REVIEW ARTICLE


Role of Genes and Treatments for Parkinson’s Disease



Falaq Naz1, Yasir Hasan Siddique1, *
1 Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India


Article Metrics

CrossRef Citations:
4
Total Statistics:

Full-Text HTML Views: 2386
Abstract HTML Views: 184
PDF Downloads: 486
Total Views/Downloads: 3056
Unique Statistics:

Full-Text HTML Views: 1168
Abstract HTML Views: 97
PDF Downloads: 328
Total Views/Downloads: 1593



Creative Commons License
© 2020 Naz and Siddique.

open-access license: This is an open access article distributed under the terms of the Creative Commons Attribution 4.0 International Public License (CC-BY 4.0), a copy of which is available at: https://creativecommons.org/licenses/by/4.0/legalcode. This license permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Correspondence: Address correspondence to this author at the Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India; Tel: 0571-2700920-3447; E-mail: yasir_hasansiddique@rediffmail.com


Abstract

Parkinson’s Disease (PD) is a complex neurodegenerative disorder that mainly results due to the loss of dopaminergic neurons in the substantia nigra of the midbrain. It is well known that dopamine is synthesized in substantia nigra and is transported to the striatum via nigrostriatal tract. Besides the sporadic forms of PD, there are also familial cases of PD and number of genes (both autosomal dominant as well as recessive) are responsible for PD. There is no permanent cure for PD and to date, L-dopa therapy is considered to be the best option besides having dopamine agonists. In the present review, we have described the genes responsible for PD, the role of dopamine, and treatment strategies adopted for controlling the progression of PD in humans.

Keywords: Parkinson’s disease, Neurodegenerative diseases, Dopamine, Vata rogas, Neurological disorder, Shaking palsy.



1. INTRODUCTION

Neurodegeneration refers to the condition which primarily affects neurons [1]. Hippocrates (460-375 BC), in his important treatise entitled ‘On The Sacred Disease’ describes epilepsy and relates it with the brain. In the ancient period, neurological disorders were classified under humor- Vata rogas (diseases) in Ayurveda. Vata refers to the energy present in the body involved in controlling both voluntary and involuntary functions. Any change in vata leads to the pathogenesis of the neurodegenerative disease. In Ayurveda, about eighty types of neurological disorders have been described, such as apasmara /apasmati (Epilepsy), kampavata (Parkinson’s disease). The clinical symptoms and treatments of neurodegenerative disease have been described by Hippocrates [2-5].

The clinical symptoms of neurodegenerative diseases are often characterized by alteration in cognitive function, movement, memory and learning, co-ordination and autonomic control [6]. Worldwide neurological disorders have more than 6% of the total burden of diseases [7]. The most common neurological diseases are Alzheimer's Disease (AD), Parkinson’s Disease (PD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS), fronto-temporal dementia and the spinocerebellar ataxia (Table 1).

Neurodegenerative diseases/disorders have different pathophysiology, along with cognitive impairments. The loss of brain cells in neurodegenerative diseases is not a rapid process but rather a very slow process that becomes worse during the progression of the disease. Sporadic, as well as familial forms of neurodegenerative diseases, are common in the human population. These forms are difficult to distinguish phenotypically as well as histologically. A number of causative genes have been discovered for the neurodegenerative disease [8]. James Parkinson was born on April 11, 1755, in the semi-urban hamlet of Hoxton, Northeast of the city of London, England. He describes PD for the first time. PD is an age-progressive disease, (second after AD), of the nervous system. James Parkinson was the first who provided a description of “Paralysis agitans” or “Shaking palsy”, in his popular monograph named “An Essay on the Shaking Palsy”, in 1817 [9, 10].

2. BASIC STRUCTURE AND FORMATION OF DOPAMINE IN DOPAMINERGIC NEURONS

PD is a complex neurological disorder of CNS that affects mainly the motor system of the body. The non-motor symptoms also become worsen as the disease progresses [11, 12]. Early symptoms of the PD are shaking, rigidity, slowness of movement, and difficulty in walking [13, 14]. The behavioural problems may also occur with the disability of thinking power. It is reported that about 3% of the affected

Table 1. Common Neurodegenerative diseases and their pathological characteristics with their founders.
Neurodegenerative Diseases
Diseases Characteristics
Dementia
Dr. Philippe Pinel,
1797
The two lobes become damaged and then shrink. The genetic mutations are thought to have a negative effect on a protein called the tau protein. All brain cells contain tau proteins that help to keep them stable but abnormal tau proteins may lead to dementia.
Parkinson's disease (PD)
Dr. James Parkinson,
1817
Normally brain neurons produce chemical messenger known as dopamine. When the neurons die or become impaired, they produce less dopamine and Lewy bodies formed unusual clumps of the protein α-synuclein in substantia nigra which causes the PD.
Spino-cerebellar Ataxia (SCA)
Dr. Nikolaus Friedreich,
1863
Mutation in several forms of SCA1 protein in brainstem cerebellum and spinal cord lead to Spino-cerebellar Ataxia.
Huntington's disease (HD)
Dr. George Huntington,
1872
Mutation in HTT gene instruct to make a protein i.e. huntingtin. DNA segment “CAG” segment is repeated normal 36 to more than 120 times which cause HD.
Amyotrophic lateral sclerosis (ALS)
Dr. Jean-Martin Charcot,
1874
Mutations in gene encoding superoxide dismutase 1 (SOD1) in nerve cells that cause ALS.
Alzheimer's disease (AD)
Dr. Alois Alzheimer,
1906
Abnormal clumps i.e. amyloid plaques and tangled bundles of fibers i.e. neurofibrillary tangles or tau.

people are above 65 years of age [15]. In PD, nerve cells of substantia-nigra of the mid-brain are degenerated. The basal ganglia help in initiating and smoothening of voluntary muscle movements, suppresses involuntary movements and problem in the coordination in changes of posture [16, 17]. Each part of the brain consists of Globus pallidus i.e. internal and external. The other part of the brain which is involved in the neurotransmitter function is the striatum, which includes caudate nucleus, putamen and substantia nigra. There are basically two pathways by which three main neurotransmitters work i.e. Glutamate, GABA and Dopamine [18, 19]. The basal ganglia release neurotransmitters (chemical messenger) for the activation of nerve impulse pathways. The main neurotransmitter in the basal ganglia is dopamine [20]. If the nerve cells in the basal ganglia are degenerated then there will be less production of dopamine, leading to the loss of control on muscle movements (Fig. 1). It leads to tremors, bradykinesia, hypokinesia, posture problems and the loss of overall coordination of functions. The substantia nigra, which is degenerated in PD plays an important role in the normal functioning of motor and non-motor activity of the body. The substantia nigra sends chemical message through axons that pass throughout the striatum and help in the movement of a particular part of the body. This is known as the nigrostriatal pathway or dopamine secretion pathway [21, 22].

The signals from one neuron to another are transferred through the active neuron refers as dopaminergic neurons. Within the striatum, there are neurons having specific receptors to recognize and respond to dopamine only. These receptors cannot be activated by any other neurotransmitters. When the cells in substantia nigra are dead, the level of dopamine decreases rapidly as a result, striatal cells are not activated and the PD symptoms increases day by day [23, 24]. The presence of dopaminergic neurons in the brainstem was first reported by Dahlstrom and Fuxe in 1964. In the pathophysiology of PD, the importance of dopamine depletion was first suggested by Carlsson in the late 1950s [25, 26]. In 1960, Ehringer and Hornykiewicz published their report after examining six cases of PD that the dopamine content in the neostriatum was found to be severely reduced. Dopamine (an amine), is synthesized by a precursor molecule L-Dopa. It is synthesized in the brain as well as in kidneys [27, 28].

It is well established that dopamine itself is a neurotransmitter that is synthesized in substantia nigra and transported to the striatum via the nigrostriatal tract. Several other functions have also been attributed to dopamine i.e. it can act as a vasodilator, it enhances sodium excretion and urine output; it reduces insulin production, gastrointestinal motility and protects intestinal mucosa. It also reduces the activity of lymphocytes [25, 29]. Dopamine is not able to cross the blood-brain barrier, so the interest was focused on the dopamine precursor levodopa. Levodopa takes entry into the brain with a large neutral amino acid transport pathway and can be decarboxylated to form dopamine. Henry Dale, a Physician, was first to examine the biological activity of dopamine in 1910. He suggested the shorter name dopamine in 1952. The Physician George Barger and James Hill synthesized 3,4-dihydroxyphenylethylamine at The Well-Come Laboratories in London [30]. At the same time, Kathleen Montagu and Arvid Carlsson, together with Lindqvist, Magnusson, and Waldeck in 1957, reported the presence of dopamine in the brain, and in 1959, the Swedish group claimed the presence of dopamine in high concentrations in the striatum [31-33]. Marthe Vogt suggests that there is a continuous discharge of dopamine but was not sure about dopamine to be a conventional neurotransmitter in the neurons of nigrostriatal regions [34]. Dahlstrom (1973) [35] was also not sure about the presence of dopamine in the Central Nervous System (CNS). Dopamine receptors play an essential role in daily life functions. There are various dopamine receptors in the CNS, specifically in the hippocampal dentate gyrus and sub-ventricular zone. The five types of dopamine receptors, namely, D1, D2, D3, D4, and D5 has been described to date. Each receptor has a different function. According to Mishra et al. (2018) [36] the functions of various dopamine receptors are as follows:

  • D1: memory, attention, impulse control, regulation of renal function and locomotion.
  • D2: locomotion, attention, sleep, memory and learning.
  • D3: cognition, impulse control, attention and sleep.
  • D4: cognition, impulse control, attention and sleep.
  • D5: decision making, cognition, attention and renin secretion.

Further, the two types of sub-families of dopamine receptors are D1-like family and D2 like family. The D1 and D5 receptors belong to D1-like family of dopamine receptors, and D2, D3 and D4 receptors belong to the D2-like family [37]. D1 and D5 receptors coupled to G stimulatory sites and activate adenylyl cyclase. The activation of adenylyl cyclase leads to the production of the second messenger cAMP, which further leads to the production of protein kinase A (PKA) and affects transcription in the nucleus [38, 39]. The D1 receptor is most abundantly distributed in the CNS, followed by D2, D3, D5 and D4 (D1>D2>D3>D5>D4) [40]. D1 receptors regulate the development of neurons are play an important role in the regulation of most of the cognitive activities as well as locomotion [41]. D1 and D5 receptors have high density in the striatum, nucleus accumbens, olfactory bulb, and substantia nigra. D1 and D5 receptors, along with stimulating adenyl cyclase, also activates phospholipase C, which leads to the induction of intracellular calcium release and activation of protein kinase C. Protein kinase C is a calcium-dependent protein kinase. Calcium is also involved in modulating the neurotransmitter released by exocytosis. D1 and D5 inhibits Na/K ATPase i.e. cyclic AMP-dependent Protein Kinase (PKA) calcium- and phospholipid-dependent Protein Kinase (PKC) pathways in the kidneys [42]. D2, D3, and D4 receptors are expressed mainly

Fig. (1). Direct and indirect dopaminergic pathway.

in the striatum, external globus pallidus, core of nucleus accumbens, hippocampus, amygdala, and cerebral cortex. These receptor affects the postsynaptic receptor-mediated extrapyramidal activity. D2-D4 receptors are important for signaling, the survival of human dopaminergic neurons and their development [43, 44]. It has been reported in mice that the dysfunction of the dopaminergic pathways leads to neurological and psychiatric disorders [45, 46]. Fig. (2) shows a comparison of the normal brain with a brain affected by various factors that are responsible for inducing PD. The dopamine signaling is important for maintaining the normal physiological processes. The disruption of pathways may lead

Fig. (2). Comparison of dopamine signaling pathway in normal brain and PD brain.

to the dysfunctions attributed to neurodegenerative disorders. Tyrosine in the presence of Tyrosine Hydroxylase (TH) is converted into L-Dopa. Then, L-Dopa is converted into the Dopamine in the presence of DOPA Decarboxylase (DDC). Dopamine is transported from the cytosol by a Vesicular Monoamine Transporter (VMAT2) into the synaptic vesicles where it is stored until its release into the synaptic cleft. Dopamine receptors are present in both post as well as pre-synaptic neurons (including Dopamine Transporter, DAT). The required dopamine reaches the postsynaptic synapse through the dendrites, which leads to the normal functioning of both motor and non-motor neurons [47].

3. GENETICS OF PARKINSON’S DISEASE (PD)

Several common cellular mechanisms of protein aggregation are involved in the progression of neurodegenerative diseases. The β-amyloid is the main part of plaques formation in AD, α-synuclein for Lewy bodies (LBs) formation in the substantia nigra in PD and dementia with LBs (DLB), and expanded repetition of polyglutamine is found as intra-nuclear and cytoplasmic inclusions in HD [48, 49]. The LBs were first described by F.H. Lewy in 1912, in the nucleus of Basalis of Meynert and the dorsal vagal nucleus in patients with PD. It was named in the honour of Friederich H. Lewy by Tretiakoff, who confirmed the presence of LBs in the substantia nigra [50-52]. LBs are rounded, eosinophilic and intra-cytoplasmic neuronal inclusion or aggregation of α-synuclein. According to some studies, the LBs are found in the selected areas of the brain i.e. substantia nigra, locus ceruleus, dorsal vagal nucleus, the nucleus of Meynert, hypothalamus cortex and sympathetic ganglia [53-55]. For the last 20 years several important discoveries have been made by researchers on the genetics of PD. The SNCA and its mutations are associated with autosomal dominant parkinsonism [56, 57]. Mutations in this gene include missense, which results in amino acid substitutions. SNCA-related PD is not so common, but identification of SNCA mutations has led to the identification of α-synuclein as the major component of Lewy bodies [58-61]. α-synuclein has 140 amino acids, that belongs to the family of related synucleins (β-synuclein of 134 amino acids) [62, 63]. α-synuclein contains an acidic stretch towards the C-terminus and there are no cysteines and tryptophans. Structurally, human α-synuclein consists of three regions i.e. an N-terminal amphipathic region, a hydrophobic central region, and an acidic C-terminal region [61, 64-66]. Six genes have been found to be associated with autosomal dominant forms of PD i.e. SNCA, LRRK2, VPS35, EIF4G1, DNAJC13, and CHCHD2 and three with autosomal recessive forms of PD i.e. Parkin, PINK 1 and DJ-1 [67, 68].

4. GENES INVOLVED IN THE AUTOSOMAL DOMINANT FORMS OF PD:

4.1. SNCA (PARK1-4)

The product of SNCA is α-synuclein protein. Its aggregation leads to the formation of Lewy bodies [69, 70]. It is localized in presynaptic terminals of the dopaminergic neurons [71]. It is reported that patients suffering from PD with SNCA mutations are associated with early-onset of PD (age of onset ≤50 year). The association of REP1 polymorphism in the promoter region of the gene and PD has been suggested by the researchers [72, 73]. The exact function of α-synuclien is still unknown, but it has been suggested that it plays an important role in the transport of synaptic vesicles thus maintaining the synaptic plasticity [74]. The abnormal form of the protein is more susceptible to fibrillogenesis and self-aggregation [58, 75, 76]. Only three different missense mutations in the form of duplications and triplications have been reported to date [77]. The first missense is p.A53T, which is most frequent and reported in various studies from Italy, Greek, Korea and Sweden [76, 78-82]. p.A53T substitution was discovered in 1997. The p.A30P and p.E46K substitutions were discovered in 1998 and 2004, respectively [53, 76].

The penetrance of the missense mutations is about 85% for p.A53T [83, 84]. However, the penetrance of SNCA duplication was surprisingly found to be low as 33.3%, in Japanese family [85]. The SNCA consist of six exons ranging in size from 42 to 1110 bp and the α-synuclein is about 140 amino acid long [86]. It has three domains: (i) the amino-terminal region (ii) a central hydrophobic domain and (iii) an acidic, negatively charged C-terminal domain. All three missense mutations are associated with the amino-terminal domain [87]. When associated with the phospholipid membranes, via its amino-terminal repeats (amino acids 71-82), it adopts a structure rich in α-helical character [88]. The mutation in the gene results in the formation of toxic oligomers of stable β-sheets and thus forming protofibrils and amyloid fibrils [89-91]. The accumulation of glucocerebrosides, due to the loss of functional glucocerebrosidase enhances the aggregation of α-synuclein. α-synuclein inhibits the lysososmal activity of glucocerebrosidase in the neurons [92, 93].

4.2. LRRK2 (PARK8)

LRRK2 (leucine-rich repeat kinase-2), is a large multidomain protein, involved in neurite outgrowth, synaptic morphogenesis, protein cargo trafficking, autophagy and protein synthesis [94]. The LRRK2 is also involved in the making of dardarin [95]. LRRK2 also play an important role in regulating the innate immune system. It has a conserved supra domain consisting of a Ras-like GTPase called Roc (Ras of complex proteins) and a C-terminal of Roc (COR) domain [96]. LRRK2 is involved in dual enzymatic functions (GTPase and kinase activity). So far 80 mutations have been reported for LRRK2, and only six mutations have been found to be associated with PD i.e. N1437H, R1441 G/H/C, Y1699C, I2012T, G2019S, and I2020T [97-99].

Mutation in the LRRK2 (PARK8) gene (leucine-rich repeated kinase-2) is the most common cause of late-onset autosomal-dominant PD (ADPD) as well as the sporadic form of PD, having a mutation frequency of 2 to 40% among different populations [100-102]. LRRK2 consists of 51 exons and encodes the 2527 amino acid cytoplasmic protein. It consists of a leucine-rich repeat towards the amino terminus and a kinase domain towards the carboxyl terminus. More than 50 different missense and nonsense mutations have been reported for LRRK2 to date among which 16 of them (including the six recurrent mutations- p.R114C, p.R1441G, p.R1441H, p.Y1699C, p.G2019S, and p.I2020T) seems to be pathogenic [103]. The mutation c.6055G >A (p.G2019S) has been reported in 40% of Arabs [101], 20% of Ashkenazi Jews [102], and 1%-7% of Europeans [104, 105]. Few researchers have described p.G2019S mutation in homozygous conditions [106]. Due to the founder effect, p.R1441G is very frequent among Basques [107, 108] and p.I2020T among Japanese patients [109]. The p.G2019S showed reduced penetrance, whereas p.R1441 mutation has been reported to be highly penetrant [110]. The exact mechanism leading to PD by LRRK2 mutations is not properly understood. However, due to its large size, any change in its domain would lead to the loss of its interaction with other proteins, thus inhibiting its function. In addition, various mutations have also been reported to affect its kinase activity [111].

4.3. VPS35, EIF4G1, DNAJC13 and CHCHD2

VPS35, EIF4G1, DNAJC13, and CHCHD2 are the most recent genes to be associated with autosomal dominant form of PD. VPS35 (vacuolar protein sorting 35), a multi subunit complex, is found to be associated with endosomes and is involved in intracellular membrane-bound compartments transportation between the plasma membrane to golgi bodies [112, 113]. EIF4G1 (eukaryotic translation initiation factor 4-gamma) mutations are also linked with PD. It may interact with FMR1, involved in the regulation of the actin cytoskeleton pathway, have shown the strongest association with PD [103, 114]. DNAJC13 [mammalian homolog to receptor-mediated endocytosis 8 (REM-8)] is a chaperone protein found in endosomes and regulates trans-membrane protein transfer. A mutation in DNAJC13 showed aberrant endosomal confinement of α-synuclein, which might stimulate the process of neurodegeneration leading to PD [115, 116]. CHCHD2 (Coiled-coil-helix-coiled-coil-helix domain containing protein 2), is a mitochondrial protein, promotes mitochondrial oxygen consumption and its mutation leads to apoptosis and production of reactive oxygen species (ROS) (mitochondrial dysfunctioning) [117-119].

5. GENES INVOLVED IN AUTOSOMAL RECESSIVE FORMS OF PD

Three genes (parkin, PINK1 and DJ1) have been identified to be associated with autosomal recessive form of inherited parkinsonism. parkin encodes for E3 ubiquitin ligase, which controls the proteasome-dependent degradation or autophagy in neurons [120]. PINK1, is a mitochondrial protein kinase plays an important role in mitophagy [121, 122]. The mutations in DJ-1 bring changes in mitochondrial morphology and leads to the high production of reactive oxygen species [123]. The most common cause of autosomal recessive PD is the mutation in parkin. Parkin mutation has been reported in about 49% of familial cases and about 15% of sporadic cases [124, 125]. Mutations in DJ1 (1-8%) and PINK1 (1-2%) are less common. The function of DJ-1 is not well understood, but it protects mitochondria against the oxidative stress [126-128]. The proteins encoded by parkin, PINK1 and DJ-1 are directly or indirectly involved in the recovery of damaged mitochondria [129-131]. The other genes associated with PD are ATP13A2 [132, 133], FBXO7 [134, 135], PLA2G6 [136, 137], POLG1 [138, 139] and SYNJ1 [140, 141]. Genetic mapping methods such as linkage analysis and genome-wide association studies have been reported to be helpful in identifying new genes responsible for PD [142].

5.1. Parkin (PARK2)

Parkin gene was discovered in 1998. It contains 12 exons.The clinical phenotypes of Parkin, PINK1 and DJ-1 linked PD are identical [143, 144]. A number of parkin mutations have been identified in all 12 exons [106, 124]. The mutations associated with parkin lead to the early onset of PD that accounts for about 77% of the familial cases with an age of onset <30 years [124]. The protein has a conserved Ubiquitin- Like domain (UBL) with Really Interesting New Gene (RING) finger motifs [145, 146]. There are two main targets for parkin (i) CDCrel - 1, belongs to the family of GTPases called septins, and is expressed in synaptic vesicles [145, 147], (ii) Pael-R (Parkin associated endothelin receptor). Parkin specifically ubiquitinates this receptor in the presence of ubiquitin-conjugating enzymes and promotes the degradation of insoluble Pael-R, resulting in the suppression of cell death. In the absence of functional parkin, Pael-R accumulates in the brains and leads to neuronal death [148, 149]. It has been reported that protein synphilin-1, ubiquitinated by parkin leads to the formation of protein aggregates [150].

5.2. PINK1 (PARK6)

PINK1 (PTEN-induced kinase 1), is a mitochondrial serine or threonine-protein kinase. PINK1 is a 581 amino acid protein kinase [151].The mitochondrial membrane protein PINK1 mutations are also found in the forms of familial PD. An amino-terminal of the protein has 34 amino acids (mitochondrial targeting motif), and the carboxy-terminal serve as an autoregulatory domain. Some recent studies have suggested that PINK1 and parkin, are associated with a common pathway, for eliminating the deformed mitochondria [152]. PINK1 attaches itself to the mitochondrial membrane and recruits parkin from the cytosol. Once recruited parkin becomes enzymatically active and initiates the process of eliminating the mitochondria (mitophagy) [153].

Mutations in the phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1) leads to the onset of early autosomal recessive PD. The frequency of PINK1 mutations is about 1-9%, varying among different ethnic groups [129, 151, 154-156]. PINK1 mutations have been reported as either missense or nonsense mutations in the Sudanese family (exons 4-8) [157], exons 6-8 deletion in a sibling family [156], and exon-7 deletion [158], and one with a splice site mutations [159]. To date, about 60 different missense and nonsense mutations have been reported in more than 170 patients, affecting all eight PINK1 exons with a nearly equal frequencies (5-10 different mutations in each exon) [160].

5.3. DJ-1 (PARK7)

The mutation of DJ-1 is reported in about 1-2% of PD cases [161]. DJ-1 has different mechanisms to protect dopaminergic neurons against neurodegeneration in PD [162]. DJ-1-linked PD is rare and so far, only ten different point mutations and exonic deletions have been found to be associated with the disease [163]. The level of oxidative stress regulates its activity [164]. The gene consists of seven exons and codes for a protein of 189 amino acids [165]. In physiological condition, DJ-1 protein forms a dimeric structure [166].

6. OTHER GENES RESPONSIBLE FOR PD

Mutations in ATP13A2 are associated with an autosomal recessive form of PD i.e. Kufor-Rakeb syndrome (KRS). It has a juvenile onset with rapid progression, followed by dementia, supranuclear gaze palsy and pyramidal signs [167, 168]. The gene consists of 29 exons, which codes for a protein of 1180 amino acids. The ATP13A2 protein is normally located in the membrane of lysosome, with 10 trans-membrane domains having an ATPase domain [169]. The first mutation in ATP13A2 (PARK9) was reported in a Chilean family [170, 171]. The lysosomal enzyme β-glucocerebrosidase is coded by GBA gene and play a role in glycolipid metabolism. Mutations in the GBA gene results in the production of non-functional β-glucocerebrosidase which leads to Gaucher disease [172]. GBA mutations are reported to enhance the progression of PD and about 8-14% of the autopsy revealed its link with PD [173-175].

The complex interactions of environmental and genetic factors may lead to PD. Such cases have been classified as sporadic [176, 177]. Damage in the substantia nigra has been reported in most of the sporadic cases [178, 179]. Some gene mutations can lead to the accumulation of unwanted protein that may alter the function of mitochondria leading to the increase in oxidative stress [180, 181]. Table 2 gives a description of genes associated with PD.

Table 2. Mutation in Genes associated with PD.
S.No. Gene with Symbol Locus/
Loci
Chromosomal Location Year
of Discovery
Recessive or Dominant Clinical Features in PD Reported Variants associated with PD Target in Cell Possible
Function(s)
Reference(s)
1. α-synuclein (SNCA) PARK1 and PARK4 4q21 1997 Dominant Hyposomnia, and sleep disorder rs2583988, rs356219, and rs2736990 Presynaptic terminals Regulating synapse, vesicle trafficking [181, 182]
2. Parkin (PRKN) PARK2 6q25.2-q27 1998 Recessive Hyper-reflexia S167N, E310D, D394N Mitochondria E3 Ubiquitin Protein Ligase [183, 184]
3. UCHL1 PARK5 4p13 1998 Dominant Multiple tumors I93M and S18Y Nerve cells Synthesize an enzyme called ubiquitin carboxyl-terminal esterase L1 [72, 185]
4. DJ1 PARK7 1p36 2003 Recessive Dyskinesia, rigidity, and tremors, C53A and DJ-1C106 Nerve cells Transcriptional regulation, antioxidative stress reaction, and chaperone, protease, and mitochondrial regulation [164, 165]
5. PINK1 PARK6 1p36-1p35 2004 Recessive Dystonia P37L, R42P, R256C, G328E, and R334C Mitochondrial membrane Mitochondrial kinase [186, 187]
6. POLG POLG1 15q26.1 2004 Dominant Bradykinesiaand rigidity, resting tremor and they respond well to levodopa treatment p.A899T Cortex CAG-repeat, encoding a polyglutamine (poly-Q) trac [188-190]
7. LRRK2 PARK8 12q12 2004 Dominant Less non-motor function, olfactory, and cognitive impairment A419V, G2019S, R1441C/G/H, G2385R, and R1628P Glial cells and Neurons Protein kinase [191-192]
8. HtrA2 PARK13 2p12 2005 Dominant Bradykinesia, tremor Omi/HtrA2, Pro143Ala Glia cells and Neurons Mitochondrial serine kinase [193-195]
9. ATP13A2 PARK9 1p36 2006 Recessive Kufor-Rakeb syndrome, pallido-pyramidal syndrome, dystonia F182L and G504R Lysosomes Improves the lysosome membrane integrity and protects against iron-induced cell damage [132, 196, 197]
10. FBOX7 PARK15 22q12-q13 2008 Recessive Rigidity, bradykinesia and postural instability Arg498Stop Cerebral cortex, Globus pallidum and the Substantia nigra Ubiquitin ligase [134, 198]
11. GIGYF2 PARK11 2q37.1 2008 Dominant Less motor symptom N56SN457Trs12328151, rs2289912, rs2305138, rs3816334, A572AH1171R Endosomes Progression in motor symptoms [199, 200]
12. GBA GBA1 1q21 2009 Dominant Hallucinations and cognitive decline E326K, T369M, N370S, D409H, and L444P Endoplasmic reticulum Regulation of the lysosomal system [201, 202]
13. PLA2G6 or iPLA2β PARK14 22q12-q13 2009 Recessive Axonal dystrophy, dystonia, dementia, visual disturbances p.R301C and p.D331N Peripheral nerves Phospholipase A2 [203, 204]
14. Vps35 PARK17 16q11.2 2011 Dominant Tremors, bradykinesia, rigidity of limbs and gait defects p. Asp620Asn Lysosomal vesicles Retromer component [205-207]
15. EIF4G1 PAERK18 3q27.1 2011 Dominant Motor dysfunction A502V, G686C, S1164R, and R1197W Presynaptic nerves Scaffold subunit of the translation initiation complex eIF4F, which binds the ribosomal 40S [114, 208]
16. DNAJC6 PARK19 1p31.3 2012 Recessive Mental retardation, pyramidal signs and epilepsy p.Q734X Cerebellum, corpus callosum, cortex, brainstem, pons, putamen, spinal cord, and substantia nigra. Cochaperone [209, 210]
17. SYNJ1 PARK20 21q22.11 2013 Recessive Dystonia, oculomotor apraxia, and seizures Nil Presynaptic terminal Polyphosphoinisitide phosphatase [211, 212]
18. DNAJC13 PARK21 3q22.1 2014 Recessive Motor dysfunction p. N855S Neurons Endosomal transport [213, 214]
19. CHCHD2 CHCHD2 7p11.2 2015 Recessive Bradykinesia and rigidity, hypomimic face and mydriatic pupils G66V and P80L Cortical neurons Mitochondrial protein [215, 216]
20. LRP10 (low-density lipoprotein receptor-related protein 10) LRP10 14p13–q12 2018 Dominant Motor fluctuations and dyskinesia, no cognitive impairment OR11H12, POTEG, and LRP10 Cerebral cortex Regulates amyloid precursor protein (APP) trafficking and processing. [217-219]

7. POST TRANSLATION MODIFICATIONS OF SNCA, DJ1, PARKIN AND LRRK2

The pathogenesis of PD mainly involves dysfunction of mitochondria, protein misfolding, aggregation and oxidative stress [220]. A number of post-translational modifications (PTMs) regulates the activity of protein; for example, nitration, arginylation and N-terminal acetylation which affects the structure and behaviour of α-synuclein to interact with other biomolecules [221]. The modification of α-synuclein, its aggregation, toxicity and targets for future therapeutics has been extensively reviewed by Barrett and Greenamyre [222]. Although PTMs are required for the normal functions of α-synuclein, abnormal modifications may promote pathogenesis of the PD [223]. The phosphorylation of S129 residue has been reported to be associated with the formation of Lewy bodies [224]. Nitration of tyrosine residues in α-synuclein at Y39, Y125, Y133 and Y136 has been reported to be associated with oxidative stress [225]. Environmental factors are also responsible for the PTMs. The exposure of 1-Methyl-4-phenyl-1,2,3,6-Tetrahydropyridine (MPTP) results in the phosphorylation of S-129 in α-synuclein [226]. It has been observed that Mn induced the aggregation of α-synuclein and the similar effect was observed in cultured cells exposed to fungicide Maneb (having Mn) showed the same results [227].

Parkin, Pink1 and DJ-1 are involved in the formation of the heteromeric cytosolic complex (PPD complex) which is involved in Ubiquitin mediated proteasomal degradation [228]. It has been reported that Parkin can mediates its own ubiquitylation and give rise to high molecular mass k49-linked polyubiquitlylated moiety [229]. Phosphorylation at two different sites (T175, T217) was reported, but has not been confirmed by other researchers [230]. There are evidences that the activity of Parkin, choice of E2, choice of substrate, localization and solubility can be regulated by post-translational modification [231]. It has been reported that reactive cysteine (cys106) is essential for the proper functioning of DJ1 in humans [232]. Due to the increase in oxidative stress either by aggregation of α-synuclein or environmental agents promotes the oxidation of cysteine to sulfenic, sulfinic and sulfonic acid [232]. The oxidation results in the loss of the activity of DJ-1 and thus enhances the progression of PD. The phosphorylation of LRRK2 can occur in two ways constitutive and autophosphorylation. These phosphorylations are modulated in PD [233]. The mutations in the ROCO domain (Ras of complex proteins) results in the decrease of phosphorylation at R1441C, R1441G, and Y1699C, which leads to the aggregation of LRRK2 in the substantia nigra [234]. Ubiquitination also regulates the activity of LRRK2. The mutations R1441C, R1441G and Y1699C and 12020T may result in the decrease of constitutive phosphorylation or hyper-ubiquitination or instability, leading to the formation of protein aggregates [235].

8. GENOME WIDE ASSOCIATION STUDIES (GWAS)

Genome wide association studies are helpful in understanding the genetic basis of PD [236]. About 24 loci have been reported to be associated significantly with the risk of disease in the individuals of European ancestry [237]. In another study, 17 novel PD loci, using a neurocentric candidate gene nomination pipeline, found that these loci play a role in lysosomal biology and autophagy [238]. The identification of such candidate genes will definitely help in understanding the role of causal genes and will also open new horizons for the therapeutic approaches.

9. TREATMENTS FOR PARKINSON’S DISEASE

The past half century of research has brought many alluring advancements in the treatment of PD. It is a fact that PD is a progressive disease with no permanent cure, but the symptoms can be suppressed or delayed by the treatment of levodopa [239-241]. To overcome the side effects of Levodopa, there are several treatment strategies for the management of PD in order to provide more improvement in combating the PD symptoms [242]. Various treatment strategies for the PD are as follows:

9.1. Levodopa

Levodopa is also known as L-Dopa (l-3,4-dihydroxy phenylalanine). For treating early symptoms of PD the levodopa is given to the PD patients [243]. It is a precursor of dopamine which is activated through decarboxylation by the cytosolic enzyme Aromatic Amino Acid Decarboxylase (AADC). It is formed by the hydroxylation of the amino acid L-tyrosine. This reaction is catalyzed by tyrosine hydroxylase and is a rate-limiting step in the biosynthesis of the neurotransmitter dopamine. Dopamine is formed by the decarboxylation of levodopa in the presence of enzyme L-amino acid decarboxylase or Dopa Decarboxylase (DDC). The absorption of levodopa takes place first in the small intestine and enters as large neutral amino acids (LNAAs). Levodopa accesses in brain by the help of LNAAs system, where it is converted to dopamine. Once the dopamine is formed it acts on dopamine receptors (D1 and D2 families) to improve the motor functions in PD. The levodopa is absorbed by the specific LNAAs transporter i.e. protien rich meals. The therapeutic efficacy of levodopa is reduced due to the over-saturation of the transporter with amino acid derived from dietary protein. Thus, it is recommended that the levodopa should be taken without the meal to ensure the full absorption [244, 245]. In commercial preparations, levodopa is combined with carbidopa or benserazide, which act as inhibitors of decarboxylase, thus reducing the risk of peripheral side effects [246, 247].

The long term use of levodopa is associated with side effects such as nausea, vomiting, constipation and hallucinations [248, 249]. L-dopa may also induce cell death in neuronal cells by increasing the activity of caspase-3 [250, 251]. L-dopa treatment may lead to alteration in D1 dopamine receptors and might form the basis for the occurrence of dyskinesia and motor fluctuations [252, 253].

9.2. Catechol-O-methyl Transferase (COMT) inhibitors

COMT is an enzyme that metabolizes or degrades neurotransmitters such as dopamine, hence the COMT inhibitors may be useful to enhance the half-life of dopamine. Tolcapone and entacapone are commonly used in COMT inhibitors. Although, both the drugs are associated with side effects such as diarrhea, hepatotoxicity, increased dyskinesia, and urine discoloration [254, 255]. Opicapone, a third-generation COMT inhibitor, is also used to treat PD at a dosage of 50 mg daily [256]. It is prescribed in Germany and the United Kingdom, but further detailed studies are required for the validation of the efficacy and long-term risks associated with it. Most of the COMT inhibitors prevents the peripheral catabolism of L-dopa and increase the half-life of L-dopa [257, 258].

9.3. Amantadine

Although the exact mechanism of amantadine is not known, it is thought that the symptomatic relief is due to its anticholinergic property [259]. It blocks the glutamatergic N-methyl-D-aspartate (NMDA) receptors in the basal ganglia and thus helps in reducing the dyskinesias associated with L-dopa therapy [260, 261].

9.4. Anti-cholinergic Drugs

Anticholinergics (anti-muscarinics) are used to reduce tremors in PD patients. Due to neuropsychiatric side effects (memory loss, confusion) and autonomic side effects (constipation, urinary retention, dry mouth), these drugs are not normally recommended to the patients above 65 years [262]. Trihexyphenidyl, benztropine, orphenadrine, procyclidine, and biperiden are commonly use anticholinergic drugs [263-265].

9.5. Monoamine Oxidase Inhibitors (MAOIs)

They inhibit the activity of one or both monoamine oxidase enzymes i.e. Monoamine Oxidase A (MAO-A) and Monoamine Oxidase B (MAO-B). They are used as a powerful anti-depressants to treat panic disorders and social phobias [266, 267]. MAO-A shows greater affinity for hydroxylated amine, i.e. noradrenaline (NA) and serotonin (5-HT), while MAO-B shows greater affinity for non-hydroxylated amines, i.e. benzylamine and β-Phenylethylamine (PEA) [268, 269]. MAO-A degrades dopamine, norepinephrine and serotonin by oxidative deamination. It is mostly expressed in neuronal cardiac cells and is localized in the outer mitochondrial membrane [270, 271]. MAO-B enzymes are mostly expressed in the brain and regulate the storage as well as free intra-neuronal concentration of dopamine. MAO-B is involved in the breakdown of dopamine [272, 273]. It has been well established that the inhibition of MAO-B increases the level of synaptic dopamine and provides symptomatic relief [274]. Two commonly used MAO-B inhibitors are selegiline and rasagiline. Selegiline is used as adjunctive therapy to L-dopa for patients exhibiting motor fluctuations [275, 276]. Rasagiline (N-propargyl-1(R)-aminoindan) is a selective

Table 3. Pharmacology of ergot and non-ergot derivatives of dopamine agonists.
Ergoline Derivatives Non-Ergoline Derivatives
Name of Derivatives Bromocriptine Cabergoline Pergolide Lisuride Apomorphine Piribedil Pramipexole Ropinirole Rotigotine
Trade Name (s) Parlodel and Cycloset Cabaser and Dostinex Permax and Prascend Dopergin, Proclacam and Revanil Apokyn, Ixense, Spontane and Uprima Trivastal, Pronoran, Trastal and Trivastan Mirapex and Mirapex ER Requip and Requip XL Neupro
Half-life (t½) 12-14 hours 63-69 hours 27 hours 2 hours 30-60 minutes 20 hours 8-12 hours 6 hours 3 hour
Peak Plasma Time 58 minutes 2-3 hours 2-3 hours 40 min 10-60 min 1 hour IR-2 hour,
ER-6 hour
IR -1-2 hour, ER 6-10 hour 15-18 hour
Highest binding affinity D2 receptors D2 receptors D2 receptors D2 receptor D2 receptor D2 and D3 receptors D3 receptor D2 and D3 receptors D3 receptor
Chemical formula C32H40BrN5O5 C26H37N5O2 C19H26N2S C20H26N4O C17H17NO2 C16H18N4O2 C10H17N3S C16H24N2O C19H25NOS
Protein Binding (%) 90-96% 40-42% 90% 15% 50% - 15% 40% bound to plasma proteins 92%
Metabolism Gastrointestinal tract and liver Hepatic Hepatic - Hepatic - Hepatic Hepatic Hepatic
Route of Administration oral, vaginal, intravenous Oral Oral Oral Subcutaneous - Oral Oral Transdermal
Route of Elimination Liver and Kidney Urine, Fecal Kidney - - - Urine Urine Urine, Fecal

inhibitor of MAO-B and therefore does not interfere with MAO-A. It has been approved to be used as an adjunct therapeutic agent for the PD patients on L-dopa and experiencing motor fluctuations [277-279]. It has a prolonged effect on the ability to reduce the motor fluctuations associated with L-dopa therapy [280]. Safinamide, an MAO-B inhibitor, has been introduced in Europe as an adjunct therapy to levodopa. It is a selective and reversible MAO-B inhibitor [281-284].

9.6. Dopamine Agonists (DAs)

DAs are synthetic drugs which directly act on striatal postsynaptic dopamine receptors. They are reported to have a better pharmacokinetic profile than levodopa and are divided into two broad classes: Ergot derived and nonergot derived. The ergot-derived includes: Bromocriptine, cabergoline, lisuride, dihydro-ergocryptine, and pergolide [285, 286]. Non ergot-derived includes: Pramipexole, ropinirole, piribedil and rotigotine [287]. Apomorphine is used in advanced stages of PD. It acts on both D1 and D2 dopamine receptors. It displays the pharmacological profile of all clinically available DAs, but due to the short half-life and poor oral bioavailability, it is not used for the treatment of early stages of PD [288]. DAs are also associated with side effects such as nausea, day time somnolence, confusion, hallucinations, leg edema, orthostatic hypotension, and erythromelalgia (particularly ergot derivatives) [285]. The pharmacological properties of ergot and non-ergot derived drugs are listed in Table 3.

10. NONPHARMACOLOGICAL THERAPIES

Non-pharmacological therapies includes: Deep Brain Stimulation (DBS), Gene therapy and Cell transplantation.

10.1. Deep Brain Stimulation (DBS)

In DBS, the two hyperactive regions during the progression of PD i.e. Subthalamic Nucleus (STN) and Globus Pallidus internus (GPi) are used as targets. DBS blocks unusual signals through the cortico-basal ganglia loop [289]. DBS of STN and GPi-DBS showed a significant improvement in dyskinesias and motor fluctuations associated with PD patients [290, 291]. A study on low frequency (60 Hz of DBS) showed great potential to improve overall motor symptoms associated with PD, such as swallowing, gait and freezing [292-294]. Some pre-clinical studies have concluded that DBS showed improvement in the survival of dopaminergic neurons and also the levels of BDNF in the substantia nigra [295, 296]. According to Wong et al. [297], STN-DBS and GPi-DBS are equally potent in reducing tremors associated with PD patients.

10.2. Gene Therapy

Gene therapy involves creating new cells that produces a specific neurotransmitter (i.e. dopamine). The modified cells are transplanted in PD patients [298]. It is well known that enzyme Aromatic Acid Decarboxylase (AADC) plays a major role in converting the levodopa to dopamine. In PD patients, the loss of neurons in the substantia nigra leads to the loss of AADC, which leads to the inability to convert levodopa to dopamine. The gene therapy has been conducted to restore the activity of AADC in the striatum, which levodopa to dopamine [299, 300]. In 2012, the gene therapy experiment was also performed on primates for Tyrosine Hydroxylase (TH) activity in astrocytes. The results showed behavioral improvement in the rats receiving transplantation by gene therapy [301, 302].

10.3. Cell Transplantation

Cell transplantation has been used for decades and the clinical trials have shown positive results improving the motor symptoms [303-305]. In 2015, dopaminergic neurons transplantation was reported to maintain healthy and functional expression of DA transporters with normal mitochondrial morphology [306].

10.4. Physical Therapy

Physical therapy plays an important role in improving walking and postural instability. The physical therapy includes some compensatory strategies such as, motor skill learning and education to optimize physical activity. According to Morris et al. [307] management of gait disorders in people with PD should follow a critically designed training. In this training PD patients should be taught how to move more easily and maintain postural stability by using cognitive strategies. It targets the primary motor control deficits in the basal ganglia, brain stem, and motor cortex. There are two forms of strategies: (1) compensatory strategies to pass the defective basal ganglia and (2) learning strategies to improve performance through practice. Worldwide, Physical therapists provides the following measures to reduce the PD symptoms [308]:

  • Lecture on self-management.
  • Routine exercise associated with improvements in mobility, quality of life and disease severity.
  • Ways to maintain safety while performing exercises.
  • Help in normal physical activity which includes walking.
  • Practicing to maintain balance or stability.
  • Getting around (daily routine work).

CONCLUSION

Since there is no permanent cure for the disease to date, a continuous efforts in the direction of treatment strategies is the need of the hour. It is difficult to differentiate between the sporadic and familial forms of PD on the basis of symptoms. Recently, the GWAS studies have paved the pathway for studying the collective role of the loci in the progression of PD. Hence, GWAS should be conducted for the better understanding of pathology as well as treatment of PD.

CONSENT FOR PUBLICATION

Not applicable.

FUNDING

None.

CONFLICT OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

ACKNOWLEDGEMENTS

Declared none.

REFERENCES

[1] Przedborski S. Neurodegeneration. Neuroimmune Pharmacology 2008.
[2] Mills J. The history of modern psychiatry in India, 1858-1947. Hist Psychiatry 2001; 12(48 Pt 4): 431-58.
[3] Ganesan U. Medicine and modernity: The ayurvedic revival movement in india Studies on Asia, Fall 2010; 4(1)
[4] Jacoby A, Snape D, Baker GA. Epilepsy and social identity: The stigma of a chronic neurological disorder. Lancet Neurol 2005; 4(3): 171-8.
[5] Basu S. History of neurodegenerative diseases and its impact on aged population in India: An assessment. Ind J His Psych 2017; 52: 106-15.
[6] Swerdlow RH, Khan SM. The Alzheimer’s disease mitochondrial cascade hypothesis: An update. Exp Neurol 2009; 218(2): 308-15.
[7] World Health Organization WHO. Neurological disorders: Public health challenges 2006.
[8] Dominguez C, Ed. Neurodegenerative diseases 2010; Vol. 6
[9] Morris AD. James Parkinson, born April 11, 1755. Lancet 1955; 268(6867): 761-3.
[10] McCall B. Young-onset Parkinson’s disease: A guide to care and support. Nurs Times 2003; 99(30): 28-31.
[11] Martinez-Martin P, Rodriguez-Blazquez C, Kurtis MM, Chaudhuri KR. The impact of non-motor symptoms on health-related quality of life of patients with Parkinson’s disease. Mov Disord 2011; 26(3): 399-406.
[12] Noyce AJ, Bestwick JP, Silveira-Moriyama L, et al. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann Neurol 2012; 72(6): 893-901.
[13] Khoo TK, Yarnall AJ, Duncan GW, et al. The spectrum of nonmotor symptoms in early Parkinson disease. Neurology 2013; 80(3): 276-81.
[14] Duncan GW, Khoo TK, Yarnall AJ, et al. Health-related quality of life in early Parkinson’s disease: The impact of nonmotor symptoms. Mov Disord 2014; 29(2): 195-202.
[15] Reeve A, Simcox E, Turnbull D. Ageing and Parkinson’s disease: Why is advancing age the biggest risk factor? Ageing Res Rev 2014; 14: 19-30.
[16] Nambu A. A new approach to understand the pathophysiology of Parkinson’s disease. J Neurol 2005; 252(4)(Suppl. 4): IV1-4.
[17] Calabresi P, Picconi B, Tozzi A, Ghiglieri V, Di Filippo M. Direct and indirect pathways of basal ganglia: A critical reappraisal. Nat Neurosci 2014; 17(8): 1022-30.
[18] Beeler JA, Frank MJ, McDaid J, et al. A role for dopamine-mediated learning in the pathophysiology and treatment of Parkinson’s disease. Cell Rep 2012; 2(6): 1747-61.
[19] Obeso I, Wilkinson L, Casabona E, et al. Deficits in inhibitory control and conflict resolution on cognitive and motor tasks in Parkinson’s disease. Exp Brain Res 2011; 212(3): 371-84.
[20] Perreault ML, Hasbi A, O’Dowd BF, George SR. The dopamine d1-d2 receptor heteromer in striatal medium spiny neurons: evidence for a third distinct neuronal pathway in Basal Ganglia. Front Neuroanat 2011; 5: 31.
[21] Hernández-Romero MC, Delgado-Cortés MJ, Sarmiento M, et al. Peripheral inflammation increases the deleterious effect of CNS inflammation on the nigrostriatal dopaminergic system. Neurotoxicology 2012; 33(3): 347-60.
[22] Machado A, Herrera AJ, Venero JL, et al. Peripheral inflammation increases the damage in animal models of nigrostriatal dopaminergic neurodegeneration: Possible implication in Parkinson’s disease incidence 2011.
[23] Surmeier DJ, Guzmán JN, Sánchez-Padilla J, Goldberg JA. What causes the death of dopaminergic neurons in Parkinson’s disease? Prog Brain Res 2010; 183: 59-77.
[24] Venda LL, Cragg SJ, Buchman VL, Wade-Martins R. α-Synuclein and dopamine at the crossroads of Parkinson’s disease. Trends Neurosci 2010; 33(12): 559-68.
[25] Björklund A, Dunnett SB. Fifty years of dopamine research. Trends Neurosci 2007; 30(5): 185-7.
[26] Carlsson A, Lindqvist M, Magnusson T. 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature 1957; 180(4596): 1200-0.
[27] Hornykiewicz O. The discovery of dopamine deficiency in the parkinsonian brain.2006.
[28] Hornykiewicz O. How L-DOPA was discovered as a drug for Parkinson's disease 40 years ago 2001.
[29] Bergh C, Eklund T, Södersten P, Nordin C. Altered dopamine function in pathological gambling. Psychol Med 1997; 27(2): 473-5.
[30] Hornykiewicz O. A quarter century of brain dopamine research. Dopaminergic systems and their regulation 1986; 3-18.
[31] Marsden CA. Dopamine: The rewarding years. Br J Pharmacol 2006; 147(Suppl. 1): S136-44.
[32] Carlsson A, Waldeck B. A fluorimetric method for the determination of dopamine (3-hydroxytyramine). Acta Physiol Scand 1958; 44(3-4): 293-8.
[33] Bertler A, Rosengren E. Occurrence and distribution of dopamine in brain and other tissues. Experientia 1959; 15(1): 10-1.
[34] Portig PJ, Vogt M. Release to the cerebral ventricles of substances with possible transmitter function in the cnadate nucleus Foresight-Brain Science, Addiction and Drugs www.foresight.gov.uk1969.
[35] Dahlström A. Aminergic transmission. Introduction and short review. Brain Res 1973; 62(2): 441-60.
[36] Mishra A, Singh S, Shukla S. Physiological and functional basis of dopamine receptors and their role in neurogenesis: Possible implication for parkinson’s disease. J Exp Neurosci 2018; 12: 1179069518779829.
[37] Contreras F, Fouillioux C, Bolívar A, et al. Dopamine, hypertension and obesity. J Hum Hypertens 2002; 16(1)(Suppl. 1): S13-7.
[38] Kebabian JW, Petzold GL, Greengard P. Dopamine-sensitive adenylate cyclase in caudate nucleus of rat brain, and its similarity to the “dopamine receptor”. Proc Natl Acad Sci USA 1972; 69(8): 2145-9.
[39] Gaspar P, Bloch B, Le Moine C. D1 and D2 receptor gene expression in the rat frontal cortex: Cellular localization in different classes of efferent neurons. Eur J Neurosci 1995; 7(5): 1050-63.
[40] Jaber M, Robinson SW, Missale C, Caron MG. Dopamine receptors and brain function. Neuropharmacology 1996; 35(11): 1503-19.
[41] Müller U, von Cramon DY, Pollmann S. D1- versus D2-receptor modulation of visuospatial working memory in humans. J Neurosci 1998; 18(7): 2720-8.
[42] Nishi A, Fisone G, Snyder GL, et al. Regulation of Na+, K+-ATPase isoforms in rat neostriatum by dopamine and protein kinase C. J Neurochem 1999; 73(4): 1492-501.
[43] Ha CM, Park D, Han JK, et al. Calcyon forms a novel ternary complex with dopamine D1 receptor through PSD-95 protein and plays a role in dopamine receptor internalization. J Biol Chem 2012; 287(38): 31813-22.
[44] Gomes P, Soares-da-Silva P. Na(+)/H(+) exchanger activity and dopamine D(1)-like receptor function in two opossum kidney cell clonal sublines. Cell Physiol Biochem 2002; 12(5-6): 259-68.
[45] Mercuri NB, Saiardi A, Bonci A, et al. Loss of autoreceptor function in dopaminergic neurons from dopamine D2 receptor deficient mice. Neuroscience 1997; 79(2): 323-7.
[46] McAllister G, Knowles MR, Ward-Booth SM, et al. Functional coupling of human D2, D3, and D4 dopamine receptors in HEK293 cells. J Recept Signal Transduct Res 1995; 15(1-4): 267-81.
[47] Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: Functions, signaling, and association with neurological diseases. Cell Mol Neurobiol 2019; 39(1): 31-59.
[48] Moore DJ, West AB, Dawson VL, Dawson TM. Molecular pathophysiology of Parkinson’s disease. Annu Rev Neurosci 2005; 28: 57-87.
[49] Lansbury PT Jr, Brice A. Genetics of Parkinson’s disease and biochemical studies of implicated gene products. Curr Opin Cell Biol 2002; 14(5): 653-60.
[50] Rocca WA, McDonnell SK, Strain KJ, et al. Familial aggregation of Parkinson’s disease: The Mayo Clinic family study. Ann Neurol 2004; 56(4): 495-502.
[51] Pearce JMS. The Lewy body. J Neurol Neurosurg Psychiatry 2001; 71(2): 214-5.
[52] Schiller F. Fritz Lewy and his bodies. J Hist Neurosci 2000; 9(2): 148-51.
[53] Zarranz JJ, Alegre J, Gómez-Esteban JC, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004; 55(2): 164-73.
[54] 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(11): 6469-73.
[55] Kon T, Tomiyama M, Wakabayashi K. Neuropathology of Lewy body disease: Clinicopathological crosstalk between typical and atypical cases. Neuropathology 2020; 40(1): 30-9.
[56] Konno T, Ross OA, Puschmann A, Dickson DW, Wszolek ZK. Autosomal dominant Parkinson’s disease caused by SNCA duplications. Parkinsonism Relat Disord 2016; 22(Suppl. 1): S1-6.
[57] Luk KC, Kehm V, Carroll J, et al. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 2012; 338(6109): 949-53.
[58] Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997; 276(5321): 2045-7.
[59] Lim KL, Dawson VL, Dawson TM. The genetics of Parkinson’s disease. Curr Neurol Neurosci Rep 2002; 2(5): 439-46.
[60] Mouradian MM. Recent advances in the genetics and pathogenesis of Parkinson disease. Neurology 2002; 58(2): 179-85.
[61] Uéda K, Fukushima H, Masliah E, et al. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc Natl Acad Sci USA 1993; 90(23): 11282-6.
[62] Jakes R, Spillantini MG, Goedert M. Identification of two distinct synucleins from human brain. FEBS Lett 1994; 345(1): 27-32.
[63] Clayton DF, George JM. The synucleins: A family of proteins involved in synaptic function, plasticity, neurodegeneration and disease. Trends Neurosci 1998; 21(6): 249-54.
[64] Uversky VN. A protein-chameleon: Conformational plasticity of α-synuclein, a disordered protein involved in neurodegenerative disorders. J Biomol Struct Dyn 2003; 21(2): 211-34.
[65] Shulman JM, De Jager PL, Feany MB. Parkinson’s disease: Genetics and pathogenesis. Annu Rev Pathol 2011; 6: 193-222.
[66] Devine MJ, Gwinn K, Singleton A, Hardy J. Parkinson’s disease and α-synuclein expression. Mov Disord 2011; 26(12): 2160-8.
[67] Kalia LV, Lang AE. Parkinson disease in 2015: Evolving basic, pathological and clinical concepts in PD. Nat Rev Neurol 2016; 12(2): 65-6.
[68] Lee S, Imai Y, Gehrke S, Liu S, Lu B. The synaptic function of LRRK2. Biochem Soc Trans 2012; 40(5): 1047-51.
[69] Spillantini MG, Aloe L, Alleva E, et al. Nerve growth factor mRNA and protein increase in hypothalamus in a mouse model of aggression.World Scientific Series in 20th Century Biology. 1997.
[70] Chartier-Harlin MC, Kachergus J, Roumier C, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 2004; 364(9440): 1167-9.
[71] Totterdell S, Meredith GE. Localization of alpha-synuclein to identified fibers and synapses in the normal mouse brain. Neuroscience 2005; 135(3): 907-13.
[72] Maraganore DM, de Andrade M, Elbaz A, et al. Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease. JAMA 2006; 296(6): 661-70.
[73] Vaughan J, Durr A, Tassin J, et al. The alpha-synuclein Ala53Thr mutation is not a common cause of familial Parkinson’s disease: A study of 230 European cases. Ann Neurol 1998; 44(2): 270-3.
[74] Jensen PH, Nielsen MS, Jakes R, Dotti CG, Goedert M. Binding of alpha-synuclein to brain vesicles is abolished by familial Parkinson’s disease mutation. J Biol Chem 1998; 273(41): 26292-4.
[75] Conway KA, Harper JD, Lansbury PT. Accelerated in vitro fibril formation by a mutant alpha-synuclein linked to early-onset Parkinson disease. Nat Med 1998; 4(11): 1318-20.
[76] Krüger R, Kuhn W, Müller T, et al. Ala30Pro mutation in the gene encoding α-synuclein in Parkinson’s disease. Nat Genet 1998; 18(2): 106-8.
[77] Klein C, Schlossmacher MG. The genetics of Parkinson disease: Implications for neurological care. Nat Clin Pract Neurol 2006; 2(3): 136-46.
[78] Athanassiadou A, Voutsinas G, Psiouri L, et al. Genetic analysis of families with Parkinson disease that carry the Ala53Thr mutation in the gene encoding alpha-synuclein. Am J Hum Genet 1999; 65(2): 555-8.
[79] Spira PJ, Sharpe DM, Halliday G, Cavanagh J, Nicholson GA. Clinical and pathological features of a Parkinsonian syndrome in a family with an Ala53Thr α-synuclein mutation. Ann Neurol 2001; 49(3): 313-9.
[80] Ki CS, Stavrou EF, Davanos N, et al. The Ala53Thr mutation in the α-synuclein gene in a Korean family with Parkinson disease. Clin Genet 2007; 71(5): 471-3.
[81] Choi JM, Woo MS, Ma HI, et al. Analysis of PARK genes in a Korean cohort of early-onset Parkinson disease. Neurogenetics 2008; 9(4): 263-9.
[82] Puschmann A, Ross OA, Vilariño-Güell C, et al. A Swedish family with de novo α-synuclein A53T mutation: Evidence for early cortical dysfunction. Parkinsonism Relat Disord 2009; 15(9): 627-32.
[83] Golbe LI. α-synuclein and Parkinson’s disease. Mov Disord 1999; 14(1): 6-9.
[84] Trinh J, Guella I, Farrer MJ. Disease penetrance of late-onset parkinsonism: A meta-analysis. JAMA Neurol 2014; 71(12): 1535-9.
[85] Nishioka K, Hayashi S, Farrer MJ, et al. Clinical heterogeneity of α-synuclein gene duplication in Parkinson’s disease. Ann Neurol 2006; 59(2): 298-309.
[86] Xia Y, Saitoh T, Uéda K, et al. Characterization of the human α-synuclein gene: Genomic structure, transcription start site, promoter region and polymorphisms. J Alzheimers Dis 2001; 3(5): 485-94.
[87] Gámez-Valero A, Beyer K. Alternative splicing of alpha- and beta-synuclein genes plays differential roles in synucleinopathies. Genes (Basel) 2018; 9(2): 63.
[88] Giasson BI, Murray IV, Trojanowski JQ, Lee VM. A hydrophobic stretch of 12 amino acid residues in the middle of α-synuclein is essential for filament assembly. J Biol Chem 2001; 276(4): 2380-6.
[89] Bitan G, Kirkitadze MD, Lomakin A, Vollers SS, Benedek GB, Teplow DB. Amyloid beta -protein (Abeta) assembly: Abeta 40 and Abeta 42 oligomerize through distinct pathways. Proc Natl Acad Sci USA 2003; 100(1): 330-5.
[90] Lashuel HA, Overk CR, Oueslati A, Masliah E. The many faces of α-synuclein: From structure and toxicity to therapeutic target. Nat Rev Neurosci 2013; 14(1): 38-48.
[91] Bertoncini CW, Jung YS, Fernandez CO, et al. Release of long-range tertiary interactions potentiates aggregation of natively unstructured alpha-synuclein. Proc Natl Acad Sci USA 2005; 102(5): 1430-5.
[92] Mazzulli JR, Xu YH, Sun Y, et al. Gaucher disease glucocerebrosidase and α-synuclein form a bidirectional pathogenic loop in synucleinopathies. Cell 2011; 146(1): 37-52.
[93] Goker-Alpan O, Giasson BI, Eblan MJ, et al. Glucocerebrosidase mutations are an important risk factor for Lewy body disorders. Neurology 2006; 67(5): 908-10.
[94] Lesage S, Leutenegger AL, Ibanez P, et al. LRRK2 haplotype analyses in European and North African families with Parkinson disease: A common founder for the G2019S mutation dating from the 13th century. Am J Hum Genet 2005; 77(2): 330-2.
[95] Wallings R, Manzoni C, Bandopadhyay R. Cellular processes associated with LRRK2 function and dysfunction. FEBS J 2015; 282(15): 2806-26.
[96] Yakhine-Diop SM, Rodríguez-Arribas M, Gómez-Sánchez R, et al. G2019S mutation of lrrk2 increases autophagy via MEK/ERK pathway autophagy: Cancer, other pathologies, inflammation, immunity, infection, and aging 2016; 123-42.
[97] Gillardon F, Schmid R, Draheim H. Parkinson’s disease-linked leucine-rich repeat kinase 2(R1441G) mutation increases proinflammatory cytokine release from activated primary microglial cells and resultant neurotoxicity. Neuroscience 2012; 208: 41-8.
[98] Chen ML, Wu RM. LRRK 2 gene mutations in the pathophysiology of the ROCO domain and therapeutic targets for Parkinson’s disease: A review. J Biomed Sci 2018; 25(1): 52.
[99] Pimentel MMG, Moura KCV, Abdalla CB, et al. A study of LRRK2 mutations and Parkinson’s disease in Brazil. Neurosci Lett 2008; 433(1): 17-21.
[100] Brice A. Genetics of Parkinson’s disease: LRRK2 on the rise. Brain 2005; 128(Pt 12): 2760-2.
[101] Lesage S, Dürr A, Tazir M, et al. LRRK2 G2019S as a cause of Parkinson’s disease in North African Arabs. N Engl J Med 2006; 354(4): 422-3.
[102] Ozelius LJ, Senthil G, Saunders-Pullman R, et al. LRRK2 G2019S as a cause of Parkinson’s disease in Ashkenazi Jews. N Engl J Med 2006; 354(4): 424-5.
[103] Nuytemans K, Bademci G, Inchausti V, et al. Whole exome sequencing of rare variants in EIF4G1 and VPS35 in Parkinson disease. Neurology 2013; 80(11): 982-9.
[104] Clark IE, Dodson MW, Jiang C, et al. Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin. Nature 2006; 441(7097): 1162-6.
[105] Zabetian CP, Hutter CM, Yearout D, et al. LRRK2 G2019S in families with Parkinson disease who originated from Europe and the Middle East: Evidence of two distinct founding events beginning two millennia ago. Am J Hum Genet 2006; 79(4): 752-8.
[106] Klein C, Lohmann-Hedrich K, Rogaeva E, Schlossmacher MG, Lang AE. Deciphering the role of heterozygous mutations in genes associated with parkinsonism. Lancet Neurol 2007; 6(7): 652-62.
[107] Simón-Sánchez J, Herranz-Pérez V, Olucha-Bordonau F, Pérez-Tur J. LRRK2 is expressed in areas affected by Parkinson’s disease in the adult mouse brain. Eur J Neurosci 2006; 23(3): 659-66.
[108] Gorostidi A, Ruiz-Martínez J, Lopez de Munain A, Alzualde A, Martí Massó JF. LRRK2 G2019S and R1441G mutations associated with Parkinson’s disease are common in the Basque Country, but relative prevalence is determined by ethnicity. Neurogenetics 2009; 10(2): 157-9.
[109] Tomiyama H, Li Y, Funayama M, et al. Clinicogenetic study of mutations in LRRK2 exon 41 in Parkinson’s disease patients from 18 countries. Mov Disord 2006; 21(8): 1102-8.
[110] Haugarvoll K, Rademakers R, Kachergus JM, et al. Lrrk2 R1441C parkinsonism is clinically similar to sporadic Parkinson disease. Neurology 2008; 70(16 Pt 2): 1456-60.
[111] MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A. The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron 2006; 52(4): 587-93.
[112] Follett J, Norwood SJ, Hamilton NA, et al. The Vps35 D620N mutation linked to Parkinson’s disease disrupts the cargo sorting function of retromer. Traffic 2014; 15(2): 230-44.
[113] Bonifacino JS, Hurley JH. Retromer. Curr Opin Cell Biol 2008; 20(4): 427-36.
[114] Deng H, Wu Y, Jankovic J. The EIF4G1 gene and Parkinson’s disease. Acta Neurol Scand 2015; 132(2): 73-8.
[115] Gustavsson EK, Trinh J, Guella I, et al. DNAJC13 genetic variants in parkinsonism. Mov Disord 2015; 30(2): 273-8.
[116] Yoshida S, Hasegawa T, Suzuki M, et al. Parkinson’s disease-linked DNAJC13 mutation aggravates alpha-synuclein-induced neurotoxicity through perturbation of endosomal trafficking. Hum Mol Genet 2018; 27(5): 823-36.
[117] Meng H, Yamashita C, Shiba-Fukushima K, et al. Loss of Parkinson’s disease-associated protein CHCHD2 affects mitochondrial crista structure and destabilizes cytochrome c. Nat Commun 2017; 8: 15500.
[118] Funayama M, Ohe K, Amo T, et al. CHCHD2 mutations in autosomal dominant late-onset Parkinson’s disease: A genome-wide linkage and sequencing study. Lancet Neurol 2015; 14(3): 274-82.
[119] Jansen IE, Bras JM, Lesage S, et al. CHCHD2 and Parkinson’s disease. Lancet Neurol 2015; 14(7): 678-9.
[120] Chan NC, Salazar AM, Pham AH, et al. Broad activation of the ubiquitin-proteasome system by Parkin is critical for mitophagy. Hum Mol Genet 2011; 20(9): 1726-37.
[121] Deas E, Plun-Favreau H, Gandhi S, et al. PINK1 cleavage at position A103 by the mitochondrial protease PARL. Hum Mol Genet 2011; 20(5): 867-79.
[122] Jin SM, Lazarou M, Wang C, Kane LA, Narendra DP, Youle RJ. Mitochondrial membrane potential regulates PINK1 import and proteolytic destabilization by PARL. J Cell Biol 2010; 191(5): 933-42.
[123] Thomas KJ, McCoy MK, Blackinton J, et al. DJ-1 acts in parallel to the PINK1/parkin pathway to control mitochondrial function and autophagy. Hum Mol Genet 2011; 20(1): 40-50.
[124] Lücking CB, Dürr A, Bonifati V, et al. Association between early-onset Parkinson’s disease and mutations in the parkin gene. N Engl J Med 2000; 342(21): 1560-7.
[125] Periquet M, Latouche M, Lohmann E, et al. Parkin mutations are frequent in patients with isolated early-onset parkinsonism. Brain 2003; 126(Pt 6): 1271-8.
[126] Djarmati A, Hedrich K, Svetel M, et al. Detection of Parkin (PARK2) and DJ1 (PARK7) mutations in early-onset Parkinson disease: Parkin mutation frequency depends on ethnic origin of patients. Hum Mutat 2004; 23(5): 525.
[127] Hedrich K, Djarmati A, Schäfer N, et al. DJ-1 (PARK7) mutations are less frequent than Parkin (PARK2) mutations in early-onset Parkinson disease. Neurology 2004; 62(3): 389-94.
[128] Singleton AB, Farrer MJ, Bonifati V. The genetics of Parkinson’s disease: Progress and therapeutic implications. Mov Disord 2013; 28(1): 14-23.
[129] Rogaeva E, Johnson J, Lang AE, et al. Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease. Arch Neurol 2004; 61(12): 1898-904.
[130] Klein C, Djarmati A, Hedrich K, et al. PINK1, Parkin, and DJ-1 mutations in Italian patients with early-onset parkinsonism. Eur J Hum Genet 2005; 13(9): 1086-93.
[131] Deng H, Le WD, Hunter CB, et al. Heterogeneous phenotype in a family with compound heterozygous parkin gene mutations. Arch Neurol 2006; 63(2): 273-7.
[132] Yang X, Xu Y. Mutations in the ATP13A2 gene and Parkinsonism: A preliminary review. BioMed Res Internat 2014.
[133] Dehay B, Ramirez A, Martinez-Vicente M, et al. Loss of P-type ATPase ATP13A2/PARK9 function induces general lysosomal deficiency and leads to Parkinson disease neurodegeneration. Proc Natl Acad Sci USA 2012; 109(24): 9611-6.
[134] Conedera S, Apaydin H, Li Y, et al. FBXO7 mutations in Parkinson’s disease and multiple system atrophy. Neurobiol Aging 2016; 40: 192.e1-5.
[135] Joseph S, Schulz JB, Stegmüller J. Mechanistic contributions of FBXO7 to Parkinson disease. J Neurochem 2018; 144(2): 118-27.
[136] Paisan-Ruiz C, Bhatia KP, Li A, et al. Characterization of PLA2G6 as a locus for dystonia-parkinsonism. Ann Neurol 2009; 65(1): 19-23.
[137] Bohlega SA, Al-Mubarak BR, Alyemni EA, et al. Clinical heterogeneity of PLA2G6-related Parkinsonism: Analysis of two Saudi families BMC Re Not 2016; 9(1): 295.
[138] Tiangyou W, Hudson G, Ghezzi D, et al. POLG1 in idiopathic Parkinson disease. Neurology 2006; 67(9): 1698-700.
[139] Gui YX, Xu ZP, Lv W, Liu HM, Zhao JJ, Hu XY. Association of mitochondrial DNA polymerase γ gene POLG1 polymorphisms with parkinsonism in Chinese populations. PLoS One 2012; 7(12): e50086.
[140] Drouet V, Lesage S. Synaptojanin 1 mutation in Parkinson’s disease brings further insight into the neuropathological mechanisms. BioMed Res Internat 2014.
[141] Fasano D, Parisi S, Pierantoni GM, et al. Alteration of endosomal trafficking is associated with early-onset parkinsonism caused by SYNJ1 mutations. Cell Death Dis 2018; 9(3): 385.
[142] Fung HC, Scholz S, Matarin M, et al. Genome-wide genotyping in Parkinson’s disease and neurologically normal controls: First stage analysis and public release of data. Lancet Neurol 2006; 5(11): 911-6.
[143] Kobayashi T, Wang M, Hattori N, Matsumine H, Kondo T, Mizuno Y. Exonic deletion mutations of the Parkin gene among sporadic patients with Parkinson’s disease. Parkinsonism Relat Disord 2000; 6(3): 129-31.
[144] Mizuno Y, Hattori N, Mori H, Suzuki T, Tanaka K. Parkin and Parkinson’s disease. Curr Opin Neurol 2001; 14(4): 477-82.
[145] Dawson TM, Dawson VL. The role of parkin in familial and sporadic Parkinson’s disease. Mov Disord 2010; 25(S1)(Suppl. 1): S32-9.
[146] Deshaies RJ, Joazeiro CA. RING domain E3 ubiquitin ligases. Annu Rev Biochem 2009; 78: 399-434.
[147] Shimura H, Schwartz D, Gygi SP, Kosik KS. CHIP-Hsc70 complex ubiquitinates phosphorylated tau and enhances cell survival. J Biol Chem 2004; 279(6): 4869-76.
[148] Bordallo J, Plemper RK, Finger A, Wolf DH. Der3p/Hrd1p is required for endoplasmic reticulum-associated degradation of misfolded lumenal and integral membrane proteins. Mol Biol Cell 1998; 9(1): 209-22.
[149] Imai Y, Soda M, Inoue H, Hattori N, Mizuno Y, Takahashi R. An unfolded putative transmembrane polypeptide, which can lead to endoplasmic reticulum stress, is a substrate of Parkin. Cell 2001; 105(7): 891-902.
[150] Chung KK, Zhang Y, Lim KL, et al. Parkin ubiquitinates the α-synuclein-interacting protein, synphilin-1: Implications for Lewy-body formation in Parkinson disease. Nat Med 2001; 7(10): 1144-50.
[151] Valente EM, Salvi S, Ialongo T, et al. PINK1 mutations are associated with sporadic early-onset parkinsonism. Ann Neurol 2004; 56(3): 336-41.
[152] Poulogiannis G, McIntyre RE, Dimitriadi M, et al. PARK2 deletions occur frequently in sporadic colorectal cancer and accelerate adenoma development in Apc mutant mice. Proc Natl Acad Sci USA 2010; 107(34): 15145-50.
[153] Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol 2011; 12(1): 9-14.
[154] Healy DG, Falchi M, O’Sullivan SS, et al. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: A case-control study. Lancet Neurol 2008; 7(7): 583-90.
[155] Bonifati V, Rohé CF, Breedveld GJ, et al. Early-onset parkinsonism associated with PINK1 mutations: Frequency, genotypes, and phenotypes. Neurology 2005; 65(1): 87-95.
[156] Li Y, Tomiyama H, Sato K, et al. Clinicogenetic study of PINK1 mutations in autosomal recessive early-onset parkinsonism. Neurology 2005; 64(11): 1955-7.
[157] Cazeneuve C, Sân C, Ibrahim SA, et al. A new complex homozygous large rearrangement of the PINK1 gene in a Sudanese family with early onset Parkinson’s disease. Neurogenetics 2009; 10(3): 265-70.
[158] Camargos ST, Dornas LO, Momeni P, et al. Familial Parkinsonism and early onset Parkinson’s disease in a Brazilian movement disorders clinic: Phenotypic characterization and frequency of SNCA, PRKN, PINK1, and LRRK2 mutations. Mov Disord 2009; 24(5): 662-6.
[159] Marongiu R, Brancati F, Antonini A, et al. Whole gene deletion and splicing mutations expand the PINK1 genotypic spectrum. Hum Mutat 2007; 28(1): 98.
[160] Klein C, Westenberger A. Genetics of Parkinson’s disease. Cold Spring Harb Perspect Med 2012; 2(1): a008888.
[161] Pankratz N, Pauciulo MW, Elsaesser VE, et al. Mutations in DJ-1 are rare in familial Parkinson disease. Neurosci Lett 2006; 408(3): 209-13.
[162] Dolgacheva LP, Berezhnov AV, Fedotova EI, Zinchenko VP, Abramov AY. Role of DJ-1 in the mechanism of pathogenesis of Parkinson’s disease. J Bioenerg Biomembr 2019; 51(3): 175-88.
[163] Takahashi-Niki K, Niki T, Taira T, Iguchi-Ariga SM, Ariga H. Reduced anti-oxidative stress activities of DJ-1 mutants found in Parkinson’s disease patients. Biochem Biophys Res Commun 2004; 320(2): 389-97.
[164] Ariga H, Takahashi-Niki K, Kato I, et al. Neuroprotective function of DJ-1 in Parkinson’s disease 2013.
[165] Canet-Avilés RM, Wilson MA, Miller DW, et al. The Parkinson’s disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization. Proc Natl Acad Sci USA 2004; 101(24): 9103-8.
[166] Macedo MG, Anar B, Bronner IF, et al. The DJ-1L166P mutant protein associated with early onset Parkinson’s disease is unstable and forms higher-order protein complexes. Hum Mol Genet 2003; 12(21): 2807-16.
[167] Williams DR, Hadeed A, al-Din ASN, Wreikat AL, Lees AJ. Kufor Rakeb disease: Autosomal recessive, levodopa-responsive parkinsonism with pyramidal degeneration, supranuclear gaze palsy, and dementia. Mov Disord 2005; 20(10): 1264-71.
[168] Ramirez A, Heimbach A, Gründemann J, et al. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat Genet 2006; 38(10): 1184-91.
[169] Selvaraj S, Piramanayagam S. Impact of gene mutation in the development of Parkinson’s disease 2019.
[170] Bras J, Verloes A, Schneider SA, et al. Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis. Hum Mol Gen 2012; 21(12): 2646-50.
[171] Hruska KS, LaMarca ME, Scott CR, Sidransky E. Gaucher disease: Mutation and polymorphism spectrum in the glucocerebrosidase gene (GBA). Hum Mutat 2008; 29(5): 567-83.
[172] Goker-Alpan O, Schiffmann R, LaMarca ME, Nussbaum RL, McInerney-Leo A, Sidransky E. Parkinsonism among Gaucher disease carriers. J Med Genet 2004; 41(12): 937-40.
[173] Lwin A, Orvisky E, Goker-Alpan O, LaMarca ME, Sidransky E. Glucocerebrosidase mutations in subjects with parkinsonism. Mol Genet Metab 2004; 81(1): 70-3.
[174] Eblan MJ, Walker JM, Sidransky E. The glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N Engl J Med 2005; 352(7): 728-31.
[175] 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 1999; 45(5): 577-82.
[176] Farrer M, Maraganore DM, Lockhart P, et al. alpha-Synuclein gene haplotypes are associated with Parkinson’s disease. Hum Mol Genet 2001; 10(17): 1847-51.
[177] Hardy J, Cai H, Cookson MR, Gwinn-Hardy K, Singleton A. Genetics of Parkinson’s disease and parkinsonism. Ann Neurol 2006; 60(4): 389-98.
[178] Chai C, Lim KL. Genetic insights into sporadic Parkinson’s disease pathogenesis. Curr Genomics 2013; 14(8): 486-501.
[179] Ahfeldt T, Ordureau A, Bell C, et al. Pathogenic pathways in early-onset autosomal recessive parkinson’s disease discovered using isogenic human dopaminergic neurons. Stem Cell Reports 2020; 14(1): 75-90.
[180] Lehtonen Š, Sonninen TM, Wojciechowski S, Goldsteins G, Koistinaho J. Dysfunction of cellular proteostasis in Parkinsons disease. Front Neurosci 2019; 13: 457.
[181] Al-Chalabi A, Dürr A, Wood NW, et al. Genetic variants of the α-synuclein gene SNCA are associated with multiple system atrophy. PLoS One 2009; 4(9)e7114
[182] Stefanis L. α-Synuclein in Parkinson’s disease. Cold Spring Harb Perspect Med 2012; 2(2)a009399
[183] Oczkowska A, Florczak-Wyspianska J, Permoda-Osip A, et al. Analysis of PRKN variants and clinical features in Polish patients with Parkinson’s disease. Curr Genomics 2015; 16(4): 215-23.
[184] Mata IF, Lockhart PJ, Farrer MJ. Parkin genetics: One model for Parkinson’s disease. Hum Mol Genet 2004; 13(Spec No 1): R127-33.
[185] Ragland M, Hutter C, Zabetian C, Edwards K. Association between the ubiquitin carboxyl-terminal esterase L1 gene (UCHL1) S18Y variant and Parkinson’s Disease: A HuGE review and meta-analysis. Am J Epidemiol 2009; 170(11): 1344-57.
[186] Siuda J, Jasinska-Myga B, Boczarska-Jedynak M, et al. Early-onset Parkinson’s disease due to PINK1 p.Q456X mutation-clinical and functional study. Parkinsonism Relat Disord 2014; 20(11): 1274-8.
[187] Geisler S, Holmström KM, Skujat D, et al. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol 2010; 12(2): 119-31.
[188] Eerola J, Luoma PT, Peuralinna T, et al. POLG1 polyglutamine tract variants associated with Parkinson’s disease. Neurosci Lett 2010; 477(1): 1-5.
[189] Bandettini di Poggio M, Nesti C, Bruno C, Meschini MC, Schenone A, Santorelli FM. Dopamine-agonist responsive Parkinsonism in a patient with the SANDO syndrome caused by POLG mutation. BMC Med Genet 2013; 14(1): 105.
[190] Luoma P, Melberg A, Rinne JO, et al. Parkinsonism, premature menopause, and mitochondrial DNA polymerase γ mutations: Clinical and molecular genetic study. Lancet 2004; 364(9437): 875-82.
[191] Shu L, Zhang Y, Sun Q, Pan H, Tang B. A comprehensive analysis of population differences in LRRK2 variant distribution in Parkinson’s disease. Front Aging Neurosci 2019; 11: 13.
[192] Higashi S, Biskup S, West AB, et al. Localization of Parkinson’s disease-associated LRRK2 in normal and pathological human brain. Brain Res 2007; 1155: 208-19.
[193] Lin CH, Chen ML, Chen GS, Tai CH, Wu RM. Novel variant Pro143Ala in HTRA2 contributes to Parkinson’s disease by inducing hyperphosphorylation of HTRA2 protein in mitochondria. Hum Genet 2011; 130(6): 817-27.
[194] Ross OA, Soto AI, Vilariño-Güell C, et al. Genetic variation of Omi/HtrA2 and Parkinson’s disease. Parkinsonism Relat Disord 2008; 14(7): 539-43.
[195] Unal Gulsuner H, Gulsuner S, Mercan FN, et al. Mitochondrial serine protease HTRA2 p.G399S in a kindred with essential tremor and Parkinson disease. Proc Natl Acad Sci USA 2014; 111(51): 18285-90.
[196] Podhajska A, Musso A, Trancikova A, et al. Common pathogenic effects of missense mutations in the P-type ATPase ATP13A2 (PARK9) associated with early-onset parkinsonism. PLoS One 2012; 7(6)e39942
[197] Najim al-Din AS, Wriekat A, Mubaidin A, Dasouki M, Hiari M. Pallido-pyramidal degeneration, supranuclear upgaze paresis and dementia: Kufor-Rakeb syndrome. Acta Neurol Scand 1994; 89(5): 347-52.
[198] Zhao T, De Graaff E, Breedveld GJ, et al. Loss of nuclear activity of the FBXO7 protein in patients with parkinsonian-pyramidal syndrome (PARK15). PLoS One 2011; 6(2)e16983
[199] Zhang Y, Sun QY, Yu RH, Guo JF, Tang BS, Yan XX. The contribution of GIGYF2 to Parkinson’s disease: A meta-analysis. Neurol Sci 2015; 36(11): 2073-9.
[200] Higashi S, Iseki E, Minegishi M, Togo T, Kabuta T, Wada K. GIGYF2 is present in endosomal compartments in the mammalian brains and enhances IGF-1-induced ERK1/2 activation. J Neurochem 2010; 115(2): 423-37.
[201] Mullin S, Hughes D, Mehta A, Schapira AHV. Neurological effects of glucocerebrosidase gene mutations. Eur J Neurol 2019; 26(3): 388-e29.
[202] Moors TE, Paciotti S, Ingrassia A, et al. Characterization of brain lysosomal activities in GBA-related and sporadic Parkinson’s disease and dementia with Lewy bodies. Mol Neurobiol 2019; 56(2): 1344-55.
[203] Tomiyama H, Yoshino H, Ogaki K, et al. PLA2G6 variant in Parkinson’s disease. J Hum Genet 2011; 56(5): 401-3.
[204] Shen T, Hu J, Jiang Y, et al. Early-onset Parkinson’s disease caused by PLA2G6 compound heterozygous mutation, a case report and literature review. Front Neurol 2019; 10: 915.
[205] Tsika E, Glauser L, Moser R, et al. Parkinson’s disease-linked mutations in VPS35 induce dopaminergic neurodegeneration. Hum Mol Genet 2014; 23(17): 4621-38.
[206] Deutschländer A, Ross OA, Wszolek ZK. VPS35-Related Parkinson Disease.GeneReviews 2017.
[207] Williams ET, Chen X, Moore DJ. VPS35, the retromer complex and Parkinson’s disease. J Parkinsons Dis 2017; 7(2): 219-33.
[208] Weng YC, Chen CM, Chen YC, et al. Eukaryotic translation initiation factor 4-γ, 1 gene mutations are rare in Parkinson’s disease among Taiwanese. J Formos Med Assoc 2016; 115(9): 728-33.
[209] Edvardson S, Cinnamon Y, Ta-Shma A, et al. A deleterious mutation in DNAJC6 encoding the neuronal-specific clathrin-uncoating co-chaperone auxilin, is associated with juvenile parkinsonism. PLoS One 2012; 7(5)e36458
[210] Köroğlu Ç, Baysal L, Cetinkaya M, Karasoy H, Tolun A. DNAJC6 is responsible for juvenile parkinsonism with phenotypic variability. Parkinsonism Relat Disord 2013; 19(3): 320-4.
[211] Bouhouche A, Tesson C, Regragui W, et al. Mutation analysis of consanguineous moroccan patients with Parkinson’s disease combining microarray and gene panel. Front Neurol 2017; 8: 567.
[212] Haffner C, Takei K, Chen H, et al. Synaptojanin 1: Localization on coated endocytic intermediates in nerve terminals and interaction of its 170 kDa isoform with Eps15. FEBS Lett 1997; 419(2-3): 175-80.
[213] Roosen DA, Blauwendraat C, Cookson MR, Lewis PA. DNAJC proteins and pathways to parkinsonism. FEBS J 2019; 286(16): 3080-94.
[214] Vilariño-Güell C, Rajput A, Milnerwood AJ, et al. DNAJC13 mutations in Parkinson disease. Hum Mol Genet 2014; 23(7): 1794-801.
[215] Imai Y, Meng H, Shiba-Fukushima K, Hattori N. Twin CHCH Proteins, CHCHD2, and CHCHD10: Key molecules of Parkinson’s disease, amyotrophic lateral sclerosis, and frontotemporal dementia. Int J Mol Sci 2019; 20(4): 908.
[216] Lee RG, Sedghi M, Salari M, et al. Early-onset Parkinson disease caused by a mutation in CHCHD2 and mitochondrial dysfunction. Neurol Genet 2018; 4(5): e276.
[217] Brodeur J, Thériault C, Lessard-Beaudoin M, Marcil A, Dahan S, Lavoie C. LDLR-related protein 10 (LRP10) regulates amyloid precursor protein (APP) trafficking and processing: Evidence for a role in Alzheimer’s disease. Mol Neurodegener 2012; 7(1): 31.
[218] Willnow TE. The low-density lipoprotein receptor gene family: Multiple roles in lipid metabolism. J Mol Med (Berl) 1999; 77(3): 306-15.
[219] Quadri M, Mandemakers W, Grochowska MM, et al. LRP10 genetic variants in familial Parkinson’s disease and dementia with Lewy bodies: A genome-wide linkage and sequencing study. Lancet Neurol 2018; 17(7): 597-608.
[220] Chakraborty J, Basso V, Ziviani E. Post translational modification of Parkin. Biol Direct 2017; 12(1): 6.
[221] Pan B, Petersson EJ, Rhoades E. Investigating the effect of alpha-synuclein post-translational modifications on synaptic vesicle trafficking. Biophys J 2019; 116(3): 64a.
[222] Barrett PJ, Timothy Greenamyre J. Post-translational modification of α-synuclein in Parkinson’s disease. Brain Res 2015; 1628(Pt B): 247-53.
[223] Pajarillo E, Rizor A, Lee J, Aschner M, Lee E. The role of posttranslational modifications of α-synuclein and LRRK2 in Parkinson’s disease: Potential contributions of environmental factors. Biochim Biophys Acta Mol Basis Dis 2019; 1865(8): 1992-2000.
[224] Fujiwara H, Hasegawa M, Dohmae N, et al. α-Synuclein is phosphorylated in synucleinopathy lesions. Nat Cell Biol 2002; 4(2): 160-4.
[225] Duda JE, Giasson BI, Chen Q, et al. Widespread nitration of pathological inclusions in neurodegenerative synucleinopathies. Am J Pathol 2000; 157(5): 1439-45.
[226] McCormack AL, Mak SK, Di Monte DA. Increased α-synuclein phosphorylation and nitration in the aging primate substantia nigra. Cell Death Dis 2012; 3(5): e315-5.
[227] Zhou Y, Shie FS, Piccardo P, Montine TJ, Zhang J. Proteasomal inhibition induced by manganese ethylene-bis-dithiocarbamate: Relevance to Parkinson’s disease. Neuroscience 2004; 128(2): 281-91.
[228] Xiong H, Wang D, Chen L, et al. Parkin, PINK1, and DJ-1 form a ubiquitin E3 ligase complex promoting unfolded protein degradation. J Clin Invest 2009; 119(3): 650-60.
[229] Imai Y, Soda M, Takahashi R. Parkin suppresses unfolded protein stress-induced cell death through its E3 ubiquitin-protein ligase activity. J Biol Chem 2000; 275(46): 35661-4.
[230] Kim Y, Park J, Kim S, et al. PINK1 controls mitochondrial localization of Parkin through direct phosphorylation. Biochem Biophys Res Commun 2008; 377(3): 975-80.
[231] Rubio de la Torre E, Gómez-Suaga P, Martínez-Salvador M, Hilfiker S. Posttranslational modifications as versatile regulators of parkin function. Curr Med Chem 2011; 18(16): 2477-85.
[232] Mussakhmetov A, Shumilin IA, Nugmanova R, et al. A transient post-translational modification of active site cysteine alters binding properties of the parkinsonism protein DJ-1. Biochem Biophys Res Commun 2018; 504(1): 328-33.
[233] Nichols RJ. LRRK2 Phosphorylation. Adv Neurobiol 2017; 14: 51-70.
[234] Nichols RJ, Dzamko N, Morrice NA, et al. 14-3-3 binding to LRRK2 is disrupted by multiple Parkinson’s disease-associated mutations and regulates cytoplasmic localization. Biochem J 2010; 430(3): 393-404.
[235] Nucifora FC Jr, Nucifora LG, Ng CH, et al. Ubiqutination via K27 and K29 chains signals aggregation and neuronal protection of LRRK2 by WSB1. Nat Commun 2016; 7(1): 11792.
[236] Nalls MA, Pankratz N, Lill CM, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet 2014; 46(9): 989-93.
[237] Lill CM, Roehr JT, McQueen MB, et al. Comprehensive research synopsis and systematic meta-analyses in Parkinson’s disease genetics: The PDGene database. PLoS Genet 2012; 8(3): e1002548.
[238] Chang D, Nalls MA, Hallgrímsdóttir IB, et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat Genet 2017; 49(10): 1511-6.
[239] Katzenschlager R, Lees AJ. Treatment of Parkinson’s disease: Levodopa as the first choice. J Neurol 2002; 249(2)(Suppl. 2): II19-24.
[240] Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med 2004; 351(24): 2498-508.
[241] Obeso JA, Olanow CW, Nutt JG. Levodopa motor complications in Parkinson's disease. Trends Neurosci 2000; 23(10 Suppl): S2-7.
[242] Jankovic J. Complications and limitations of drug therapy for Parkinson’s disease. Neurology 2000; 55(12)(Suppl. 6): S2-6.
[243] Fahn S. The history of dopamine and levodopa in the treatment of Parkinson’s disease. Mov Disord 2008; 23(S3)(Suppl. 3): S497-508.
[244] Black KJ, Hershey T, Hartlein JM, Carl JL, Perlmutter JS. Levodopa challenge neuroimaging of levodopa-related mood fluctuations in Parkinson’s disease. Neuropsychopharmacology 2005; 30(3): 590-601.
[245] Koller WC, Rueda MG. Mechanism of action of dopaminergic agents in Parkinson’s disease. Neurology 1998; 50(6)(Suppl. 6): S11-4.
[246] Münchau A, Bhatia KP. Pharmacological treatment of Parkinson’s disease. Postgrad Med J 2000; 76(900): 602-10.
[247] Müller T, Welnic J, Muhlack S. Acute levodopa administration reduces cortisol release in patients with Parkinson’s disease. J Neural Transm (Vienna) 2007; 114(3): 347-50.
[248] Poirier AA, Aubé B, Côté M, et al. Gastrointestinal dysfunctions in Parkinson’s disease: Symptoms and treatments. Parkinson’s Disease 2016.
[249] Varanese S, Birnbaum Z, Rossi R. Treatment of advanced Parkinson's disease. Parkinson’s disease 2010.
[250] Gupta HV, Lyons KE, Wachter N, Pahwa R. Long term response to levodopa in parkinson’s disease. J Parkinsons Dis 2019; 9(3): 525-9.
[251] Müller T, Möhr JD. Long-term management of Parkinson’s disease using levodopa combinations. Expert Opin Pharmacother 2018; 19(9): 1003-11.
[252] Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord 2001; 16(3): 448-58.
[253] Bordia T, Perez XA, Heiss J, Zhang D, Quik M. Optogenetic activation of striatal cholinergic interneurons regulates L-dopa-induced dyskinesias. Neurobiol Dis 2016; 91: 47-58.
[254] Martínez-Martín P, O’Brien CF. Extending levodopa action: COMT inhibition. Neurology 1998; 50(6)(Suppl. 6): S27-32.
[255] Lv X, Wang XX, Hou J, et al. Comparison of the inhibitory effects of tolcapone and entacapone against human UDP-glucuronosyltransferases. Toxicol Appl Pharmacol 2016; 301: 42-9.
[256] Ferreira JJ, Lees A, Rocha JF, Poewe W, Rascol O, Soares-da-Silva P. Opicapone as an adjunct to levodopa in patients with Parkinson’s disease and end-of-dose motor fluctuations: A randomised, double-blind, controlled trial. Lancet Neurol 2016; 15(2): 154-65.
[257] Smith Y, Wichmann T, Factor SA, DeLong MR. Parkinson’s disease therapeutics: New developments and challenges since the introduction of levodopa. Neuropsychopharmacology 2012; 37(1): 213-46.
[258] Heikkinen H, Varhe A, Laine T, et al. Entacapone improves the availability of L-dopa in plasma by decreasing its peripheral metabolism independent of L-dopa/carbidopa dose. Br J Clin Pharmacol 2002; 54(4): 363-71.
[259] Ossola B, Schendzielorz N, Chen SH, et al. Amantadine protects dopamine neurons by a dual action: Reducing activation of microglia and inducing expression of GDNF in astroglia [corrected]. Neuropharmacology 2011; 61(4): 574-82.
[260] Naderi S, Faghih H, Aqamolaei A, et al. Amantadine as adjuvant therapy in the treatment of moderate to severe obsessive-compulsive disorder: A double-blind randomized trial with placebo control. Psychiatry Clin Neurosci 2019; 73(4): 169-74.
[261] Aoki FY. Antiviral drugs for influenza and other respiratory virus infections Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases 8th ed. 2015.
[262] Kempster PA, Frankel JP, Stern GM, Lees AJ. Comparison of motor response to apomorphine and levodopa in Parkinson’s disease. J Neurol Neurosurg Psychiatry 1990; 53(11): 1004-7.
[263] Lees AJ. Drugs for Parkinson’s disease. J Neurol Neurosurg Psychiatry 2002; 73(6): 607-10.
[264] Brocks DR. Anticholinergic drugs used in Parkinson’s disease: An overlooked class of drugs from a pharmacokinetic perspective. J Pharm Pharm Sci 1999; 2(2): 39-46.
[265] Luginger E, Wenning GK, Bösch S, Poewe W. Beneficial effects of amantadine on L-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord 2000; 15(5): 873-8.
[266] Bieck PR, Antonin KH. Tyramine potentiation during treatment with MAOIs.Clinical Advances in Monoamine Oxidase Inhibitor Therapies 1994; 83-110.
[267] Kang SJ, Scott WK, Li YJ, et al. Family-based case-control study of MAOA and MAOB polymorphisms in Parkinson disease. Mov Disord 2006; 21(12): 2175-80.
[268] Finberg JP, Rabey JM. Inhibitors of MAO-A and MAO-B in psychiatry and neurology. Front Pharmacol 2016; 7: 340.
[269] De Colibus L, Li M, Binda C, Lustig A, Edmondson DE, Mattevi A. Three-dimensional structure of human monoamine oxidase A (MAO A): Relation to the structures of rat MAO A and human MAO B. Proc Natl Acad Sci USA 2005; 102(36): 12684-9.
[270] Denney RM, Denney CB. An update on the identity crisis of monoamine oxidase: New and old evidence for the independence of MAO A and B. Pharmacol Ther 1985; 30(3): 227-58.
[271] Gupta V, Khan AA, Sasi BK, Mahapatra NR. Molecular mechanism of monoamine oxidase A gene regulation under inflammation and ischemia-like conditions: Key roles of the transcription factors GATA2, Sp1 and TBP. J Neurochem 2015; 134(1): 21-38.
[272] Zahoor I, Shafi A, Haq E. Pharmacological treatment of Parkinson’s disease.2018.
[273] Goldenberg MM. Medical management of Parkinson’s disease. P&T 2008; 33(10): 590-606.
[274] Fernandez HH, Chen JJ. Monoamine oxidase‐B inhibition in the treatment of Parkinson's disease. Pharmacother 2007; 27(12P2): 174S-85S.
[275] Riederer P, Lachenmayer L, Laux G. Clinical applications of MAO-inhibitors. Curr Med Chem 2004; 11(15): 2033-43.
[276] Kaakkola S, Männistö PT, Nissinen E. Striatal membrane-bound and soluble catechol-O-methyl-transferase after selective neuronal lesions in the rat. J Neural Transm (Vienna) 1987; 69(3-4): 221-8.
[277] Gaines KD, Hinson VK. Adjunctive therapy in Parkinson’s disease: The role of rasagiline. Neuropsychiatr Dis Treat 2012; 8: 285-94.
[278] Zhang Z, Shao M, Chen S, et al. Adjunct rasagiline to treat Parkinson’s disease with motor fluctuations: A randomized, double-blind study in China. Transl Neurodegener 2018; 7(1): 14.
[279] Rascol O, Brooks DJ, Melamed E, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): A randomised, double-blind, parallel-group trial. Lancet 2005; 365(9463): 947-54.
[280] Sharma SK, Carlson EC, Ebadi M. Neuroprotective actions of Selegiline in inhibiting 1-methyl, 4-phenyl, pyridinium ion (MPP+)-induced apoptosis in SK-N-SH neurons. J Neurocytol 2003; 32(4): 329-43.
[281] Schmeltz L, Metzger B. Therapeutic Areas I: Central nervous system, pain, metabolic syndrome, urology, gastrointestinal and cardiovascular comprehensive medicinal chemistry II 2007.
[282] Müller T. Safinamide: An add-on treatment for managing Parkinson’s disease. Clin Pharmacol 2018; 10: 31-41.
[283] Bette S, Shpiner DS, Singer C, Moore H. Safinamide in the management of patients with Parkinson’s disease not stabilized on levodopa: A review of the current clinical evidence. Ther Clin Risk Manag 2018; 14: 1737-45.
[284] Borgohain R, Szasz J, Stanzione P, et al. Two-year, randomized, controlled study of safinamide as add-on to levodopa in mid to late Parkinson’s disease. Mov Disord 2014; 29(10): 1273-80.
[285] Zanettini R, Antonini A, Gatto G, Gentile R, Tesei S, Pezzoli G. Valvular heart disease and the use of dopamine agonists for Parkinson’s disease. N Engl J Med 2007; 356(1): 39-46.
[286] Delgado-Rosas F, Gómez R, Ferrero H, et al. The effects of ergot and non-ergot-derived dopamine agonists in an experimental mouse model of endometriosis. Reproduction 2011; 142(5): 745-55.
[287] Kailash P, Bhatia K, Chaudhuri R, et al. Parkinson’s disease edited. Int Rev Neurobiol 2017; 132: 2-523.
[288] Colosimo C, Merello M, Albanese A. Clinical usefulness of apomorphine in movement disorders. Clin Neuropharmacol 1994; 17(3): 243-59.
[289] Chiken S, Nambu A. Mechanism of deep brain stimulation: Inhibition, excitation, or disruption? Neuroscientist 2016; 22(3): 313-22.
[290] Weaver FM, Follett KA, Stern M, et al. Randomized trial of deep brain stimulation for Parkinson disease: Thirty-six-month outcomes. Neurology 2012; 79(1): 55-65.
[291] Rizzone MG, Fasano A, Daniele A, et al. Long-term outcome of subthalamic nucleus DBS in Parkinson’s disease: From the advanced phase towards the late stage of the disease? Parkinsonism Relat Disord 2014; 20(4): 376-81.
[292] Sidiropoulos C, Xie T, Vigil J, et al. Low-frequency stimulation of STN-DBS reduces aspiration and freezing of gait in patients with PD. Neurology 2015; 85(6): 557-7.
[293] Eusebio A, Chen CC, Lu CS, et al. Effects of low-frequency stimulation of the subthalamic nucleus on movement in Parkinson’s disease. Exp Neurol 2008; 209(1): 125-30.
[294] Khoo HM, Kishima H, Hosomi K, et al. Low-Frequency subthalamic nucleus stimulation in parkinson’s disease: A randomized clinical trial. Mov Disord 2014; 1-5.
[295] Kumar R, Lozano AM, Sime E, Halket E, Lang AE. Comparative effects of unilateral and bilateral subthalamic nucleus deep brain stimulation. Neurology 1999; 53(3): 561-6.
[296] Herzog J, Weiss PH, Assmus A, et al. Subthalamic stimulation modulates cortical control of urinary bladder in Parkinson’s disease. Brain 2006; 129(Pt 12): 3366-75.
[297] Wong JK, Cauraugh JH, Ho KWD, et al. STN vs. GPi deep brain stimulation for tremor suppression in Parkinson disease: A systematic review and meta-analysis. Parkinsonism Relat Disord 2019; 58: 56-62.
[298] Axelsen TM, Woldbye DPD. Gene therapy for Parkinson’s disease, An update. J Parkinsons Dis 2018; 8(2): 195-215.
[299] Hadaczek P, Eberling JL, Pivirotto P, Bringas J, Forsayeth J, Bankiewicz KS. Eight years of clinical improvement in MPTP-lesioned primates after gene therapy with AAV2-hAADC. Mol Ther 2010; 18(8): 1458-61.
[300] Cellini B, Montioli R, Oppici E, Voltattorni CB. Biochemical and computational approaches to improve the clinical treatment of dopa decarboxylase-related diseases: An overview. Open Biochem J 2012; 6: 131-8.
[301] Campos-Romo A, Ojeda-Flores R, Moreno-Briseño P, et al. Behavioral improvement in MPTP-treated nonhuman primates in the HALLWAY task after transfer of TH cDNA to host astrocytes. Acta Neurobiol Exp (Warsz) 2012; 72(2): 166-76.
[302] Denyer R, Douglas MR. Gene therapy for Parkinson’s disease. Parkinsons Dis 2012; 2012: 757305.
[303] Allan LE, Petit GH, Brundin P. Cell transplantation in Parkinson’s disease: Problems and perspectives. Curr Opin Neurol 2010; 23(4): 426-32.
[304] Soldner F, Hockemeyer D, Beard C, et al. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 2009; 136(5): 964-77.
[305] Newman MB, Bakay RA. Therapeutic potentials of human embryonic stem cells in Parkinson’s disease. Neurotherapeutics 2008; 5(2): 237-51.
[306] Lindvall O. Treatment of Parkinson's disease using cell transplantation. Philoso Transac Royal Soc B: Biol Sci 2015; 370(1680): 20140370.
[307] Morris ME, Martin CL, Schenkman ML. Striding out with Parkinson disease: Evidence-based physical therapy for gait disorders. Phys Ther 2010; 90(2): 280-8.
[308] Radder DLM, Sturkenboom IH, van Nimwegen M, Keus SH, Bloem BR, de Vries NM. Physical therapy and occupational therapy in Parkinson’s disease. Int J Neurosci 2017; 127(10): 930-43.