Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 9.
Published in final edited form as: CNS Neurol Disord Drug Targets. 2012 Jun 1;11(4):395–409. doi: 10.2174/187152712800792785

A Synopsis on the Role of Tyrosine Hydroxylase in Parkinson's Disease

Shams Tabrez 1, Nasimudeen R Jabir 1, Shazi Shakil 1, Nigel H Greig 2, Qamre Alam 1, Adel M Abuzenadah 1, Ghazi A Damanhouri 1, Mohammad A Kamal 1,*
PMCID: PMC4978221  NIHMSID: NIHMS738762  PMID: 22483313

Abstract

Parkinson's disease (PD) is a common chronic progressive neurodegenerative disorder in elderly people. A consistent neurochemical abnormality in PD is degeneration of dopaminergic neurons in substantia nigra pars compacta, leading to a reduction of striatal dopamine (DA) levels. As tyrosine hydroxylase (TH) catalyses the formation of L-dihydroxyphenylalanine (L-DOPA), the rate-limiting step in the biosynthesis of DA, the disease can be considered as a TH-deficiency syndrome of the striatum. Problems related to PD usually build up when vesicular storage of DA is altered by the presence of either α-synuclein protofibrils or oxidative stress. Phosphorylation of three physiologically-regulated specific sites of N-terminal domain of TH is vital in regulating its kinetic and protein interaction. The concept of physiological significance of TH isoforms is another interesting aspect to be explored further for a comprehensive understanding of its role in PD. Thus, a logical and efficient strategy for PD treatment is based on correcting or bypassing the enzyme deficiency by the treatment with L-DOPA, DA agonists, inhibitors of DA metabolism or brain grafts with cells expressing a high level of TH. Neurotrophic factors are also attracting the attention of neuroscientists because they provide the essential neuroprotective and neurorestorative properties to the nigrostriatal DA system. PPAR-γ, a key regulator of immune responses, is likewise a promising target for the treatment of PD, which can be achieved by the use of agonists with the potential to impact the expression of pro- and anti-inflammatory cytokines at the transcriptional level in immune cells via expression of TH. Herein, we review the primary biochemical and pathological features of PD, and describe both classical and developing approaches aimed to ameliorate disease symptoms and its progression.

Keywords: Parkinson disease, tyrosine hydroxylase, dopamine, α-synuclein, neuroprotection, neurotrophic factors, L-DOPA

INTRODUCTION

Parkinson's disease is a chronic progressive neurodegenerative disorder. First identified by James Parkinson in 1817, who wrote an essay on the shaking palsy for the description of disease. Currently, it represents the second most common neurodegenerative disorder, after Alzheimer's disease [1, 2], and affects approximately 6 million people worldwide [3]. Patients exhibit a range of clinical symptoms, with the most common affecting motor function, including the development of resting tremor, rigidity, akinesia, bradykinesia and postural instability. However, and non-motor symptoms, like dementia, depression, visual hallucination and autonomic dysfunction, are also common [4, 5].

PD classically involves the degeneration of dopaminergic neurons within the substantia nigra pars compacta (SNC), leading to a reduction of striatal dopamine (DA) levels that are critical in the coordination and control of motor activity [6]. The enzyme, tyrosine hydroxylase (TH) [EC 1.14.16.2] sometimes known as tyrosine 3-monooxygenase) catalyses the formation of L-DOPA, the rate-limiting step in the biosynthesis of DA, thereby directly linking PD with TH. L-DOPA is a precursor for DA synthesis, which, in turn, is a precursor for both norepinephrine (noradrenaline) and epinephrine (adrenaline). The loss of ability to optimally synthesize catecholamines is an important step in the progression of PD and other neurodegenerative diseases [7]. Indeed, early loss of TH activity followed by a decline in TH protein is considered to contribute towards DA deficiency and phenotypic expression in PD [7, 8].

The prevalence of PD varies among ethnic and geographic groups around the world, being particularly low in China and high in Argentina [9]. In general, however, its highest prevalence is found in Caucasians, and in men versus women; nevertheless, across numerous studies prevalence and annual incidence appear to increase continuously into very old age. Albeit that a number of studies have reported that rural versus urban living is a risk factor for PD, whereas others have not, it is clear that geographic ‘disease belts’ of high prevalence/incidence exist [10], and this nonrandom disease distribution reasons compellingly for an environmental impact on the pathogenesis of PD. However, the main etiology of the disease has yet to be identified, albeit occupational and genetic factor, in addition to environment, seem to play important roles [10-12]. In the light of this introduction, research elucidating underlying mechanisms and towards the development of new therapies for PD has become a highly active area within neuroscience and continuous progress warrants frequent analysis and review.

TH is present in all dopaminergic cells and catalyzes the formation of L-DOPA, a deficiency in TH is a hallmark of PD [13]. This enzyme is a highly specific, non-heme iron, tetrahydrobiopterin-dependent protein that catalyzes the conversion of tyrosine to L-DOPA, and represents the rate-limiting step in the biosynthesis of catecholamines (CA) functioning as hormones and neurotransmitters both in invertebrates and vertebrates [14]. In mammals, TH is found in diverse tissues comprising the central and sympathetic nervous system: brain, adrenal medulla and other peripheral sympathetic neurons [15-17]. It is considered as a cytosolic protein, however some investigators have also reported it as a membranous fraction [18, 19]. The physiological significance of this fraction remains to be elucidated, but it has been proposed to be associated with the coupling of synthesis and storage of neurotransmitters into their synaptic vesicles [20]. A largely unexplored and interesting field has hence emerged in relation to TH binding to the synaptic vesicle membrane [21]. TH is found in the neuro-endocrine system, mainly in brain and in chromaffin cells of adrenal medulla, predominantly in the cytoplasm [22]. The emergence of simpler organisms as animal models to study neurological functions and development, epitomized by D. melanogaster or Caenorhabditis elegans, opens the possibility of exploring these new roles of DA, adrenaline and nor adrenaline [23-25].

CHEMISTRY OF TH IN PD VIA DA BIOSYNTHESIS

A primary problem in PD is insufficient DA, whatever the cause may be. DA is generated within dopaminergic neurons via the following pathway (Fig. 1).

Fig. (1).

Fig. (1)

Generation of DA from L-tyrosine.

The initial step is catalyzed by the enzyme, TH. Within the full reaction of L-DOPA formation from L-tyrosine, a coenzyme THFA (tetrahydrofolic acid) and ferrous ions are necessary [26]. The activity of TH is often as low as 25% of healthy age-matched controls in parkinsonism, and in severe cases can fall as low as 10%. This suggests that one or more key elements essential for the formation of L-DOPA likely is in insufficient quantity in PD. The second step for DA generation involves the enzyme DOPA decarboxylase (EC 4.1.1.28 known also as aromatic L-amino acid decarboxylase), in pyridoxal phosphate is essential. The activity of the enzyme varies in accord with the amount of pyridoxal phosphate present [26].

DA AS AN ENDOGENOUS TOXIN

DA neurons of the nigrostriatal pathway are a primary site of neuronal degeneration in PD, whereas neuronal alterations observed in PD are not restricted to DA neurons only [27]. The presence of mitochondrial dysfunction and oxidative stress in these highly metabolic neural cells, does not fully explain the heightened vulnerability of dopaminergic neurons. The fact that substantia nigra DA neurons degenerate in PD to a greater extent than other neurons might be due, in part, to the presence of DA itself [28]. The unique chemical structure of DA has the potential to form an endogenous toxin that may contribute further to mitochondrial dysfunction and oxidative damage, accelerating dopaminergic cell death in PD in response to oxidative insult [29]. Under normal conditions, DA is synthesized from tyrosine via L-DOPA. DA, once generated by the enzymatic activities of TH and DOPA decarboxylase, is rapidly stored at a high millimolar concentration and stabilized at a low pH within synaptic vesicles following uptake by the vesicular monoamine transporter, thereby limiting its cytoplasmic concentration [30]. Cytoplasmic (non-vesicular) DA is subject to oxidation, on exceeding physiological concentrations, via monoamine oxidase type A (MAO-A) and aldehyde dehydrogenase (ADH) to generate non-toxic 3,4-dihydroxyphenylacetic acid and hydrogen peroxide. It can be additionally sequestered into lyposomes where it can be oxidized to neuromelanin (NM).

Problems can potentially develop when vesicular storage of DA is altered by the presence of α-synuclein protofibrils, aberrant oxidative stress and weak base compounds [31], resulting in elevated cytoplasmic DA levels. Under such circumstances secondary oxidative pathways can be triggered that divert the typical non-toxic metabolic sequence and NM formation to allow the generation of the intermediates, such as dopamine-o-quinone and dopaminochrome, which can subsequently generate further metabolites, such as leukodopamineochrome-o-semiquinone that are considered powerful endogenous neurotoxins consequent to an ability to reduce oxygen to a superoxide radical anion, giving rise to redox cycling and producing acute toxicity [26].

INVOLVEMENT OF TH IN THE PATHOGENESIS OF PD AT GENETIC LEVEL

The identification of three genes and several additional loci linked with inherited forms of PD signifies that it is not a single disorder. The analyses of structure and function of these gene products implicates the significant role of protein aggregation in dopaminergic neurons of the substantia nigra as the common mechanism leading to neurodegeneration in all known forms of PD. The three specific genes identified thus far are α-synuclein, Parkin and ubiquitin C terminal hydrolase, which are either closely implicated in the appropriate functioning of the ubiquitin-proteasome pathway or are degraded by this protein-clearing machinery of cells [32]. The biochemical and molecular cascades characterized from genetic studies in PD may provide novel targets for curative therapies [33].

STRUCTURE AND MOLECULAR GENETICS OF TH

Mammalian TH is a highly homologous oligomeric protein that are composed of four subunits, having three well-differentiated domains for each subunit [34]. These domains include a N-terminal regulatory domain that varies in length depending on the enzyme. In the case of TH4, the central catalytic domain is composed of some 280 residues and the C-terminal oligomerization domain contains 45-50 residues [15]. The N-terminal domain of TH is quite complex, with 4 to 5 phosphorylation sites [35]. These multiple phosphorylation sites have been associated to the tight regulation of this enzyme, by controlling either kinetic activity and/or binding to protein partners [22, 36]. The boundary between the regulatory and the catalytic domain is located in the region of residues 165-179, and the last 50 residues in the C-terminal constitute the oligomerization domain [37].

The activity of TH is hence regulated by site-specific phosphorylation of three physiologically-regulated sites in the central nervous system, namely Ser19, Ser31 and Ser40 [38], which impact the function of TH. It has been reported that the phosphorylation of Ser19 does not directly influence TH activity [39] but increased phosphorylation of Ser40 can augment TH activity within a certain threshold and is associated with elevated DA turnover in neurodegenerative disorders [36, 40-42]. Ser31 phosphorylation alone can increase L-DOPA biosynthesis. Salvatore et al. [40] reported that Ser31 or Ser40 phosphorylation modulates DA availability and is necessary to define the molecular basis for DA dependent behaviors.

The human TH gene (hTH) is located on chromosome 11p15.5, spanning approximately 8 kilo bases and containing 14 codifying exons [43]. It codes for four different isoforms, created by alternative splicing (hTH1-4), albeit that later studies have revealed that more mRNA species may be expressed within the cell [44-46]. The hTH1-4 isoforms are expressed in same tissue, although in different proportions and share similar kinetic properties, with hTH1 being the more active and abundant form [47]. The physiological significance of the TH isoforms remains to be fully elucidated and remains an interesting area of current research [48]. Crystal structure analyses of catalytic and tetramerization domains of TH have revealed a novel alpha-helical basket that supports catalytic iron and a 40A0 long anti-parallel coiled coil, which forms the core of tetramer. The catalytic iron is located 10A0 below the enzyme surface in a 17A0 deep active site pocket and is coordinated by the conserved residues His331, His336 and Glu376. The structure provides a basis for understanding the effect of point mutations in TH [37, 49].

ROLE OF α-SYNUCLEIN IN THE REGULATION OF TH ACTIVITY AND DA BIOSYNTHESIS

PD is characterized by the loss of DA containing neurons projecting from the substantia nigra to the dorsal striatum [50, 51]. A key role for α-synuclein has been postulated, albeit with largely undetermined function in the pathogenesis of PD [52-55]. α-Synuclein is a major component of Lewy bodies, a neuropathological hallmark of PD [56-59]. This small and highly conserved protein is enriched in presynaptic terminals and is implicated in both normal as well as pathogenic brain function [60-64]. Whereas the physiological function of α-synuclein remains to be fully characterized [63-65]. Several studies have established that interaction of α-synuclein and TH is of functional relevance to dopaminergic neurotransmission and the pathophysiology of PD [49, 66, 67]. Indeed, α-synuclein gene mutations and PD pathogenesis are well documented [49]. In this regard, mutations such as A53T or A30P and reduced α-synuclein expression have been reported in familial and sporadic form of PD, respectively [49, 68-70]. An interaction between α-synuclein and TH in PD pathogenesis has been confirmed by the use of immuneoprecipitation, whereby both proteins co-precipitate in brain homogenate, and immunoelectron microscopy techniques, whereby they co-localize [54]. Several cell-free studies and animal models signify that α-synuclein mutations accelerate its aggregation [49, 71, 72]. Earlier studies by Kim et al. [73] and Souza et al. [74] have revealed that α-synuclein may be a chaperone protein, based on its ability to inhibit thermally induced protein precipitation and it structural homology with 14-3-3 proteins [49, 75]. Research studies indicate that the binding and activation of TH by 14-3-3 proteins preferentially to phosphoserines [76, 77] and short-term TH activity is regulated by serine phosphorylation [78, 79]. Furthermore, 14-3-3 interacts with TH through an association with Ser19 and Ser40 and the interaction of 14-3-3 with TH Ser19 is required for its activation [49, 54, 80, 81].

Even though the function of α-synuclein remains obscure, it may bind to TH and inhibit its activity, thereby serve as a key regulator of DA synthesis. A loss of α-synuclein by its aggregation or decreased expression may selectively disrupt DA homeostasis and negatively impact DA neuronal survival [49, 54, 70]. Perez et al. [54] described the association of α-synuclein and inhibition of TH activity in a cell-free assay; reporting that over expression of α-synuclein in a dopaminergic cell line dramatically reduces TH activity, TH phosphorylation and DA synthesis in brain as well as in a dopaminergic cell line. Hence, elevated levels of α-synuclein, due to a specific disease or the normal ageing process could be associated with dopaminergic neuronal dysfunction through TH regulation interference [49, 63].

ENVIRONMENTAL FACTORS ASSOCIATED WITH THE ONSET OF PD

Epidemiological studies have suggested several possible environmental factors, associated with PD such as exposure to heavy metals, pesticides and farming [10, 82-85]. The mechanism(s) underpinning the actions of pesticides on the dopaminergic nigrostriatal pathway are far from understood, but the sporadic form of PD has been associated with an increased exposure to pesticides [86-88]. Furthermore, there is convincing support that several environmental chemicals, including rotenone [89], paraquat [90-92] and heptachlor [93], act directly or generate neurotoxicants that selectively induce damage to the nigrostriatal pathway and instigate PD symptoms in experimental animals. The mechanism of dopamine neuropathy induced by rotenone has been reported to be mediated selectively via the inhibition of mitochondrial complex I, which resulted in elevated oxidative stress. Interestingly, rotenone affected complex I uniformly throughout the brain, which results into highly selective degeneration of the nigrostriatal dopaminergic system [89]. The systemic exposure to paraquat provoked a dose-dependent loss of DA neurons in the SNC coupled to a reduction in the density of striatal DA fibers expressing TH in animal models [94-96]. Furthermore, an exposure to pesticides paraquat and maneb during early postnatal days has been reported to produce permanent and progressive lesions of the nigrostriatal DA system and enhance adult susceptibility to pesticides [92, 97]. Pesticides and metals can also produce synergistic effects on the fibrillization of α-synuclein [49, 97]. Deltamethrin, one of the most potent pyrethroid insecticides, selectively inhibits synthesis of DA while increasing the turnover of DA in the striatum. It also inhibits the activity and mRNA/protein expression of TH in striatum [97]; thereby implicating TH as a molecular target of pesticides within the nigrostriatal pathway.

A single nucleotide polymorphism (SNP) in the gene was found to be associated with PD with an early age of onset. Genetic component plays a much more important role in the pathogenesis of PD. Polymorphisms in several genes such as α-synuclein, Parkin, UCH-L1 (ubiquitin-C terminal hydrolase-L1) and DJ-1 have previously been identified in PD [98, 99]. The more common, sporadic form of Parkinson's disease appears to result from an interaction between genetic and environmental factors [100]. SNP in the gene for the pro-inflammatory cytokine interleukin 6 potentiate the susceptibility to PD at early age [101]. In one study, Ohtake et al. (2004) suggested that mutations in β-synuclein gene may predispose to dementia with Lewy bodies in which the substitutions of amino acid from valine to methionine at codon 70 and proline to histidine at codon 123 was present. A meta-analysis of 84 association studies of 14 genes showed that polymorphisms in four genes are significantly associated with PD [102]. ATP binding cassette superfamily of transporter genes have been also reported to influence susceptibility to Parkinson's disease [103].

USE OF TH IN THE TREATMENT OF PD

A decline in DA level, formed by hydroxylation of L-tyrosine to L-DOPA by TH, is a classical pathological feature of PD patients. The most common therapeutic strategy for PD is to restore DA levels by the administration of L-DOPA, the precursor of DA, which can access brain [104] consequent to an affinity for the blood-brain barrier. However, the brain penetration of L-DOPA is limited by the presence of enzymes L-DOPA decarboxylase and monoamine oxidase within brain capillary endothelial cells, forming a “enzymatic blood-brain barrier” to limit L-DOPA passage into brain. Hence, PD therapy is enhanced by concurrent treatment with an inhibitor of L-DOPA decarboxylase. Classical pharmacological studies during 1950s initially suggested that the decline in DA levels in forebrain of experimental animals and proposed motor symptoms might be due to DA dysfunction [105]. More recent studies indicate that the intravenous/oral administration of L-DOPA minimizes PD symptoms at earlier stages [106-108]. In comparatively advanced PD patients, L-DOPA does not appear to impact disease progression, loses its potency over time and leads to non-physiological intermittent stimulation of striatal neurons that express DA receptors [109].

Therapeutic means that induce an elevation in the intrastriatal activity of TH by continuously maintaining appropriate cerebral DA concentrations are expected to palliate PD symptoms. Recently various strategies, such as neuroprotective approaches through the use of antioxidants, different neurotrophic factors, cell replacement therapies including stem cells as well as gene transfer approaches are under analysis for the treatment of PD [110-114].

TREATMENT OF PD BY GENE THERAPY

Gene therapy can be broadly described as the transfer of specific genetic material into a defined target cell for the ultimate purpose of preventing or altering a particular disease state. Strategies for the application of gene therapy techniques for PD treatment have broadened beyond classical DA replacement towards the use of neurotrophic factors. Theoretically, gene therapy in patients with PD has potential utility to replace a defective gene, introduce a potentially neuroprotective or neuro-restorative protein or to permit the physiological delivery of a deficient neurotransmitter. For example, it could be undertaken to focus the enzymes responsible for the DA biosynthesis within the affected striatum. As described earlier, the rate limiting step in DA synthesis is the conversion of L-tyrosine to L-DOPA by the enzyme TH [115], with subsequent conversion to DA by DOPA decarboxylase. Enzyme turnover is so rapid that L-DOPA levels in the brain are negligible under normal conditions. As TH is the rate-limiting enzyme in DA biosynthesis, it is a prime target for physiological regulation and hence pharmacological manipulation.

A variety of viral vectors are used in gene transfer and, based on their specific advantages and disadvantages in their ability to safely transfer genes to the cells they recognize and whether they alter the cell's DNA permanently or temporarily, have provided targeted protein expression in specific regions of the brain. Specific examples of gene therapy using viral vectors include:

ADENO ASSOCIATED VIRUS – GLUTAMIC ACID DECARBOXYLASE (AAV-GAD) GENE THERAPY

Decreased GABA input to the subthalamic nucleus (SN) results in dis-inhibition of this structure in PD. A Phase II human trial involving bilateral gene transfer of GAD into the SN has been completed using an AAV vector. The gene transfer enhanced local synthesis of inhibitory neurotransmitter GABA in the SN and suppressed excessive activity [116].

AAV2-NEURTURIN GENE THERAPY OR CERE-120

Neurturin is a naturally occurring homologue of glial cell line-derived neurotrophic factor (GDNF). Within the basal ganglia, it promotes the sprouting and survival of dopaminergic neurons. CERE-120 is a modified form of human neurturin. Following up on an encouraging unblended Phase I trial, a Phase II double blind randomized trial was conducted in which patients received either bilateral intraputamin injection of AAV2-neurturin or sham surgery [117]. However, as assessed at 12 months, there was no difference between treatment and sham groups, with 13/38 subjects within the AAV2-neuroturin group presented serious adverse effects versus 4/20 in the sham group [112]. A further trial of neurturin, utilizing nigral and putaminal injection both, as well as a longer observation period, is currently underway in PD [118].

AROMATIC L-AMINO ACID DECARBOXYLASE GENE THERAPY

This therapy involves injecting an adenovirus vector containing DNA encoding the aromatic amino acid decarboxylase gene into the striatum of PD patients to increase local conversion of exogenous L-DOPA (administered as levodopa) to DA. The degree of conversion can then be controlled by changing the levodopa dosage. As assessed at 6 months, this gene therapy strategy proved safe, but longer-term analysis is required to define efficacy [119].

PROSAVIN

This approach utilized intrastriatal injections of a tricistronic lentivirus encoding TH, aromatic amino acid decarboxylase, and GPT cyclohydrolase [120]. These three enzymes were reported to result in an enhanced production of DA at the site of injection, but avoided excessive production in other areas where DA was not depleted.

Compared to transplantation of transfected cells, direct gene transfer is less invasive, inducing minimal disturbance of synaptic circuitry and does not involve risk of unregulated cellular proliferation or undesired delivery of cell byproducts. Several non-viral methods to deliver genes, such as liposomes or particle bombardment, have provided interesting results but often difficult to interpret [121-123]. Therefore, most direct gene transfer has been performed by means of viral vectors that serve as vehicles for transfer of genes to the central nervous system. In vivo gene therapy for DA replacement attempts to convert endogenous striatal cells into L-DOPA producing cells [124].

IN VIVO GENE TRANSFER OF TH

Adeno associated viral vectors (AAV), utilized to introduce the TH gene into the striatum of 6-OHDA lesioned rats, have resulted in significant behavioral recovery and were accompanied by TH immunoreactivity in striatal neurons and glia [125]. These effects were observed without signs of toxicity. During et al. [126] also observed similar results.

TH AND GTP CYCLOHYDROLASE

GTP cyclohydrolase is a critical enzyme in catecholamine function and is rate limiting for the synthesis of catecholamine co-factor tetrahydrobiopterin (BH4). Mandel et al. [127] co-transfected cells to express the genes for both TH and GTP cyclohydrolase using an AAV vector system in a rat model to achieve continuous L-DOPA delivery in the striatum. Rats that received the TH AAV vector alone produced measurable L-DOPA only after receiving exogenous BH4. Moreover, elevated L-DOPA was observed in animals that received mixed TH and GTP AAV vectors [124]. Fan et al. [128] reported a similar result while using transduced TH and AADC in rat striatal cells, using two separate AAV vectors.

TH, GTP CYCLOHYDROLASE AND (LEVO AROMATIC AMINOACID DECARBOXYLASE) AADC

Following the success by co-transfection of TH and AADC with two different AAV vectors, Shen et al. [129] attempted a triple transfection combining AAV vectors expressing TH, GTP cyclohydrolase and AADC. In vitro experiments showed that triple transfection results into greater DA production than double transduction with AAV-TH and AAV-AADC. Triple transfection also enhanced BH4 and DA production in the denervated striatum of parkinsonian rats and improved the rotational behavior of rats more effectively than double transfection. Their results thus indicate that GTP cyclohydrolase, in addition to TH and AADC, can be useful for effective gene therapy of PD [124].

CELL REPLACEMENT THERAPY OF PD

Several reports have suggested that injection of cells transfected with TH or dopaminergic in origin can improve PD symptoms [130-133]. In this regard, genetic manipulation and transplantation of cells with the ability to synthesize DA has become an increasingly important area of translational research in PD [134-136]. The need to maximize the early survival of newly transplanted cells to allow them the opportunity to function, integrate into the neural network and gain neurotrophic support has also been recognized [137] and is a current focus to optimize the transplantation approach with the aim of developing and optimizing an alternative or complementary strategy to L-DOPA treatment. Moreover, as yet, relatively little is known about possible modifications to cell homeostasis and the nature of cellular responses when TH is transfected into cells that normally do not express or produce only relatively low levels of this enzyme. For future research, such knowledge will be relevant for the development for truly effective cell transplantation therapy in PD.

INVOLVEMENT OF TH IN THE PATHOGENESIS OF PD VIA OXIDATIVE STRESS

1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) is a synthetic DA neuronal toxin that causes PD when ingested by humans or animals [138]. It is well documented in scientific literature that the actions of MPTP are mediated, in part, by free radicals [139]. In vivo and in vitro MPTP models of PD showed that key enzymes involved in the production of reactive oxygen species (ROS)/reactive nitrogen species (RNS), are upregulated in damaged areas and contribute to DA neuronal death [140, 141]. Additionally, TNF-α and IL-1β are also reported to be increased and contribute to DA neuronal death in the MPTP model of PD [142].

Mitochondria have been shown to play a role in multiple forms of cell death in PD. Mitochondrial dysfunction in PD has been identified as a systemic deficiency of the electron transport chain Complex I activity [143]. The deficiency or partial inhibition of Complex I have been shown to result in increased mitochondrial reactive oxygen species (ROS) production and subsequently increased oxidative stress [144].

GSH is the major reducing agent in the cell and its reduction is particularly important, especially because it has been reported to be significantly decreased early in PD [145]. Mitochondrial dysfunction and oxidative stress have been increasingly implicated in the pathology associated with genetic models of familial PD [146].

TH INCREASES THE RESISTANCE OF HUMAN NEUROBLASTOMA CELLS TO OXIDATIVE INSULTS

Several studies have reported that injection into dopaminergic brain regions of a wide variety of cells tranfected with TH, such as stem cells, astrocytes, fibroblast and myoblast, can ameliorate PD symptoms in animal models [130-133]. Such action can be potentially mediated at several points, in addition to directly elevating DA levels. As described, the neurotransmitter DA under specific conditions can generate ROS spontaneously or via enzymatic processing, and may trigger or exacerbate dopaminergic neurodegeneration by creating potentially harmful hydroxyl radicals, which can damage proteins, lipids and nucleic acids [147-149]. Despite the potentially severe deleterious effects of high levels of ROS to cell viability, it has been proposed that low level exposure can mediate an up regulation of endogenous defense mechanisms that may protect cells against a higher oxidative burst [150]. Preconditioning by oxidative stress is reported to be similar to ischemic preconditioning, and requires the production of nontoxic amounts of ROS, which modulate signaling cascades important in the acquisition of resistance for further harmful insults [151].

Recent studies suggest that exposure to low levels of DA provides preconditioning in different models of PD in cell culture studies in which oxidative stress acts as an initiator [152-154]. Furthermore, in culture studies Franco et al. [155] reported that neuronal cells transfected with elevated levels of TH were more resistant than their wild-type counterparts due to the establishment of a preconditioning mechanism, resulting in an elevation in the activity of the antioxidant glutathione-metabolizing enzymes glutathione reductase and glutathione peroxidase, generated by low levels of DA exposure [155]. In accord with other studies, the transfection of elevated TH into cells led to an elevated production of DA and its metabolites [155]. In the light of their study, they proposed that a therapy using cells expressing TH transcripts may provide multiple beneficial effects in PD-damaged brain areas: elevated DA levels to support neurotransmission and alleviate motor symptoms, as well as an improved antioxidant ability of these cells to withstand potential endogenous and exogenous physiological insults. McCormack et al. [156] demonstrated that specific populations of dopaminergic neurons within the brain, such as within the substantia nigra, are more vulnerable to the herbicide paraquat-induced toxicity than other dopaminergic cells within the midbrain consequent to a decreased resistance to oxidative stress. Interestingly, reduced vulnerability appeared related to presence of the calcium-binding protein, calbindin-D28k, which is greater in midbrain regions and within the ventral tegmental area that are not as impacted by PD. Thus there appears to be a level of TH expression together with associated proteins in both normal and transplanted cells that confers optimal provision of DA for neurotransmission and for preconditioning against oxidative insults.

STEM CELL THERAPY FOR PD TREATMENT

It is well documented that pharmacological treatment with L-DOPA generally provides initial positive results, but gradually over the time efficacy is lost and motor complications can occur [157]. The transplantation of DA cells from human embryonic mesencephalon (7–8 weeks after conception) has been reported to improve motor function in people with advanced PD [158-160]. However, this approach has its own limitations, the least of which involves the ethics of using embryonic tissue, difficulty in obtaining it and the poor survival of the transplanted DA neurons.

Transplantation of human embryonic stem cells (hES cells) or their partially differentiated progeny, embryoid bodies leads to the development of some DA neurons and teratomas [161, 162] and can potentially be used for the treatment of patients with PD, but is not ideal [160]. Kimberley et al. [160] produced TH-positive cells from hES cells by co culturing them with PA6 cells (mouse stromal cells possessing stromal cell-derived inducing activity), thereby revealing that PA6 cells can cause differentiation of hES cells, as has previously been shown for both mouse and nonhuman primate ES cells [163, 164]. Importantly, the yield of TH-positive cells could be dramatically increased by co-incubation with soluble factors produced by human embryonic striatal tissue as well as by the addition of glial-derived neurotrophic factor (GDNF) [160], which has been widely shown to reduce apoptosis of dopaminergic neurons. Interestingly, GDNF increased the number as well as size of differentiating hES cell colonies and approximately doubled the number of TH-positive cells. Since numerous components of the cultures were determined to be augmented, GDNF and alike factors can therefore be considered as global survival factors that optimize the differentiation of hES and additionally amplify the expression of transcription factors important in the development of dopaminergic neurons [160]. The underlying mechanism(s) via which stromal cell co-cultures and related factors promote differentiation of hES cells to TH-positive cells remains to be elucidated and represents an interesting area of ongoing research [160]. In a comparable approach, Zhou et al. [165] reported the efficacy of injecting adult bone marrow derived stem cells transfected with a pEGFP-C2 plasmid containing the gene for TH into the lateral ventricle for the treatment of rats with PD. Albeit more research is required to fully characterize this approach, it clearly remains a further future option in the potential utility of genetically engineered BMSCs in the treatment of PD [165].

NEUROTROPHIC FACTORS IN PD TREATMENT

Developing as well as mature neurons require the presence of neurotrophic factors to maintain their function and viability, and these, epitomized by BDNF, are very often synthesized and secreted by the target tissue. The augmentation of endogenous neurotrophic factors as well as exogenous administration of others represents a promising therapeutic strategy for PD treatment [166-169]. Among the various neurotrophic factors studied, the glial cell line-derived neurotrophic factor released by striatal neurons is more closely associated with PD [168, 170]. It is widely appreciated that glial cell line-derived neurotrophic factor is an agent necessary for maintenance of nigrostriatal DA neurons [171, 172]. In view of its role in the maintenance of DA neurons, it can be considered as a potential therapeutic target of PD treatment [173, 174]. Similarly, fibroblast growth factor 2 is a trophic factor for neurons and its potential neuroprotective actions in DA neurons have been reported [175, 176]. Involved in peripheral nerve development and regeneration, its exogenous application promotes neuronal survival and neurite outgrowth in cellular and animal models [177].

The neuroprotective effect of numerous drugs appears to be mediated, in part or in whole, via up regulation of endogenous neurotrophic factors and, in particular, BDNF [178] but also GDNF [179]. As a recent example among many is catalpol, an active component within a number of Chinese medicinal herbs, including Rehmannia glutinosa, that are widely used in treating a variety of neurodegenerative conditions in traditional Chinese medicine. Catalpol has been reported to attenuate amyloid-β peptide-induced toxicity of cholinergic neurons by elevating BDNF levels [180] and, additionally, has been reported to mitigate the toxicity of the dopaminergic toxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), by up regulating GDNF levels [181]. The compound, thereby, elevate TH neuron number and DA levels without binding to either DA receptors or its transporter, and thus ameliorated MPTP-induced motor effects in rodents [179]. Finally, cerebral DA neurotrophic factor (CDNF), a recently discovered neurotrophic factor, has recently been reported to have neuroprotective and neurorestorative properties for the nigrostriatal DA system following administration to mice [181-183]. TH-immunoreactivity as well as the number of TH-positive cells were reportedly increased by CDNF treatment, supporting the development of CDNF-based treatment strategies for PD [181,183].

A further neurotrophic strategy in PD involves the incretin glucagon-like peptide-1 (GLP-1). This 30-amino acid endogenous insulinotropic peptide is best known for its action to regulate blood glucose levels via activation of the GLP-1 receptor (R) on pancreatic β-cells to induce insulin secretion. It additionally acts as a trophic agent, inducing pancreatic β-cell proliferation and neogenesis, as well as inhibiting β-cell apoptosis [184, 185]. Long-acting GLP-1R agonists, including exendin-4 (Ex-4) and liraglutide are approved and now widely used in the effective treatment of type 2 diabetes mellitus [186]; a disease that is known to be associated with an increased incidence of PD as well as of Alzheimer's disease (AD) [187]. Not only is the GLP-1R found throughout the brain but also GLP-1, exendin-4 and liraglutide, despite being peptides, readily cross the blood-brain barrier [188]. Recent studies demonstrate that both GLP-1 and exendin-4 are neurotrophic [189, 190] and increase TH expression in dopaminergic cultures [181] and, additionally, are neuroprotective and fully ameliorate TH loss, DA and metabolite reductions, as well as behavioral impairments induced by MPTP in rodents [191, 192]. Likewise, exendin-4 has been found to provide neurotrophic, neuroprotective and neuroregenerative actions in 6-hydroxydopamine and LPS animal models of PD, mitigating locomotor deficits, ameliorating neuroinflammation and elevating both TH and DA levels in dopaminergic brain regions [193-195]. Such neurotrophic/protective actions appear to translate across neuronal cell types as well neurodegenerative disorders, as GLP-1R agonists have now been reported as effective in cellular and animal models of AD, stroke, ALS and peripheral neuropathy [191, 196-202], spurring repositioning of exendin-4 in recently started clinical trials of AD, PD and peripheral neuropathy (ClinicalTrials.gov identifier: NCT01255163, NCT01174810 and NCT00855439, respectively).

PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR GAMMA (PPAR-γ) MEDIATED TREATMENT OF PD

Neuroinflammation and the dysregulation of central and peripheral immune systems in PD have been reported in earlier studies. The specific targeting of immune functions contributing to neuroinflammation represents a promising therapeutic approach for PD treatment [203]. In this regard, peroxisome proliferator-activated receptors are members of the nuclear hormone receptor superfamily that regulate the expression of genes involved in a wide variety of biological processes. Members of the PPAR family, in particular PPAR-γ , are key regulators of immune responses, highly expressed within cells of the immune system, and play a pivotal role in microglial activation as well as in monocyte and T cell differentiation. This provides a promising strategy for the treatment of PD, which can be achieved by using PPAR-γ agonists consequent to their ability to modulate the expression of pro- and anti-inflammatory cytokines at the transcriptional level in both central and peripheral immune cells [204]. Preclinical evidence of PPAR-γ-induced neuroprotection in numerous experimental PD models has been also described [194, 195]. As an example, beneficial effects of thiazolidinediones, a class of PPAR-γ agonists, in PD treatment have been recently reviewed by Carta and colleagues [205]. As thiazolidinediones, particularly rosiglitazone and pioglitazone, are currently effectively used clinically as insulin-sensitizing agents in the treatment of type 2 diabetes melitus [206], their repositioning to assess clinical efficacy in PD represents a feasible option. This, however, would clearly require a cautious approach in light of the recent warning on the safety of rosiglitazone in diabetes [207]; nevertheless, pioglitazone remains clinically available and has recently shown tolerability in initial clinical studies in AD [208].

ADDITIONAL AGENTS IMPACTING TH AND OF POTENTIAL IN PD

Numerous endogenous and exogenous neurotoxin pathways have been elucidated that may contribute to the pathogenesis of PD [209-212]. β-Carbolines (BCs) are endogenously produced pyridoindoles, that can be synthesized from tryptophan or tryptophan-like indolamines. Specific BCs have been found in various fruits and meats, and appear within the body. Some bear structural similarity to MPTP and have been demonstrated to induce dopaminergic neuron toxicity. In this regard, methylated BC cations, such as 2,9-dimethyl-β the -carbolinium ion (2,9-dime-BC+), have been reported to exert highly damaging actions on dopaminergic neurons; impacting mitochondrial function at the level of respiratory chain, elevating ROS, increasing caspases-3 levels and ultimately inducing a decline in cell survival [213] in a manner reminiscent of MPTP. As 2,9-dime-BC+ has been found elevated in the lumbar CSF idiopathic PD subjects and increased with the progression of the disease [214] bioactivated BC+s, might represent causative factors underlying PD. Interestingly, a different but closely related BC, 9-methyl-β-carboline (9-me-BC), has been described to be beneficial in cellular and animal models of PD by providing a plethora of potentially valuable actions [211, 215]. It has been reported to provide stimulatory effects to dopaminergic neurons by increasing the expression of TH and multiple of its transcription factors, by reducing inflammation and by lowering α-synuclein levels. These actions appeared to translate to in vivo in a rat MPTP model of PD, wherein dopaminergic function was restored [211, 212]; supporting the agent as potentially interesting candidate treatment for PD [211, 216]. Given the close structural resemblance between potentially detrimental BCs (e.g., 2,9-dime-BC+) and potentially valuable ones (9-me-BC) further preclinical research is warranted to ensure that potential metabolites from the latter retain valuable rather than detrimental actions prior to translation.

An agent that already appears to have effectively translated to the clinic for the treatment of PD is the sulfonamide anticonvulsant drug, zonisamide, whose mechanism of action – despite over two decade of clinical use – remains to be fully elucidated. The agent was serendipitously discovered to have beneficial actions in the treatment of both convulsive attacks and parkinsonian symptoms in a patient [217]. Zonisamide was approved in Japan [2009] as adjunctive therapy with levodopa in previously treated adult patients with PD. In animal models it appears to elevate TH levels and reduce neuroinflammation, and thereby mitigate the toxicity of MPTP [218, 219].

ESTRADIOL AND TH

The sex steroids are implicated in roles that extend well beyond reproduction alone. As examples, estrogen actions have been linked to memory and cognition, synaptic plasticity, neurogenesis and neuroprotection [211]. This, in part, may account for the lower incidence of PD in women versus men [220, 221]. Extensive evidence suggests that estradiol induces the expression of TH [222-224] and this is mediated by estrogen receptors α and β [225]. Several studies have documented the neuroprotective actions of estrogens in both cellular and animal models of PD [220, 226]. Estradiol appears to impact TH promoter activity at a transcriptional level as the promoter contains an array of response elements that are sensitive to estradiol [227], as well as an estrogen response element site [224].

The sex hormones are largely generated from the backbone of cholesterol, and it is feasible that abnormalities in cholesterol metabolism contribute to PD pathogenesis. Furthermore, instabilities in the generation of cholesterol oxidation products (oxysterols) may, likewise, contribute to PD vulnerability and onset [228, 229]. In this regard, oxysterol 27-hydroxycholesterol (27-OHC) is a major oxidized cholesterol metabolite within the circulation that has access to the brain [229]. Recent studies suggest that 27-OHC induces a loss of DA content and an elevation in α-synuclein accumulation in neuronal cells, mediated by reducing the expression of TH and increasing α-synuclein levels via estrogen receptors and liver X receptors [228, 229], in particular β receptors. These hence may represent potentially interesting targets via which TH as well as α-synuclein may be regulated to delay PD [229]. The regulation of TH by α-synuclein has been detailed at the level of phosphorylation [230, 231] and at the level of its promoter region [232].

NADH AS A TOOL IN PD TREATMENT THROUGH TH

The majority of PD treatment strategies are based on mitigating its symptoms by impacting nigrostriatal dopaminergic neurons. Largely, such approaches attempt to stimulate the natural DA production capabilities of the brain, and L-DOPA continues to be the primary choice of therapy. As L-DOPA is an endogenous intermediate catecholamine molecule its exogenous administration can induce dramatic changes in the catecholamine system, the activities of key components of which already are reduced in PD patients [233-234]. In light of this, a need was envisioned to overcome the DA deficit without altering or inhibiting key components, such as TH activity, whilst increasing L-DOPA biosynthesis.

An approach to achieve this goal was the use of the coenzyme of TH, BH4; which is reduced in PD. Therapeutic application of BH4 was considered, but it lacked beneficial clinical effects in PD [235]; a limitation potentially due to its difficulty in crossing the blood-brain barrier [234]. However, reduced nicotinamide adenine di-nucleotide (NADH) has been described to effectively boost endogenous production of DA, since NADH can indirectly supply reducing equivalents to the rate-limiting, TH-catalyzed step of DA synthesis [236]. NADH enhances BH4 by increasing a key enzyme in the BH4 pathway, quinoid H2 pteridin reductase [237]. Hence, NADH has been reported to enhance endogenous L-DOPA biosynthesis and increase in L-DOPA production to provide an improvement of the clinical symptoms in PD patients [234]. How such treatment would compare to well established L-DOPA therapy, particularly over an extended duration, remains an important question and may warrant assessment.

CONCLUSION

There is a clear current medical need of new and effective treatment strategies for PD that impact disease progression, and go beyond the present logical and efficient strategy of L-DOPA symptomatic treatment. A number of approaches to correct or bypass the described enzyme deficiency with DA agonists, inhibitors of DA metabolism or brain grafts utilizing cells expressing a high level of TH have long remained areas of interest. Neurotrophic factors, likewise, have long attracted the attention of the neuroscientists consequent to their neuroprotective and neurorestorative properties [238, 239] by attempting to dial in on key regulators of the immune response, as epitomized by targeting PPAR-γ as well as TNF-α [240, 241]. Clearly evident in recent years is the application of cutting edge molecular biology to optimize use of viral vectors to deliver single or combinations of deficient human proteins to specific cells at defined locations. Such approaches are not only build on prior PD research as they move to assessment of clinical utility, but have valuable broad applicability to a variety of prevalent degenerative disorders in which their target may be associated with a different form of neuronal cell death in a different location within the nervous system (motor neurons in ALS, cholinergic and glutamatergic neurons in early AD). Common features between targets across diseases permit the repositioning of already developed drugs effective for one disease to be assessed in another, as epitomized by exendin-4 – an efficacious drug in type 2 diabetes mellitus [184-186, 188] presently being assessed in early efficacy studies in PD and AD. Even for drugs with a different target, such as the experimental AD agents posiphen and phenserine that lower the rate of synthesis of amyloid-β precursor protein and thereby reduces the generation of the AD toxic species amyloid-β peptide [242], the molecular mechanism via it is achieved (translational regulation mediated via an iron response element) within the 5’-untranslated region of amyloid-β precursor protein mRNA [242, 243] is present within the 5’-untranslated region of α-synuclein [244, 245] and thereby providing a basis for the assessment of these agents in PD [246, 247]. Pioglitazone represents a further potential example [248]. Whereas, the translation of drug across diseases, although based on sound science, is not invariably successful, this avenue [249-251] together with the other described basic and translational approaches are providing continuous incremental improvements in our understanding of the basis of PD and our search for targets will hopefully impact disease progression.

ACKNOWLEDGEMENTS

The authors, ST, JNR, SS, QA, GAD and MAK, gratefully acknowledge the research facility provided by the King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia. NG is supported by the Intramural Research Program of the National Institute on Aging, National Institutes of Health, Baltimore, MD, USA, and has received additional funding from the Michael J Fox Foundation.

ABBREVIATIONS

CDNF

Cerebral DA neurotrophic factor

DA

Dopamine

hES

Human embryonic stem

NTFs

Neurotrophic factors

NADH

Nicotinamide adenine di-nucleotide

PD

Parkinson's disease

PPAR-γ

Peroxisome proliferator-activated receptor gamma

TH

Tyrosine hydroxylase

ROS

Reactive oxygen species

Footnotes

CONFLICT OF INTEREST

Declared none.

REFERENCES

  • 1.Kawajiri S, Saiki S, Sato S, Hattori N. Genetic mutations and functions of PINK1. Trends Pharmacol. Sci. 2011;32(10):573–580. doi: 10.1016/j.tips.2011.06.001. [DOI] [PubMed] [Google Scholar]
  • 2.Bralten J, Aria VA, Makkinje R, Veltman JA, Brunner HG, Fernández G, Rijpkema M, Franke B. Association of the Alzheimer's gene SORL1 with hippocampal volume in young, healthy adults. Am. J. Psychiatry. 2011;168(10):1083–1189. doi: 10.1176/appi.ajp.2011.10101509. [DOI] [PubMed] [Google Scholar]
  • 3.Litteljohn D, Mangano E, Clarke M, Bobyn J, Moloney K, Hayley S. Inflammatory mechanisms of neurodegeneration in toxin-based models of Parkinson's disease. Parkinsons Dis. 2011;2011:713517. doi: 10.4061/2011/713517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ruiz PJ, Catalán MJ, Carril JM. Initial motor symptoms of Parkinson disease. Neurologist. 2011;17:18–20. doi: 10.1097/NRL.0b013e31823966b4. [DOI] [PubMed] [Google Scholar]
  • 5.Stacy MA, Murck H, Kroenke K. Responsiveness of motor and nonmotor symptoms of Parkinson disease to dopaminergic therapy. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2010;34(1):57–61. doi: 10.1016/j.pnpbp.2009.09.023. [DOI] [PubMed] [Google Scholar]
  • 6.Hwang DY, Ardayfio P, Kang UJ, Semina EV, Kim KS. Selective loss of dopaminergic neurons in the substantia nigra of Pitx3-deficient aphakia mice. Brain Res. Mol. Brain Res. 2003;114(2):123–131. doi: 10.1016/s0169-328x(03)00162-1. [DOI] [PubMed] [Google Scholar]
  • 7.Blanchard-Fillion B, Souza JM, Friel T, Jiang GC, Vrana K, Sharov V, Barrón L, Schoneich C, Quijano C, Alvarez B, Radi R, Przedborski S, Fernando GS, Horwitz J, Ischiropoulos H. Nitration and inactivation of tyrosine hydroxylase by peroxynitrite. J. Biol. Chem. 2001;276(49):46017–46023. doi: 10.1074/jbc.M105564200. [DOI] [PubMed] [Google Scholar]
  • 8.Nagatsu T. Change of tyrosine hydroxylase in the parkinsonian brain and in the brain of MPTP-treated mice as revealed by homospecific activity. Neurochem. Res. 1990;15(4):425–429. doi: 10.1007/BF00969928. [DOI] [PubMed] [Google Scholar]
  • 9.Masalha R, Kordysh E, Alpert G, Hallak M, Morad M, Mahajnah M, Farkas P, Herishanu Y. The prevalence of Parkinson's disease in an Arab population, Wadi Ara, Israel. Isr. Med. Assoc. J. 2010;12(1):32–35. [PubMed] [Google Scholar]
  • 10.Wright Willis A, Evanoff BA, Lian M, Criswell SR, Racette BA. Geographic and ethnic variation in Parkinson disease: a population-based study of US Medicare beneficiaries. Neuroepidemiology. 2010;34(3):143–151. doi: 10.1159/000275491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Muangpaisan W, Mathews A, Hori H, Seidel DA. systematic review of the worldwide prevalence and incidence of Parkinson's disease. J. Med. Assoc. Thai. 2011;94(6):749–755. [PubMed] [Google Scholar]
  • 12.Sha D, Chin LS, Li L. Phosphorylation of parkin by Parkinson disease-linked kinase PINK1 activates parkin E3 ligase function and NF-kappaB signaling. Hum. Mol. Genet. 2010;19(2):352–363. doi: 10.1093/hmg/ddp501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Adams JD, Jr., Chang ML, Klaidman L. Parkinson's disease-redox mechanisms. Curr. Med. Chem. 2001;8(7):809–814. doi: 10.2174/0929867013372995. [DOI] [PubMed] [Google Scholar]
  • 14.Haavik J, Toska K. Tyrosine hydroxylase and parkinson's disease. Mol. Neurobiol. 1998;16(3):285–309. doi: 10.1007/BF02741387. [DOI] [PubMed] [Google Scholar]
  • 15.Flatmark T, Stevens RC. Structural insight into the aromatic amino acid hydroxylases and their disease-related mutant forms. Chem. Rev. 1999;99:2137–2160. doi: 10.1021/cr980450y. [DOI] [PubMed] [Google Scholar]
  • 16.Fitzpatrick PF. The aromatic amino acid hydroxylases. Adv. Enzymol. Relat. Areas Mol. Biol. 2000;74:235–294. doi: 10.1002/9780470123201.ch6. [DOI] [PubMed] [Google Scholar]
  • 17.Flatmark T, Almas B, Ziegler MG. Catecholamine metabolism: an update on key biosynthetic enzymes and vesicular monoamine transporters. Ann. NY Acad. Sci. 2002;971:69–75. doi: 10.1111/j.1749-6632.2002.tb04436.x. [DOI] [PubMed] [Google Scholar]
  • 18.Fitzpatrick PF. Allosteric regulation of phenylalanine hydroxylase. Arch. Biochem. Biophys. 2011;519(2):194–201. doi: 10.1016/j.abb.2011.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Descarries L, Bérubé-Carrière N, Riad M, Bo GD, Mendez JA, Trudeau LE. Glutamate in dopamine neurons: synaptic versus diffuse transmission. Brain Res. Rev. 2008;58(2):290–302. doi: 10.1016/j.brainresrev.2007.10.005. [DOI] [PubMed] [Google Scholar]
  • 20.Tsudzuki T, Tsujita M. Isoosmotic isolation of rat brain synaptic vesicles, some of which contain tyrosine hydroxylase. J. Biochem. 2004;136:239–243. doi: 10.1093/jb/mvh113. [DOI] [PubMed] [Google Scholar]
  • 21.Cartier EA, Parra LA, Baust TB, Quiroz M, Salazar G, Faundez V, Egana L, Torres GE. A biochemical and functional protein complex involving dopamine synthesis and transport into synaptic vesicles. J. Biol. Chem. 2010;285:1957–1966. doi: 10.1074/jbc.M109.054510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Haycock J;W, Wakade AR. Activation and multiple-site phosphorylation of tyrosine hydroxylase in perfused rat adrenal glands. J. Neurochem. 1992;58:57–64. doi: 10.1111/j.1471-4159.1992.tb09276.x. [DOI] [PubMed] [Google Scholar]
  • 23.Tierney AJ, Kim T, Abrams R. Dopamine in crayfish and other crustaceans: distribution in the central nervous system and physiological functions. Microsc. Res. Tech. 2003;60:325–335. doi: 10.1002/jemt.10271. [DOI] [PubMed] [Google Scholar]
  • 24.Sanyal S, Wintle RF, Kindt KS, Nuttley WM, Arvan R, Fitzmaurice P, Bigras E, Merz DC, Hebert TE, Van Der Kooy D, Schafer WR, Culotti JG, Van Tol HH. Dopamine modulates the plasticity of mechanosensory responses in Caenorhabditis elegans. Eur. Mol. Biol. Organ J. 2004;23(2):473–482. doi: 10.1038/sj.emboj.7600057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kindt KS, Quast KB, Giles AC, De S, Hendrey D, Nicastro I, Rankin CH, Schafer WR. Dopamine mediates context-dependent modulation of sensory plasticity in C. elegans. Neuron. 2007;55:662–676. doi: 10.1016/j.neuron.2007.07.023. [DOI] [PubMed] [Google Scholar]
  • 26.Napolitano A, Manini P, d'Ischia M. Oxidation chemistry of catecholamines and neuronal degeneration: an update. Curr. Med. Chem. 2011;18:1832–1845. doi: 10.2174/092986711795496863. [DOI] [PubMed] [Google Scholar]
  • 27.Sulzer D. Multiple hit hypotheses for dopamine neuron loss in Parkinson's disease. Trends Neurosci. 2007;30:244–250. doi: 10.1016/j.tins.2007.03.009. [DOI] [PubMed] [Google Scholar]
  • 28.Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin-induced mitophagy in the pathogenesis of Parkinson diseas. J. Cell Biol. 2008;183:795–803. [Google Scholar]
  • 29.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: 10.1016/s0306-4522(00)00293-1. [DOI] [PubMed] [Google Scholar]
  • 30.Staal RG, Mosharov EV, Sulzer D. Dopamine neurons release transmitter via a flickering fusion pore. Nat. Neurosci. 2004;7:341–346. doi: 10.1038/nn1205. [DOI] [PubMed] [Google Scholar]
  • 31.Caudle WM, Richardson JR, Wang MZ, Taylor TN, Guillot TS, McCormack AL, Colebrooke RE, Di Monte DA, Emson PC, Miller GW. Reduced vesicular storage of dopamine causes progressive nigrostriatal neurodegeneration. J. Neurosci. 2007;27:8138–8148. doi: 10.1523/JNEUROSCI.0319-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gasser T. Mendelian forms of Parkinson's disease. Biochem. Biophys. Acta. 2009;1792:587–596. doi: 10.1016/j.bbadis.2008.12.007. [DOI] [PubMed] [Google Scholar]
  • 33.Martin HL, Teismann P, Piccini P, Burn DJ, Ceravolo R, Maraganore D, Brooks DJ. The role of inheritance in sporadic Parkinson's disease: evidence from a longitudinal study of dopaminergic function in twins. Ann. Neurol. 2009;45:577–582. doi: 10.1002/1531-8249(199905)45:5<577::aid-ana5>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 34.Siltberg-Liberles J, Steen IH, Svebak RM, Martinez A. The phylogeny of the aromatic amino acid hydroxylases revisited by characterizing phenylalanine hydroxylase from Dictyostelium discoideum. Gene. 2008;427:86–92. doi: 10.1016/j.gene.2008.09.005. [DOI] [PubMed] [Google Scholar]
  • 35.Dunkley PR, Bobrovskaya L, Graham ME, von Nagy-Felsobuki EI, Dickson PW. Tyrosine hydroxylase phosphorylation:regulation and consequences. J. Neurochem. 2004;91:1025–1043. doi: 10.1111/j.1471-4159.2004.02797.x. [DOI] [PubMed] [Google Scholar]
  • 36.Calvo Ana C. PhD Thesis. The University of Bergen; Jun, 2010. Function and regulation of phenylalanine and tyrosine hydroxylases from human and Caenorhabditis elegans. With focus in evolutionary aspects and development of new therapies. [Google Scholar]
  • 37.Goodwill KE, Sabatier C, Marks C, Raag R, Fitzpatrick PF, Stevens RC. Crystal structure of tyrosine hydroxylase at 2.3 Ao and its implications for inherited neurodegenerative diseases. Nat. Struct. Biol. 1997;4:578–585. doi: 10.1038/nsb0797-578. [DOI] [PubMed] [Google Scholar]
  • 38.Haycock JW, Haycock DA. Tyrosine hydroxylase in rat brain dopaminergic nerve terminals: Multiple-site phosphorylation in vivo and in synaptosomes. J. Biol. Chem. 1991;266:5650–5657. [PubMed] [Google Scholar]
  • 39.Haycock JW, Lew JY, Garcia-Espana A, Lee KY, Harada K, Meller E, Goldstein M. Role of Serine-19 phosphorylation in regulating tyrosine hydroxylase studied with site- and phosphospecific antibodies and site-directed mutagenesis. J. Neurochem. 1998;71(4):1670–1675. doi: 10.1046/j.1471-4159.1998.71041670.x. [DOI] [PubMed] [Google Scholar]
  • 40.Salvatore MF, Waymire JC, Haycock JW. Depolarization-stimulated catecholamine biosynthesis: involvement of protein kinases and tyrosine hydroxylase phosphorylation sites in situ. J. Neurochem. 2001;79(2):349–360. doi: 10.1046/j.1471-4159.2001.00593.x. [DOI] [PubMed] [Google Scholar]
  • 41.Conner JR, Wang XS, Allen RP, Beard JL, Wiesinger JA, Felt BT, Earley CJ. Altered dopaminergic profile in the putamen and substantia nigra in restless leg syndrome. Brain. 2009;132(9):2403–2412. doi: 10.1093/brain/awp125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Salvatore MF, Fisher B, Surgener SP, Gerhardt GA, Rouault T. Neurochemical investigations of dopamine neuronal systems in iron-regulatory protein 2(IRP-2) knockout mice. Brain Res. Mol. Brain. Res. 2005;139(2):341–347. doi: 10.1016/j.molbrainres.2005.06.002. [DOI] [PubMed] [Google Scholar]
  • 43.Nagatsu T. The human tyrosine hydroxylase gene. Cell. Mol. Neurobiol. 1989;9(3):313–321. doi: 10.1007/BF00711412. [DOI] [PubMed] [Google Scholar]
  • 44.Dumas S, Le Hir H, Bodeau-Pean S, Hirsch E, Thermes C, Mallet J. New species of human tyrosine hydroxylase mRNA are produced in variable amounts in adrenal medulla and are overexpressed in progressive supranuclear palsy. J. Neurochem. 1996;67:19–25. doi: 10.1046/j.1471-4159.1996.67010019.x. [DOI] [PubMed] [Google Scholar]
  • 45.Ohye T, Ichinose H, Yoshizawa T, Kanazawa I, Nagatsu T. A new splicing variant for human tyrosine hydroxylase in the adrenal medulla. Neurosci. Lett. 2001;312:157–160. doi: 10.1016/s0304-3940(01)02210-8. [DOI] [PubMed] [Google Scholar]
  • 46.Roma J, Saus E, Cuadros M, Reventos J, Sanchez de Toledo J, Gallego S. Characterisation of novel splicing variants of the tyrosine hydroxylase C-terminal domain in human neuroblastic tumours. Biol. Chem. 2007;388:419–426. doi: 10.1515/BC.2007.041. [DOI] [PubMed] [Google Scholar]
  • 47.Kaufman S. Tyrosine hydroxylase. Adv. Enzymol. Relat. Areas Mol. Biol. 1995;70:103–220. doi: 10.1002/9780470123164.ch3. [DOI] [PubMed] [Google Scholar]
  • 48.Daubner SC, Le T, Wang S. Tyrosine hydroxylase and regulation of dopamine synthesis. Arch. Biochem. Biophys. 2011;508(1):1–12. doi: 10.1016/j.abb.2010.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Corti O, Lesage S, Brice A. What genetics tells us about the causes and mechanisms of Parkinson's disease. Physiol. Rev. 2011;91(4):1161–1218. doi: 10.1152/physrev.00022.2010. [DOI] [PubMed] [Google Scholar]
  • 50.Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F. Brain DA and the syndromes of Parkinson and Huntington: clinical, morphological, and neurochemical correlations. J. Neurol. Sci. 1973;20:415–455. doi: 10.1016/0022-510x(73)90175-5. [DOI] [PubMed] [Google Scholar]
  • 51.Hornykiewicz O, Kish SJ. Biochemical pathophysiology of Parkinson's disease. Adv. Neurol. 1986;45:19–34. [PubMed] [Google Scholar]
  • 52.Martin FL, Williamson SJ, Paleologou KE, Allsop D, El Agnaf OM. Alpha-synuclein and the pathogenesis of Parkinson's disease. Protein Pept. Lett. 2004;11(3):229–237. doi: 10.2174/0929866043407138. [DOI] [PubMed] [Google Scholar]
  • 53.Galvin JE, Lee VM, Trojanowski JQ. Synucleinopathies: clinical and pathological implications. Arch. Neurol. 2001;58:186–190. doi: 10.1001/archneur.58.2.186. [DOI] [PubMed] [Google Scholar]
  • 54.Perez RG, Waymire JC, Lin E, Liu JJ, Guo F, Zigmond MJ. A role for alpha-synuclein in the regulation of dopamine biosynthesis. J. Neurosci. 2002;22(8):3090–3099. doi: 10.1523/JNEUROSCI.22-08-03090.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Takeda A, Hashimoto M, Mallory M, Sundsumo M, Hansen L, Sisk A, Masliah E. Abnormal distribution of the non-Abeta component of Alzheimer's disease amyloid precursor/alpha-synuclein in Lewy body disease as revealed by proteinase K and formic acid pretreatment. Lab. Invest. 1998;78:1169–1177. [PubMed] [Google Scholar]
  • 56.Jenner P, Olanow CW. Oxidative stress and the pathogenesis of Parkinson's disease. Neurology. 1996;47:161–170. doi: 10.1212/wnl.47.6_suppl_3.161s. [DOI] [PubMed] [Google Scholar]
  • 57.Markopoulou K, Wszolek ZK, Pfeiffer RF, Chase BA. Reduced expression of the G209A alpha-synuclein allele in familial Parkinsonism. Ann. Neurol. 1999;46:374–381. doi: 10.1002/1531-8249(199909)46:3<374::aid-ana13>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
  • 58.Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Synuclein in Lewy bodies. Nature. 1997;388:839–840. doi: 10.1038/42166. [DOI] [PubMed] [Google Scholar]
  • 59.Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. α-Synuclein in filamentous inclusions of Lewy bodies from Parkinson's disease and dementia with Lewy bodies. Proc. Natl. Acad. Sci. USA. 1998;95:6469–6473. doi: 10.1073/pnas.95.11.6469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Maroteaux L, Campanelli JT, Scheller RH. Synuclein: a neuronspecific protein localized to the nucleus and presynaptic nerve terminal. J. Neurosci. 1988;8:2804–2815. doi: 10.1523/JNEUROSCI.08-08-02804.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Xiangmin M, Peng R, Tehranian PD, Leonidas S, Ruth G. α-Synuclein activation of protein phosphatase 2A reduces tyrosine hydroxylase phosphorylation in dopaminergic cells. J. Cell Sci. 2005;118:3523–3530. doi: 10.1242/jcs.02481. [DOI] [PubMed] [Google Scholar]
  • 62.George RJ, Haycock JW, Johnston JP, Craviso GL, Waymire JC. In vitro phosphorylation of bovine adrenal chromaffin cell tyrosine hydroxylase by endogenous protein kinases. J. Neurochem. 1989;52(1):274–284. doi: 10.1111/j.1471-4159.1989.tb10928.x. [DOI] [PubMed] [Google Scholar]
  • 63.Kim SS, Moon KR, Choi HJ. Interference of alpha-synuclein with cAMP/PKA-dependent CREB signaling for tyrosine hydroxylase gene expression in SK-N-BE(2)C cells. Arch. Pharm. Res. 2011;34(5):837–845. doi: 10.1007/s12272-011-0518-0. [DOI] [PubMed] [Google Scholar]
  • 64.Perez RG, Hastings TG. Could a loss of alpha-synuclein function put dopaminergic neurons at risk? J. Neurochem. 2004;89:1318–1324. doi: 10.1111/j.1471-4159.2004.02423.x. [DOI] [PubMed] [Google Scholar]
  • 65.Mori F, Nishie M, Kakita A, Yoshimoto M, Takahashi H, Wakabayashi K. Relationship among alpha-synuclein accumulation, dopamine synthesis, and neurodegeneration in Parkinson disease substantia nigra. J. Neuropathol. Exp. Neurol. 2006;65(8):808–815. doi: 10.1097/01.jnen.0000230520.47768.1a. [DOI] [PubMed] [Google Scholar]
  • 66.Gao N, Li YH, Li X, Yu S, Fu GL, Chen B. Effect of alpha-synuclein on the promoter activity of tyrosine hydroxylase gene. Neurosci. Bull. 2007;23(1):53–57. doi: 10.1007/s12264-007-0008-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Liu D, Jin L, Wang H, Zhao H, Zhao C, Duan C, Lu L, Wu B, Yu S, Chan P, Li Y, Yang H. Silencing alpha-synuclein gene expression enhances tyrosine hydroxylase activity in MN9D cells. Neurochem. Res. 2008;33(7):1401–1409. doi: 10.1007/s11064-008-9599-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio G, Golbe LI, Nussbaum RL. Mutation in the synuclein gene identified in families with Parkinson's disease. Science. 1997;276:2045–2047. doi: 10.1126/science.276.5321.2045. [DOI] [PubMed] [Google Scholar]
  • 69.Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S, Przuntek H, Epplen JT, Schols L, Riess O. Ala30Pro mutation in the gene encoding synuclein in Parkinson's disease. Nat. Genet. 1998;18:106–108. doi: 10.1038/ng0298-106. [DOI] [PubMed] [Google Scholar]
  • 70.Neystat M, Lynch T, Przedborski S, Kholodilov N, Rzhetskaya M, Burke R. Synuclein expression in substantia nigra and cortex in Parkinson's disease. Mov. Disord. 1999;14:417–422. doi: 10.1002/1531-8257(199905)14:3<417::aid-mds1005>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
  • 71.Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A, Sagara Y, Sisk A, Mucke L. Dopaminergic loss and inclusion body formation in alpha-synuclein mice: implications for neurodegenerative disorders. Science. 2000;287:1265–1269. doi: 10.1126/science.287.5456.1265. [DOI] [PubMed] [Google Scholar]
  • 72.Conway KA, Lee SJ, Rochet JC, Ding TT, Harper JD, Williamson RE, Lansbury PT., Jr. Accelerated oligomerization by Parkinson's disease linked alpha-synuclein mutants. Acad. Sci. 2000;920:42–45. doi: 10.1111/j.1749-6632.2000.tb06903.x. [DOI] [PubMed] [Google Scholar]
  • 73.Kim TD, Paik SR, Yang CH, Kim J. Structural changes in alphasynuclein affect its chaperone-like activity in vitro. Prot. Sci. 2000;9:2489–2496. doi: 10.1110/ps.9.12.2489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Souza JM, Giasson BI, Lee VMY, Ischiropoulos H. Chaperonelike activity of synucleins. FEBS Lett. 2000;474:116–119. doi: 10.1016/s0014-5793(00)01563-5. [DOI] [PubMed] [Google Scholar]
  • 75.Ostrerova N, Petrucelli L, Farrer M, Mehta N, Choi P, Hardy J, Wolozin B. Synuclein shares physical and functional homology with 14-3-3. J. Neurosci. 1999;19:5782–5791. doi: 10.1523/JNEUROSCI.19-14-05782.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ichimura T, Isobe T, Okuyama T, Takahashi N, Araki K, Kuwano R, Takahashi Y. Molecular cloning of cDNA coding for brainspecific 14-3-3 protein, a protein kinase-dependent activator of tyrosine and tryptophan hydroxylases. Proc. Natl. Acad. Sci. USA. 1998;85:7084–7088. doi: 10.1073/pnas.85.19.7084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Muslin AJ, Xing H. 14-3-3 proteins: regulation of subcellular localization by molecular interference. Cell Signal. 2000;12:703–709. doi: 10.1016/s0898-6568(00)00131-5. [DOI] [PubMed] [Google Scholar]
  • 78.Kumer SC, Vrana KE. Intricate regulation of tyrosine hydroxylase activity and gene expression. J. Neurochem. 1996;67(2):443–462. doi: 10.1046/j.1471-4159.1996.67020443.x. [DOI] [PubMed] [Google Scholar]
  • 79.Zhang L, Wang H, Liu D, Liddington R, Fu H. Raf-1 kinase and exoenzyme S interact with 14-3-3 zeta through a common site involving lysine 49. J. Biol. Chem. 1997;272:13717–13724. doi: 10.1074/jbc.272.21.13717. [DOI] [PubMed] [Google Scholar]
  • 80.Kleppe R, Toska K, Haavik J. Interaction of phosphorylated tyrosine hydroxylase with 14-3-3 proteins: evidence for a phosphoserine 40-dependent association. J. Neurochem. 2001;77:1097–1107. doi: 10.1046/j.1471-4159.2001.00318.x. [DOI] [PubMed] [Google Scholar]
  • 81.Itagaki C, Isobe T, Taoka M, Natsume T, Nomura N, Horigome T, Omata S, Ichinose H, Nagatsu T, Greene LA, Ichimura T. Stimulus coupled interaction of tyrosine hydroxylase with 14-3-3 proteins. Biochemistry. 1999;38:15673–15680. doi: 10.1021/bi9914255. [DOI] [PubMed] [Google Scholar]
  • 82.Butterfield PG, Valanis BG, Spencer PS, Lindeman CA, Nutt JG. Environmental antecedents of young-onset Parkinson's disease. Neurology. 1993;43(6):1150–1158. doi: 10.1212/wnl.43.6.1150. [DOI] [PubMed] [Google Scholar]
  • 83.Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Kortsha GX, Brown GG, Richardson RJ. Occupational exposures to metals as risk factors for Parkinson's disease. Neurology. 1997;48(3):650–658. doi: 10.1212/wnl.48.3.650. [DOI] [PubMed] [Google Scholar]
  • 84.Menegon A, Board PG, Blackburn AC, Mellick GD, Fracp DGLC. Parkinson's disease, pesticides, and glutathione transferase polymorphisms. Lancet. 1998;352(9137):1344–1346. doi: 10.1016/s0140-6736(98)03453-9. [DOI] [PubMed] [Google Scholar]
  • 85.Ho SC, Woo J, Lee CM. Epidemiologic study of Parkinson's disease in Hong Kong. Neurology. 1989;39(10):1314–1318. doi: 10.1212/wnl.39.10.1314. [DOI] [PubMed] [Google Scholar]
  • 86.Cranmer JM. Parkinson's disease, environment and genes. Neurotoxicology. 2001;22(6):829–832. doi: 10.1016/s0161-813x(01)00091-2. [DOI] [PubMed] [Google Scholar]
  • 87.Di Monte DA, Lavasani M, Manning-Bog AB. Environmental factors in Parkinson's disease. Neurotoxicology. 2002;23(4-5):487–502. doi: 10.1016/s0161-813x(02)00099-2. [DOI] [PubMed] [Google Scholar]
  • 88.Langston JW. Parkinson's disease: current and future challenges. Neurotoxicology. 2002;23(4-5):443–450. doi: 10.1016/s0161-813x(02)00098-0. [DOI] [PubMed] [Google Scholar]
  • 89.Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat. Neurosci. 2000;3(12):1301–1306. doi: 10.1038/81834. [DOI] [PubMed] [Google Scholar]
  • 90.Costello S, Cockburn M, Bronstein J, Zhang X, Ritz B. Parkinson's disease and residential exposure to maneb and paraquat from agricultural applications in the central valley of California. Am. J. Epidemiol. 2009;169(8):919–926. doi: 10.1093/aje/kwp006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Firestone JA, Smith-Weller T, Franklin G, Swanson P, Longstreth WT, Jr., Checkoway H. Pesticides and risk of Parkinson disease: a population-based case-control study. Arch. Neurol. 2005;62(1):91–95. doi: 10.1001/archneur.62.1.91. [DOI] [PubMed] [Google Scholar]
  • 92.Thiruchelvam M, Richfield EK, Goodman BM, Baggs RB, Dory-Slechta DA. Developmental exposure to the pesticides paraquat and maneb and the Parkinson's disease phenotype. Neurotoxicology. 2002;23(4-5):621–633. doi: 10.1016/s0161-813x(02)00092-x. [DOI] [PubMed] [Google Scholar]
  • 93.Kirby ML, Barlow RL, Bloomquist JR. Neurotoxicity of the organochlorine insecticide heptachlor to murine striatal dopaminergic pathways. Toxicol. Sci. 2001;61(1):100–106. doi: 10.1093/toxsci/61.1.100. [DOI] [PubMed] [Google Scholar]
  • 94.Thrash B, Uthayathas S, Karuppagounder SS, Suppiramaniam V, Dhanasekaran M. Paraquat and maneb induced neurotoxicity. Proc. West Pharmacol. Soc. 2007;50:31–42. [PubMed] [Google Scholar]
  • 95.Li X, Yin J, Cheng CM, Sun JL, Li Z, Wu YL. Paraquat induces selective dopaminergic nigrostriatal degeneration in aging C57BL/6 mice. Chin. Med. J. (Engl) 2005;118(16):1357–1361. [PubMed] [Google Scholar]
  • 96.Brooks AI, Chadwick CA, Gelbard HA, Cory-Slechta DA, Federoff HJ. Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res. 1999;823(1-2):1–10. doi: 10.1016/s0006-8993(98)01192-5. [DOI] [PubMed] [Google Scholar]
  • 97.Gong-Ping L, Qiang MA, Nian S. Tyrosine hydroxylase as a target for deltamethrin in the nigrostriatal dopaminergic pathway1. Biomed. Env. Sci. 2006;19:27–34. [PubMed] [Google Scholar]
  • 98.Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van Duijn CM, Oostra BA, Heutink P. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science. 2003;299:256–259. doi: 10.1126/science.1077209. [DOI] [PubMed] [Google Scholar]
  • 99.Mouradian MM. Recent advances in the genetics and pathogenesis of Parkinson disease. Neurology. 2002;58:179–185. doi: 10.1212/wnl.58.2.179. [DOI] [PubMed] [Google Scholar]
  • 100.Maimone D, Dominici R, Grimaldi LM. Pharmacogenomics of neurodegenerative diseases. Eur. J. Pharmacol. 2001;413:11–29. doi: 10.1016/s0014-2999(00)00939-0. [DOI] [PubMed] [Google Scholar]
  • 101.Zettergren A. Ph.D Thesis. University of Gothenburg; Sweden: 2010. Genetics of parkinson's disease with focus on genes of relevance for inflammation and dopamine neuron development. ISBN 978-91-628-8022-4. [Google Scholar]
  • 102.Tan EK, Khajavi M, Thornby JI, Nagamitsu S, Jankovic J, Ashizawa T. Variability and validity of polymorphism association studies in Parkinson's disease. Neurology. 2000;55:533–538. doi: 10.1212/wnl.55.4.533. [DOI] [PubMed] [Google Scholar]
  • 103.Lee CGL, Tang K, Cheung YB, Wong LP, Tan C, Shen H, Zhao Y, Pavanni R, Lee EJD, Wong MC, Chong J. Med. Genet. 2004;41:e60. doi: 10.1136/jmg.2003.013003. http://jmg.bmj.com/content/41/5/e60.full - aff-6 S.S.; Tan, E.K. MDR1, the blood-brain barrier transporter, is associated with Parkinson's disease in ethnic Chinese. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Jingzhong Z, Hui Y, Deyi D, Chunli D, Chunli Z, Xiaohong S, Qunyuan X. Long-term therapeutic effects on parkinsonian rats of intrastriatal co-grafts with genetically engineered fibroblasts expressing tyrosine hydroxylase and glial cell line-derived neurotrophic factor. Int. J. Neurosci. 2005;115:769–779. doi: 10.1080/00207450590881542. [DOI] [PubMed] [Google Scholar]
  • 105.Dunnett SB, Björklund A. Prospects for new restorative and neuroprotective treatments in Parkinson's disease. Nature. 1999;399(6738):A32–A39. doi: 10.1038/399a032. [DOI] [PubMed] [Google Scholar]
  • 106.Calon F, Grondin R, Morissette M, Goulet M, Blanchet PJ, Di Paolo T, Bedard PJ. Molecular basis of levodopa-induced dyskinesias. Ann. Neurol. 2000;47(4):70–78. [PubMed] [Google Scholar]
  • 107.Schapira AH, Bezard E, Brotchie J, Calon F, Collingridge GL, Ferger B, Hengerer B, Hirsch E, Jenner P, Le Novère N, Obeso JA, Schwarzschild MA, Spampinato U, Davidai G. Novel pharmacological targets for the treatment of Parkinson's disease. Nat. Rev. Drug. Discov. 2006;5(10):845–854. doi: 10.1038/nrd2087. [DOI] [PubMed] [Google Scholar]
  • 108.Marsden CD. Problems with long-term levodopa therapy for Parkinson's disease. Clin. Neuropharmacol. 1994;17(2):S32–S44. [PubMed] [Google Scholar]
  • 109.Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov. Disord. 2001;16(3):448–458. doi: 10.1002/mds.1090. [DOI] [PubMed] [Google Scholar]
  • 110.Burton EA, Glorioso JC, Fink DJ. Gene therapy progress and prospects: Parkinson's disease. Gene Ther. 2003;10:1721–1727. doi: 10.1038/sj.gt.3302116. [DOI] [PubMed] [Google Scholar]
  • 111.Manuchair E, Shashi K, Srinivasan Mayur DB. Oxidative stress and antioxidant therapy in parkinson's disease. Prog. Neurobiol. 1996;48:1–19. doi: 10.1016/0301-0082(95)00029-1. [DOI] [PubMed] [Google Scholar]
  • 112.Gupta A, Dawson TM. The role of stem cells in Parkinson's disease. Neurosurg. Clin. N. Am. 2007;18(1):129–142. doi: 10.1016/j.nec.2006.10.015. [DOI] [PubMed] [Google Scholar]
  • 113.Rangasamy SB, Soderstrom K, Bakay RA, Kordower JH. Neurotrophic factor therapy for Parkinson's disease. Prog. Brain Res. 2010;184:237–264. doi: 10.1016/S0079-6123(10)84013-0. [DOI] [PubMed] [Google Scholar]
  • 114.Wijeyekoon R, Barker RA. Cell replacement therapy for Parkinson's disease. Biochim. Biophys. Acta. 2009;1792(7):688–702. doi: 10.1016/j.bbadis.2008.10.007. [DOI] [PubMed] [Google Scholar]
  • 115.Cooper JR, Bloom FE, Roth RH. The Biochemical basis of Neuropharmacology. Oxford University Press; New York: 1991. pp. 285–337. [Google Scholar]
  • 116.Lewitt PA, Rezai AR, Leehey MA, Ojemann SG, Flaherty AW, Eskandar EN, Kostyk SK, Thomas K, Sarkar A, Siddiqui MS, Tatter SB, Schwalb JM, Poston KL, Henderson JM, Kurlan RM, Richard IH, Van Meter L, Sapan CV, During MJ, Kaplitt MG, Feigin A. AAV2-GAD gene therapy for advanced Parkinson's disease: a doubleblind, sham-surgery controlled, randomised trial. Lancet Neurol. 2011;10(4):309–319. doi: 10.1016/S1474-4422(11)70039-4. [DOI] [PubMed] [Google Scholar]
  • 117.Marks WJ, Bartus RT, Siffert J, Davis CS, Lozano A, Boulis N, Vitek J, Stacy M, Turner D, Verhagen L, Bakay R, Watts R, Guthrie B, Jankovic J, Simpson R, Tagliati M, Alterman R, Stern M, Baltuch G, Starr PA, Larson PS, Ostrem JL, Nutt J, Kieburtz K, Kordower JH, Olanow CW. Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial. Lancet Neurol. 2010;9(12):1164–1172. doi: 10.1016/S1474-4422(10)70254-4. [DOI] [PubMed] [Google Scholar]
  • 118.Rodnitzky RL. Upcoming treatments in Parkinson's disease, including gene therapy. Parkinsonism. Relat. Disord. 2012;18(1):37–40. doi: 10.1016/S1353-8020(11)70014-1. [DOI] [PubMed] [Google Scholar]
  • 119.Muramatsu S, Fujimoto K, Kato S, Mizukami H, Asari S, Ikeguchi K, Kawakami T, Urabe M, Kume A, Sato T, Watanabe E, Ozawa K, Nakano I. A phase I study of aromatic L-amino acid decarboxylase gene therapy for Parkinson's disease. Mol. Ther. 2010;18:1731–1735. doi: 10.1038/mt.2010.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA, Grünblatt E, Moran LB, Mandel SA, Riederer P, Miller RM, Federoff HJ, Wüllner U, Papapetropoulos S, Youdim MB, Cantuti-Castelvetri I, Young AB, Vance JM, Davis RL, Hedreen JC, Adler CH, Beach TG, Graeber MB, Middleton FA, Rochet JC, Scherzer CR, Global PD, Gene Expression (GPEX) Consortium PGC-1alpha, a potential therapeutic target for early intervention in Parkinson's disease. Sci. Transl. Med. 2010;2(52):52–73. doi: 10.1126/scitranslmed.3001059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Jiao S, Cheng L, Wolff J, Yang NS. Particle bombardment-mediated gene transfer and expression in rat brain tissues. Biotechnology. 1993;11:497–502. doi: 10.1038/nbt0493-497. [DOI] [PubMed] [Google Scholar]
  • 122.Cheng L, Ziegelhoffer P, Yang NS. In vivo promoter activity and transgene expression in mammalian somatic tissues evaluated by using particle bombardment. Proc. Natl. Acad. Sci. USA. 1993;90:4455–4459. doi: 10.1073/pnas.90.10.4455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Roessler B, Davidson B. Direct plasmid mediated transfection of adult murine brain cells in vivo using cationic liposomes. Neurosci. Lett. 1994;167:5–10. doi: 10.1016/0304-3940(94)91015-4. [DOI] [PubMed] [Google Scholar]
  • 124.Emborga ME, Deglonb N, Leventhala L, Aebischerb PC, Kordowera JH. Viral vector-mediated gene therapy for Parkinson's disease. Clin. Neurosci. Res. 2001;1(6):496–506. [Google Scholar]
  • 125.Kaplitt MG, Leone P, Samulski RJ, Xiao X, Pfaff DW, O'Malley KL, During MJ. Long-term gene expression and phenotypic correction using adeno-associated virus vectors in the mammalian brain. Nat. Genet. 1994;8:148–154. doi: 10.1038/ng1094-148. [DOI] [PubMed] [Google Scholar]
  • 126.During MJ, Naegele JR, Geller AI. Long-term behavioral recovery in parkinsonian rats by an HSV vector expressing tyrosine hydroxylase. Science. 1994;266:1399–1403. doi: 10.1126/science.266.5189.1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Mandel RJ, Rendahl KG, Spratt SK, Snyder RO, Cohen LK, Leff SE. Characterization of intrastriatal recombinant adeno-associated virus-mediated gene transfer of human tyrosine hydroxylase and human GTP-cyclohydrolase I in a rat model of Parkinson's disease. J. Neurosci. 1998;18:4271–4284. doi: 10.1523/JNEUROSCI.18-11-04271.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Fan DS, Ogawa M, Fujimoto KI, Ikeguchi K, Ogasawara Y, Urabe M, Nishizawa M, Nakano I, Yoshida M, Nagatsu I, Ichinose H, Nagatsu T, Kurtzman GJ, Ozawa K. Behavioral recovery in 6- hydroxydopamine-lesioned rats by cotransduction of striatum with tyrosine hydroxylase and aromatic l-amino acid decarboxylase genes using two separate adeno-associated virus vectors. Hum. Gene Ther. 1998;9:2527–2535. doi: 10.1089/hum.1998.9.17-2527. [DOI] [PubMed] [Google Scholar]
  • 129.Shen Y, Muramatsu SI, Ikeguchi K, Fujimoto KI, Fan DS, Ogawa M, Mizukami H, Urabe M, Kume A, Nagatsu I, Urano F, Suzuki T, Ichinose H, Nagatsu T, Monahan J, Nakano I, Ozawa K. Triple transduction with adeno-associated virus vectors expressing tyrosine hydroxylase, aromatic-l-amino-acid decarboxylase, and GTP cyclohydrolase I for gene therapy of Parkinson's disease. Hum. Gene Ther. 2000;11:1509–1519. doi: 10.1089/10430340050083243. [DOI] [PubMed] [Google Scholar]
  • 130.Fisher LJ, Jinnah HA, Kale LC, Gage FH. Survival and function of intrastriatally grafted primary fibroblasts genetically modified to produce L-dopa. Neuron. 1991;6:371–380. doi: 10.1016/0896-6273(91)90246-v. [DOI] [PubMed] [Google Scholar]
  • 131.Guerrero-Cazares H, Alatorre-Carranza MP, Delgado-Rizo V, Duenas-Jimenez JM, Mendoza-Magana ML, Morales-Villagran A, Ramirez-Herrera MA, Guerrero-Hernández A, Segovia J, Duenas-Jimenez SH. Dopamine release modifies intracellular calcium levels in tyrosine hydroxylase-transfected C6 cells. Brain Res. Bull. 2007;74(1-3):113–118. doi: 10.1016/j.brainresbull.2007.05.008. [DOI] [PubMed] [Google Scholar]
  • 132.Lundberg C, Horellou P, Bjorklund A. Generation of DOPA producing astrocytes by retroviral transduction of the human tyrosine hydroxylase gene: in vitro characterization and in vivo effects in the rat Parkinson model. Exp. Neurol. 1996;139:39–53. doi: 10.1006/exnr.1996.0079. [DOI] [PubMed] [Google Scholar]
  • 133.Zhang S, Zou Z, Jiang X, Xu R, Zhang W, Ke Y. The therapeutic effects of tyrosine hydroxylase gene transfected hematopoetic stem cells in a rat model of Parkinson's disease. Cell Mol. Neurobiol. 2008;28:529–543. doi: 10.1007/s10571-007-9191-8. [DOI] [PubMed] [Google Scholar]
  • 134.Ishida A, Yasuzumi F. Approach to ex vivo gene therapy in the treatment of Parkinson's disease. Brain Dev. 2000;22:143–147. doi: 10.1016/s0387-7604(00)00138-8. [DOI] [PubMed] [Google Scholar]
  • 135.McCoy MK, Ruhn KA, Martinez TN, McAlpine FE, Tansey MG. Intranigral lentiviral delivery of dominant-negative TNF attenuates neurodegeneration and behavioral deficits in hemiparkinsonian rats. Mol. Ther. 2008;16:1572–1579. doi: 10.1038/mt.2008.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Park HJ, Lee PH, Bang OY, Ahn YH. Mesenchymal stem cells therapy exerts neuroprotection in a progressive animal model of Parkinson's disease. J. Neurochem. 2008;107:141–151. doi: 10.1111/j.1471-4159.2008.05589.x. [DOI] [PubMed] [Google Scholar]
  • 137.Chou J, Greig NH, Reiner D, Hoffer BJ, Wang Y. Enhanced survival of dopaminergic neuronal transplants in hemiparkinsonian rats by the p53 inactivator PFT-α. Cell Transplant. 2011;20:1351–1359. doi: 10.3727/096368910X557173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Przedborski S, Jackson-Lewis V, Naini AB, Jakowec M, Petzinger G, Miller R, Akram M. The parkinsonian toxin 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine(MPTP): a technical review of its utility and safety. J. Neurochem. 2001;76(5):1265–1274. doi: 10.1046/j.1471-4159.2001.00183.x. [DOI] [PubMed] [Google Scholar]
  • 139.Przedborski S, Jackson-Lewis V. Mechanisms of MPTP toxicity. Mov. Disord. 1998;13:35–38. [PubMed] [Google Scholar]
  • 140.Chung YC, Kim SR, Park JY, Chung ES, Park KW, Won SY, Bok E, Jin M, Park ES, Yoon SH, Ko HW, Kim YS, Jin BK. Fluoxetine prevents MPTP-induced loss of dopaminergic neurons by inhibiting micriglial activation. Neuropharmacology. 2011;60(6):963–974. doi: 10.1016/j.neuropharm.2011.01.043. [DOI] [PubMed] [Google Scholar]
  • 141.Huh SH, Chung YC, Piao Y, Jin MY, Son HJ, Yoon NS, Hong JY, Pak YK, Kim YS, Hong JK, Hwang O, Jin BK. Ethyl pyruvate rescues nigrostriatal dopaminergic neurons by regulating glial activation in a mouse model of Parkinson's disease. J. Immunol. 2011;187(2):960–969. doi: 10.4049/jimmunol.1100009. [DOI] [PubMed] [Google Scholar]
  • 142.Chung YC, Kim SR, Jin BK. Paroxetine Prevents loss of nigrostriatal dopaminergic neurons by inhibiting brain inflammation and oxidative stress in an experimental model of parkinson's disease. J. Immunol. 2010;185:1230–1237. doi: 10.4049/jimmunol.1000208. [DOI] [PubMed] [Google Scholar]
  • 143.Schapira AH. Evidence for mitochondrial dysfunction in Parkinson's disease - a critical appraisal. Movem. Disord. 1994;9:125–138. doi: 10.1002/mds.870090202. [DOI] [PubMed] [Google Scholar]
  • 144.Haas RH, Nasirian F, Nakano K, Ward D, Pay M, Hill R, Shults CW. Low platelet mitochondrial complex I and complex II/III activity in early untreated Parkinson's disease. Ann. Neurol. 1995;37:714–722. doi: 10.1002/ana.410370604. [DOI] [PubMed] [Google Scholar]
  • 145.Martin HL, Teismann P. Glutathione-a review on its role and significance in Parkinson's disease. FASEB. J. 2009;23(10):3263–3272. doi: 10.1096/fj.08-125443. [DOI] [PubMed] [Google Scholar]
  • 146.Banerjee R, Starkov AA, Beal MF, Thomas B. Mitochondrial dysfunction in the limelight of Parkinson's disease pathogenesis. Biochim. Biophys. Acta. 2009;1792:651–663. doi: 10.1016/j.bbadis.2008.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Hastings TG, Zigmond MJ. Loss of dopaminergic neurons in parkinsonism: possible role of reactive dopamine metabolites. J. Neural Transm. 1997;49:103–110. doi: 10.1007/978-3-7091-6844-8_11. [DOI] [PubMed] [Google Scholar]
  • 148.Halliwell B. Reactive oxygen species and the central nervous system. J. Neurochem. 1992;59:1609–1623. doi: 10.1111/j.1471-4159.1992.tb10990.x. [DOI] [PubMed] [Google Scholar]
  • 149.Miyazaki I, Asanuma M. Dopaminergic neuron-specific oxidative stress caused by dopamine itself. Acta. Med. Okayama. 2008;62:141–150. doi: 10.18926/AMO/30942. [DOI] [PubMed] [Google Scholar]
  • 150.Calabrese EJ. Pharmacological enhancement of neuronal survival. Crit. Rev. Toxicol. 2008;38:349–389. doi: 10.1080/10408440801981973. [DOI] [PubMed] [Google Scholar]
  • 151.Otani H. Reactive oxygen species as mediators of signal transduction in ischemic preconditioning. Antioxid. Redox. Signal. 2004;6:449–469. doi: 10.1089/152308604322899521. [DOI] [PubMed] [Google Scholar]
  • 152.Jia Z, Zhu H, Misra BR, Misra HP. Dopamine as a potent inducer of cellular glutathione and NAD(P)H:quinone oxidoreductase 1 in PC12 neuronal cells: a potential adaptive mechanism for dopaminergic neuroprotection. Neurochem. Res. 2008;33:2197–2205. doi: 10.1007/s11064-008-9670-4. [DOI] [PubMed] [Google Scholar]
  • 153.Lazou A, Markou T, Zioga M, Vasara E, Gaitanaki C. Dopamine mimics the cardioprotective effect of ischemic preconditioning via activation of alpha1-adrenoceptors in the isolated rat heart. Physiol. Res. 2006;55:1–8. doi: 10.33549/physiolres.930694. [DOI] [PubMed] [Google Scholar]
  • 154.Shih AY, Murphy TH. Dopamine activates Nrf2-regulated neuroprotective pathways in astrocytes and meningeal cells. J. Neurochem. 2007;101:109–119. doi: 10.1111/j.1471-4159.2006.04345.x. [DOI] [PubMed] [Google Scholar]
  • 155.Franco JL, Posser T, Gordon SL, Bobrovskaya L, Schneider JJ, Farina M, Dafre AL, Dickson PW, Dunkley PR. Expression of tyrosine hydroxylase increases the resistance of human neuroblastoma cells to oxidative insults. Toxicol. Sci. 2010;113(1):150–157. doi: 10.1093/toxsci/kfp245. [DOI] [PubMed] [Google Scholar]
  • 156.McCormack AL, Atienza JG, Langston JW, Di Monte DA. Decreased susceptibility to oxidative stress underlies the resistance of specific dopaminergic cell populations to paraquat-induced degeneration. Neuroscience. 2006;141:929–937. doi: 10.1016/j.neuroscience.2006.03.069. [DOI] [PubMed] [Google Scholar]
  • 157.Olanow CW, Obeso JA. Preventing levodopa-induced dyskinesias. Ann. Neurol. 2000;47:167–176. [PubMed] [Google Scholar]
  • 158.Freed CR, Breeze RE, Rosenberg NL. Transplantation of human fetal dopamine cells for Parkinson's disease: results at 1 year. Arch. Neurol. 1990;47:505–512. doi: 10.1001/archneur.1990.00530050021007. [DOI] [PubMed] [Google Scholar]
  • 159.Freed CR, Greene PE, Breeze RE. Transplantation of embryonic dopamine neurons for severe Parkinson's disease. N. Engl. J. Med. 2001;344:710–719. doi: 10.1056/NEJM200103083441002. [DOI] [PubMed] [Google Scholar]
  • 160.Kimberley A, Buytaert-Hoefen E, Alvarez, Curt RF. Generation of Tyrosine Hydroxylase Positive Neurons from Human Embryonic Stem Cells after Coculture with Cellular Substrates and Exposure to GDNF. Stem Cells. 2004;22:669–674. doi: 10.1634/stemcells.22-5-669. [DOI] [PubMed] [Google Scholar]
  • 161.Bjorklund LM, Sanchez-Pernaute R, Chung S. Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model. Proc. Natl. Acad. Sci. USA. 2002;99:2344–2349. doi: 10.1073/pnas.022438099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Deacon T, Dinsmore J, Costantini LC. Blastula-stage stem cells can differentiate into dopaminergic and serotonergic neurons after transplantation. Exp. Neurol. 1998;149:28–41. doi: 10.1006/exnr.1997.6674. [DOI] [PubMed] [Google Scholar]
  • 163.Kawasaki H, Mizuseki K, Nishikawa S. Induction of midbrain dopaminergic neurons from ES cells by stromal cell-derived inducing activity. Neuron. 2000;28:31–40. doi: 10.1016/s0896-6273(00)00083-0. [DOI] [PubMed] [Google Scholar]
  • 164.Kawasaki H, Suemori H, Mizuseki K. Generation of dopaminergic neurons and pigmented epithelia from primate ES cells by stromal cell-derived inducing activity. Proc. Natl. Acad. Sci. USA. 2002;99:1580–1585. doi: 10.1073/pnas.032662199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Zou Z, Jiang X, Zhang W, Zhou Y, Ke Y, Zhang S, Xu R. Efficacy of Tyrosine Hydroxylase gene modified neural stem cells derived from bone marrow on Parkinson's disease a rat model study. Brain Res. 2010;1346:279–286. doi: 10.1016/j.brainres.2010.05.071. [DOI] [PubMed] [Google Scholar]
  • 166.Kirik D, Georgievska B, Bjorklund A. Localized striatal delivery of GDNF as a treatment for Parkinson disease. Nat. Neurosci. 2004;7:105–110. doi: 10.1038/nn1175. [DOI] [PubMed] [Google Scholar]
  • 167.Lang AE, Gill S, Patel NK, Lozano A, Nutt JG, Penn R, Brooks DJ, Hotton G, Moro E, Heywood P. Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann. Neurol. 2006;59:459–466. doi: 10.1002/ana.20737. [DOI] [PubMed] [Google Scholar]
  • 168.Pascual A, Hidalgo FM, Gomez DR, Lopez BJ. GDNF and protection of adult central catecholaminergic neurons. J. Mol. Endocrinol. 2011;46(3):R83–R92. doi: 10.1530/JME-10-0125. [DOI] [PubMed] [Google Scholar]
  • 169.Ibanez CF. Message in a bottle: long-range retrograde signaling in the nervous system. Trend. Cell Biol. 2007;17:519–528. doi: 10.1016/j.tcb.2007.09.003. [DOI] [PubMed] [Google Scholar]
  • 170.Lin LF, Doherty DH, Lile JD, Bektesh S, Collins F. GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science. 1993;260:1130–1132. doi: 10.1126/science.8493557. [DOI] [PubMed] [Google Scholar]
  • 171.Pascual A, Hidalgo-Figueroa M, Piruat JI, Pintado CO, Gomez-Diaz R, Lopez-Barneo J. Absolute requirement of GDNF for adult catecholaminergic neuron survival. Nat. Neurosci. 2008;11:755–761. doi: 10.1038/nn.2136. [DOI] [PubMed] [Google Scholar]
  • 172.Tomac A, Lindqvist E, Lin LF, Ogren SO, Young D, Hoffer BJ, Olson L. Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo. Nature. 1995;373:335–339. doi: 10.1038/373335a0. [DOI] [PubMed] [Google Scholar]
  • 173.Gill SS, Patel NK, Hotton GR, O'Sullivan K, McCarter R, Bunnage M, Brooks DJ, Svendsen CN, Heywood P. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat. Med. 2003;9:589–595. doi: 10.1038/nm850. [DOI] [PubMed] [Google Scholar]
  • 174.Vastag B. Biotechnology: crossing the barrier. Nature. 2010;466:916–918. doi: 10.1038/466916a. [DOI] [PubMed] [Google Scholar]
  • 175.Chadi G, Moller A, Rosen L, Janson AM, Agnati LA, Goldstein M, Ogren SO, Pettersson RF, Fuxe K. Protective actions of human recombinant basic fibroblast growth factor on MPTP-lesioned nigrostriatal dopamine neurons after intraventricular infusion. Exp. Brain Res. 1993;97(1):145–158. doi: 10.1007/BF00228825. [DOI] [PubMed] [Google Scholar]
  • 176.Duobles T, Lima TS, Levy BF, Chadi GS. 100 beta and fibroblast growth factor-2 are present in cultured Schwann cells and may exert paracrine actions on the peripheral nerve injury. Acta Cir. Braz. 2008;23(6):555–560. doi: 10.1590/s0102-86502008000600014. [DOI] [PubMed] [Google Scholar]
  • 177.Grothe C, Haastert K, Jungnickel J. Physiological function and putative therapeutic impact of the FGF-2 system in peripheral nerve regeneration-lessons from in vivo studies in mice and rats. Brain Res. Rev. 2006;51(2):293–299. doi: 10.1016/j.brainresrev.2005.12.001. [DOI] [PubMed] [Google Scholar]
  • 178.Mattson MP. Glutamate and neurotrophic factors in neuronal plasticity and disease. Ann. NY Acad. Sci. 2008;1144:97–112. doi: 10.1196/annals.1418.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Xu G, Xiong Z, Yong Y, Wang Z, Ke Z, Xia Z, Hu Y. Catalpol attenuates MPTP induced neuronal degeneration of nigralstriatal dopaminergic pathway in mice through elevating glial cell derived neurotrophic factor in striatum. Neuroscience. 2010;167(1):174–184. doi: 10.1016/j.neuroscience.2010.01.048. [DOI] [PubMed] [Google Scholar]
  • 180.Wang Z, Liu Q, Zhang R, Liu S, Xia Z, Hu Y. Catalpol ameliorates beta amyloid-induced degeneration of cholinergic neurons by elevating brain-derived neurotrophic factors. Neuroscience. 2009;163(4):1363–1372. doi: 10.1016/j.neuroscience.2009.07.041. [DOI] [PubMed] [Google Scholar]
  • 181.Airavaara M, Harvey BK, Voutilainen MH, Shen H, Chou J, Lindholm P, Lindahl M, Tuominen RK, Saarma M, Wang Y, Hoffer B. CDNF protects the nigrostriatal DA system and promotes recovery after MPTP treatment in mice. Cell Transplant. 2011 doi: 10.3727/096368911X600948. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Lindholm P, Voutilainen MH, Laurén J, Peranen J, Leppanen VM, Andressoo JO, Lindahl M, Janhunen S, Kalkkinen N, Timmusk T, Tuominen RK, Saarma M. Novel neurotrophic factor CDNF protects and rescues midbrain dopamine neurons in vivo. Nature. 2007;448(7149):73–77. doi: 10.1038/nature05957. [DOI] [PubMed] [Google Scholar]
  • 183.Voutilainen MH, Back S, Peranen J, Lindholm P, Raasmaja A, Mannisto PT, Saarma M, Tuominen RK. Chronic infusion of CDNF prevents 6-OHDA-induced deficits in a rat model of Parkinson's disease. Exp. Neurol. 2011;228(1):99–108. doi: 10.1016/j.expneurol.2010.12.013. [DOI] [PubMed] [Google Scholar]
  • 184.Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132:2131–2157. doi: 10.1053/j.gastro.2007.03.054. [DOI] [PubMed] [Google Scholar]
  • 185.Garber AJ. Novel GLP-1 receptor agonists for diabetes. Expert Opin. Investig. Drugs. 2012;21(1):45–57. doi: 10.1517/13543784.2012.638282. [DOI] [PubMed] [Google Scholar]
  • 186.Shyangdan DS, Royle P, Clar C, Sharma P, Waugh N, Snaith A. Glucagon-like peptide analogues for type 2 diabetes mellitus. Cochrane Database Syst. Rev. 2011;(10):CD006423. doi: 10.1002/14651858.CD006423.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Craft S, Watson GS. Insulin and neurodegenerative disease: Shared and specific mechanisms. Lancet. 2004;3:169–178. doi: 10.1016/S1474-4422(04)00681-7. [DOI] [PubMed] [Google Scholar]
  • 188.Perry T, Greig NH. The glucagon-like peptides: A double-edged therapeutic sword? Trends Pharmacol. Sci. 2003;24:377–83. doi: 10.1016/S0165-6147(03)00160-3. [DOI] [PubMed] [Google Scholar]
  • 189.Perry T, Lahiri DK, Chen D, Zhou J, Shaw KT, Egan JM, Greig NH. A novel neurotrophic property of glucagon-like peptide 1: A promoter of nerve growth factor–mediated differentiation in PC12 cells. J. Pharmacol. Exp. Ther. 2002;300:958–966. doi: 10.1124/jpet.300.3.958. [DOI] [PubMed] [Google Scholar]
  • 190.Perry T, Haughey NJ, Mattson MP, Egan JM, Greig NH. Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin-4. J. Pharmacol. Exp. Ther. 2002;302:881–888. doi: 10.1124/jpet.102.037481. [DOI] [PubMed] [Google Scholar]
  • 191.Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, Powers K, Shen H, Egan JM, Sambamurti K, Brossi A, Lahiri DK, Mattson MP, Hoffer BJ, Wang Y, Greig NH. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc. Natl. Acad. Sci. USA. 2009;106:1285–1290. doi: 10.1073/pnas.0806720106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Li Y, Tweedie D, Mattson MP, Holloway HW, Greig NH. Enhancing the GLP-1 receptor signaling pathway leads to proliferation and neuroprotection in human neuroblastoma cells. J. Neurochem. 2010;113:1621–1631. doi: 10.1111/j.1471-4159.2010.06731.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Harkavyi A, Abuirmeileh A, Lever R, Kingsbury AE, Biggs CS, Whitton PS. Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson's disease. J. Neuroinflammation. 2008;5:19. doi: 10.1186/1742-2094-5-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Kim S, Moon M, Park S. Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson's disease. J. Endocrinol. 2009;202:431–439. doi: 10.1677/JOE-09-0132. [DOI] [PubMed] [Google Scholar]
  • 195.Bertilsson G, Patrone C, Zachrisson O, Andersson A, Dannaeus K, Heidrich J, Kortesmaa J, Mercer A, Nielsen E, Rönnholm H, Wikström L. Peptide hormone exendin-4 stimulates subventricular zone neurogenesis in the adult rodent brain and induces recovery in an animal model of Parkinson's disease. J. Neurosci. Res. 2008;86:326–338. doi: 10.1002/jnr.21483. [DOI] [PubMed] [Google Scholar]
  • 196.Li Y, Duffy K, Ottinger MA, Ray B, Bailey JA, Holloway HW, Tweedie D, Perry T, Mattson MP, Kapogiannis D, Sambamurti K, Lahiri DK, Greig NH. GLP-1 receptor stimulation reduces amyloid-beta peptide accumulation and cytotoxicity in cellular and animal models of Alzheimer's disease. J. Alzheimers Dis. 2010;19:1205–1219. doi: 10.3233/JAD-2010-1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Perry T, Holloway HW, Weerasuriya A, Mouton PR, Duffy K, Mattison JA, Greig NH. Evidence of GLP-1-mediated neuroprotection in an animal model of pyridoxine-induced peripheral sensory neuropathy. Exp. Neurol. 2007;203:293–301. doi: 10.1016/j.expneurol.2006.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Perry T, Lahiri DK, Sambamurti K, Chen D, Mattson MP, Egan JM, Greig NH. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. J. Neurosci. Res. 2003;72:603–612. doi: 10.1002/jnr.10611. [DOI] [PubMed] [Google Scholar]
  • 199.McClean PL, Parthsarathy V, Faivre E, Hölscher C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer's disease. J. Neurosci. 2011;31(17):6587–6594. doi: 10.1523/JNEUROSCI.0529-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Martin B, Golden E, Carlson OD, Pistell P, Zhou J, Kim W, Frank BP, Thomas S, Chadwick WA, Greig NH, Bates GP, Sathasivam K, Bernier M, Maudsley S, Mattson MP, Egan JM. Exendin-4 improves glycemic control, ameliorates brain and pancreatic pathologies, and extends survival in a mouse model of Huntington's disease. Diabetes. 2009;58(2):318–328. doi: 10.2337/db08-0799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Hölscher C. The role of GLP-1 in neuronal activity and neurodegeneration. Vitam. Horm. 2010;84:331–354. doi: 10.1016/B978-0-12-381517-0.00013-8. [DOI] [PubMed] [Google Scholar]
  • 202.Salcedo I, Tweedie D, Li Y, Greig NH. Glucagon-like peptide-1: an emerging opportunity to treat neurodegenerative disorders. Br. J. Pharmacol. 2012 doi: 10.1111/j.1476-5381.2012.01971.x. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Carta AR, Pisanu A, Carboni E. Do PPAR-Gamma Agonists Have a Future in Parkinson's Disease Therapy? Parkinsons Dis. 2011;2011:689181. doi: 10.4061/2011/689181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Schintu N, Frau L, Ibba M, Caboni P, Garau A, Carboni E, Carta AR. PPAR-gamma-mediated neuroprotection in a chronic mouse model of Parkinson's disease. Eur. J. Neurosci. 2009;29(5):954–963. doi: 10.1111/j.1460-9568.2009.06657.x. [DOI] [PubMed] [Google Scholar]
  • 205.Carta AR, Frau L, Pisanu A, Wardas J, Spiga S, Carboni E. Rosiglitazone decreases peroxisome proliferator receptor-gamma levels in microglia and inhibits TNF-alpha production: new evidences on neuroprotection in a progressive Parkinson's disease model. Neuroscience. 2011;194:250–261. doi: 10.1016/j.neuroscience.2011.07.046. [DOI] [PubMed] [Google Scholar]
  • 206.Germino FW. Noninsulin treatment of type 2 diabetes mellitus in geriatric patients: a review. Clin. Ther. 2011;33(12):1868–1882. doi: 10.1016/j.clinthera.2011.10.020. [DOI] [PubMed] [Google Scholar]
  • 207.Cheung BM. Behind the rosiglitazone controversy. Expert Rev. Clin. Pharmacol. 2010;3(6):723–725. doi: 10.1586/ecp.10.126. [DOI] [PubMed] [Google Scholar]
  • 208.Geldmacher DS, Fritsch T, McClendon MJ, Landreth G. A randomized pilot clinical trial of the safety of pioglitazone in treatment of patients with Alzheimer disease. Arch. Neurol. 2011;68(1):45–50. doi: 10.1001/archneurol.2010.229. [DOI] [PubMed] [Google Scholar]
  • 209.Martinez TN, Greenamyre JT. Toxin models of mitochondrial dysfunction in Parkinson's disease. Antioxid. Redox Signal. 2011;16(9):920–934. doi: 10.1089/ars.2011.4033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Nagatsu T, Sawada M. Cellular and molecular mechanisms of Parkinson's disease: neurotoxins, causative genes, and inflammatory cytokines. Cell. Mol. Neurobiol. 2006;26:781–802. doi: 10.1007/s10571-006-9061-9. [DOI] [PubMed] [Google Scholar]
  • 211.Polanski W, Reichmann H, Gille G. Stimulation, protection and regeneration of dopaminergic neurons by 9-methyl-β-carboline: a new anti-Parkinson drug? Exp. Rev. Neurother. 2011;11(6):845–860. doi: 10.1586/ern.11.1. [DOI] [PubMed] [Google Scholar]
  • 212.Polanski W, Enzensperger C, Reichmann H, Gille G. The exceptional properties of 9-methyl-beta-carboline: stimulation, protection and regeneration of dopaminergic neurons coupled with anti-inflammatory effects. J. Neurochem. 2010;113(6):1659–1675. doi: 10.1111/j.1471-4159.2010.06725.x. [DOI] [PubMed] [Google Scholar]
  • 213.Hamann J, Rommelspacher H, Storch A, Reichmann H, Gille G. Neurotoxic mechanisms of 2, 9-dimethyl-beta-carbolinium ion in primary dopaminergic culture. J. Neurochem. 2006;98:1185–1199. doi: 10.1111/j.1471-4159.2006.03940.x. [DOI] [PubMed] [Google Scholar]
  • 214.Matsubara K, Kobayashi S, Kobayashi Y, Yamashita K, Koide H, Hatta M, Iwamoto K, Tanaka O, Kimura K. beta-Carbolinium cations, endogenous MPP+ analogs, in the lumbar cerebrospinal fluid of patients with Parkinson's disease. Neurology. 1995;45:2240–2245. doi: 10.1212/wnl.45.12.2240. [DOI] [PubMed] [Google Scholar]
  • 215.Hamann J, Wernicke C, Lehmann J, Reichmann H, Rommelspacher H, Gille G. 9-Methyl-beta-carboline up-regulates the appearance of differentiated dopaminergic neurones in primary mesencephalic culture. Neurochem. Int. 2008;52:688–700. doi: 10.1016/j.neuint.2007.08.018. [DOI] [PubMed] [Google Scholar]
  • 216.Wernicke C, Hellmann J, Zieba B, Kuter K, Ossowska K, Frenzel M, Dencher NA, Rommelspacher H. 9-Methyl-beta-carboline has restorative effects in an animal model of Parkinson's disease. Pharmacol. Rep. 2010;62(1):35–53. doi: 10.1016/s1734-1140(10)70241-3. [DOI] [PubMed] [Google Scholar]
  • 217.Murata M. The discovery of an antiparkinsonian drug, zonisamide. Clin. Neurol. 2010;50(11):780–782. doi: 10.5692/clinicalneurol.50.780. [DOI] [PubMed] [Google Scholar]
  • 218.Yokoyama H, Yano R, Kuroiwa H, Tsukada T, Uchida H, Kato H, Kasahara J, Araki T. Therapeutic effect of a novel anti-parkinsonian agent zonisamide against MPTP(1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine) neurotoxicity in mice. Metab. Brain Dis. 2010;25(2):135–143. doi: 10.1007/s11011-010-9191-0. [DOI] [PubMed] [Google Scholar]
  • 219.Choudhury ME, Moritoyo T, Kubo M, Kyaw WT, Yabe H, Nishikawa N, Nagai M, Matsuda S, Nomoto M. Zonisamide-induced long-lasting recovery of dopaminergic neurons from MPTP-toxicity. Brain Res. 2011;1384:170–178. doi: 10.1016/j.brainres.2011.02.017. [DOI] [PubMed] [Google Scholar]
  • 220.Bourque M, Dluzen DE, Di Paolo T. Neuroprotective actions of sex steroids in Parkinson's disease. Front. Neuroendocrinol. 2009;30(2):142–157. doi: 10.1016/j.yfrne.2009.04.014. [DOI] [PubMed] [Google Scholar]
  • 221.Gillies GE, McArthur S. Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol. Rev. 2010;62(2):155–198. doi: 10.1124/pr.109.002071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Liaw JJ, He JR, Hartman RD, Barraclough CA. Changes in tyrosine hydroxylase mRNA levels in medullary A1 and A2 neurons and locus coeruleus following castration and estrogen replacement in rats. Brain Res. Mol. Brain Res. 1992;13:231–238. doi: 10.1016/0169-328x(92)90031-6. [DOI] [PubMed] [Google Scholar]
  • 223.Gayrard V, Malpaux B, Tillet Y, Thiery JC. Estradiol increases tyrosine hydroxylase activity of the A15 nucleus dopaminergic neurons during long days in the ewe. Biol. Reprod. 1994;50:1168–1177. doi: 10.1095/biolreprod50.5.1168. [DOI] [PubMed] [Google Scholar]
  • 224.Serova L, Rivkin M, Nakashima A, Sabban EL. Estradiol stimulates gene expression of norepinephrine biosynthetic enzymes in rat locus coeruleus. Neuroendocrinology. 2002;75:193–200. doi: 10.1159/000048237. [DOI] [PubMed] [Google Scholar]
  • 225.Ivanova T, Beyer C. Estrogen regulates tyrosine hydroxylase expression in the neonate mouse midbrain. J. Neurobiol. 2003;54:638–647. doi: 10.1002/neu.10193. [DOI] [PubMed] [Google Scholar]
  • 226.Brann DW, Dhandapani K, Wakade C, Mahesh V, Khan MM. Neurotrophic and neuroprotective actions of estrogen: basic mechanisms and clinical implications. Steroids. 2007;72:381–405. doi: 10.1016/j.steroids.2007.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Nakashima A, Ota A, Sabban EL. Interactions between Egr1 and AP1 factors in regulation of tyrosine hydroxylase transcription. Brain Res. Mol. Brain Res. 2003;112:61–69. doi: 10.1016/s0169-328x(03)00047-0. [DOI] [PubMed] [Google Scholar]
  • 228.Rantham Prabhakara JP, Feist G, Thomasson S, Thompson A, Schommer E, Ghribi O. Differential effects of 24-hydroxycholesterol and 27-hydroxycholesterol on tyrosine hydroxylase and alpha-synuclein in human neuroblastoma SHSY5Y cells. J. Neurochem. 2008;107(6):1722–1729. doi: 10.1111/j.1471-4159.2008.05736.x. [DOI] [PubMed] [Google Scholar]
  • 229.Marwarha G, Rhen T, Schommer T, Ghribi O. The oxysterol 27-hydroxycholesterol regulates α-synuclein and tyrosine hydroxylase expression levels in human neuroblastoma cells through modulation of liver X receptors and estrogen receptors-Relevance to Parkinson's disease. J. Neurochem. 2011;119(5):1119–1136. doi: 10.1111/j.1471-4159.2011.07497.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Alerte TN, Akinfolarin AA, Friedrich EE, Mader SA, Hong CS, Perez RG. Alpha-synuclein aggregation alters tyrosine hydroxylase phosphorylation and immunoreactivity: lessons from viral transduction of knockout mice. Neurosci. Lett. 2008;435(1):24–29. doi: 10.1016/j.neulet.2008.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Perez RG, Waymire JC, Lin E, Liu JJ, Guo F, Zigmond MJ. A role for alpha-synuclein in the regulation of dopamine biosynthesis. J. Neurosci. 2002;22(8):3090–3099. doi: 10.1523/JNEUROSCI.22-08-03090.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Li YH, Gao N, Ye YW, Li X, Yu S, Yang H, Uéda K, Chan P. Alpha-synuclein functions as a negative regulator for expression of tyrosine hydroxylase. Acta Neurol. Belg. 2011;111(2):130–135. [PubMed] [Google Scholar]
  • 233.Ames M, Lerner P, Lovenberg W. Tyrosine hydroxylase activation by protein phosphorylation and end product inhibition. J. Biol. Chem. 1978;253:27–31. [PubMed] [Google Scholar]
  • 234.Birkmayer JGD, Vrecko C, Volc D, Birkmayer W. Nicotinamide adenine dinucleotide(NADH)-a new therapeutic approach to Parkinson's disease. Acta Neurol. Scand. 1993;87(146):32–35. [PubMed] [Google Scholar]
  • 235.Birkmayer GJ, Birkmayer W. Stimulation of endogenous L-dopa biosynthesis--a new principle for the therapy of Parkinson's disease. The clinical effect of nicotinamide adenine dinucleotide(NADH) and nicotinamide adenine dinucleotidephosphate(NADPH). Acta Neurol. Scand. Suppl. 1989;126:183–187. doi: 10.1111/j.1600-0404.1989.tb01800.x. [DOI] [PubMed] [Google Scholar]
  • 236.Swerdlow RH. Is NADH effective in the treatment of Parkinson's disease? Drugs Ageing. 1998;13(4):263–268. doi: 10.2165/00002512-199813040-00002. [DOI] [PubMed] [Google Scholar]
  • 237.Nichol CA, Smith GK, Duch DS. Biosynthesis and metabolism of tetrahydrobiopterin and molybdopterin. Ann. Rev. Biochem. 1985;54:729–764. doi: 10.1146/annurev.bi.54.070185.003501. [DOI] [PubMed] [Google Scholar]
  • 238.Schwab C, Klegeris A, McGeer PL. Inflammation in transgenic mouse models of neurodegenerative disorders. Biochim. Biophys. Acta. 2010;1802(10):889–902. doi: 10.1016/j.bbadis.2009.10.013. [DOI] [PubMed] [Google Scholar]
  • 239.McGeer EG, McGeer PL. The role of anti-inflammatory agents in Parkinson's disease. CNS Drugs. 2007;21(10):789–797. doi: 10.2165/00023210-200721100-00001. [DOI] [PubMed] [Google Scholar]
  • 240.Tansey MG, Goldberg MS. Neuroinflammation in Parkinson's disease: its role in neuronal death and implications for therapeutic intervention. Neurobiol. Dis. 2010;37(3):510–518. doi: 10.1016/j.nbd.2009.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Frankola KA, Greig NH, Luo W, Tweedie D. Targeting TNF-α to elucidate and ameliorate neuroinflammation in neurodegenerative diseases. CNS Neurol. Disord. Drug Targets. 2011;10(3):391–403. doi: 10.2174/187152711794653751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Lahiri DK, Chen D, Maloney B, Holloway HW, Yu QS, Utsuki T, Giordano T, Sambamurti K, Greig NH. The experimental Alzheimer's disease drug posiphen [(+)-phenserine] lowers amyloid-beta peptide levels in cell culture and mice. J. Pharmacol. Exp. Ther. 2007;320(1):386–396. doi: 10.1124/jpet.106.112102. [DOI] [PubMed] [Google Scholar]
  • 243.Shaw KT, Utsuki T, Rogers J, Yu QS, Sambamurti K, Brossi A, Ge YW, Lahiri DK, Greig NH. Phenserine regulates translation of beta -amyloid precursor protein mRNA by a putative interleukin-1 responsive element, a target for drug development. Proc. Natl. Acad. Sci. USA. 2001;98(13):7605–7610. doi: 10.1073/pnas.131152998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Friedlich AL, Tanzi RE, Rogers JT. The 5'-untranslated region of Parkinson's disease alpha-synuclein messenger RNA contains a predicted iron responsive element. Mol. Psychiatry. 2007;12(3):222–223. doi: 10.1038/sj.mp.4001937. [DOI] [PubMed] [Google Scholar]
  • 245.Olivares D, Huang X, Branden L, Greig NH, Rogers JT. Physiological and pathological role of alpha-synuclein in Parkinson's disease through iron mediated oxidative stress; The role of a putative iron-responsive element. Int. J. Mol. Sci. 2009;10(3):1226–1260. doi: 10.3390/ijms10031226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Rogers JT, Mikkilineni S, Cantuti-Castelvetri I, Smith DH, Huang X, Bandyopadhyay S, Cahill CM, Maccecchini ML, Lahiri DK, Greig NH. The alpha-synuclein 5'untranslated region targeted translation blockers: anti-alpha synuclein efficacy of cardiac glycosides and Posiphen. J. Neural. Transm. 2011;118:493–507. doi: 10.1007/s00702-010-0513-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Mikkilineni S, Cantuti-Castelvetri I, Cahill C, Greig NH, Rogers JT. The anti-cholinesterase phenserine and its enantiomer posiphen as 5’untranslated region directed translation blockers of the Parkinson's alpha synuclein expression. Parkinson's Dis. 2012 doi: 10.1155/2012/142372. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Swanson CR, Joers V, Bondarenko V, Brunner K, Simmons HA, Ziegler TE, Kemnitz JW, Johnson JA, Emborg ME. The PPAR-γ agonist pioglitazone modulates inflammation and induces neuroprotection in parkinsonian monkeys. J. Neuroinflammation. 2011;5:91. doi: 10.1186/1742-2094-8-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat. Rev. Drug Discov. 2004;3(8):673–683. doi: 10.1038/nrd1468. [DOI] [PubMed] [Google Scholar]
  • 250.Dolgin E. Nonprofit disease groups earmark grants for drug repositioning. Nat. Med. 2011;17(9):1027. doi: 10.1038/nm0911-1027. [DOI] [PubMed] [Google Scholar]
  • 251.Sardana D, Zhu C, Zhang M, Gudivada RC, Yang L, Jegga AG. Drug repositioning for orphan diseases. Brief Bioinform. 2011;12(4):346–56. doi: 10.1093/bib/bbr021. [DOI] [PubMed] [Google Scholar]

RESOURCES