of mitochondrial complex I and the proteasome: Interactions between two biochemical deficits in a cellular model of Parkinson's disease. J Neuroche 2003;86:1297–1307. [DOI] [PubMed] [Google Scholar]
  • 41. Klivenyi P, Kekesi KA, Hartai Z, Juhasz G, Vecsei L. Effects of mitochondrial toxins on the brain amino acid concentrations. Neurochem Res 2005;30:1421–1427. [DOI] [PubMed] [Google Scholar]
  • 42. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA‐DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology 1988;38:1285–1291. [DOI] [PubMed] [Google Scholar]
  • 43. Imamura K, Hishikawa N, Sawada M, Nagatsu T, Yoshida M, Hashizume Y. Distribution of major histocompatibility complex class II‐positive microglia and cytokine profile of Parkinson's disease brains. Acta Neuropathol 2003;106:518–526. [DOI] [PubMed] [Google Scholar]
  • 44. Mirza B, Hadberg H, Thomsen P, Moos T. The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson's disease. Neuroscience 2000;95:425–432. [DOI] [PubMed] [Google Scholar]
  • 45. Brochard V, Combadiere B, Prigent A, et al Infiltration of CD4 + lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest 2009;119:182–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Mogi M, Harada M, Narabayashi H, Inagaki H, Minami M, Nagatsu T. Interleukin (IL)‐1 beta, IL‐2, IL‐4, IL‐6 and transforming growth factor‐alpha levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson's disease. Neurosci Lett 1996;211:13–16. [DOI] [PubMed] [Google Scholar]
  • 47. Mogi M, Harada M, Kondo T, Riederer P, Nagatsu T. Interleukin‐2 but not basic fibroblast growth factor is elevated in parkinsonian brain. Short communication. J Neural Transm 1996;103:1077–1081. [DOI] [PubMed] [Google Scholar]
  • 48. Mogi M, Harada M, Kondo T, Narabayashi H, Riederer P, Nagatsu T. Transforming growth factor‐beta 1 levels are elevated in the striatum and in ventricular cerebrospinal fluid in Parkinson's disease. Neurosci Lett 1995;193:129–132. [DOI] [PubMed] [Google Scholar]
  • 49. Mogi M, Harada M, Kondo T, Riederer P, Nagatsu T. Brain beta 2‐microglobulin levels are elevated in the striatum in Parkinson's disease. J Neural Transm Park Dis Dement Sect 1995;9:87–92. [DOI] [PubMed] [Google Scholar]
  • 50. Mogi M, Harada M, Kondo T, et al Interleukin‐1 beta, interleukin‐6, epidermal growth factor and transforming growth factor‐alpha are elevated in the brain from parkinsonian patients. Neurosci Lett 1994;180:147–150. [DOI] [PubMed] [Google Scholar]
  • 51. Mogi M, Harada M, Riederer P, Narabayashi H, Fujita K, Nagatsu T. Tumor necrosis factor‐alpha (TNF‐alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci Lett 1994;165:208–210. [DOI] [PubMed] [Google Scholar]
  • 52. Mogi M, Harada M, Kojima K, et al Beta 2‐microglobulin decrease in cerebrospinal fluid from parkinsonian patients. Neurosci Lett 1989;104:241–246. [DOI] [PubMed] [Google Scholar]
  • 53. Stypula G, Kunert‐Radek J, Stepien H, Zylinska K, Pawlikowski M. Evaluation of interleukins, ACTH, cortisol and prolactin concentrations in the blood of patients with parkinson's disease. Neuroimmunomodulation 1996;3:131–134. [DOI] [PubMed] [Google Scholar]
  • 54. Dobbs RJ, Charlett A, Purkiss AG, Dobbs SM, Weller C, Peterson DW. Association of circulating TNF‐alpha and IL‐6 with ageing and parkinsonism. Acta Neurol Scand 1999;100:34–41. [DOI] [PubMed] [Google Scholar]
  • 55. Rentzos M, Nikolaou C, Andreadou E, et al Circulating interleukin‐15 and RANTES chemokine in Parkinson's disease. Acta Neurol Scand 2007;116:374–379. [DOI] [PubMed] [Google Scholar]
  • 56. Rowe DB, Le W, Smith RG, Appel SH. Antibodies from patients with Parkinson's disease react with protein modified by dopamine oxidation. J Neurosci Res 1998;53:551–558. [DOI] [PubMed] [Google Scholar]
  • 57. Blum‐Degen D, Muller T, Kuhn W, Gerlach M, Przuntek H, Riederer P. Interleukin‐1 beta and interleukin‐6 are elevated in the cerebrospinal fluid of Alzheimer's and de novo Parkinson's disease patients. Neurosci Lett 1995;202:17–20. [DOI] [PubMed] [Google Scholar]
  • 58. Gerhard A, Pavese N, Hotton G, et al In vivo imaging of microglial activation with [11C](R)‐PK11195 PET in idiopathic Parkinson's disease. Neurobiol Dis 2006;21:404–412. [DOI] [PubMed] [Google Scholar]
  • 59. Ouchi Y, Yoshikawa E, Sekine Y, et al Microglial activation and dopamine terminal loss in early Parkinson's disease. Ann Neurol 2005;57:168–175. [DOI] [PubMed] [Google Scholar]
  • 60. Kruger R, Hardt C, Tschentscher F, et al Genetic analysis of immunomodulating factors in sporadic Parkinson's disease. J Neural Transm 2000;107:553–562. [DOI] [PubMed] [Google Scholar]
  • 61. Wu YR, Feng IH, Lyu RK, et al Tumor necrosis factor‐alpha promoter polymorphism is associated with the risk of Parkinson's disease. Am J Med Genet B Neuropsychiatr Genet 2007;144B:300–304. [DOI] [PubMed] [Google Scholar]
  • 62. Nishimura M, Mizuta I, Mizuta E, Yamasaki S, Ohta M, Kuno S. Influence of interleukin‐1beta gene polymorphisms on age‐at‐onset of sporadic Parkinson's disease. Neurosci Lett 2000;284:73–76. [DOI] [PubMed] [Google Scholar]
  • 63. Bialecka M, Klodowska‐Duda G, Kurzawski M, et al Interleukin‐10 (IL10) and tumor necrosis factor alpha (TNF) gene polymorphisms in Parkinson's disease patients. Parkinsonism Relat Disord 2008;14:636–640. [DOI] [PubMed] [Google Scholar]
  • 64. Nishimura M, Mizuta I, Mizuta E, et al Tumor necrosis factor gene polymorphisms in patients with sporadic Parkinson's disease. Neurosci Lett 2001;311:1–4. [DOI] [PubMed] [Google Scholar]
  • 65. Lin JJ, Chen CH, Yueh KC, Chang CY, Lin SZ. A CD14 monocyte receptor polymorphism and genetic susceptibility to Parkinson's disease for females. Parkinsonism Relat Disord 2006;12:9–13. [DOI] [PubMed] [Google Scholar]
  • 66. Klivenyi P, Ferrante RJ, Matthews RT, et al Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat Med 1999;5:347–350. [DOI] [PubMed] [Google Scholar]
  • 67. Choi DK, Pennathur S, Perier C, et al Ablation of the inflammatory enzyme myeloperoxidase mitigates features of Parkinson's disease in mice. J Neurosci 2005;25:6594–6600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Wu DC, Teismann P, Tieu K, et al NADPH oxidase mediates oxidative stress in the 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine model of Parkinson's disease. Proc Natl Acad Sci USA 2003;100:6145–6150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Furuya T, Hayakawa H, Yamada M, et al Caspase‐11 mediates inflammatory dopaminergic cell death in the 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine mouse model of Parkinson's disease. J Neurosci 2004;24:1865–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Block ML, Hong JS. Microglia and inflammation‐mediated neurodegeneration: Multiple triggers with a common mechanism. Prog Neurobiol 2005;76:77–98. [DOI] [PubMed] [Google Scholar]
  • 71. Qian L, Hong JS, Flood PM. Role of microglia in inflammation‐mediated degeneration of dopaminergic neurons: Neuroprotective effect of interleukin 10. J Neural Transm 2006;(Suppl 70):367–371. [DOI] [PubMed] [Google Scholar]
  • 72. Chau KY, Ching HL, Schapira AH, Cooper JM. Relationship between alpha synuclein phosphorylation, proteasomal inhibition and cell death: Relevance to Parkinson's disease pathogenesis. J Neurochem 2009;110:1005–1013. [DOI] [PubMed] [Google Scholar]
  • 73. Maguire‐Zeiss KA. alpha‐Synuclein: A therapeutic target for Parkinson's disease? Pharmacol Res 2008;58:271–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Olanow CW, Prusiner SB. Is Parkinson's disease a prion disorder? Proc Natl Acad Sci USA 2009;106:12571–12572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Kordower JH, Chu Y, Hauser RA, Olanow CW, Freeman TB. Transplanted dopaminergic neurons develop PD pathologic changes: A second case report. Mov Disord 2008;23:2303–2306. [DOI] [PubMed] [Google Scholar]
  • 76. Li JY, Englund E, Holton JL, et al Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host‐to‐graft disease propagation. Nat Med 2008;14:501–503. [DOI] [PubMed] [Google Scholar]
  • 77. Desplats P, Lee HJ, Bae EJ, et al Inclusion formation and neuronal cell death through neuron‐to‐neuron transmission of alpha‐synuclein. Proc Natl Acad Sci USA 2009;106:13010–13015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Dingledine R, Borges K, Bowie D, Traynelis SF. The glutamate receptor ion channels. Pharmacol Rev 1999;51:7–61. [PubMed] [Google Scholar]
  • 79. Greenamyre JT, Porter RH. Anatomy and physiology of glutamate in the CNS. Neurology 1994;44(11 Suppl 8):S7–S13. [PubMed] [Google Scholar]
  • 80. Hetz CA. ER stress signaling and the BCL‐2 family of proteins: From adaptation to irreversible cellular damage. Antioxid Redox Signal 2007;9:2345–2355. [DOI] [PubMed] [Google Scholar]
  • 81. Lindholm D, Wootz H, Korhonen L. ER stress and neurodegenerative diseases. Cell Death Differ 2006;13:385–392. [DOI] [PubMed] [Google Scholar]
  • 82. McCormack JG, Halestrap AP, Denton RM. Role of calcium ions in regulation of mammalian intramitochondrial metabolism. Physiol Rev 1990;70:391–425. [DOI] [PubMed] [Google Scholar]
  • 83. Chan CS, Gertler TS, Surmeier DJ. Calcium homeostasis, selective vulnerability and Parkinson's disease. Trends Neurosci 2009;32:249–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Rodriguez MC, Obeso JA, Olanow CW. Subthalamic nucleus‐mediated excitotoxicity in Parkinson's disease: A target for neuroprotection. Ann Neurol 1998;44(3 Suppl 1):S175–188. [DOI] [PubMed] [Google Scholar]
  • 85. Bonci A, Grillner P, Mercuri NB, Bernardi G. L‐Type calcium channels mediate a slow excitatory synaptic transmission in rat midbrain dopaminergic neurons. J Neurosci 1998;18:6693–6703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Ping HX, Shepard PD. Apamin‐sensitive Ca(2+)‐activated K+ channels regulate pacemaker activity in nigral dopamine neurons. Neuroreport 1996;7:809–814. [DOI] [PubMed] [Google Scholar]
  • 87. Kerr JF, Wyllie AH, Currie AR. Apoptosis: A basic biological phenomenon with wide‐ranging implications in tissue kinetics. Br J Cancer 1972;26:239–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Sartorius U, Schmitz I, Krammer PH. Molecular mechanisms of death‐receptor‐mediated apoptosis. Chembiochem 2001;2:20–29. [DOI] [PubMed] [Google Scholar]
  • 89. Hirsch EC, Hunot S. Neuroinflammation in Parkinson's disease: A target for neuroprotection? Lancet Neurol 2009;8:382–397. [DOI] [PubMed] [Google Scholar]
  • 90. Ferger B, Leng A, Mura A, Hengerer B, Feldon J. Genetic ablation of tumor necrosis factor‐alpha (TNF‐alpha) and pharmacological inhibition of TNF‐synthesis attenuates MPTP toxicity in mouse striatum. J Neurochem 2004;89:822–833. [DOI] [PubMed] [Google Scholar]
  • 91. Leng A, Mura A, Feldon J, Ferger B. Tumor necrosis factor‐alpha receptor ablation in a chronic MPTP mouse model of Parkinson's disease. Neurosci Lett 2005;375:107–111. [DOI] [PubMed] [Google Scholar]
  • 92. Rousselet E, Callebert J, Parain K, et al Role of TNF‐alpha receptors in mice intoxicated with the parkinsonian toxin MPTP. Exp Neurol 2002;177:183–192. [DOI] [PubMed] [Google Scholar]
  • 93. Sriram K, Matheson JM, Benkovic SA, Miller DB, Luster MI, O’Callaghan JP. Mice deficient in TNF receptors are protected against dopaminergic neurotoxicity: Implications for Parkinson's disease. FASEB J 2002;16:1474–1476. [DOI] [PubMed] [Google Scholar]
  • 94. Sriram K, Matheson JM, Benkovic SA, Miller DB, Luster MI, O’Callaghan JP. Deficiency of TNF receptors suppresses microglial activation and alters the susceptibility of brain regions to MPTP‐induced neurotoxicity: Role of TNF‐alpha. FASEB J 2006;20:670–682. [DOI] [PubMed] [Google Scholar]
  • 95. Ferrer I, Blanco R, Cutillas B, Ambrosio S. Fas and Fas‐L expression in Huntington's disease and Parkinson's disease. Neuropathol Appl Neurobiol 2000;26:424–433. [DOI] [PubMed] [Google Scholar]
  • 96. Hayley S, Crocker SJ, Smith PD, et al Regulation of dopaminergic loss by Fas in a 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydro‐pyridine model of Parkinson's disease. J Neurosci 2004;24:2045–2053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Landau AM, Luk KC, Jones ML, et al Defective Fas expression exacerbates neurotoxicity in a model of Parkinson's disease. J Exp Med 2005;202:575–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Gomez C, Reiriz J, Pique M, Gil J, Ferrer I, Ambrosio S. Low concentrations of 1‐methyl‐4‐phenylpyridinium ion induce caspase‐mediated apoptosis in human SH‐SY5Y neuroblastoma cells. J Neurosci Res 2001;63:421–428. [DOI] [PubMed] [Google Scholar]
  • 99. Hunot S, Vila M, Teismann P, et al JNK‐mediated induction of cyclooxygenase 2 is required for neurodegeneration in a mouse model of Parkinson's disease. Proc Natl Acad Sci USA 2004;101:665–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Saporito MS, Brown EM, Miller MS, Carswell S. CEP‐1347/KT‐7515, an inhibitor of c‐jun N‐terminal kinase activation, attenuates the 1‐methyl‐4‐phenyl tetrahydropyridine‐mediated loss of nigrostriatal dopaminergic neurons In vivo. J Pharmacol Exp Ther 1999;288:421–427. [PubMed] [Google Scholar]
  • 101. Xia XG, Harding T, Weller M, Bieneman A, Uney JB, Schulz JB. Gene transfer of the JNK interacting protein‐1 protects dopaminergic neurons in the MPTP model of Parkinson's disease. Proc Natl Acad Sci USA 2001;98:10433–10438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Baltriukiene D, Kalvelyte A, Bukelskiene V. Induction of apoptosis and activation of JNK and p38 MAPK pathways in deoxynivalenol‐treated cell lines. Altern Lab Anim 2007;35:53–59. [DOI] [PubMed] [Google Scholar]
  • 103. Investigators TPSGP . Mixed lineage kinase inhibitor CEP‐1347 fails to delay disability in early Parkinson disease. Neurology 2007;69:1480–1490. [DOI] [PubMed] [Google Scholar]
  • 104. Degli Esposti M, Dive C. Mitochondrial membrane permeabilisation by Bax/Bak. Biochem Biophys Res Commun 2003;304:455–461. [DOI] [PubMed] [Google Scholar]
  • 105. Caroppi P, Sinibaldi F, Fiorucci L, Santucci R. Apoptosis and human diseases: Mitochondrion damage and lethal role of released cytochrome C as proapoptotic protein. Curr Med Chem 2009;16:4058–4065. [DOI] [PubMed] [Google Scholar]
  • 106. Er E, Oliver L, Cartron PF, Juin P, Manon S, Vallette FM. Mitochondria as the target of the pro‐apoptotic protein Bax. Biochim Biophys Acta 2006;1757:1301–1311. [DOI] [PubMed] [Google Scholar]
  • 107. Newman EJ, Grosset KA, Grosset DG. Geographical difference in Parkinson's disease prevalence within West Scotland. Mov Disord 2009;24:401–406. [DOI] [PubMed] [Google Scholar]
  • 108. Zorzon M, Capus L, Pellegrino A, Cazzato G, Zivadinov R. Familial and environmental risk factors in Parkinson's disease: A case‐control study in north‐east Italy. Acta Neurol Scand 2002;105:77–82. [DOI] [PubMed] [Google Scholar]
  • 109. Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Richardson RJ. The risk of Parkinson's disease with exposure to pesticides, farming, well water, and rural living. Neurology 1998;50:1346–1350. [DOI] [PubMed] [Google Scholar]
  • 110. Gorell JM, Peterson EL, Rybicki BA, Johnson CC. Multiple risk factors for Parkinson's disease. J Neurol Sci 2004;217:169–174. [DOI] [PubMed] [Google Scholar]
  • 111. Elbaz A, Moisan F. Update in the epidemiology of Parkinson's disease. Curr Opin Neurol 2008;21:454–460. [DOI] [PubMed] [Google Scholar]
  • 112. Elbaz A, Tranchant C. Epidemiologic studies of environmental exposures in Parkinson's disease. J Neurol Sci 2007;262:37–44. [DOI] [PubMed] [Google Scholar]
  • 113. Polymeropoulos MH, Lavedan C, Leroy E, et al Mutation in the alpha‐synuclein gene identified in families with Parkinson's disease. Science 1997;276:2045–2047. [DOI] [PubMed] [Google Scholar]
  • 114. Windisch M, Wolf H, Hutter‐Paier B, Wronski R. The role of alpha‐synuclein in neurodegenerative diseases: A potential target for new treatment strategies? Neurodegener Dis 2008;5:218–221. [DOI] [PubMed] [Google Scholar]
  • 115. Kruger R, Kuhn W, Muller T, et al Ala30Pro mutation in the gene encoding alpha‐synuclein in Parkinson's disease. Nat Genet 1998;18:106–108. [DOI] [PubMed] [Google Scholar]
  • 116. Zarranz JJ, Alegre J, Gomez‐Esteban JC, et al The new mutation, E46K, of alpha‐synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004;55:164–173. [DOI] [PubMed] [Google Scholar]
  • 117. Chartier‐Harlin MC, Kachergus J, Roumier C, et al Alpha‐synuclein locus duplication as a cause of familial Parkinson's disease. Lancet 2004;364:1167–1169. [DOI] [PubMed] [Google Scholar]
  • 118. Singleton AB, Farrer M, Johnson J, et al alpha‐Synuclein locus triplication causes Parkinson's disease. Science 2003;302:841. [DOI] [PubMed] [Google Scholar]
  • 119. Bonifati V, Dekker MC, Vanacore N, et al Autosomal recessive early onset parkinsonism is linked to three loci: PARK2, PARK6, and PARK7. Neurol Sci 2002;23(Suppl 2):S59–S60. [DOI] [PubMed] [Google Scholar]
  • 120. Darios F, Corti O, Lucking CB, et al Parkin prevents mitochondrial swelling and cytochrome c release in mitochondria‐dependent cell death. Hum Mol Genet 2003;12:517–26. [DOI] [PubMed] [Google Scholar]
  • 121. Kitada T, Asakawa S, Hattori N, et al Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998;392:605–608. [DOI] [PubMed] [Google Scholar]
  • 122. Sun M, Latourelle JC, Wooten GF, et al Influence of heterozygosity for parkin mutation on onset age in familial Parkinson disease: The GenePD study. Arch Neurol 2006;63:826–832. [DOI] [PubMed] [Google Scholar]
  • 123. Foroud T, Uniacke SK, Liu L, et al Heterozygosity for a mutation in the parkin gene leads to later onset Parkinson disease. Neurology 2003;60:796–801. [DOI] [PubMed] [Google Scholar]
  • 124. Lesage S, Lohmann E, Tison F, Durif F, Durr A, Brice A. Rare heterozygous parkin variants in French early‐onset Parkinson disease patients and controls. J Med Genet 2008;45:43–46. [DOI] [PubMed] [Google Scholar]
  • 125. Leroy E, Boyer R, Auburger G, et al The ubiquitin pathway in Parkinson's disease. Nature 1998;395:451–452. [DOI] [PubMed] [Google Scholar]
  • 126. Momose Y, Murata M, Kobayashi K, et al Association studies of multiple candidate genes for Parkinson's disease using single nucleotide polymorphisms. Ann Neurol 2002;51:133–136. [DOI] [PubMed] [Google Scholar]
  • 127. Carmine Belin A, Westerlund M, Bergman O, et al S18Y in ubiquitin carboxy‐terminal hydrolase L1 (UCH‐L1) associated with decreased risk of Parkinson's disease in Sweden. Parkinsonism Relat Disord 2007;13:295–298. [DOI] [PubMed] [Google Scholar]
  • 128. Tan EK, Puong KY, Fook‐Chong S, et al Case‐control study of UCHL1 S18Y variant in Parkinson's disease. Mov Disord 2006;21:1765–1768. [DOI] [PubMed] [Google Scholar]
  • 129. Larsen CN, Price JS, Wilkinson KD. Substrate binding and catalysis by ubiquitin C‐terminal hydrolases: Identification of two active site residues. Biochemistry 1996;35:6735–6744. [DOI] [PubMed] [Google Scholar]
  • 130. Valente EM, Abou‐Sleiman PM, Caputo V, et al Hereditary early‐onset Parkinson's disease caused by mutations in PINK1. Science 2004;304:1158–1160. [DOI] [PubMed] [Google Scholar]
  • 131. Silvestri L, Caputo V, Bellacchio E, et al Mitochondrial import and enzymatic activity of PINK1 mutants associated to recessive parkinsonism. Hum Mol Genet 2005;14:3477–3492. [DOI] [PubMed] [Google Scholar]
  • 132. Beilina A, Van Der Brug M, Ahmad R, et al 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] [PMC free article] [PubMed] [Google Scholar]
  • 133. Valente EM, Bentivoglio AR, Dixon PH, et al Localization of a novel locus for autosomal recessive early‐onset parkinsonism, PARK6, on human chromosome 1p35‐p36. Am J Hum Genet 2001;68:895–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Valente EM, Brancati F, Ferraris A, et al PARK6‐linked parkinsonism occurs in several European families. Ann Neurol 2002;51:14–18. [PubMed] [Google Scholar]
  • 135. Shendelman S, Jonason A, Martinat C, Leete T, Abeliovich A. DJ‐1 is a redox‐dependent molecular chaperone that inhibits alpha‐synuclein aggregate formation. PLoS Biol 2004;2:1764–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Bonifati V, Rizzu P, van Baren MJ, et al Mutations in the DJ‐1 gene associated with autosomal recessive early‐onset parkinsonism. Science 2003;299:256–259. [DOI] [PubMed] [Google Scholar]
  • 137. Mata IF, Wedemeyer WJ, Farrer MJ, Taylor JP, Gallo KA. LRRK2 in Parkinson's disease: Protein domains and functional insights. Trends Neurosci 2006;29:286–293. [DOI] [PubMed] [Google Scholar]
  • 138. West AB, Moore DJ, Biskup S, et al Parkinson's disease‐associated mutations in leucine‐rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci USA 2005;102:16842–16847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. West AB, Moore DJ, Choi C, et al Parkinson's disease‐associated mutations in LRRK2 link enhanced GTP‐binding and kinase activities to neuronal toxicity. Hum Mol Genet 2007;16:223–232. [DOI] [PubMed] [Google Scholar]
  • 140. Funayama M, Hasegawa K, Kowa H, Saito M, Tsuji S, Obata F. A new locus for Parkinson's disease (PARK8) maps to chromosome 12p11.2‐q13.1. Ann Neurol 2002;51:296–301. [DOI] [PubMed] [Google Scholar]
  • 141. Paisan‐Ruiz C, Jain S, Evans EW, et al Cloning of the gene containing mutations that cause PARK8‐linked Parkinson's disease. Neuron 2004;44:595–600. [DOI] [PubMed] [Google Scholar]
  • 142. Ozelius LJ, Senthil G, Saunders‐Pullman R, et al LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews. N Engl J Med 2006;354:424–425. [DOI] [PubMed] [Google Scholar]
  • 143. Albani D, Peverelli E, Rametta R, et al Protective effect of TAT‐delivered alpha‐synuclein: Relevance of the C‐terminal domain and involvement of HSP70. FASEB J 2004;18:1713–1715. [DOI] [PubMed] [Google Scholar]
  • 144. Bodles AM, El‐Agnaf OM, Greer B, Guthrie DJ, Irvine GB. Inhibition of fibril formation and toxicity of a fragment of alpha‐synuclein by an N‐methylated peptide analogue. Neurosci Lett 2004;359:89–93. [DOI] [PubMed] [Google Scholar]
  • 145. El‐Agnaf OM, Paleologou KE, Greer B, et al A strategy for designing inhibitors of alpha‐synuclein aggregation and toxicity as a novel treatment for Parkinson's disease and related disorders. FASEB J 2004;18:1315–1317. [DOI] [PubMed] [Google Scholar]
  • 146. Paleologou KE, Irvine GB, El‐Agnaf OM. Alpha‐synuclein aggregation in neurodegenerative diseases and its inhibition as a potential therapeutic strategy. Biochem Soc Trans 2005;33(Pt 5S):1106–1110. [DOI] [PubMed] [Google Scholar]
  • 147. Emadi S, Liu R, Yuan B, et al Inhibiting aggregation of alpha‐synuclein with human single chain antibody fragments. Biochemistry 2004;43:2871–2878. [DOI] [PubMed] [Google Scholar]
  • 148. Masliah E, Rockenstein E, Adame A, et al Effects of alpha‐synuclein immunization in a mouse model of Parkinson's disease. Neuron 2005;46:857–868. [DOI] [PubMed] [Google Scholar]
  • 149. Bar‐On P, Crews L, Koob AO, et al Statins reduce neuronal alpha‐synuclein aggregation in in vitro models of Parkinson's disease. J Neurochem 2008;105:1656–1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Bar‐On P, Rockenstein E, Adame A, Ho G, Hashimoto M, Masliah E. Effects of the cholesterol‐lowering compound methyl‐beta‐cyclodextrin in models of alpha‐synucleinopathy. J Neurochem 2006;98:1032–1045. [DOI] [PubMed] [Google Scholar]
  • 151. Imai Y, Soda M, Inoue H, Hattori N, Mizuno Y, Takahashi R. An unfolded putative transmembrane polypeptide, which can lead to endoplasmic reticulum stress, is a substrate of Parkin. Cell 2001;105:891–902. [DOI] [PubMed] [Google Scholar]
  • 152. Zhang Y, Gao J, Chung KK, Huang H, Dawson VL, Dawson TM. Parkin functions as an E2‐dependent ubiquitin‐protein ligase and promotes the degradation of the synaptic vesicle‐associated protein, CDCrel‐1. Proc Natl Acad Sci USA 2000;97:13354–13359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Chung KK, Zhang Y, Lim KL, et al Parkin ubiquitinates the alpha‐synuclein‐interacting protein, synphilin‐1: Implications for Lewy‐body formation in Parkinson disease. Nat Med 2001;7:1144–1150. [DOI] [PubMed] [Google Scholar]
  • 154. Shimura H, Schlossmacher MG, Hattori N, et al Ubiquitination of a new form of alpha‐synuclein by parkin from human brain: Implications for Parkinson's disease. Science 2001;293:263–269. [DOI] [PubMed] [Google Scholar]
  • 155. Mochizuki H. Parkin gene therapy. Parkinsonism Relat Disord 2009;15 (Suppl 1):S43–45. [DOI] [PubMed] [Google Scholar]
  • 156. Lo Bianco C, Schneider BL, Bauer M, et al 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] [PMC free article] [PubMed] [Google Scholar]
  • 157. Zhang ZJ, Burgunder JM, An XK, et al Lack of evidence for association of a UCH‐L1 S18Y polymorphism with Parkinson's disease in a Han‐Chinese population. Neurosci Lett 2008;442:200–202. [DOI] [PubMed] [Google Scholar]
  • 158. Lansbury PT Jr., Brice A. Genetics of Parkinson's disease and biochemical studies of implicated gene products. Curr Opin Cell Biol 2002;14:653–660. [DOI] [PubMed] [Google Scholar]
  • 159. Gautier CA, Kitada T, Shen J. Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc Natl Acad Sci USA 2008;105:11364–11369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Pridgeon JW, Olzmann JA, Chin LS, Li L. PINK1 protects against oxidative stress by phosphorylating mitochondrial chaperone TRAP1. PLoS Biol 2007;5:1494–1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Petit A, Kawarai T, Paitel E, et al Wild‐type PINK1 prevents basal and induced neuronal apoptosis, a protective effect abrogated by Parkinson disease‐related mutations. J Biol Chem 2005;280:34025–34032. [DOI] [PubMed] [Google Scholar]
  • 162. Blackinton J, Lakshminarasimhan M, Thomas KJ, et al Formation of a stabilized cysteine sulfinic acid is critical for the mitochondrial function of the parkinsonism protein DJ‐1. J Biol Chem 2009;284:6476–6485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Zucchelli S, Vilotti S, Calligaris R, et al Aggresome‐forming TTRAP mediates pro‐apoptotic properties of Parkinson's disease‐associated DJ‐1 missense mutations. Cell Death Differ 2009;16:428–438. [DOI] [PubMed] [Google Scholar]
  • 164. Paterna JC, Leng A, Weber E, Feldon J, Bueler H. DJ‐1 and Parkin modulate dopamine‐dependent behavior and inhibit MPTP‐induced nigral dopamine neuron loss in mice. Mol Ther 2007;15:698–704. [DOI] [PubMed] [Google Scholar]
  • 165. Inden M, Taira T, Kitamura Y, et al PARK7 DJ‐1 protects against degeneration of nigral dopaminergic neurons in Parkinson's disease rat model. Neurobiol Dis 2006;24:144–158. [DOI] [PubMed] [Google Scholar]
  • 166. Miyazaki S, Yanagida T, Nunome K, et al DJ‐1‐binding compounds prevent oxidative stress‐induced cell death and movement defect in Parkinson's disease model rats. J Neurochem 2008;105:2418–2434. [DOI] [PubMed] [Google Scholar]
  • 167. Greggio E, Jain S, Kingsbury A, et al Kinase activity is required for the toxic effects of mutant LRRK2/dardarin. Neurobiol Dis 2006;23:329–341. [DOI] [PubMed] [Google Scholar]
  • 168. Smith WW, Pei Z, Jiang H, Dawson VL, Dawson TM, Ross CA. Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nat Neurosci 2006;9:1231–1233. [DOI] [PubMed] [Google Scholar]
  • 169. Gloeckner CJ, Kinkl N, Schumacher A, et al The Parkinson disease causing LRRK2 mutation I2020T is associated with increased kinase activity. Hum Mol Genet 2006;15:223–232. [DOI] [PubMed] [Google Scholar]
  • 170. Iaccarino C, Crosio C, Vitale C, Sanna G, Carri MT, Barone P. Apoptotic mechanisms in mutant LRRK2‐mediated cell death. Hum Mol Genet 2007;16:1319–1326. [DOI] [PubMed] [Google Scholar]
  • 171. Dachsel JC, Taylor JP, Mok SS, et al Identification of potential protein interactors of Lrrk2. Parkinsonism Relat Disord 2007;13:382–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Wang L, Xie C, Greggio E, et al The chaperone activity of heat shock protein 90 is critical for maintaining the stability of leucine‐rich repeat kinase 2. J Neurosci 2008;28:3384–3391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Tanner CM, Ottman R, Goldman SM, et al Parkinson disease in twins: An etiologic study. JAMA 1999;281:341–346. [DOI] [PubMed] [Google Scholar]
  • 174. Hoehn MM. The natural history of Parkinson's disease in the pre‐levodopa and post‐levodopa eras. Neurol Clin 1992;10:331–339. [PubMed] [Google Scholar]
  • 175. Myllyla V, Haapaniemi T, Kaakkola S, et al Patient satisfaction with switching to Stalevo: An open‐label evaluation in PD patients experiencing wearing‐off (Simcom Study). Acta Neurol Scand 2006;114:181–186. [DOI] [PubMed] [Google Scholar]
  • 176. Klivenyi P, Vecsei L. Long term experience with Stalevo in Szeged, Hungary. Ideggyogy Sz 2009;62:178–180. [PubMed] [Google Scholar]
  • 177. Klivenyi P, Vecsei L. The use of Stalevo in Hungary for patients with Parkinson disease and its effect on the quality of life. Ideggyogy Sz 2008;61:42–48. [PubMed] [Google Scholar]
  • 178. Lyons KE, Pahwa R. Conversion from sustained release carbidopa/levodopa to carbidopa/levodopa/entacapone (stalevo) in Parkinson disease patients. Clin Neuropharmacol 2006;29:73–76. [DOI] [PubMed] [Google Scholar]
  • 179. Huot P, Parent A. Dopaminergic neurons intrinsic to the striatum. J Neurochem 2007;101:1441–1447. [DOI] [PubMed] [Google Scholar]
  • 180. Kitahama K, Geffard M, Araneda S, et al Localization of L‐DOPA uptake and decarboxylating neuronal structures in the cat brain using dopamine immunohistochemistry. Brain Res 2007;1167:56–70. [DOI] [PubMed] [Google Scholar]
  • 181. Yamada H, Aimi Y, Nagatsu I, Taki K, Kudo M, Arai R. Immunohistochemical detection of L‐DOPA‐derived dopamine within serotonergic fibers in the striatum and the substantia nigra pars reticulata in Parkinsonian model rats. Neurosci Res 2007;59:1–7. [DOI] [PubMed] [Google Scholar]
  • 182. Caudle WM, Richardson JR, Wang MZ, et al Reduced vesicular storage of dopamine causes progressive nigrostriatal neurodegeneration. J Neurosci 2007;27:8138–8148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Glatt CE, Wahner AD, White DJ, Ruiz‐Linares A, Ritz B. Gain‐of‐function haplotypes in the vesicular monoamine transporter promoter are protective for Parkinson disease in women. Hum Mol Genet 2006;15:299–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Rabinovic AD, Lewis DA, Hastings TG. Role of oxidative changes in the degeneration of dopamine terminals after injection of neurotoxic levels of dopamine. Neuroscience 2000;101:67–76. [DOI] [PubMed] [Google Scholar]
  • 185. Fahn S. Does levodopa slow or hasten the rate of progression of Parkinson's disease? J Neurol 2005;252(Suppl 4):IV37–IV42. [DOI] [PubMed] [Google Scholar]
  • 186. Muralikrishnan D, Mohanakumar KP. Neuroprotection by bromocriptine against 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine‐induced neurotoxicity in mice. FASEB J 1998;12:905–912. [DOI] [PubMed] [Google Scholar]
  • 187. 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] [PubMed] [Google Scholar]
  • 188. Ogawa N, Tanaka K, Asanuma M, et al Bromocriptine protects mice against 6‐hydroxydopamine and scavenges hydroxyl free radicals in vitro. Brain Res 1994;657:207–213. [DOI] [PubMed] [Google Scholar]
  • 189. Finotti N, Castagna L, Moretti A, Marzatico F. Reduction of lipid peroxidation in different rat brain areas after cabergoline treatment. Pharmacol Res 2000;42:287–291. [DOI] [PubMed] [Google Scholar]
  • 190. Yoshioka M, Tanaka K, Miyazaki I, et al The dopamine agonist cabergoline provides neuroprotection by activation of the glutathione system and scavenging free radicals. Neurosci Res 2002;43:259–267. [DOI] [PubMed] [Google Scholar]
  • 191. Nishibayashi S, Asanuma M, Kohno M, Gomez‐Vargas M, Ogawa N. Scavenging effects of dopamine agonists on nitric oxide radicals. J Neurochem 1996;67:2208–2211. [DOI] [PubMed] [Google Scholar]
  • 192. 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] [PubMed] [Google Scholar]
  • 193. Sawada H, Ibi M, Kihara T, et al Dopamine D2‐type agonists protect mesencephalic neurons from glutamate neurotoxicity: Mechanisms of neuroprotective treatment against oxidative stress. Ann Neurol 1998;44:110–119. [DOI] [PubMed] [Google Scholar]
  • 194. Iida M, Miyazaki I, Tanaka K, Kabuto H, Iwata‐Ichikawa E, Ogawa N. Dopamine D2 receptor‐mediated antioxidant and neuroprotective effects of ropinirole, a dopamine agonist. Brain Res 1999;838:51–59. [DOI] [PubMed] [Google Scholar]
  • 195. Hartmann A, Hunot S, Michel PP, et al Caspase‐3: A vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson's disease. Proc Natl Acad Sci USA 2000;97:2875–2880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Hashimoto M, Takeda A, Hsu LJ, Takenouchi T, Masliah E. Role of cytochrome c as a stimulator of alpha‐synuclein aggregation in Lewy body disease. J Biol Chem 1999;274:28849–28852. [DOI] [PubMed] [Google Scholar]
  • 197. Kitamura Y, Kohno Y, Nakazawa M, Nomura Y. Inhibitory effects of talipexole and pramipexole on MPTP‐induced dopamine reduction in the striatum of C57BL/6N mice. Jpn J Pharmacol 1997;74:51–57. [DOI] [PubMed] [Google Scholar]
  • 198. Vu TQ, Ling ZD, Ma SY, et al Pramipexole attenuates the dopaminergic cell loss induced by intraventricular 6‐hydroxydopamine. J Neural Transm 2000;107:159–176. [DOI] [PubMed] [Google Scholar]
  • 199. Kitamura Y, Kakimura J, Taniguchi T. Protective effect of talipexole on MPTP‐treated planarian, a unique parkinsonian worm model. Jpn J Pharmacol 1998;78:23–29. [DOI] [PubMed] [Google Scholar]
  • 200. Kitamura Y, Kosaka T, Kakimura JI, et al Protective effects of the antiparkinsonian drugs talipexole and pramipexole against 1‐methyl‐4‐phenylpyridinium‐induced apoptotic death in human neuroblastoma SH‐SY5Y cells. Mol Pharmacol 1998;54:1046–1054. [PubMed] [Google Scholar]
  • 201. Iravani MM, Haddon CO, Cooper JM, Jenner P, Schapira AH. Pramipexole protects against MPTP toxicity in non‐human primates. J Neurochem 2006;96:1315–1321. [DOI] [PubMed] [Google Scholar]
  • 202. Kakimura J, Kitamura Y, Takata K, Kohno Y, Nomura Y, Taniguchi T. Release and aggregation of cytochrome c and alpha‐synuclein are inhibited by the antiparkinsonian drugs, talipexole and pramipexole. Eur J Pharmacol 2001;417:59–67. [DOI] [PubMed] [Google Scholar]
  • 203. Gu M, Iravani MM, Cooper JM, King D, Jenner P, Schapira AH. Pramipexole protects against apoptotic cell death by non‐dopaminergic mechanisms. J Neurochem 2004;91:1075–1081. [DOI] [PubMed] [Google Scholar]
  • 204. Izumi Y, Sawada H, Yamamoto N, et al Novel neuroprotective mechanisms of pramipexole, an anti‐Parkinson drug, against endogenous dopamine‐mediated excitotoxicity. Eur J Pharmacol 2007;557:132–140. [DOI] [PubMed] [Google Scholar]
  • 205. Nair VD, Olanow CW, Sealfon SC. Activation of phosphoinositide 3‐kinase by D2 receptor prevents apoptosis in dopaminergic cell lines. Biochem J 2003;373(Pt 1):25–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Du F, Li R, Huang Y, Li X, Le W. Dopamine D3 receptor‐preferring agonists induce neurotrophic effects on mesencephalic dopamine neurons. Eur J Neurosci 2005;22:2422–2430. [DOI] [PubMed] [Google Scholar]
  • 207. Spina MB, Squinto SP, Miller J, Lindsay RM, Hyman C. Brain‐derived neurotrophic factor protects dopamine neurons against 6‐hydroxydopamine and N‐methyl‐4‐phenylpyridinium ion toxicity: Involvement of the glutathione system. J Neurochem 1992;59:99–106. [DOI] [PubMed] [Google Scholar]
  • 208. Howells DW, Porritt MJ, Wong JY, et al Reduced BDNF mRNA expression in the Parkinson's disease substantia nigra. Exp Neurol 2000;166:127–135. [DOI] [PubMed] [Google Scholar]
  • 209. Kohno R, Sawada H, Kawamoto Y, Uemura K, Shibasaki H, Shimohama S. BDNF is induced by wild‐type alpha‐synuclein but not by the two mutants, A30P or A53T, in glioma cell line. Biochem Biophys Res Commun 2004;318:113–118. [DOI] [PubMed] [Google Scholar]
  • 210. Porritt MJ, Batchelor PE, Howells DW. Inhibiting BDNF expression by antisense oligonucleotide infusion causes loss of nigral dopaminergic neurons. Exp Neurol 2005;192:226–234. [DOI] [PubMed] [Google Scholar]
  • 211. Karamohamed S, Latourelle JC, Racette BA, et al BDNF genetic variants are associated with onset age of familial Parkinson disease: GenePD Study. Neurology 2005;65:1823–1825. [DOI] [PubMed] [Google Scholar]
  • 212. Toda T, Momose Y, Murata M, et al Toward identification of susceptibility genes for sporadic Parkinson's disease. J Neurol 2003;250(Suppl 3):III40–III43. [DOI] [PubMed] [Google Scholar]
  • 213. Hong CJ, Liu HC, Liu TY, Lin CH, Cheng CY, Tsai SJ. Brain‐derived neurotrophic factor (BDNF) Val66Met polymorphisms in Parkinson's disease and age of onset. Neurosci Lett 2003;353:75–77. [DOI] [PubMed] [Google Scholar]
  • 214. Hakansson A, Melke J, Westberg L, et al Lack of association between the BDNF Val66Met polymorphism and Parkinson's disease in a Swedish population. Ann Neurol 2003;53:823. [DOI] [PubMed] [Google Scholar]
  • 215. Group PS . Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002;287:1653–1661. [DOI] [PubMed] [Google Scholar]
  • 216. Whone AL, Watts RL, Stoessl AJ, et al Slower progression of Parkinson's disease with ropinirole versus levodopa: The REAL‐PET study. Ann Neurol 2003;54:93–101. [DOI] [PubMed] [Google Scholar]
  • 217. Dostert PL, Strolin Benedetti M, Tipton KF. Interactions of monoamine oxidase with substrates and inhibitors. Med Res Rev 1989;9:45–89. [DOI] [PubMed] [Google Scholar]
  • 218. Magyar K, Vizi ES, Ecseri Z, Knoll J. Comparative pharmacological analysis of the optical isomers of phenyl‐isopropyl‐methyl‐propinylamine (E‐250). Acta Physiol Acad Sci Hung 1967;32:377–387. [PubMed] [Google Scholar]
  • 219. Knoll J, Ecseri Z, Kelemen K, Nievel J, Knoll B. Phenylisopropylmethylpropinylamine (E‐250), a new spectrum psychic energizer. Arch Int Pharmacodyn Ther 1965;155:154–164. [PubMed] [Google Scholar]
  • 220. Finberg JP, Youdim MB. Pharmacological properties of the anti‐Parkinson drug rasagiline; modification of endogenous brain amines, reserpine reversal, serotonergic and dopaminergic behaviours. Neuropharmacology 2002;43:1110–1118. [DOI] [PubMed] [Google Scholar]
  • 221. Bar Am O, Amit T, Youdim MB. Contrasting neuroprotective and neurotoxic actions of respective metabolites of anti‐Parkinson drugs rasagiline and selegiline. Neurosci Lett 2004;355:169–172. [DOI] [PubMed] [Google Scholar]
  • 222. Kupsch A, Sautter J, Gotz ME, et al Monoamine oxidase‐inhibition and MPTP‐induced neurotoxicity in the non‐human primate: Comparison of rasagiline (TVP 1012) with selegiline. J Neural Transm 2001;108:985–1009. [DOI] [PubMed] [Google Scholar]
  • 223. Muralikrishnan D, Samantaray S, Mohanakumar KP. D‐deprenyl protects nigrostriatal neurons against 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine‐induced dopaminergic neurotoxicity. Synapse 2003;50:7–13. [DOI] [PubMed] [Google Scholar]
  • 224. Chiba K, Trevor A, Castagnoli N Jr. Metabolism of the neurotoxic tertiary amine, MPTP, by brain monoamine oxidase. Biochem Biophys Res Commun 1984;120:574–578. [DOI] [PubMed] [Google Scholar]
  • 225. Maruyama W, Youdim MB, Naoi M. Antiapoptotic properties of rasagiline, N‐propargylamine‐1(R)‐aminoindan, and its optical (S)‐isomer, TV1022. Ann N Y Acad Sci 2001;939:320–329. [DOI] [PubMed] [Google Scholar]
  • 226. Yogev‐Falach M, Amit T, Bar‐Am O, Youdim MB. The importance of propargylamine moiety in the anti‐Parkinson drug rasagiline and its derivatives in MAPK‐dependent amyloid precursor protein processing. FASEB J 2003;17:2325–2327. [DOI] [PubMed] [Google Scholar]
  • 227. Akao Y, Maruyama W, Yi H, Shamoto‐Nagai M, Youdim MB, Naoi M. An anti‐Parkinson's disease drug, N‐propargyl‐1(R)‐aminoindan (rasagiline), enhances expression of anti‐apoptotic bcl‐2 in human dopaminergic SH‐SY5Y cells. Neurosci Lett 2002;326:105–108. [DOI] [PubMed] [Google Scholar]
  • 228. Akao Y, Maruyama W, Shimizu S, et al Mitochondrial permeability transition mediates apoptosis induced by N‐methyl(R)salsolinol, an endogenous neurotoxin, and is inhibited by Bcl‐2 and rasagiline, N‐propargyl‐1(R)‐aminoindan. J Neurochem 2002;82:913–923. [DOI] [PubMed] [Google Scholar]
  • 229. Sagi Y, Mandel S, Amit T, Youdim MB. Activation of tyrosine kinase receptor signaling pathway by rasagiline facilitates neurorescue and restoration of nigrostriatal dopamine neurons in post‐MPTP‐induced parkinsonism. Neurobiol Dis 2007;25:35–44. [DOI] [PubMed] [Google Scholar]
  • 230. Weinreb O, Bar‐Am O, Amit T, Chillag‐Talmor O, Youdim MB. Neuroprotection via pro‐survival protein kinase C isoforms associated with Bcl‐2 family members. FASEB J 2004;18:1471–1473. [DOI] [PubMed] [Google Scholar]
  • 231. Weinreb O, Amit T, Bar‐Am O, Youdim MB. Induction of neurotrophic factors GDNF and BDNF associated with the mechanism of neurorescue action of rasagiline and ladostigil: New insights and implications for therapy. Ann N Y Acad Sci 2007;1122:155–168. [DOI] [PubMed] [Google Scholar]
  • 232. Bar‐Am O, Amit T, Youdim MB. Aminoindan and hydroxyaminoindan, metabolites of rasagiline and ladostigil, respectively, exert neuroprotective properties in vitro. J Neurochem 2007;103:500–508. [DOI] [PubMed] [Google Scholar]
  • 233. Tazik S, Johnson S, Lu D, et al Comparative neuroprotective effects of rasagiline and aminoindan with selegiline on dexamethasone‐induced brain cell apoptosis. Neurotox Res 2009;15:284–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Youdim MB, Fridkin M, Zheng H. Novel bifunctional drugs targeting monoamine oxidase inhibition and iron chelation as an approach to neuroprotection in Parkinson's disease and other neurodegenerative diseases. J Neural Transm 2004;111:1455–1471. [DOI] [PubMed] [Google Scholar]
  • 235. Gal S, Zheng H, Fridkin M, Youdim MB. Restoration of nigrostriatal dopamine neurons in post‐MPTP treatment by the novel multifunctional brain‐permeable iron chelator‐monoamine oxidase inhibitor drug, M30. Neurotox Res 2010;17:15–27. [DOI] [PubMed] [Google Scholar]
  • 236. 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 2006:447–456. [DOI] [PubMed] [Google Scholar]
  • 237. 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] [PubMed] [Google Scholar]
  • 238. Group TPS . 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] [PubMed] [Google Scholar]
  • 239. Olanow CW, Hauser RA, Gauger L, et al The effect of deprenyl and levodopa on the progression of Parkinson's disease. Ann Neurol 1995;38:771–777. [DOI] [PubMed] [Google Scholar]
  • 240. Rascol O, Olanow CW. ADAGIO: A prospective, double‐blind, delayed‐start study to examine the disease‐modifying effect of rasagiline in early Parkinsons disease (PD). Eur J Neurology 2008;15(Suppl 3):413. [Google Scholar]
  • 241. Olanow CW, Rascol O, Hauser R, et al A double‐blind, delayed‐start trial of rasagiline in Parkinson's disease. N Engl J Med 2009;361:1268–1278. [DOI] [PubMed] [Google Scholar]
  • 242. Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of Parkinson's disease: Systematic review and meta‐analysis of observational studies. Drugs Aging 2009;26:769–779. [DOI] [PubMed] [Google Scholar]
  • 243. Wahner AD, Bronstein JM, Bordelon YM, Ritz B. Nonsteroidal anti‐inflammatory drugs may protect against Parkinson disease. Neurology 2007;69:1836–1842. [DOI] [PubMed] [Google Scholar]
  • 244. Etminan M, Suissa S. NSAID use and the risk of Parkinson's disease. Curr Drug Saf 2006;1:223–225. [DOI] [PubMed] [Google Scholar]
  • 245. Ton TG, Heckbert SR, Longstreth WT Jr., et al Nonsteroidal anti‐inflammatory drugs and risk of Parkinson's disease. Mov Disord 2006;21:964–969. [DOI] [PubMed] [Google Scholar]
  • 246. Asanuma M, Nishibayashi‐Asanuma S, Miyazaki I, Kohno M, Ogawa N. Neuroprotective effects of non‐steroidal anti‐inflammatory drugs by direct scavenging of nitric oxide radicals. J Neurochem 2001;76:1895–1904. [DOI] [PubMed] [Google Scholar]
  • 247. Du Y, Ma Z, Lin S, et al Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson's disease. Proc Natl Acad Sci USA 2001;98:14669–14674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. He Y, Appel S, Le W. Minocycline inhibits microglial activation and protects nigral cells after 6‐hydroxydopamine injection into mouse striatum. Brain Res 2001;909:187–193. [DOI] [PubMed] [Google Scholar]
  • 249. Investigators TNN‐P . A randomized, double‐blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 2006;66:664–671. [DOI] [PubMed] [Google Scholar]
  • 250. Investigators TNN‐P . A pilot clinical trial of creatine and minocycline in early Parkinson disease: 18‐month results. Clin Neuropharmacol 2008;31:141–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Aharoni R, Kayhan B, Eilam R, Sela M, Arnon R. Glatiramer acetate‐specific T cells in the brain express T helper 2/3 cytokines and brain‐derived neurotrophic factor in situ. Proc Natl Acad Sci USA 2003;100:14157–14162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Aharoni R, Teitelbaum D, Sela M, Arnon R. Copolymer 1 induces T cells of the T helper type 2 that crossreact with myelin basic protein and suppress experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA 1997;94:10821–10826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Ziemssen T, Kumpfel T, Schneider H, Klinkert WE, Neuhaus O, Hohlfeld R. Secretion of brain‐derived neurotrophic factor by glatiramer acetate‐reactive T‐helper cell lines: Implications for multiple sclerosis therapy. J Neurol Sci 2005;233:109–112. [DOI] [PubMed] [Google Scholar]
  • 254. Ziemssen T, Kumpfel T, Klinkert WE, Neuhaus O, Hohlfeld R. Glatiramer acetate‐specific T‐helper 1‐ and 2‐type cell lines produce BDNF: Implications for multiple sclerosis therapy. Brain‐derived neurotrophic factor. Brain 2002;125(Pt 11):2381–2391. [DOI] [PubMed] [Google Scholar]
  • 255. Laurie C, Reynolds A, Coskun O, Bowman E, Gendelman HE, Mosley RL. CD4+ T cells from Copolymer‐1 immunized mice protect dopaminergic neurons in the 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine model of Parkinson's disease. J Neuroimmunol 2007;183:60–68. [DOI] [PubMed] [Google Scholar]
  • 256. Benner EJ, Mosley RL, Destache CJ, et al Therapeutic immunization protects dopaminergic neurons in a mouse model of Parkinson's disease. Proc Natl Acad Sci USA 2004;101:9435–9440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Boska MD, Lewis TB, Destache CJ, et al Quantitative 1H magnetic resonance spectroscopic imaging determines therapeutic immunization efficacy in an animal model of Parkinson's disease. J Neurosci 2005;25:1691–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Matthews RT, Ferrante RJ, Klivenyi P, et al Creatine and cyclocreatine attenuate MPTP neurotoxicity. Exp Neurol 1999;157:142–149. [DOI] [PubMed] [Google Scholar]
  • 259. Brewer GJ, Wallimann TW. Protective effect of the energy precursor creatine against toxicity of glutamate and beta‐amyloid in rat hippocampal neurons. J Neurochem 2000;74:1968–1978. [DOI] [PubMed] [Google Scholar]
  • 260. Ferrante RJ, Andreassen OA, Jenkins BG, et al Neuroprotective effects of creatine in a transgenic mouse model of Huntington's disease. J Neurosci 2000;20:4389–4397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. O’Gorman E, Beutner G, Dolder M, Koretsky AP, Brdiczka D, Wallimann T. The role of creatine kinase in inhibition of mitochondrial permeability transition. FEBS Lett 1997;414:253–257. [DOI] [PubMed] [Google Scholar]
  • 262. Dolder M, Walzel B, Speer O, Schlattner U, Wallimann T. Inhibition of the mitochondrial permeability transition by creatine kinase substrates. Requirement for microcompartmentation. J Biol Chem 2003;278:17760–17766. [DOI] [PubMed] [Google Scholar]
  • 263. Klivenyi P, Calingasan NY, Starkov A, et al Neuroprotective mechanisms of creatine occur in the absence of mitochondrial creatine kinase. Neurobiol Dis 2004;15:610–617. [DOI] [PubMed] [Google Scholar]
  • 264. Lawler JM, Barnes WS, Wu G, Song W, Demaree S. Direct antioxidant properties of creatine. Biochem Biophys Res Commun 2002;290:47–52. [DOI] [PubMed] [Google Scholar]
  • 265. Packer L, Weber SU, Rimbach G. Molecular aspects of alpha‐tocotrienol antioxidant action and cell signalling. J Nutr 2001;131:369S–373S. [DOI] [PubMed] [Google Scholar]
  • 266. Adams JD Jr., Odunze IN, Sevanian A. Induction by 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine of lipid peroxidation in vivo in vitamin E deficient mice. Biochem Pharmacol 1990;39:R5–R8. [DOI] [PubMed] [Google Scholar]
  • 267. Dexter DT, Nanayakkara I, Goss‐Sampson MA, et al Nigral dopaminergic cell loss in vitamin E deficient rats. Neuroreport 1994;5:1773–1776. [DOI] [PubMed] [Google Scholar]
  • 268. Lan J, Jiang DH. Desferrioxamine and vitamin E protect against iron and MPTP‐induced neurodegeneration in mice. J Neural Transm 1997;104:469–481. [DOI] [PubMed] [Google Scholar]
  • 269. Perry TL, Yong VW, Clavier RM, et al 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] [PubMed] [Google Scholar]
  • 270. Chi DS, Gong L, Daigneault EA, Kostrzewa RM. Effects of MPTP and vitamin E treatments on immune function in mice. Int J Immunopharmacol 1992;14:739–746. [DOI] [PubMed] [Google Scholar]
  • 271. 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] [PubMed] [Google Scholar]
  • 272. Ren YR, Nishida Y, Yoshimi K, et al Genetic vitamin E deficiency does not affect MPTP susceptibility in the mouse brain. J Neurochem 2006;98:1810–1816. [DOI] [PubMed] [Google Scholar]
  • 273. Fahn S. A pilot trial of high‐dose alpha‐tocopherol and ascorbate in early Parkinson's disease. Ann Neurol 1992;32(Suppl):S128–S132. [DOI] [PubMed] [Google Scholar]
  • 274. Ernster L, Dallner G. Biochemical, physiological and medical aspects of ubiquinone function. Biochim Biophys Acta 1995;1271:195–204. [DOI] [PubMed] [Google Scholar]
  • 275. Lass A, Sohal RS. Electron transport‐linked ubiquinone‐dependent recycling of alpha‐tocopherol inhibits autooxidation of mitochondrial membranes. Arch Biochem Biophys 1998;352:229–236. [DOI] [PubMed] [Google Scholar]
  • 276. Kozlov AV, Gille L, Staniek K, Nohl H. Dihydrolipoic acid maintains ubiquinone in the antioxidant active form by two‐electron reduction of ubiquinone and one‐electron reduction of ubisemiquinone. Arch Biochem Biophys 1999;363:148–154. [DOI] [PubMed] [Google Scholar]
  • 277. Gotz ME, Dirr A, Burger R, et al Effect of lipoic acid on redox state of coenzyme Q in mice treated with 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine and diethyldithiocarbamate. Eur J Pharmacol 1994;266:291–300. [DOI] [PubMed] [Google Scholar]
  • 278. Ostrowski RP. Effect of coenzyme Q(10) on biochemical and morphological changes in experimental ischemia in the rat brain. Brain Res Bull 2000;53:399–407. [DOI] [PubMed] [Google Scholar]
  • 279. Beal MF, Henshaw DR, Jenkins BG, Rosen BR, Schulz JB. Coenzyme Q10 and nicotinamide block striatal lesions produced by the mitochondrial toxin malonate. Ann Neurol 1994;36:882–888. [DOI] [PubMed] [Google Scholar]
  • 280. 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] [PubMed] [Google Scholar]
  • 281. Somayajulu‐Nitu M, Sandhu JK, Cohen J, et al Paraquat induces oxidative stress, neuronal loss in substantia nigra region and Parkinsonism in adult rats: Neuroprotection and amelioration of symptoms by water‐soluble formulation of Coenzyme Q10. BMC Neurosci 2009;10:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Borowy‐Borowski H, Sodja C, Docherty J, Walker PR, Sikorska M. Unique technology for solubilization and delivery of highly lipophilic bioactive molecules. J Drug Target 2004;12:415–424. [DOI] [PubMed] [Google Scholar]
  • 283. Shults CW, Oakes D, Kieburtz K, et al Effects of coenzyme Q10 in early Parkinson disease: Evidence of slowing of the functional decline. Arch Neurol 2002;59:1541–1550. [DOI] [PubMed] [Google Scholar]
  • 284. Investigators TNN‐P . A randomized clinical trial of coenzyme Q10 and GPI‐1485 in early Parkinson disease. Neurology 2007;68:20–28. [DOI] [PubMed] [Google Scholar]
  • 285. Vamos E, Pardutz A, Klivenyi P, Toldi J, Vecsei L. The role of kynurenines in disorders of the central nervous system: Possibilities for neuroprotection. J Neurol Sci 2009;283:21–27. [DOI] [PubMed] [Google Scholar]
  • 286. Sas K, Robotka H, Toldi J, Vecsei L. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with focus on neurodegenerative disorders. J Neurol Sci 2007;257:221–239. [DOI] [PubMed] [Google Scholar]
  • 287. Zadori D, Klivenyi P, Vamos E, Fulop F, Toldi J, Vecsei L. Kynurenines in chronic neurodegenerative disorders: Future therapeutic strategies. J Neural Transm 2009;116:1403–1409. [DOI] [PubMed] [Google Scholar]
  • 288. Kessler M, Terramani T, Lynch G, Baudry M. A glycine site associated with N‐methyl‐D‐aspartic acid receptors: Characterization and identification of a new class of antagonists. J Neurochem 1989;52:1319–1328. [DOI] [PubMed] [Google Scholar]
  • 289. Birch PJ, Grossman CJ, Hayes AG. Kynurenic acid antagonises responses to NMDA via an action at the strychnine‐insensitive glycine receptor. Eur J Pharmacol 1988;154:85–87. [DOI] [PubMed] [Google Scholar]
  • 290. Birch PJ, Grossman CJ, Hayes AG. Kynurenate and FG9041 have both competitive and non‐competitive antagonist actions at excitatory amino acid receptors. Eur J Pharmacol 1988;151:313–315. [DOI] [PubMed] [Google Scholar]
  • 291. Hilmas C, Pereira EF, Alkondon M, Rassoulpour A, Schwarcz R, Albuquerque EX. The brain metabolite kynurenic acid inhibits alpha7 nicotinic receptor activity and increases non‐alpha7 nicotinic receptor expression: Physiopathological implications. J Neurosci 2001;21:7463–7473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Rozsa E, Robotka H, Vecsei L, Toldi J. The Janus‐face kynurenic acid. J Neural Transm 2008;115:1087–1091. [DOI] [PubMed] [Google Scholar]
  • 293. Schwarcz R, Whetsell WO Jr., Mangano RM. Quinolinic acid: An endogenous metabolite that produces axon‐sparing lesions in rat brain. Science 1983;219:316–318. [DOI] [PubMed] [Google Scholar]
  • 294. Stone TW, Perkins MN. Quinolinic acid: A potent endogenous excitant at amino acid receptors in CNS. Eur J Pharmacol 1981;72:411–412. [DOI] [PubMed] [Google Scholar]
  • 295. Tavares RG, Tasca CI, Santos CE, et al Quinolinic acid stimulates synaptosomal glutamate release and inhibits glutamate uptake into astrocytes. Neurochem Int 2002;40:621–627. [DOI] [PubMed] [Google Scholar]
  • 296. Rios C, Santamaria A. Quinolinic acid is a potent lipid peroxidant in rat brain homogenates. Neurochem Res 1991;16:1139–1143. [DOI] [PubMed] [Google Scholar]
  • 297. Eastman CL, Guilarte TR. Cytotoxicity of 3‐hydroxykynurenine in a neuronal hybrid cell line. Brain Res 1989;495:225–231. [DOI] [PubMed] [Google Scholar]
  • 298. Knyihar‐Csillik E, Csillik B, Pakaski M, et al Decreased expression of kynurenine aminotransferase‐I (KAT‐I) in the substantia nigra of mice after 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP) treatment. Neuroscience 2004;126:899–914. [DOI] [PubMed] [Google Scholar]
  • 299. Knyihar‐Csillik E, Chadaide Z, Mihaly A, Krisztin‐Peva B, Fenyo R, Vecsei L. Effect of 6‐hydroxydopamine treatment on kynurenine aminotransferase‐I (KAT‐I) immunoreactivity of neurons and glial cells in the rat substantia nigra. Acta Neuropathol 2006;112:127–137. [DOI] [PubMed] [Google Scholar]
  • 300. Hartai Z, Klivenyi P, Janaky T, Penke B, Dux L, Vecsei L. Kynurenine metabolism in plasma and in red blood cells in Parkinson's disease. J Neurol Sci 2005;239:31–35. [DOI] [PubMed] [Google Scholar]
  • 301. Turski L, Bressler K, Rettig KJ, Loschmann PA, Wachtel H. Protection of substantia nigra from MPP+ neurotoxicity by N‐methyl‐D‐aspartate antagonists. Nature 1991;349:414–418. [DOI] [PubMed] [Google Scholar]
  • 302. Merino M, Vizuete ML, Cano J, Machado A. The non‐NMDA glutamate receptor antagonists 6‐cyano‐7‐nitroquinoxaline‐2,3‐dione and 2,3‐dihydroxy‐6‐nitro‐7‐sulfamoylbenzo(f)quinoxaline, but not NMDA antagonists, block the intrastriatal neurotoxic effect of MPP+. J Neurochem 1999;73:750–757. [DOI] [PubMed] [Google Scholar]
  • 303. Weiss G, Murr C, Zoller H, et al Modulation of neopterin formation and tryptophan degradation by Th1‐ and Th2‐derived cytokines in human monocytic cells. Clin Exp Immunol 1999;116:435–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304. Widner B, Leblhuber F, Fuchs D. Increased neopterin production and tryptophan degradation in advanced Parkinson's disease. J Neural Transm 2002;109:181–189. [DOI] [PubMed] [Google Scholar]
  • 305. Nemeth H, Toldi J, Vecsei L. Kynurenines, Parkinson's disease and other neurodegenerative disorders: Preclinical and clinical studies. J Neural Transm Suppl 2006:285–304. [DOI] [PubMed] [Google Scholar]
  • 306. Miranda AF, Boegman RJ, Beninger RJ, Jhamandas K. Protection against quinolinic acid‐mediated excitotoxicity in nigrostriatal dopaminergic neurons by endogenous kynurenic acid. Neuroscience 1997;78:967–975. [DOI] [PubMed] [Google Scholar]
  • 307. Moroni F, Cozzi A, Carpendo R, Cipriani G, Veneroni O, Izzo E. Kynurenine 3‐mono‐oxygenase inhibitors reduce glutamate concentration in the extracellular spaces of the basal ganglia but not in those of the cortex or hippocampus. Neuropharmacology 2005;48:788–795. [DOI] [PubMed] [Google Scholar]
  • 308. Gregoire L, Rassoulpour A, Guidetti P, et al Prolonged kynurenine 3‐hydroxylase inhibition reduces development of levodopa‐induced dyskinesias in parkinsonian monkeys. Behav Brain Res 2008;186:161–167. [DOI] [PubMed] [Google Scholar]
  • 309. Foster AC, Willis CL, Tridgett R. Protection against N‐methyl‐D‐aspartate receptor‐mediated neuronal degeneration in rat brain by 7‐chlorokynurenate and 3‐amino‐1‐hydroxypyrrolid‐2‐one, antagonists at the allosteric site for glycine. Eur J Neurosci 1990;2:270–277. [DOI] [PubMed] [Google Scholar]
  • 310. Hartley DM, Monyer H, Colamarino SA, Choi DW. 7‐Chlorokynurenate blocks NMDA receptor‐mediated neurotoxicity in murine cortical culture. Eur J Neurosci 1990;2:291–295. [DOI] [PubMed] [Google Scholar]
  • 311. Wu HQ, Salituro FG, Schwarcz R. Enzyme‐catalyzed production of the neuroprotective NMDA receptor antagonist 7‐chlorokynurenic acid in the rat brain in vivo. Eur J Pharmacol 1997;319:13–20. [DOI] [PubMed] [Google Scholar]
  • 312. Wu HQ, Lee SC, Schwarcz R. Systemic administration of 4‐chlorokynurenine prevents quinolinate neurotoxicity in the rat hippocampus. Eur J Pharmacol 2000;390:267–274. [DOI] [PubMed] [Google Scholar]

Articles from CNS Neuroscience & Therapeutics are provided here courtesy of Wiley

RESOURCES