Your activity: 7808 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: [email protected]

Etiology and pathogenesis of Parkinson disease

Etiology and pathogenesis of Parkinson disease
Author:
Joseph Jankovic, MD
Section Editor:
Howard I Hurtig, MD
Deputy Editor:
April F Eichler, MD, MPH
Literature review current through: Feb 2022. | This topic last updated: Dec 21, 2021.

INTRODUCTION — Parkinson disease (PD) is the most common cause of parkinsonism, a syndrome manifested by rest tremor, rigidity, bradykinesia, and postural instability. The disorder was first described by James Parkinson in his 1817 Essay on the Shaking Palsy. Although it has been proposed that PD emerged as a result of the industrial revolution, there is some evidence that a disease known as "kampavata," consisting of shaking (kampa) and lack of muscular movement (vata), existed in the ancient Indian medical system, Ayurveda, as long as 4500 years ago [1]. The Mucuna pruriens plant was used in ancient times to treat the symptoms, and was later discovered to contain levodopa [2].

The pathology of PD was not well understood until the early 20th century, when the German pathologist Frederick Lewy in 1912 reported neuronal cytoplasmic inclusions in a variety of brain regions. In 1919, Tretiakoff observed that the most critical abnormality in PD was the loss of neurons in the substantia nigra pars compacta (SNc) of the midbrain. In the 1950s, investigators discovered the importance of dopamine and its depletion from the basal ganglia as the key to understanding the pathophysiology and pathologic biochemistry of PD [3].

While the cause of PD is still unknown, remarkable advances have been made in understanding the possible underlying mechanisms [4]. This extraordinary progress has been fueled by new discoveries about the anatomy and function of the basal ganglia, by improved characterization of neuropathologic and neurochemical abnormalities in PD, and by studies of genetic and experimental forms of parkinsonism.

This topic will review the etiology and pathogenesis of PD. Clinical issues related to PD are discussed separately. (See "Clinical manifestations of Parkinson disease" and "Diagnosis and differential diagnosis of Parkinson disease" and "Initial pharmacologic treatment of Parkinson disease".)

PATHOPHYSIOLOGY — Dopamine depletion from the basal ganglia results in major disruptions in the connections to the thalamus and motor cortex, and leads to parkinsonian signs such as bradykinesia.

A number of compensatory mechanisms may operate to mask or reduce the deleterious effects of dopamine depletion, particularly in the presymptomatic phase of PD, but they are eventually overwhelmed by disease progression.

Basal ganglia circuits — The basal ganglia, sometimes referred to as the extrapyramidal system, include the substantia nigra, striatum (caudate and putamen), globus pallidus (GP), subthalamic nucleus (STN), and thalamus.

The cortical input to the basal ganglia from the prefrontal supplementary motor area, amygdala, and hippocampus is excitatory, mediated by the neurotransmitter glutamate. Neurons in the substantia nigra pars compacta (SNc) provide major dopaminergic input to the striatum and exert both excitatory and inhibitory influences on the striatal output neurons. The interaction between the afferent and efferent pathways is mediated by striatal interneurons, which utilize acetylcholine as the main neurotransmitter.

The striatal output system is mediated by the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). The connection between the STN and the internal (medial) globus pallidus (GPi) and between STN and the lateral (or external) globus pallidus (GPe) is excitatory, mediated by glutamate.

Five distinct dopamine receptors (D1 through D5) have been cloned and characterized; they are found throughout the basal ganglia and limbic system. The D1 and D2 receptors are highly concentrated in the dorsal (motor) striatum and are the most relevant to the pathophysiology of PD because they are activated by the dopaminergic pathway originating in the SNc and terminating in the caudate and putamen. Receptors designated as D3, D4, and D5 are more abundant in the mesolimbic or emotional part of the brain (D3, D4) and hippocampus/hypothalamus (D5) [5].

Dopamine deficiency in the nigrostriatal pathway, such as seen in PD, causes denervation hypersensitivity of D1 and D2 receptors [6]. When compared with normal matched controls, D2 receptors in the dorsal putamen are increased by 15 percent in patients with PD, whereas D3 receptors in the mesolimbic system are decreased by 40 to 45 percent [7]. These results may explain the basis for the hypersensitivity of the nigrostriatal (D2) dopaminergic receptors that is observed in PD.

There are two output pathways from the striatum (figure 1):

The indirect pathway is mediated chiefly via dopamine's inhibitory influence on striatal D2 dopamine receptors. In the indirect pathway, the striatum projects to the neurons in the GPe utilizing GABA, and the GPe in turn projects to the STN, which provides excitatory input via glutamate to the GPi and substantia nigra pars reticulata (SNr). GPi neurons are GABAergic and synapse in the ventrolateral nucleus of the thalamus. Thalamic input to the cortex is excitatory.

The direct pathway is mediated via dopamine's excitatory influence on striatal D1 dopamine receptors. In the direct pathway, the striatum projects directly to the GPi and SNr.

In PD, a reduction of dopamine-producing neurons from the normal complement of approximately 550,000 to the critically low level of 100,000 leads to dopamine depletion in the substantia nigra and in the nigrostriatal pathway to the caudate and putamen. This, in turn, results in relative overactivity of the indirect pathway, functionally disinhibiting the STN. Decreased inhibition of the direct pathway causes additional disinhibition of the output nuclei (GPi and SNr). Increased output from GPi causes increased inhibition of the thalamus and reduced excitatory input to the motor cortex, which is ultimately expressed as bradykinesia and other parkinsonian signs.

In PD, synchronized oscillatory activity in the 10 to 50 Hz band (often termed the beta-band), prevalent in the basal ganglia thalamocortical circuit, may be important in mediating certain parkinsonian features, including bradykinesia and tremor, and can be reduced by dopaminergic treatments [8]. Therefore, surgical treatments of PD, such as lesion placement within or stimulation of GPi or STN, may act by desynchronizing the oscillatory basal ganglia-thalamo-cortical network activity.

Models of basal ganglia dysfunction (figure 1) are useful for conceptualizing how the motor symptoms of PD arise. However, the actual pathophysiology of the basal ganglia associated with PD is much more complex than indicated by the current models [9]. Existing models should be constantly reevaluated as new findings become available.

Compensatory mechanisms — The brain has a remarkable capacity to compensate for the presynaptic dopamine depletion by increasing the synthesis of dopamine in surviving neurons and by increasing the afferents to the dendrites of dopaminergic neurons. Furthermore, dopaminergic denervation has been shown to result in a proliferation of D2 receptors, as well as a co-localization of D1 and D2 receptors [10]. Similarly, gap junctions, which allow rapid communications between striatal neurons, increase dramatically after dopaminergic denervation [11].

In the brains of patients with PD, the number of tyrosine hydroxylase-staining neurons in the striatum is markedly decreased [12]. Since tyrosine hydroxylase, the rate-limiting enzyme in the synthesis of dopamine, is still present in surviving neurons, the synthesis of dopamine could be increased in these striatal neurons, thereby compensating for the presynaptic dopamine loss [13,14]. Another compensatory mechanism may be downregulation of the dopamine transporter, resulting in less dopamine reuptake and higher synaptic dopamine levels [15].

Three stages of compensation during the presymptomatic period of PD have been proposed [14]:

An early period during which the dopamine homeostatic compensatory mechanisms discussed above are capable of "masking" the disease

Increased activity of the basal ganglia output nuclei (eg, internal segment of the GP) as striatal dopamine homeostasis breaks down

Increased intensity of compensation in structures outside of the basal ganglia (eg, supplementary motor area of the cortex) as parkinsonian motor abnormalities emerge

PATHOLOGY — Depigmentation, neuronal loss, and gliosis, particularly in the substantia nigra pars compacta (SNc) and in the pontine locus ceruleus, are typical abnormalities found in the brains of patients with PD. Neuronal degeneration is also present in the dorsal nucleus of the vagus in the medulla and other brainstem nuclei.

Using a quantitative method, one study of seven patients with PD and seven controls found that the number of pigmented neurons in the substantia nigra, normally 550,000, was reduced by 66 percent in those with PD [16]. In addition, the number of nonpigmented neurons, normally 260,000, was reduced by 24 percent. By the time the first symptoms of PD emerge, approximately 60 percent of the neurons in the SNc have been lost [3].

The ventrolateral portion of the substantia nigra that projects to the dorsal putamen is preferentially affected early in the course of PD, resulting in the gradual loss of dopaminergic neurons in the SNc and a nearly complete depletion of dopamine, particularly in the putamen [17]. This contrasts with normal aging, which is usually associated with neuronal loss in the dorsal tier of the SNc, and depletion of dopamine, predominantly in the caudate nucleus [18].

Because of the apparent discrepancy between loss of striatal dopamine (>80 percent) and the degree of loss of neurons in the substantia nigra (50 to 60 percent), some have suggested that the initial site of pathology is in the striatum and that retrograde degeneration may be responsible for the neuronal loss in the substantia nigra [3]. An alternative explanation is that each dopaminergic neuron has multiple projections that terminate in the striatum, so that death of the cell body has a multiplying effect on loss of terminals.

In addition to the degeneration of the SNc, other nuclei are affected by the pathology of PD, including the internal segment of the globus pallidus (GPi), the center median-parafascicular complex, the pedunculopontine tegmental nucleus, and the glutamatergic caudal intralaminar thalamic nuclei [19]. Moreover, volumetric magnetic resonance imaging (MRI) studies have found significant hippocampal atrophy in patients with PD, with or without cognitive impairment [20].

Lewy bodies and other intracellular inclusions — There is no consensus as to what pathologic criteria are necessary for the diagnosis of PD [4], but most investigators believe that Lewy bodies, named for Frederick Lewy, constitute the pathologic hallmark of PD.

Lewy bodies are round, eosinophilic, intracytoplasmic neuronal inclusions. They are 3 to 25 nm in diameter with a dense granular core (1 to 8 nm) and loosely arranged fibrillary elements extending towards a peripheral "halo." Immunohistochemical studies have demonstrated that Lewy bodies are made up mainly of alpha-synuclein and ubiquitin, and also contain calbindin, complement proteins, microfilament subunits, tubulin, microtubule associated protein 1 and 2, and a parkin substrate protein called Pael-R [21]. However, Lewy bodies do not contain the tau protein [21].

As noted, Lewy bodies stain for alpha-synuclein, a 140 amino acid presynaptic protein found mutated in rare families with PD. Lewy bodies also contain synphilin-1 and other cytoskeletal proteins associated with alpha-synuclein [22]. (See 'SNCA-associated PD' below.)

In patients with PD, Lewy bodies are seen in the substantia nigra, the basal nucleus of Meynert, the locus ceruleus, the cerebral cortex, the sympathetic ganglia, the dorsal vagal nucleus, the myenteric plexus of the intestines, and even in the cardiac sympathetic plexus. Lewy bodies appear to arise from the peripheral portion of other inclusions known as pale bodies, which are found in the substantia nigra and locus ceruleus [23].

Lewy bodies are not specific for PD, since they are found in as many as 10 percent of brains of normal older adults, and in patients with other neurodegenerative diseases, such as neurodegeneration with brain iron accumulation (NBIA), ataxia-telangiectasia, progressive supranuclear palsy, corticobasal degeneration, Down syndrome, and Alzheimer disease. There is growing evidence that Lewy bodies occur not only in synucleinopathies such as PD, or amyloidopathies such as Alzheimer disease, but also in tauopathies, such as frontotemporal dementia [24].

The lack of specificity of the pathologic findings raises the possibility that PD may not be a specific disease entity, but rather a clinically prototypical syndrome with different clinical subtypes and pathogenic causes (table 1) [25].

Inclusions such as Lewy bodies have traditionally been considered toxic. However, some studies suggest that they may actually be neuroprotective, and that compounds that promote the formation of inclusions lessen the pathology of PD [23,26].

Braak staging — In the traditional view, the pathologic process of PD starts with degeneration of dopaminergic neurons in the substantia nigra. This view has been challenged by the neuropathologist Heiko Braak, who has proposed that the pathologic changes of PD start in the medulla of the brainstem and in the olfactory bulb, progressing rostrally over many years to the cerebral cortex in a predictable six-stage process (figure 2) [27,28].

According to Braak staging, the progression of pathologic changes occurs as follows [27]:

During presymptomatic stages 1 and 2, the pathologic changes are found in the medulla oblongata and olfactory bulb.

In stages 3 and 4, the pathology has migrated rostrally to the SNc and other neuronal clusters of the midbrain and basal forebrain, at which time the classic motor symptoms of PD first appear.

In end-stages 5 and 6, the pathologic process encroaches upon the telencephalic cortex of the temporal and frontal lobes.

However, the validity and predictive utility of Braak staging has been questioned, as there are no cell counts to correlate with the described synuclein pathology and no observed asymmetry in the pathologic findings that correlate with the well-recognized asymmetry of clinical findings [29,30].

In addition, there is controversy as to the classification of dementia with Lewy bodies (DLB), considered by some to be a separate entity from PD. Braak did not include DLB his observations on the progression of PD. (See "Epidemiology, pathology, and pathogenesis of dementia with Lewy bodies" and "Clinical features and diagnosis of dementia with Lewy bodies".)

PATHOGENESIS OF CELL DEGENERATION — Irrespective of the initial trigger (etiology) of the neuronal degeneration in PD, the pathogenesis of neurodegeneration probably involves either programmed cell death (apoptosis) or necrosis [4,31-33].

Apoptosis is characterized by condensation of cytoplasm and chromatin, DNA fragmentation, and cell fragmentation into apoptotic bodies, followed by lysosome-mediated phagocytosis. The other mechanism of cell death, called autophagy, is characterized by accumulation of autophagic vesicles (autophagosomes and autophagolysosomes), and also plays an important role in neurodegeneration in PD [34].

It has been suggested that only 0.5 percent of substantia nigra neurons in normal brains are undergoing apoptosis, but this number is increased fourfold to 2 percent in those with PD. Some experimental models of PD suggest that apoptosis is the primary mechanism of substantia nigra neuronal degeneration in PD, but convincing evidence from careful neuropathologic studies is lacking [35,36].

Although the precise mechanisms of neurodegeneration in PD are not yet understood, they most likely involve a cascade of events that include interaction between genetic factors and abnormalities in protein processing, oxidative stress, mitochondrial dysfunction, excitotoxicity, inflammation, immune regulation, glial-specific factors, lack of trophic factors, and other, yet unknown, mechanisms (figure 3 and figure 4).

One of the emerging hypotheses is that neurodegeneration in PD is due to disruption of intracellular vesicular transport as a result of destabilizing of microtubules [37]. Another area of increasing interest is the role of astrocytes in various neurodegenerative disorders [38].

Some have suggested that PD arises from a prenatal event or process that predisposes some individuals to have fewer dopaminergic neurons at the time of birth because of abnormalities in the genes that code for factors important in the development of the dopaminergic system [39].

As an example, it is possible that prenatal or early postnatal exposure to certain dopaminergic neuronal toxins, such as the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-like herbicide paraquat and the manganese-containing fungicide maneb, may reduce the number of dopamine neurons in the substantia nigra in early development and enhance vulnerability to these toxins when individuals are subsequently exposed in adulthood [40]. This is consistent with the so-called "multiple-hit hypothesis."

Protein misfolding, aggregation, and toxicity — Mutations in the gene on chromosome 4q21.3-q22 that codes for alpha-synuclein (SNCA) have emerged as one of the most important elements of cell death in various neurodegenerative disorders, together known as synucleinopathies [4,41]. These include not only PD, but also dementia with Lewy bodies (DLB), multiple system atrophy, and neurodegeneration with brain iron accumulation (NBIA).

Alpha-synuclein is quite abundant in the central nervous system (CNS), accounting for 1 percent of total CNS protein. Its physiologic role is not fully understood, though it appears to be involved in synaptic function and plasticity [42]. A number of observations suggest that abnormal alpha-synuclein processing plays a role in the pathogenesis of PD:

Mutations in the SNCA gene may cause the natively unfolded alpha-synuclein protein to alter its secondary structure and self-aggregate after being targeted for proteasomal degradation by ubiquitin [43]. Misfolding of proteins and subsequent formation of insoluble aggregates can occur due to gene mutations that result in abnormal structure of the gene product, or as a result of age-related phenomenon [44].

An aggregated and insoluble form of alpha-synuclein is a major component of Lewy bodies, intracellular inclusions that are the pathologic hallmark of PD [45]. (See 'Lewy bodies and other intracellular inclusions' above.)

A number of observations in transgenic and normal mice and in humans suggest that misfolded forms of alpha-synuclein can somehow be transmitted from diseased neurons to healthy ones [46].

In normal mice, a single injection of synthetic misfolded alpha-synuclein fibrils into the striatum leads to cell-to-cell transmission of pathologic alpha-synuclein and Lewy body-like pathology with progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) and impaired motor coordination [47,48]. Pathologic alpha-synuclein appears to act as a template that corrupts normal alpha-synuclein, so it too becomes pathologic and thereby spreads the disease from an affected neuron to a normal one, which in turn becomes diseased.

Human fetal nigral neurons transplanted into the brains of patients with PD developed Lewy bodies that are demonstrable at autopsy in long-term survivors [49,50].

The hydrophobic portion of alpha-synuclein can spontaneously form fibrillar protein aggregates, and SNCA mutations may promote the development of these aggregates [51]. Furthermore, alpha-synuclein protoaggregates or oligomers can disrupt cell membranes, including dopamine vesicles and mitochondria, possibly by causing pores in the membranes [51]. A protofibrillar form of alpha-synuclein appears to be more toxic than the normal or fibrillar form. Several anti-synuclein strategies are currently being investigated in clinical trials as potential disease-modifying therapies [52].

Recombinant alpha-synuclein preformed fibrils (PFF) similar to those found in PD cause poly(adenosine 5’-diphosphate-ribose) [PAR] polymerase-1 (PARP-1) activation and cell death when injected into mouse brain, and PAR generated by PARP-1 activation binds to alpha-synuclein PFF and accelerates fibrillization, misfolding, and in vivo spread [53]. Further, transmission and neurotoxicity of pathologic alpha-synuclein in this model system were attenuated by PARP-1 deletion and by clinically available PARP inhibitors (developed as anticancer drugs).

It should be noted that the Braak hypothesis and the notion of aggregated synuclein as the primary pathogenic mechanism of neurodegeneration have been increasingly challenged. Some investigators have proposed that abnormal soluble oligomers and fibrils of alpha-synuclein that aggregate into Lewy bodies are merely byproducts and may actually serve a protective rather than toxic function [54,55].

Defective proteolysis — Cellular protein homeostasis is normally maintained primarily by three coordinated pathways (molecular chaperones, the ubiquitin-proteasome system, and the autophagy-lysosomal pathway) that mediate the repair or removal of abnormal proteins [56-59]. While the data are not entirely consistent, it appears that all three pathways are involved in the processing of alpha-synuclein. When these systems are inhibited or impaired, abnormal proteins such as mutated alpha-synuclein can misfold, aggregate, and clog the normal molecular traffic of the cell, leading to cell death.

Of particular interest is the finding in mice that the proteins parkin, PTEN-induced putative kinase 1 (PINK1), and DJ-1 (encoded by the PARK7 gene) bind to each other to form a complex that promotes degradation of unfolded or misfolded proteins via the ubiquitin-proteasome system [60]. This observation is notable because gene mutations of parkin (PARK2), PINK1, and DJ-1 (PARK7) are individually associated with autosomal recessive forms of PD. Furthermore, Atp13a2 deficiency can cause lysosomal dysfunction and enhance the accumulation and toxicity of alpha-synuclein in vitro [57,61]. This finding may reflect the pathogenesis of neurodegeneration associated with loss-of-function ATP13A2 gene mutations that cause an early-onset form of parkinsonism. (See 'Genetics' below.)

Mitochondrial dysfunction — The role of mitochondria in the pathogenesis of PD was first suggested by discovery of the association between the meperidine analogue MPTP and parkinsonism [62,63]. The oxidation of MPTP produces 1-methyl-4-phenylpyridium (MPP+), which is taken up by dopaminergic terminals, selectively inhibits mitochondrial complex I activity, disrupts calcium homeostasis, and induces endoplasmic reticulum stress, resulting in cell damage [63,64]. Direct evidence of mitochondrial dysfunction is supported by the finding that complex I activity is decreased by 32 to 38 percent in the substantia nigra of patients with sporadic PD [65,66], and by the finding that mitochondrial membrane potential and intracellular adenosine triphosphate (ATP) levels are significantly decreased in skin fibroblasts of patients with PD who carry the leucine-rich repeat kinase-2 (LRRK2) G2019S mutation [67].

The following cascade of intracellular events has been postulated to lead to neurodegeneration [68,69]:

A cellular insult (eg, oxidative stress, excitotoxicity, DNA damage) increases cytosolic calcium and oxidative radicals, and activates nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) leading to the formation of poly(ADP-ribose PAR)

This leads to decreased mitochondrial membrane potential, which in turn opens mitochondrial permeability transition pores (PTP)

Release of nicotinamide adenine dinucleotide (NAD+) through the PTP leads to NAD+ depletion

Release of mitochondrial apoptosis initiating factors promotes release of cytochrome c, which leads to activation of the "executioner" enzyme caspase and to apoptosis

Based on epidemiologic studies supporting an association between exposure to pesticides and PD (see 'Risk factors' below), and evidence of mitochondrial complex I deficiency in PD, an animal model of PD pathogenesis has been proposed [70]. The lipophilic pesticide rotenone is a potent inhibitor of mitochondrial complex I. Continuous infusion of rotenone into the jugular vein of rats for several weeks produced highly selective degeneration of the nigrostriatal dopaminergic pathway, associated clinically with bradykinesia and rigidity, and pathologically with fibrillary cytoplasmic inclusions staining for ubiquitin and alpha-synuclein.

Unlike MPTP, rotenone does not require the dopamine transporter for neuronal uptake. The degeneration actually begins in the dopaminergic nerve terminals in the central and dorsolateral striatum, leading to retrograde apoptotic death of the cell body. Rotenone infusion is associated with marked and uniform depletion of complex I in the brain and generation of reactive oxygen species, or oxygen-free radicals, resulting in oxidative damage [70]. This can also lead to release of cytochrome c from the mitochondria into the cytoplasm and aggregation of proteins, such as alpha-synuclein, and formation of cytoplasmic inclusions.

The rotenone model provides support for the theory that neurodegeneration results from an interaction between environmental exposure and mitochondrial dysfunction. Further evidence for the role of mitochondria in the pathogenesis of PD is provided by the increasing number of genes (such as PINK1) coding for mitochondrial proteins, and implicated in cellular protection against oxidative damage, that have been associated with the PD phenotype [71,72]. (See 'PINK1-associated PD' below.)

Oxidative stress — The oxidative stress hypothesis postulates that inappropriate production of reactive oxygen species leads to neurodegeneration [66,73]. Dopamine is normally metabolized not only by monoamine oxidase-mediated enzymatic oxidation, but also by auto-oxidation to neuromelanin.

Intraneuronal neuromelanin appears to have a dual role [74]. First, it may be neuroprotective, preventing toxic accumulation of metabolites of catechol amines and scavenging reactive metals, pesticides, and other oxidants. Second, dying neurons may release neuromelanin, leading to chronic inflammation.

These metabolic pathways generate byproducts, including hydrogen peroxide, superoxide anions, and hydroxyradicals. Free radicals, through interaction with membrane lipids, cause toxic lipid peroxidation, which has been found to be increased in the substantia nigra of PD brains. The oxidative products may cause neurotoxicity and, therefore, may play an important role in the development of PD.

It is possible that increased oxidative stress also contributes to misfolding of proteins. This notion is supported by the finding that nitric oxide, a free radical increased in the brains of patients with PD, attacks disulfide isomerase, an aggregation-preventing chaperone protein localized to the endoplasmic reticulum and normally responsible for unfolding and transport of proteins [75].

Iron metabolism — Elemental iron plays a critical role in oxidative metabolism, and it also serves as a cofactor in the synthesis of neurotransmitters [76]. It is increased by approximately 50 percent in substantia nigra of PD brains relative to controls [77], suggesting that abnormal iron metabolism plays a pathologic role in the development of PD [78]. One study found that mice lacking the tau protein developed parkinsonism due to toxic iron accumulation and neuronal loss in the substantia nigra [79]. In addition, loss of tau in neuronal culture caused intracellular iron retention. Brain permeable iron chelators prevent experimentally induced degeneration of nigrostriatal dopamine neurons [79,80].

Immunologic and inflammatory mechanisms — Immunologic mechanisms have been implicated in the pathogenesis of PD [81,82]. Supporting evidence comes from the finding of elevated levels of the proinflammatory cytokines TNF-alpha, interleukin-1 beta, and interferon-gamma in patients with PD.

The role of inflammatory processes in the pathogenesis of PD is further supported by the following observations:

Cyclooxygenase-2, the rate-limiting enzyme in prostaglandin E2 synthesis, appears to be upregulated in patients with PD and in the MPTP mouse model of PD; cyclooxygenase-2 inhibition prevents the formation of potentially toxic dopamine-quinones in MPTP mice and presumably in patients with PD [83].

In a positron emission tomography (PET) study that used markers for activated microglia and for dopamine transporter, microglial activity in patients with PD correlated with decreased density of dopamine transporter [84].

Infiltration of CD4+ T lymphocytes contributed to neuronal cell death in a mouse model of PD [85].

EPIDEMIOLOGY

Incidence and prevalence — PD is a growing source of disability and mortality among neurologic disorders. Based on a meta-analysis of 47 studies, the worldwide prevalence of PD is estimated to be 0.3 percent in the general population 40 years of age and older [86]. In the year 2016, the estimated global prevalence of PD was 6.1 million people, increased from 2.5 million in 1990 [87]. A similar trend has been observed in age-adjusted mortality from PD [88]. The increase over time is largely although not entirely accounted for by increasing numbers of older people.

Estimates of the incidence of PD (number of new cases per year) range from 8 to 18.6 per 100,000 person-years [89].

A male preponderance of PD has been observed in many but not all epidemiologic studies [86,89,90], suggesting that men have a higher risk than women for developing PD.

Associated factors — Epidemiologic studies may provide important clues to potential risk factors associated with PD. Data for most putative PD risk factors are conflicting, but there is consistent evidence that older age and a family history of PD are associated with an increased risk of developing PD, while cigarette smoking is associated with a decreased risk [91,92].

Protective factors — An inverse correlation between PD and smoking is supported by the findings of large cohort studies and meta-analyses [4,91,93-96]. In a 2012 meta-analysis of 26 observational studies, the risk of PD was more than twofold lower for current smokers compared with never smokers (relative risk [RR] 0.44, 95% CI 0.39-0.50) [91]. In addition, the risk of PD was lower for ever smokers compared with never smokers (RR 0.64, 95% CI 0.60-0.69). A neuroprotective effect of nicotine has been proposed as one possible explanation for these observations [97]. The relatively high frequency of nonsmokers among patients with PD does not explain the relatively low prevalence of most cancers, except for melanoma and possibly breast cancer, reported in patients with PD [98].

An alternative hypothesis is that patients who develop PD are less likely to smoke in the first place, or more likely to quit smoking than those who do not develop PD. This alternative explanation posits that since dopamine is an integral component of the brain's reward system, people who will later develop signs of PD do not engage in reward-seeking behaviors, such as smoking, because dopamine is significantly depleted in the basal ganglia years before symptoms of PD appear [99-101]. Others hypothesize that the tendency to smoke represents a risk-taking behavior, and that general risk tolerance as a genetically determined trait is causally related to PD [102].

Other factors associated with a reduced risk of PD in at least some studies include coffee and caffeine intake, exercise, and certain medications.

Caffeine – Coffee and caffeine intake have been associated with lower risk of PD in meta-analyses and large cohort studies [91,95,103,104].

Exercise – Aerobic exercise and physical activity may be protective against development of PD [105-111]. In a meta-analysis of eight prospective studies in more than 500,000 individuals, moderate to vigorous physical activity was associated with an approximately 30 percent reduction in the RR of PD [112]. However, an alternative explanation for the association is reversed causality, given that reduced physical activity may be a preclinical sign of PD.

Ibuprofen – There is evidence from several meta-analyses that ibuprofen may be associated with a reduced risk of PD [113-116]. The data regarding other nonsteroidal anti-inflammatory drugs (NSAIDs) are conflicting, with some meta-analyses finding that NSAIDs are associated with a reduced risk of PD [91,114], and others finding no significant association [113,115,116].

Statins – The relationship between statin use and lipid levels with PD is unsettled. Several studies suggest that statins are associated with a lower risk of PD [117,118]. Other studies suggest that the apparent protective effect of statin use is explained, at least in part, by failure to adjust for confounders [119], or that statin use is associated with a higher risk of PD incidence and progression [120,121].

Glycolysis-enhancing drugs – Terazosin, doxazosin, and alfuzosin are alpha-1-adrenergic receptor antagonists used to treat hypertension and benign prostatic hyperplasia that bind to phosphoglycerate kinase 1 (PGK1) and increase energy metabolism. In cellular and animal models, enhancing glycolysis reduces PD progression [122]. In several large database studies, use of terazosin, doxazosin, or alfuzosin has been associated with reduced risk of PD or PD progression compared with nonuse or use of tamsulosin, an alpha-1-adrenergic receptor antagonist that does not have PGK1 activity [122,123]. Further studies are needed to explore a possible protective effect prospectively.

Risk factors — A family history of PD is an important risk factor for developing PD [91]. The risk for PD associated with specific genes and genetic loci is discussed below. (See 'Genetics' below.)

A number of reports, including several large population-based case-control studies, have found an association between depression and the subsequent development of PD [124-129]. These findings suggest that depression is either a risk factor for PD or a prodromal symptom of PD. Similarly, meta-analyses of observational studies suggest a preceding history of constipation is either an early manifestation of PD or a risk factor for PD [91,130].

A large number of environmental and other potentially modifiable risk factors have been identified in epidemiologic studies. Examples include the following:

Exposure to pesticides [91,131-137]

Exposure to nitrogen dioxide in air pollution [138]

High consumption of dairy products [139,140]

History of traumatic brain injury [141-143]

Reduced levels of dietary and sunlight-derived vitamin D [144-146]

History of midlife migraine with aura [147]

Living in urban or industrial areas with high release of copper, manganese, or lead [148]

Exposure to hydrocarbon solvents, particularly trichloroethylene [149]

Living in rural areas [91]

Farming or agriculture work [91]

The use of well water [91,150]

High dietary intake of iron, especially in combination with high manganese intake [151]

Excess body weight, type 2 diabetes, and metabolic syndrome [152-155]

Paradoxically, other data suggest that mortality from PD is increased among people with higher socioeconomic occupations (eg, education, computer and mathematical, legal, architecture and engineering) where exposure to toxins is unlikely, while PD mortality is decreased among people with lower socioeconomic occupations (eg, mining and drilling, transportation and material moving, construction) where exposure to toxins is more likely [156,157].

Although the prevalence of most cancers among patients with PD is relatively low [98], there is evidence that a history of melanoma or prostate cancer is associated with an increased risk for PD, and vice versa [158-162].

GENETICS — Although the majority of cases of PD appear to be sporadic, there is increasing evidence that genetic factors play a role in the pathogenesis of PD, particularly when the age at symptom onset is younger than 50 years [163-165].

The most cogent evidence for a genetic contribution to the pathogenesis of PD comes from the study of large multicase kindreds with a PD phenotype of monogenetic origin. These familial forms of parkinsonism (designated PARK1 through PARK13) have been associated with causative mutations in a number of nuclear genes (table 2) [166]. Autosomal dominant, autosomal recessive, and possible X-linked forms of PD have been identified [163,166].

The reported frequency of monogenetic forms of PD varies considerably across studies, depending on the genes tested, the racial and ethnic background of the cohort, and age at disease onset. In a population-based cohort in the United Kingdom that included analysis of four genes (PRKN, PINK1, LRRK2, and SNCA) in over 2000 patients with PD, monogenetic forms of PD accounted for 1.4 percent of all PD cases and approximately 3 percent of young-onset PD (≤50 years old at onset) [167]. In a multicenter cross-sectional study of 953 patients with young-onset PD that included analysis of six relevant genes (including glucocerebrosidase), the mutation carrier frequency was 17 percent [168].

Several of the most important genetic risk factors and monogenetic forms of PD are discussed in greater detail in the following sections.

SNCA-associated PD — Alpha-synuclein (SNCA) gene missense mutations (PARK1) or multiplications (PARK4) are associated with autosomal dominant parkinsonism; the phenotype varies from classic PD to dementia with Lewy bodies (DLB) [166,169,170]. However, SNCA mutations or multiplications appear to be rare causes of PD [171].

The mechanism of SNCA-associated neurodegeneration is not yet fully understood, but is probably due to a toxic gain-of-function effect [166]. As discussed earlier, there is evidence that impaired processing of alpha-synuclein leads to abnormal protein aggregation and misfolding, Lewy body formation, cellular oxidative stress, and energy depletion. (See 'Protein misfolding, aggregation, and toxicity' above.)

The SNCA gene was the first to be associated with parkinsonism when a genome scan in the Greek-Italian Contursi kindred identified a genetic marker on chromosome 4q21-q23 linked to the PD phenotype [172]. This finding led to the discovery of a mutation in the single base pair (Ala53Thr) of the SNCA gene, designated PARK1 in the hierarchy of genes associated with PD [172,173].

In the Contursi kindred, the clinical features of the disease are similar to otherwise typical PD, except for younger age at onset (mean 46), greater cognitive decline, and more rapid progression, with a mean time from onset to death of nine years [174-176]. The pattern of nigrostriatal degeneration, with preservation of D2 receptors, as demonstrated by positron emission tomography (PET), is similar to that seen in idiopathic PD.

Subsequently, a second mutation in the SNCA gene involving an alanine for proline substitution at amino acid 30 (Ala30Pro) was found in a German family [169], and a third mutation involving a glutamic acid-to-lysine substitution at position 46 (E46K) was identified in a Spanish family [170].

With reports of additional families, the phenotype of PARK1 has expanded to include not only typical PD features, but also dementia, hallucinations, central hypoventilation, orthostatic hypotension, myoclonus, and urinary incontinence, with pathologic involvement of the brainstem pigmented nuclei, hippocampus, and temporal neocortex. Thus, the clinical and pathologic features of families with SNCA mutations and parkinsonism overlap the features of multiple system atrophy and DLB.

Similarly, overexpression of SNCA may lead to neurodegenerative disease with features that overlap those of PD, DLB, and multiple system atrophy, as observed in families with parkinsonism and whole gene duplication or triplication of SNCA [177]. Duplication of SNCA appears to be associated with late-onset parkinsonism and dysautonomia, while triplication (designated PARK4) leads to early-onset PD and dementia [178]. SNCA triplication was found in one large family with autosomal dominant, young-onset parkinsonism, dysautonomia, cardiac denervation, DLB, and glial cytoplasmic inclusions at autopsy, features typical of multiple system atrophy [179].

Mounting evidence supports the role of common SNCA polymorphisms in sporadic PD [180-183]. In particular, several large GWAS and a meta-analysis of GWAS studies in PD found that SNCA was a risk locus for PD [182-185].

LRRK2-associated PD — The most common form of monogenic PD is PARK8, caused by mutations in the leucine-rich repeat kinase-2 (LRRK2) gene on chromosome 12p11.2-q13.1 [186,187].

The LRRK2 gene product is a protein called dardarin (from the Basque word "dardara," meaning tremor) that probably functions as a cytoplasmic kinase involved in phosphorylation of proteins, such as alpha-synuclein and microtubule-associated protein tau [188-190]. Dardarin is a large molecule, encoded by 51 exons and containing 2527 amino acids. This contrasts with the much smaller alpha-synuclein protein, which contains 140 amino acids.

Dardarin is closely associated with a variety of membrane and vesicular structures, membrane-bound organelles and microtubules, suggesting its role in vesicular transport and in membrane and protein turnover, including lysosomal degradation pathway.

LRRK2-associated PD may account for a significant proportion of familial PD cases, and a smaller proportion of sporadic PD cases. It is estimated to cause up to 8 percent of autosomal dominant PD in the Basque population and up to 50 percent of familial PD in people of North African and Middle Eastern origin [189,191-194]. In addition, LRRK2 mutations have been found in 0.4 to 1.9 percent of patients with idiopathic PD [195-199], although such cases could also be explained by reduced penetrance in familial disease [192].

Genetic screening studies suggest that the G2019S mutation, the most common of the LRRK2 mutations, accounts for 3 to 13 percent of autosomal dominant PD in Europe [171,193,200-202], and 10 to 18 percent of autosomal dominant PD in Ashkenazi Jews [194,203]. The G2019S mutation has also been identified in asymptomatic carriers, suggesting reduced or age-dependent penetrance [192,201]. Evidence of age-dependent penetrance was found in a study of 19 families with the G2019S mutation, where the cumulative incidence of PD at ages 60, 70, and 80 years was 15, 21, and 32 percent [204].

The LRRK2-associated PD phenotype is often, but not always, associated with late-onset (mean age 65 years) disease [190,201,205-207]. However, the typical features of PD associated with LRRK2 G2019S mutations are indistinguishable from idiopathic PD [176,199]. The course of the disease is relatively benign, usually presenting with unilateral hand or leg tremor without cognitive deficit. Patients respond well to levodopa and have a slower decline in motor function compared with patients without an LRRK2 mutation [208]. Several studies have suggested that LRRK2-PD patients may have an increased risk of certain types of cancer compared with idiopathic PD and healthy controls, such as leukemia, colon cancer, and possibly breast cancer [209-212].

Other clinical phenotypes associated with LRRK2 mutations have included parkinsonism with dementia or amyotrophy or both, typical essential tremor, dysautonomia, familial progressive supranuclear palsy, familial multiple system atrophy, corticobasal degeneration, and primary progressive aphasia [213,214].

Autopsy findings in patients with LRRK2-associated PD are heterogeneous and range from pure nigral degeneration without Lewy bodies, to pathology consistent with typical PD, diffuse Lewy body disease, and neurofibrillary tangle and other tau pathology [213,215,216].

Parkin-associated PD — Patients with parkin (PARK2) gene mutations usually have a family history consistent with autosomal recessive inheritance. The disease is characterized by early onset of symptoms (before age 50), a slowly progressive course, a symmetric presentation at onset of parkinsonian signs and symptoms (unlike classic PD, which is asymmetric), and early dystonia and postural instability. Additional features include leg tremor, freezing, festination, retropulsion, hyperreflexia, sensory axonal neuropathy, and autonomic involvement [217-220]. Dementia is uncommon (<3 percent) among PARK2 mutation carriers [221]. However, on an individual basis, patients with early-onset PD who have parkin mutations are clinically indistinguishable from those with early-onset PD who lack parkin mutations [222].

There is typically a good response to levodopa with early development of motor fluctuations and dyskinesia. Fluorodopa PET shows a marked reduction in the fluorodopa uptake, similar to idiopathic PD, but asymptomatic carriers may also have abnormal PET studies [223].

Parkin, the protein product of the PARK2 gene, is expressed in the substantia nigra and other brain regions, as well as in Lewy bodies. Normal parkin strongly binds to microtubules and is involved in ubiquitination and subsequent degradation of certain proteins by proteasomes [37,224]. However, mutated parkin protein loses this proteasome-enhancing activity, and the result is a hastening of neuronal cell death because the disabled proteasome cannot clear the cell of accumulating aggregated protein. Neurodegeneration associated with the parkin mutation is not usually accompanied by formation of Lewy bodies, although there are exceptions [225].

Over 180 mutations, variants, and polymorphisms have been identified in the PARK2 gene [221]. However, it is not clear which of these changes leads to functional deficits. Several studies have demonstrated that heterozygous PARK2 variants may not necessarily be disease-causing [226-228].

The incomplete penetrance and variability of clinical and pathologic expression of PARK2-associated PD may be due to an interaction between the mutated parkin gene and other genes, including SNCA [229].

PINK1-associated PD — Mutations of mitochondrial PTEN-induced putative kinase 1 gene (PINK1; PARK6) are associated with autosomal recessive familial PD, age younger than 50 at onset, slow progression, and excellent response to levodopa [230-232], similar to parkin (PARK2) and DJ-1 (PARK7) mutations. PINK1 mutations have been found worldwide with a frequency that ranges from 1 to 8 percent of patients, most of whom had early-onset and/or familial PD [166,231,233-235].

Postmortem brain examinations of patients with PD and PINK1 mutations are limited to a few cases. One showed Lewy body pathology in the brainstem and Meynert nucleus while the locus ceruleus and the amygdala were spared [236]; another case revealed nigral degeneration without Lewy bodies [237].

PINK1 mutations may cause disease through a loss-of-function effect resulting in mitochondrial dysfunction [238,239].

DJ-1-associated PD — Mutations of the mitochondrial DJ-1 gene (PARK7) are associated with autosomal recessive inheritance, age younger than 40 at onset, slow progression, and good response to levodopa [240,241]. Wildtype DJ-1 is thought to be neuroprotective against oxidative stress.

Other monogenic forms of PD — Other monogenic forms of PD are reviewed in the table (table 2) [242-247]. Novel genetic forms of parkinsonism are likely to emerge from future research, thereby shrinking the large majority of PD cases currently regarded as sporadic.

Glucocerebrosidase gene — Heterozygous mutations in the glucocerebrosidase (GBA) gene, which when homozygous are the cause of Gaucher disease, are an important genetic risk factor for PD [4,248-251].

Across various populations, the risk of PD among GBA variant carriers is increased by two- to sevenfold over noncarriers [248,252-255]. The estimated penetrance of heterozygous GBA mutations for PD over a lifetime varies fairly widely, from approximately 10 to 30 percent depending on the population studied [256-260]. Genotype-phenotype relationships have not been well established, but GBA variant carriers as a group appear to have more cognitive decline compared with patients with idiopathic PD.

Mutations in GBA were initially linked with PD and other movement disorders in people of Ashkenazi Jewish decent, who have an increased prevalence of GBA mutations [261]. One of the largest studies analyzed data from 16 centers around the world, including 5691 patients with PD and 4898 controls [248]. All centers screened for two relatively frequent GBA mutations (N370S and L444P). Compared with controls, either GBA mutation was more common in patients with PD, both among Ashkenazi Jewish subjects (15 percent, versus 3 percent of controls) and among non-Ashkenazi Jewish subjects (3 percent, versus <1 percent of controls). Overall, the likelihood of finding any GBA mutation was significantly higher in patients with PD than in controls (odds ratio 5.43, 95% CI 3.89-7.57).

Among all patients with PD, the clinical profile was generally similar. However, when compared with patients who had PD but lacked a GBA mutation, those with PD who carried a GBA mutation were significantly more likely to have the following features [248]:

Younger age at onset

Less prominent tremor, bradykinesia, and rigidity

Lower frequency of asymmetric onset

Higher frequency of a family history of PD

Greater likelihood of cognitive impairment

Other reports have also found that patients with PD who are GBA mutation carriers have a younger age at onset, faster motor progression, and an increased prevalence of cognitive dysfunction compared with noncarriers [261-267].

Other lysosomal genes — There is emerging evidence that mutations in other lysosomal enzyme-encoding genes also affect PD risk through effects on alpha-synuclein [268-271]. For example, several genetic variants in the acid sphingomyelinase gene SMPD1, the gene associated with Niemann-Pick type A, have been identified with increased prevalence in patients of Ashkenazi Jewish ancestry (and other populations) with PD compared with controls [272-275]. Furthermore, experimentally, SMPD1 knockdown results in alpha-synuclein accumulation [272].

Genetic testing — Molecular genetic testing is available for many of the genetic forms of PD, including SNCA, parkin (PARK2), PINK1, DJ-1 (PARK7), and LRRK2 (PARK8) gene mutations [276], but the interpretation of these tests is difficult, and there are no established disease-modifying therapies yet available for PD. Genetic testing is therefore not recommended in routine clinical practice at this time [166,277,278].

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Parkinson disease (The Basics)")

Beyond the Basics topics (see "Patient education: Parkinson disease symptoms and diagnosis (Beyond the Basics)")

PATIENT PERSPECTIVE TOPIC — Patient perspectives are provided for selected disorders to help clinicians better understand the patient experience and patient concerns. These narratives may offer insights into patient values and preferences not included in other UpToDate topics. (See "Patient perspective: Parkinson disease".)

SUMMARY

In Parkinson disease (PD), dopamine depletion in the substantia nigra and in the nigrostriatal pathway to the caudate and putamen ultimately results in increased inhibition of the thalamus and reduced excitatory input to the motor cortex (figure 1), which is expressed as bradykinesia and other parkinsonian signs. (See 'Basal ganglia circuits' above.)

Compensatory mechanisms may operate to mask or reduce the deleterious effects of dopamine depletion, particularly in the presymptomatic phase of PD. (See 'Compensatory mechanisms' above.)

Typical abnormalities found in the brains of patients with PD include depigmentation (from loss of neuromelanin), neuronal loss, and gliosis, particularly in the substantia nigra pars compacta (SNc) and in the pontine locus ceruleus. Lewy bodies are round, eosinophilic, intracytoplasmic inclusions in the nuclei of neurons that constitute the pathologic hallmark of PD. (See 'Pathology' above.)

The precise mechanisms of neurodegeneration in PD are not yet understood, but they most likely involve a cascade of events that include interaction between genetic and environmental factors, and abnormalities in protein processing, oxidative stress, mitochondrial dysfunction, excitotoxicity, inflammation, immune regulation, and other mechanisms (figure 3 and figure 4). (See 'Pathogenesis of cell degeneration' above.)

The prevalence of PD is approximately 0.3 percent in the general population 40 years of age and older. The prevalence of PD rises with age. Worldwide, there are approximately 6.1 million people with PD. (See 'Epidemiology' above.)

There is consistent evidence that older age is associated with increased risk of PD, while cigarette smoking is a protective factor. Evidence supporting other potential risk factors for PD, including occupational exposures (eg, pesticides, herbicides, heavy metals), dietary factors, and body weight is inconclusive. (See 'Associated factors' above.)

Although the majority of cases of PD appear to be sporadic, there are genetic forms of parkinsonism (designated PARK1 through PARK13) related to nuclear and mitochondrial genes. Novel genetic forms of parkinsonism are likely to emerge from future research, thereby shrinking the large majority of PD cases currently regarded as sporadic. (See 'Genetics' above.)

REFERENCES

  1. Manyam BV. Paralysis agitans and levodopa in "Ayurveda": ancient Indian medical treatise. Mov Disord 1990; 5:47.
  2. Katzenschlager R, Evans A, Manson A, et al. Mucuna pruriens in Parkinson's disease: a double blind clinical and pharmacological study. J Neurol Neurosurg Psychiatry 2004; 75:1672.
  3. Hornykiewicz O. The discovery of dopamine deficiency in the parkinsonian brain. J Neural Transm Suppl 2006; :9.
  4. Jankovic J, Tan EK. Parkinson's disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry 2020; 91:795.
  5. Gerfen CR. Molecular effects of dopamine on striatal-projection pathways. Trends Neurosci 2000; 23:S64.
  6. Bamford NS, Robinson S, Palmiter RD, et al. Dopamine modulates release from corticostriatal terminals. J Neurosci 2004; 24:9541.
  7. Ryoo HL, Pierrotti D, Joyce JN. Dopamine D3 receptor is decreased and D2 receptor is elevated in the striatum of Parkinson's disease. Mov Disord 1998; 13:788.
  8. Gatev P, Darbin O, Wichmann T. Oscillations in the basal ganglia under normal conditions and in movement disorders. Mov Disord 2006; 21:1566.
  9. Obeso JA, Rodríguez-Oroz MC, Rodríguez M, et al. Pathophysiology of the basal ganglia in Parkinson's disease. Trends Neurosci 2000; 23:S8.
  10. Calabresi P, Centonze D, Bernardi G. Electrophysiology of dopamine in normal and denervated striatal neurons. Trends Neurosci 2000; 23:S57.
  11. Moore H, Grace AA. A role for electrotonic coupling in the striatum in the expression of dopamine receptor-mediated stereotypies. Neuropsychopharmacology 2002; 27:980.
  12. Huot P, Lévesque M, Parent A. The fate of striatal dopaminergic neurons in Parkinson's disease and Huntington's chorea. Brain 2007; 130:222.
  13. Huot P, Parent A. Dopaminergic neurons intrinsic to the striatum. J Neurochem 2007; 101:1441.
  14. Bezard E, Gross CE, Brotchie JM. Presymptomatic compensation in Parkinson's disease is not dopamine-mediated. Trends Neurosci 2003; 26:215.
  15. Adams JR, van Netten H, Schulzer M, et al. PET in LRRK2 mutations: comparison to sporadic Parkinson's disease and evidence for presymptomatic compensation. Brain 2005; 128:2777.
  16. Pakkenberg B, Møller A, Gundersen HJ, et al. The absolute number of nerve cells in substantia nigra in normal subjects and in patients with Parkinson's disease estimated with an unbiased stereological method. J Neurol Neurosurg Psychiatry 1991; 54:30.
  17. Porritt M, Stanic D, Finkelstein D, et al. Dopaminergic innervation of the human striatum in Parkinson's disease. Mov Disord 2005; 20:810.
  18. Fearnley JM, Lees AJ. Ageing and Parkinson's disease: substantia nigra regional selectivity. Brain 1991; 114 ( Pt 5):2283.
  19. Henderson JM, Carpenter K, Cartwright H, Halliday GM. Degeneration of the centré median-parafascicular complex in Parkinson's disease. Ann Neurol 2000; 47:345.
  20. Camicioli R, Moore MM, Kinney A, et al. Parkinson's disease is associated with hippocampal atrophy. Mov Disord 2003; 18:784.
  21. Murakami T, Shoji M, Imai Y, et al. Pael-R is accumulated in Lewy bodies of Parkinson's disease. Ann Neurol 2004; 55:439.
  22. Wakabayashi K, Engelender S, Yoshimoto M, et al. Synphilin-1 is present in Lewy bodies in Parkinson's disease. Ann Neurol 2000; 47:521.
  23. Wakabayashi K, Tanji K, Mori F, Takahashi H. The Lewy body in Parkinson's disease: molecules implicated in the formation and degradation of alpha-synuclein aggregates. Neuropathology 2007; 27:494.
  24. Popescu A, Lippa CF, Lee VM, Trojanowski JQ. Lewy bodies in the amygdala: increase of alpha-synuclein aggregates in neurodegenerative diseases with tau-based inclusions. Arch Neurol 2004; 61:1915.
  25. Thenganatt MA, Jankovic J. Parkinson disease subtypes. JAMA Neurol 2014; 71:499.
  26. Bodner RA, Outeiro TF, Altmann S, et al. Pharmacological promotion of inclusion formation: a therapeutic approach for Huntington's and Parkinson's diseases. Proc Natl Acad Sci U S A 2006; 103:4246.
  27. Goedert M, Spillantini MG, Del Tredici K, Braak H. 100 years of Lewy pathology. Nat Rev Neurol 2013; 9:13.
  28. Braak H, Del Tredici K. Invited Article: Nervous system pathology in sporadic Parkinson disease. Neurology 2008; 70:1916.
  29. Burke RE, Dauer WT, Vonsattel JP. A critical evaluation of the Braak staging scheme for Parkinson's disease. Ann Neurol 2008; 64:485.
  30. Jellinger KA. Neuropathology of sporadic Parkinson's disease: evaluation and changes of concepts. Mov Disord 2012; 27:8.
  31. Savitt JM, Dawson VL, Dawson TM. Diagnosis and treatment of Parkinson disease: molecules to medicine. J Clin Invest 2006; 116:1744.
  32. Lang AE. The progression of Parkinson disease: a hypothesis. Neurology 2007; 68:948.
  33. Atkin G, Paulson H. Ubiquitin pathways in neurodegenerative disease. Front Mol Neurosci 2014; 7:63.
  34. Pan T, Kondo S, Le W, Jankovic J. The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson's disease. Brain 2008; 131:1969.
  35. Jellinger KA. Cell death mechanisms in Parkinson's disease. J Neural Transm (Vienna) 2000; 107:1.
  36. Tatton WG, Chalmers-Redman R, Brown D, Tatton N. Apoptosis in Parkinson's disease: signals for neuronal degradation. Ann Neurol 2003; 53 Suppl 3:S61.
  37. Feng J. Microtubule: a common target for parkin and Parkinson's disease toxins. Neuroscientist 2006; 12:469.
  38. Maragakis NJ, Rothstein JD. Mechanisms of Disease: astrocytes in neurodegenerative disease. Nat Clin Pract Neurol 2006; 2:679.
  39. Logroscino G. The role of early life environmental risk factors in Parkinson disease: what is the evidence? Environ Health Perspect 2005; 113:1234.
  40. Cory-Slechta DA, Thiruchelvam M, Barlow BK, Richfield EK. Developmental pesticide models of the Parkinson disease phenotype. Environ Health Perspect 2005; 113:1263.
  41. Maries E, Dass B, Collier TJ, et al. The role of alpha-synuclein in Parkinson's disease: insights from animal models. Nat Rev Neurosci 2003; 4:727.
  42. Calo L, Wegrzynowicz M, Santivañez-Perez J, Grazia Spillantini M. Synaptic failure and α-synuclein. Mov Disord 2016; 31:169.
  43. Feany MB. New genetic insights into Parkinson's disease. N Engl J Med 2004; 351:1937.
  44. Meredith GE, Halliday GM, Totterdell S. A critical review of the development and importance of proteinaceous aggregates in animal models of Parkinson's disease: new insights into Lewy body formation. Parkinsonism Relat Disord 2004; 10:191.
  45. Spillantini MG, Schmidt ML, Lee VM, et al. Alpha-synuclein in Lewy bodies. Nature 1997; 388:839.
  46. Dehay B, Vila M, Bezard E, et al. Alpha-synuclein propagation: New insights from animal models. Mov Disord 2016; 31:161.
  47. Luk KC, Kehm V, Carroll J, et al. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 2012; 338:949.
  48. Luk KC, Kehm VM, Zhang B, et al. Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice. J Exp Med 2012; 209:975.
  49. Kordower JH, Chu Y, Hauser RA, et al. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease. Nat Med 2008; 14:504.
  50. Li JY, Englund E, Holton JL, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med 2008; 14:501.
  51. Lashuel HA, Petre BM, Wall J, et al. Alpha-synuclein, especially the Parkinson's disease-associated mutants, forms pore-like annular and tubular protofibrils. J Mol Biol 2002; 322:1089.
  52. Jankovic J. Pathogenesis-targeted therapeutic strategies in Parkinson's disease. Mov Disord 2019; 34:41.
  53. Kam TI, Mao X, Park H, et al. Poly(ADP-ribose) drives pathologic α-synuclein neurodegeneration in Parkinson's disease. Science 2018; 362.
  54. Rietdijk CD, Perez-Pardo P, Garssen J, et al. Exploring Braak's Hypothesis of Parkinson's Disease. Front Neurol 2017; 8:37.
  55. Espay AJ, Vizcarra JA, Marsili L, et al. Revisiting protein aggregation as pathogenic in sporadic Parkinson and Alzheimer diseases. Neurology 2019; 92:329.
  56. Lim KL, Zhang CW. Molecular events underlying Parkinson's disease - an interwoven tapestry. Front Neurol 2013; 4:33.
  57. Dehay B, Martinez-Vicente M, Caldwell GA, et al. Lysosomal impairment in Parkinson's disease. Mov Disord 2013; 28:725.
  58. Ghavami S, Shojaei S, Yeganeh B, et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol 2014; 112:24.
  59. Xilouri M, Brekk OR, Stefanis L. Autophagy and Alpha-Synuclein: Relevance to Parkinson's Disease and Related Synucleopathies. Mov Disord 2016; 31:178.
  60. 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; 119:650.
  61. Usenovic M, Tresse E, Mazzulli JR, et al. Deficiency of ATP13A2 leads to lysosomal dysfunction, α-synuclein accumulation, and neurotoxicity. J Neurosci 2012; 32:4240.
  62. Bové J, Prou D, Perier C, Przedborski S. Toxin-induced models of Parkinson's disease. NeuroRx 2005; 2:484.
  63. Przedborski S, Tieu K, Perier C, Vila M. MPTP as a mitochondrial neurotoxic model of Parkinson's disease. J Bioenerg Biomembr 2004; 36:375.
  64. Selvaraj S, Sun Y, Watt JA, et al. Neurotoxin-induced ER stress in mouse dopaminergic neurons involves downregulation of TRPC1 and inhibition of AKT/mTOR signaling. J Clin Invest 2012; 122:1354.
  65. Schapira AH, Cooper JM, Dexter D, et al. Mitochondrial complex I deficiency in Parkinson's disease. Lancet 1989; 1:1269.
  66. Greenamyre JT, Hastings TG. Biomedicine. Parkinson's--divergent causes, convergent mechanisms. Science 2004; 304:1120.
  67. Mortiboys H, Johansen KK, Aasly JO, Bandmann O. Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2. Neurology 2010; 75:2017.
  68. Yu SW, Wang H, Poitras MF, et al. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 2002; 297:259.
  69. Krantic S, Mechawar N, Reix S, Quirion R. Molecular basis of programmed cell death involved in neurodegeneration. Trends Neurosci 2005; 28:670.
  70. Sherer TB, Betarbet R, Testa CM, et al. Mechanism of toxicity in rotenone models of Parkinson's disease. J Neurosci 2003; 23:10756.
  71. Abou-Sleiman PM, Muqit MM, McDonald NQ, et al. A heterozygous effect for PINK1 mutations in Parkinson's disease? Ann Neurol 2006; 60:414.
  72. Schapira AH. Mitochondria in the aetiology and pathogenesis of Parkinson's disease. Lancet Neurol 2008; 7:97.
  73. Ahlskog JE. Challenging conventional wisdom: the etiologic role of dopamine oxidative stress in Parkinson's disease. Mov Disord 2005; 20:271.
  74. Hirsch EC, Hunot S, Hartmann A. Neuroinflammatory processes in Parkinson's disease. Parkinsonism Relat Disord 2005; 11 Suppl 1:S9.
  75. Uehara T, Nakamura T, Yao D, et al. S-nitrosylated protein-disulphide isomerase links protein misfolding to neurodegeneration. Nature 2006; 441:513.
  76. Benarroch EE. Brain iron homeostasis and neurodegenerative disease. Neurology 2009; 72:1436.
  77. Oakley AE, Collingwood JF, Dobson J, et al. Individual dopaminergic neurons show raised iron levels in Parkinson disease. Neurology 2007; 68:1820.
  78. Dusek P, Jankovic J, Le W. Iron dysregulation in movement disorders. Neurobiol Dis 2012; 46:1.
  79. Lei P, Ayton S, Finkelstein DI, et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat Med 2012; 18:291.
  80. Zhu W, Xie W, Pan T, et al. Prevention and restoration of lactacystin-induced nigrostriatal dopamine neuron degeneration by novel brain-permeable iron chelators. FASEB J 2007; 21:3835.
  81. Hirsch EC, Hunot S. Neuroinflammation in Parkinson's disease: a target for neuroprotection? Lancet Neurol 2009; 8:382.
  82. Hirsch EC, Jenner P, Przedborski S. Pathogenesis of Parkinson's disease. Mov Disord 2013; 28:24.
  83. Teismann P, Tieu K, Choi DK, et al. Cyclooxygenase-2 is instrumental in Parkinson's disease neurodegeneration. Proc Natl Acad Sci U S A 2003; 100:5473.
  84. Ouchi Y, Yoshikawa E, Sekine Y, et al. Microglial activation and dopamine terminal loss in early Parkinson's disease. Ann Neurol 2005; 57:168.
  85. Brochard V, Combadière B, Prigent A, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest 2009; 119:182.
  86. Pringsheim T, Jette N, Frolkis A, Steeves TD. The prevalence of Parkinson's disease: a systematic review and meta-analysis. Mov Disord 2014; 29:1583.
  87. GBD 2016 Parkinson's Disease Collaborators. Global, regional, and national burden of Parkinson's disease, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 2018; 17:939.
  88. Rong S, Xu G, Liu B, et al. Trends in Mortality From Parkinson Disease in the United States, 1999-2019. Neurology 2021; 97:e1986.
  89. de Lau LM, Breteler MM. Epidemiology of Parkinson's disease. Lancet Neurol 2006; 5:525.
  90. Moisan F, Kab S, Mohamed F, et al. Parkinson disease male-to-female ratios increase with age: French nationwide study and meta-analysis. J Neurol Neurosurg Psychiatry 2016; 87:952.
  91. Noyce AJ, Bestwick JP, Silveira-Moriyama L, et al. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann Neurol 2012; 72:893.
  92. Kieburtz K, Wunderle KB. Parkinson's disease: evidence for environmental risk factors. Mov Disord 2013; 28:8.
  93. Chen H, Huang X, Guo X, et al. Smoking duration, intensity, and risk of Parkinson disease. Neurology 2010; 74:878.
  94. Ritz B, Ascherio A, Checkoway H, et al. Pooled analysis of tobacco use and risk of Parkinson disease. Arch Neurol 2007; 64:990.
  95. Liu R, Guo X, Park Y, et al. Caffeine intake, smoking, and risk of Parkinson disease in men and women. Am J Epidemiol 2012; 175:1200.
  96. Mappin-Kasirer B, Pan H, Lewington S, et al. Tobacco smoking and the risk of Parkinson disease: A 65-year follow-up of 30,000 male British doctors. Neurology 2020; 94:e2132.
  97. Quik M. Smoking, nicotine and Parkinson's disease. Trends Neurosci 2004; 27:561.
  98. Inzelberg R, Jankovic J. Are Parkinson disease patients protected from some but not all cancers? Neurology 2007; 69:1542.
  99. Stamey W, Jankovic J. Impulse control disorders and pathological gambling in patients with Parkinson disease. Neurologist 2008; 14:89.
  100. Ritz B, Lee PC, Lassen CF, Arah OA. Parkinson disease and smoking revisited: ease of quitting is an early sign of the disease. Neurology 2014; 83:1396.
  101. Evans AH, Lawrence AD, Potts J, et al. Relationship between impulsive sensation seeking traits, smoking, alcohol and caffeine intake, and Parkinson's disease. J Neurol Neurosurg Psychiatry 2006; 77:317.
  102. Grover S, Lill CM, Kasten M, et al. Risky behaviors and Parkinson disease: A mendelian randomization study. Neurology 2019; 93:e1412.
  103. Palacios N, Gao X, McCullough ML, et al. Caffeine and risk of Parkinson's disease in a large cohort of men and women. Mov Disord 2012; 27:1276.
  104. Costa J, Lunet N, Santos C, et al. Caffeine exposure and the risk of Parkinson's disease: a systematic review and meta-analysis of observational studies. J Alzheimers Dis 2010; 20 Suppl 1:S221.
  105. Logroscino G, Sesso HD, Paffenbarger RS Jr, Lee IM. Physical activity and risk of Parkinson's disease: a prospective cohort study. J Neurol Neurosurg Psychiatry 2006; 77:1318.
  106. Chen H, Zhang SM, Schwarzschild MA, et al. Physical activity and the risk of Parkinson disease. Neurology 2005; 64:664.
  107. Thacker EL, Chen H, Patel AV, et al. Recreational physical activity and risk of Parkinson's disease. Mov Disord 2008; 23:69.
  108. Xu Q, Park Y, Huang X, et al. Physical activities and future risk of Parkinson disease. Neurology 2010; 75:341.
  109. Ahlskog JE. Does vigorous exercise have a neuroprotective effect in Parkinson disease? Neurology 2011; 77:288.
  110. Hughes KC, Gao X, Molsberry S, et al. Physical activity and prodromal features of Parkinson disease. Neurology 2019; 93:e2157.
  111. Yoon SY, Suh JH, Yang SN, et al. Association of Physical Activity, Including Amount and Maintenance, With All-Cause Mortality in Parkinson Disease. JAMA Neurol 2021; 78:1446.
  112. Fang X, Han D, Cheng Q, et al. Association of Levels of Physical Activity With Risk of Parkinson Disease: A Systematic Review and Meta-analysis. JAMA Netw Open 2018; 1:e182421.
  113. Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of Parkinson's disease: systematic review and meta-analysis of observational studies. Drugs Aging 2009; 26:769.
  114. Gagne JJ, Power MC. Anti-inflammatory drugs and risk of Parkinson disease: a meta-analysis. Neurology 2010; 74:995.
  115. Gao X, Chen H, Schwarzschild MA, Ascherio A. Use of ibuprofen and risk of Parkinson disease. Neurology 2011; 76:863.
  116. Rees K, Stowe R, Patel S, et al. Non-steroidal anti-inflammatory drugs as disease-modifying agents for Parkinson's disease: evidence from observational studies. Cochrane Database Syst Rev 2011; :CD008454.
  117. Bai S, Song Y, Huang X, et al. Statin Use and the Risk of Parkinson's Disease: An Updated Meta-Analysis. PLoS One 2016; 11:e0152564.
  118. Sheng Z, Jia X, Kang M. Statin use and risk of Parkinson's disease: A meta-analysis. Behav Brain Res 2016; 309:29.
  119. Bykov K, Yoshida K, Weisskopf MG, Gagne JJ. Confounding of the association between statins and Parkinson disease: systematic review and meta-analysis. Pharmacoepidemiol Drug Saf 2017; 26:294.
  120. Liu G, Sterling NW, Kong L, et al. Statins may facilitate Parkinson's disease: Insight gained from a large, national claims database. Mov Disord 2017; 32:913.
  121. Jeong SH, Lee HS, Chung SJ, et al. Effects of statins on dopamine loss and prognosis in Parkinson's disease. Brain 2021; 144:3191.
  122. Cai R, Zhang Y, Simmering JE, et al. Enhancing glycolysis attenuates Parkinson's disease progression in models and clinical databases. J Clin Invest 2019; 129:4539.
  123. Simmering JE, Welsh MJ, Liu L, et al. Association of Glycolysis-Enhancing α-1 Blockers With Risk of Developing Parkinson Disease. JAMA Neurol 2021; 78:407.
  124. Gustafsson H, Nordström A, Nordström P. Depression and subsequent risk of Parkinson disease: A nationwide cohort study. Neurology 2015; 84:2422.
  125. Shen CC, Tsai SJ, Perng CL, et al. Risk of Parkinson disease after depression: a nationwide population-based study. Neurology 2013; 81:1538.
  126. Fang F, Xu Q, Park Y, et al. Depression and the subsequent risk of Parkinson's disease in the NIH-AARP Diet and Health Study. Mov Disord 2010; 25:1157.
  127. Jacob EL, Gatto NM, Thompson A, et al. Occurrence of depression and anxiety prior to Parkinson's disease. Parkinsonism Relat Disord 2010; 16:576.
  128. Alonso A, Rodríguez LA, Logroscino G, Hernán MA. Use of antidepressants and the risk of Parkinson's disease: a prospective study. J Neurol Neurosurg Psychiatry 2009; 80:671.
  129. Ishihara L, Brayne C. A systematic review of depression and mental illness preceding Parkinson's disease. Acta Neurol Scand 2006; 113:211.
  130. Adams-Carr KL, Bestwick JP, Shribman S, et al. Constipation preceding Parkinson's disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 2016; 87:710.
  131. Ascherio A, Chen H, Weisskopf MG, et al. Pesticide exposure and risk for Parkinson's disease. Ann Neurol 2006; 60:197.
  132. Frigerio R, Sanft KR, Grossardt BR, et al. Chemical exposures and Parkinson's disease: a population-based case-control study. Mov Disord 2006; 21:1688.
  133. Costello S, Cockburn M, Bronstein J, et al. Parkinson's disease and residential exposure to maneb and paraquat from agricultural applications in the central valley of California. Am J Epidemiol 2009; 169:919.
  134. Firestone JA, Smith-Weller T, Franklin G, et al. Pesticides and risk of Parkinson disease: a population-based case-control study. Arch Neurol 2005; 62:91.
  135. Elbaz A, Clavel J, Rathouz PJ, et al. Professional exposure to pesticides and Parkinson disease. Ann Neurol 2009; 66:494.
  136. Weisskopf MG, Knekt P, O'Reilly EJ, et al. Persistent organochlorine pesticides in serum and risk of Parkinson disease. Neurology 2010; 74:1055.
  137. Pezzoli G, Cereda E. Exposure to pesticides or solvents and risk of Parkinson disease. Neurology 2013; 80:2035.
  138. Jo S, Kim YJ, Park KW, et al. Association of NO2 and Other Air Pollution Exposures With the Risk of Parkinson Disease. JAMA Neurol 2021; 78:800.
  139. Jiang W, Ju C, Jiang H, Zhang D. Dairy foods intake and risk of Parkinson's disease: a dose-response meta-analysis of prospective cohort studies. Eur J Epidemiol 2014; 29:613.
  140. Ascherio A, Schwarzschild MA. The epidemiology of Parkinson's disease: risk factors and prevention. Lancet Neurol 2016; 15:1257.
  141. Jafari S, Etminan M, Aminzadeh F, Samii A. Head injury and risk of Parkinson disease: a systematic review and meta-analysis. Mov Disord 2013; 28:1222.
  142. Gardner RC, Burke JF, Nettiksimmons J, et al. Traumatic brain injury in later life increases risk for Parkinson disease. Ann Neurol 2015; 77:987.
  143. Gardner RC, Byers AL, Barnes DE, et al. Mild TBI and risk of Parkinson disease: A Chronic Effects of Neurotrauma Consortium Study. Neurology 2018; 90:e1771.
  144. Lv Z, Qi H, Wang L, et al. Vitamin D status and Parkinson's disease: a systematic review and meta-analysis. Neurol Sci 2014; 35:1723.
  145. Wang L, Evatt ML, Maldonado LG, et al. Vitamin D from different sources is inversely associated with Parkinson disease. Mov Disord 2015; 30:560.
  146. Shrestha S, Lutsey PL, Alonso A, et al. Serum 25-hydroxyvitamin D concentrations in Mid-adulthood and Parkinson's disease risk. Mov Disord 2016; 31:972.
  147. Scher AI, Ross GW, Sigurdsson S, et al. Midlife migraine and late-life parkinsonism: AGES-Reykjavik study. Neurology 2014; 83:1246.
  148. Willis AW, Evanoff BA, Lian M, et al. Metal emissions and urban incident Parkinson disease: a community health study of Medicare beneficiaries by using geographic information systems. Am J Epidemiol 2010; 172:1357.
  149. Goldman SM, Quinlan PJ, Ross GW, et al. Solvent exposures and Parkinson disease risk in twins. Ann Neurol 2012; 71:776.
  150. Petrovitch H, Ross GW, Abbott RD, et al. Plantation work and risk of Parkinson disease in a population-based longitudinal study. Arch Neurol 2002; 59:1787.
  151. Powers KM, Smith-Weller T, Franklin GM, et al. Parkinson's disease risks associated with dietary iron, manganese, and other nutrient intakes. Neurology 2003; 60:1761.
  152. Hu G, Jousilahti P, Nissinen A, et al. Body mass index and the risk of Parkinson disease. Neurology 2006; 67:1955.
  153. De Pablo-Fernandez E, Goldacre R, Pakpoor J, et al. Association between diabetes and subsequent Parkinson disease: A record-linkage cohort study. Neurology 2018; 91:e139.
  154. Nam GE, Kim SM, Han K, et al. Metabolic syndrome and risk of Parkinson disease: A nationwide cohort study. PLoS Med 2018; 15:e1002640.
  155. Chohan H, Senkevich K, Patel RK, et al. Type 2 Diabetes as a Determinant of Parkinson's Disease Risk and Progression. Mov Disord 2021; 36:1420.
  156. Frigerio R, Elbaz A, Sanft KR, et al. Education and occupations preceding Parkinson disease: a population-based case-control study. Neurology 2005; 65:1575.
  157. Beard JD, Steege AL, Ju J, et al. Mortality from Amyotrophic Lateral Sclerosis and Parkinson's Disease Among Different Occupation Groups - United States, 1985-2011. MMWR Morb Mortal Wkly Rep 2017; 66:718.
  158. Liu R, Gao X, Lu Y, Chen H. Meta-analysis of the relationship between Parkinson disease and melanoma. Neurology 2011; 76:2002.
  159. Kareus SA, Figueroa KP, Cannon-Albright LA, Pulst SM. Shared predispositions of parkinsonism and cancer: a population-based pedigree-linked study. Arch Neurol 2012; 69:1572.
  160. Pan T, Zhu J, Hwu WJ, Jankovic J. The role of alpha-synuclein in melanin synthesis in melanoma and dopaminergic neuronal cells. PLoS One 2012; 7:e45183.
  161. Inzelberg R, Flash S, Friedman E, Azizi E. Cutaneous malignant melanoma and Parkinson disease: Common pathways? Ann Neurol 2016; 80:811.
  162. Dalvin LA, Damento GM, Yawn BP, et al. Parkinson Disease and Melanoma: Confirming and Reexamining an Association. Mayo Clin Proc 2017; 92:1070.
  163. Singleton AB, Farrer MJ, Bonifati V. The genetics of Parkinson's disease: progress and therapeutic implications. Mov Disord 2013; 28:14.
  164. Marder K, Tang MX, Mejia H, et al. Risk of Parkinson's disease among first-degree relatives: A community-based study. Neurology 1996; 47:155.
  165. Tanner CM, Ottman R, Goldman SM, et al. Parkinson disease in twins: an etiologic study. JAMA 1999; 281:341.
  166. Klein C, Schlossmacher MG. Parkinson disease, 10 years after its genetic revolution: multiple clues to a complex disorder. Neurology 2007; 69:2093.
  167. Tan MMX, Malek N, Lawton MA, et al. Genetic analysis of Mendelian mutations in a large UK population-based Parkinson's disease study. Brain 2019; 142:2828.
  168. Alcalay RN, Caccappolo E, Mejia-Santana H, et al. Frequency of known mutations in early-onset Parkinson disease: implication for genetic counseling: the consortium on risk for early onset Parkinson disease study. Arch Neurol 2010; 67:1116.
  169. Krüger R, Kuhn W, Müller T, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat Genet 1998; 18:106.
  170. 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; 55:164.
  171. Berg D, Schweitzer KJ, Leitner P, et al. Type and frequency of mutations in the LRRK2 gene in familial and sporadic Parkinson's disease*. Brain 2005; 128:3000.
  172. Polymeropoulos MH, Higgins JJ, Golbe LI, et al. Mapping of a gene for Parkinson's disease to chromosome 4q21-q23. Science 1996; 274:1197.
  173. Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science 1997; 276:2045.
  174. Golbe LI, Di Iorio G, Sanges G, et al. Clinical genetic analysis of Parkinson's disease in the Contursi kindred. Ann Neurol 1996; 40:767.
  175. Bostantjopoulou S, Katsarou Z, Papadimitriou A, et al. Clinical features of parkinsonian patients with the alpha-synuclein (G209A) mutation. Mov Disord 2001; 16:1007.
  176. Trinh J, Zeldenrust FMJ, Huang J, et al. Genotype-phenotype relations for the Parkinson's disease genes SNCA, LRRK2, VPS35: MDSGene systematic review. Mov Disord 2018; 33:1857.
  177. Nishioka K, Hayashi S, Farrer MJ, et al. Clinical heterogeneity of alpha-synuclein gene duplication in Parkinson's disease. Ann Neurol 2006; 59:298.
  178. Fuchs J, Nilsson C, Kachergus J, et al. Phenotypic variation in a large Swedish pedigree due to SNCA duplication and triplication. Neurology 2007; 68:916.
  179. Singleton A, Gwinn-Hardy K, Sharabi Y, et al. Association between cardiac denervation and parkinsonism caused by alpha-synuclein gene triplication. Brain 2004; 127:768.
  180. Shulman JM, De Jager PL. Evidence for a common pathway linking neurodegenerative diseases. Nat Genet 2009; 41:1261.
  181. Maraganore DM, de Andrade M, Elbaz A, et al. Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease. JAMA 2006; 296:661.
  182. Satake W, Nakabayashi Y, Mizuta I, et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson's disease. Nat Genet 2009; 41:1303.
  183. Simón-Sánchez J, Schulte C, Bras JM, et al. Genome-wide association study reveals genetic risk underlying Parkinson's disease. Nat Genet 2009; 41:1308.
  184. International Parkinson Disease Genomics Consortium, Nalls MA, Plagnol V, et al. Imputation of sequence variants for identification of genetic risks for Parkinson's disease: a meta-analysis of genome-wide association studies. Lancet 2011; 377:641.
  185. Pihlstrøm L, Blauwendraat C, Cappelletti C, et al. A comprehensive analysis of SNCA-related genetic risk in sporadic parkinson disease. Ann Neurol 2018; 84:117.
  186. Funayama M, Hasegawa K, Kowa H, et al. A new locus for Parkinson's disease (PARK8) maps to chromosome 12p11.2-q13.1. Ann Neurol 2002; 51:296.
  187. Paisán-Ruíz C, Jain S, Evans EW, et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron 2004; 44:595.
  188. Zimprich A, Biskup S, Leitner P, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004; 44:601.
  189. Schapira AH. The importance of LRRK2 mutations in Parkinson disease. Arch Neurol 2006; 63:1225.
  190. Whaley NR, Uitti RJ, Dickson DW, et al. Clinical and pathologic features of families with LRRK2-associated Parkinson's disease. J Neural Transm Suppl 2006; :221.
  191. Deng H, Le W, Guo Y, et al. Genetic analysis of LRRK2 mutations in patients with Parkinson disease. J Neurol Sci 2006; 251:102.
  192. Brice A. How much does dardarin contribute to Parkinson's disease? Lancet 2005; 365:363.
  193. Lesage S, Ibanez P, Lohmann E, et al. G2019S LRRK2 mutation in French and North African families with Parkinson's disease. Ann Neurol 2005; 58:784.
  194. Clark LN, Wang Y, Karlins E, et al. Frequency of LRRK2 mutations in early- and late-onset Parkinson disease. Neurology 2006; 67:1786.
  195. Gilks WP, Abou-Sleiman PM, Gandhi S, et al. A common LRRK2 mutation in idiopathic Parkinson's disease. Lancet 2005; 365:415.
  196. Farrer M, Stone J, Mata IF, et al. LRRK2 mutations in Parkinson disease. Neurology 2005; 65:738.
  197. Williams-Gray CH, Goris A, Foltynie T, et al. Prevalence of the LRRK2 G2019S mutation in a UK community based idiopathic Parkinson's disease cohort. J Neurol Neurosurg Psychiatry 2006; 77:665.
  198. Lesage S, Janin S, Lohmann E, et al. LRRK2 exon 41 mutations in sporadic Parkinson disease in Europeans. Arch Neurol 2007; 64:425.
  199. 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; 7:583.
  200. Nichols WC, Pankratz N, Hernandez D, et al. Genetic screening for a single common LRRK2 mutation in familial Parkinson's disease. Lancet 2005; 365:410.
  201. Di Fonzo A, Rohé CF, Ferreira J, et al. A frequent LRRK2 gene mutation associated with autosomal dominant Parkinson's disease. Lancet 2005; 365:412.
  202. Gaig C, Ezquerra M, Marti MJ, et al. LRRK2 mutations in Spanish patients with Parkinson disease: frequency, clinical features, and incomplete penetrance. Arch Neurol 2006; 63:377.
  203. Ozelius LJ, Senthil G, Saunders-Pullman R, et al. LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews. N Engl J Med 2006; 354:424.
  204. Goldwurm S, Zini M, Mariani L, et al. Evaluation of LRRK2 G2019S penetrance: relevance for genetic counseling in Parkinson disease. Neurology 2007; 68:1141.
  205. Hedrich K, Winkler S, Hagenah J, et al. Recurrent LRRK2 (Park8) mutations in early-onset Parkinson's disease. Mov Disord 2006; 21:1506.
  206. Kay DM, Zabetian CP, Factor SA, et al. Parkinson's disease and LRRK2: frequency of a common mutation in U.S. movement disorder clinics. Mov Disord 2006; 21:519.
  207. Paisàn-Ruìz C, Sàenz A, Lòpez de Munain A, et al. Familial Parkinson's disease: clinical and genetic analysis of four Basque families. Ann Neurol 2005; 57:365.
  208. Saunders-Pullman R, Mirelman A, Alcalay RN, et al. Progression in the LRRK2-Asssociated Parkinson Disease Population. JAMA Neurol 2018; 75:312.
  209. Agalliu I, Ortega RA, Luciano MS, et al. Cancer outcomes among Parkinson's disease patients with leucine rich repeat kinase 2 mutations, idiopathic Parkinson's disease patients, and nonaffected controls. Mov Disord 2019; 34:1392.
  210. Inzelberg R, Cohen OS, Aharon-Peretz J, et al. The LRRK2 G2019S mutation is associated with Parkinson disease and concomitant non-skin cancers. Neurology 2012; 78:781.
  211. Agalliu I, San Luciano M, Mirelman A, et al. Higher frequency of certain cancers in LRRK2 G2019S mutation carriers with Parkinson disease: a pooled analysis. JAMA Neurol 2015; 72:58.
  212. Warø BJ, Aasly JO. Exploring cancer in LRRK2 mutation carriers and idiopathic Parkinson's disease. Brain Behav 2018; 8:e00858.
  213. Ross OA, Toft M, Whittle AJ, et al. Lrrk2 and Lewy body disease. Ann Neurol 2006; 59:388.
  214. Chen-Plotkin AS, Yuan W, Anderson C, et al. Corticobasal syndrome and primary progressive aphasia as manifestations of LRRK2 gene mutations. Neurology 2008; 70:521.
  215. Spanaki C, Latsoudis H, Plaitakis A. LRRK2 mutations on Crete: R1441H associated with PD evolving to PSP. Neurology 2006; 67:1518.
  216. Kalia LV, Lang AE, Hazrati LN, et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol 2015; 72:100.
  217. 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; 342:1560.
  218. Lohmann E, Periquet M, Bonifati V, et al. How much phenotypic variation can be attributed to parkin genotype? Ann Neurol 2003; 54:176.
  219. Khan NL, Graham E, Critchley P, et al. Parkin disease: a phenotypic study of a large case series. Brain 2003; 126:1279.
  220. Doherty KM, Silveira-Moriyama L, Parkkinen L, et al. Parkin disease: a clinicopathologic entity? JAMA Neurol 2013; 70:571.
  221. Grünewald A, Kasten M, Ziegler A, Klein C. Next-generation phenotyping using the parkin example: time to catch up with genetics. JAMA Neurol 2013; 70:1186.
  222. Lohmann E, Thobois S, Lesage S, et al. A multidisciplinary study of patients with early-onset PD with and without parkin mutations. Neurology 2009; 72:110.
  223. Hilker R, Klein C, Ghaemi M, et al. Positron emission tomographic analysis of the nigrostriatal dopaminergic system in familial parkinsonism associated with mutations in the parkin gene. Ann Neurol 2001; 49:367.
  224. Burke RE. Recent advances in research on Parkinson disease: synuclein and parkin. Neurologist 2004; 10:75.
  225. Pramstaller PP, Schlossmacher MG, Jacques TS, et al. Lewy body Parkinson's disease in a large pedigree with 77 Parkin mutation carriers. Ann Neurol 2005; 58:411.
  226. Kay DM, Moran D, Moses L, et al. Heterozygous parkin point mutations are as common in control subjects as in Parkinson's patients. Ann Neurol 2007; 61:47.
  227. Langston JW, Tanner CM, Schüle B. Parkin gene variations and parkinsonism: association does not imply causation. Ann Neurol 2007; 61:4.
  228. Kay DM, Stevens CF, Hamza TH, et al. A comprehensive analysis of deletions, multiplications, and copy number variations in PARK2. Neurology 2010; 75:1189.
  229. Deng H, Le WD, Hunter CB, et al. Heterogeneous phenotype in a family with compound heterozygous parkin gene mutations. Arch Neurol 2006; 63:273.
  230. Valente EM, Abou-Sleiman PM, Caputo V, et al. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science 2004; 304:1158.
  231. 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; 61:1898.
  232. Valente EM, Salvi S, Ialongo T, et al. PINK1 mutations are associated with sporadic early-onset parkinsonism. Ann Neurol 2004; 56:336.
  233. Hatano Y, Sato K, Elibol B, et al. PARK6-linked autosomal recessive early-onset parkinsonism in Asian populations. Neurology 2004; 63:1482.
  234. Bonifati V, Rohé CF, Breedveld GJ, et al. Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, and phenotypes. Neurology 2005; 65:87.
  235. Ishihara-Paul L, Hulihan MM, Kachergus J, et al. PINK1 mutations and parkinsonism. Neurology 2008; 71:896.
  236. Samaranch L, Lorenzo-Betancor O, Arbelo JM, et al. PINK1-linked parkinsonism is associated with Lewy body pathology. Brain 2010; 133:1128.
  237. Takanashi M, Li Y, Hattori N. Absence of Lewy pathology associated with PINK1 homozygous mutation. Neurology 2016; 86:2212.
  238. Morais VA, Haddad D, Craessaerts K, et al. PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling. Science 2014; 344:203.
  239. Subramaniam SR, Chesselet MF. Mitochondrial dysfunction and oxidative stress in Parkinson's disease. Prog Neurobiol 2013; 106-107:17.
  240. Bonifati V, Rizzu P, van Baren MJ, et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003; 299:256.
  241. Hague S, Rogaeva E, Hernandez D, et al. Early-onset Parkinson's disease caused by a compound heterozygous DJ-1 mutation. Ann Neurol 2003; 54:271.
  242. Wider C, Ross OA, Wszolek ZK. Genetics of Parkinson disease and essential tremor. Curr Opin Neurol 2010; 23:388.
  243. Deng H, Liang H, Jankovic J. F-box only protein 7 gene in parkinsonian-pyramidal disease. JAMA Neurol 2013; 70:20.
  244. Deng H, Gao K, Jankovic J. The VPS35 gene and Parkinson's disease. Mov Disord 2013; 28:569.
  245. Lin MK, Farrer MJ. Genetics and genomics of Parkinson's disease. Genome Med 2014; 6:48.
  246. Park JS, Blair NF, Sue CM. The role of ATP13A2 in Parkinson's disease: Clinical phenotypes and molecular mechanisms. Mov Disord 2015; 30:770.
  247. 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; 17:597.
  248. Sidransky E, Nalls MA, Aasly JO, et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson's disease. N Engl J Med 2009; 361:1651.
  249. Brockmann K, Srulijes K, Hauser AK, et al. GBA-associated PD presents with nonmotor characteristics. Neurology 2011; 77:276.
  250. Sidransky E, Lopez G. The link between the GBA gene and parkinsonism. Lancet Neurol 2012; 11:986.
  251. Murphy KE, Gysbers AM, Abbott SK, et al. Reduced glucocerebrosidase is associated with increased α-synuclein in sporadic Parkinson's disease. Brain 2014; 137:834.
  252. den Heijer JM, Cullen VC, Quadri M, et al. A Large-Scale Full GBA1 Gene Screening in Parkinson's Disease in the Netherlands. Mov Disord 2020; 35:1667.
  253. Gan-Or Z, Amshalom I, Kilarski LL, et al. Differential effects of severe vs mild GBA mutations on Parkinson disease. Neurology 2015; 84:880.
  254. Ruskey JA, Greenbaum L, Roncière L, et al. Increased yield of full GBA sequencing in Ashkenazi Jews with Parkinson's disease. Eur J Med Genet 2019; 62:65.
  255. Lesage S, Anheim M, Condroyer C, et al. Large-scale screening of the Gaucher's disease-related glucocerebrosidase gene in Europeans with Parkinson's disease. Hum Mol Genet 2011; 20:202.
  256. Balestrino R, Tunesi S, Tesei S, et al. Penetrance of Glucocerebrosidase (GBA) Mutations in Parkinson's Disease: A Kin Cohort Study. Mov Disord 2020; 35:2111.
  257. Anheim M, Elbaz A, Lesage S, et al. Penetrance of Parkinson disease in glucocerebrosidase gene mutation carriers. Neurology 2012; 78:417.
  258. Alcalay RN, Dinur T, Quinn T, et al. Comparison of Parkinson risk in Ashkenazi Jewish patients with Gaucher disease and GBA heterozygotes. JAMA Neurol 2014; 71:752.
  259. McNeill A, Duran R, Hughes DA, et al. A clinical and family history study of Parkinson's disease in heterozygous glucocerebrosidase mutation carriers. J Neurol Neurosurg Psychiatry 2012; 83:853.
  260. Rosenbloom B, Balwani M, Bronstein JM, et al. The incidence of Parkinsonism in patients with type 1 Gaucher disease: data from the ICGG Gaucher Registry. Blood Cells Mol Dis 2011; 46:95.
  261. Inzelberg R, Hassin-Baer S, Jankovic J. Genetic movement disorders in patients of Jewish ancestry. JAMA Neurol 2014; 71:1567.
  262. Alcalay RN, Caccappolo E, Mejia-Santana H, et al. Cognitive performance of GBA mutation carriers with early-onset PD: the CORE-PD study. Neurology 2012; 78:1434.
  263. Chahine LM, Qiang J, Ashbridge E, et al. Clinical and biochemical differences in patients having Parkinson disease with vs without GBA mutations. JAMA Neurol 2013; 70:852.
  264. Mata IF, Leverenz JB, Weintraub D, et al. GBA Variants are associated with a distinct pattern of cognitive deficits in Parkinson's disease. Mov Disord 2016; 31:95.
  265. Liu G, Boot B, Locascio JJ, et al. Specifically neuropathic Gaucher's mutations accelerate cognitive decline in Parkinson's. Ann Neurol 2016; 80:674.
  266. Cilia R, Tunesi S, Marotta G, et al. Survival and dementia in GBA-associated Parkinson's disease: The mutation matters. Ann Neurol 2016; 80:662.
  267. Maple-Grødem J, Dalen I, Tysnes OB, et al. Association of GBA Genotype With Motor and Functional Decline in Patients With Newly Diagnosed Parkinson Disease. Neurology 2021; 96:e1036.
  268. Clark LN, Chan R, Cheng R, et al. Gene-wise association of variants in four lysosomal storage disorder genes in neuropathologically confirmed Lewy body disease. PLoS One 2015; 10:e0125204.
  269. Robak LA, Jansen IE, van Rooij J, et al. Excessive burden of lysosomal storage disorder gene variants in Parkinson's disease. Brain 2017; 140:3191.
  270. Ysselstein D, Shulman JM, Krainc D. Emerging links between pediatric lysosomal storage diseases and adult parkinsonism. Mov Disord 2019; 34:614.
  271. Lee JS, Kanai K, Suzuki M, et al. Arylsulfatase A, a genetic modifier of Parkinson's disease, is an α-synuclein chaperone. Brain 2019; 142:2845.
  272. Alcalay RN, Mallett V, Vanderperre B, et al. SMPD1 mutations, activity, and α-synuclein accumulation in Parkinson's disease. Mov Disord 2019; 34:526.
  273. Dagan E, Schlesinger I, Ayoub M, et al. The contribution of Niemann-Pick SMPD1 mutations to Parkinson disease in Ashkenazi Jews. Parkinsonism Relat Disord 2015; 21:1067.
  274. Deng S, Deng X, Song Z, et al. Systematic Genetic Analysis of the SMPD1 Gene in Chinese Patients with Parkinson's Disease. Mol Neurobiol 2016; 53:5025.
  275. Gan-Or Z, Orr-Urtreger A, Alcalay RN, et al. The emerging role of SMPD1 mutations in Parkinson's disease: Implications for future studies. Parkinsonism Relat Disord 2015; 21:1294.
  276. Pankratz ND, Wojcieszek J, Foroud T. Parkinson Disease Overview. In: GeneReviews. www.ncbi.nlm.nih.gov./books/NBK1223/ (Accessed on January 15, 2013).
  277. Tan EK, Jankovic J. Genetic testing in Parkinson disease: promises and pitfalls. Arch Neurol 2006; 63:1232.
  278. Grimes D, Fitzpatrick M, Gordon J, et al. Canadian guideline for Parkinson disease. CMAJ 2019; 191:E989.
Topic 4906 Version 82.0

References

1 : Paralysis agitans and levodopa in "Ayurveda": ancient Indian medical treatise.

2 : Mucuna pruriens in Parkinson's disease: a double blind clinical and pharmacological study.

3 : The discovery of dopamine deficiency in the parkinsonian brain.

4 : Parkinson's disease: etiopathogenesis and treatment.

5 : Molecular effects of dopamine on striatal-projection pathways.

6 : Dopamine modulates release from corticostriatal terminals.

7 : Dopamine D3 receptor is decreased and D2 receptor is elevated in the striatum of Parkinson's disease.

8 : Oscillations in the basal ganglia under normal conditions and in movement disorders.

9 : Pathophysiology of the basal ganglia in Parkinson's disease.

10 : Electrophysiology of dopamine in normal and denervated striatal neurons.

11 : A role for electrotonic coupling in the striatum in the expression of dopamine receptor-mediated stereotypies.

12 : The fate of striatal dopaminergic neurons in Parkinson's disease and Huntington's chorea.

13 : Dopaminergic neurons intrinsic to the striatum.

14 : Presymptomatic compensation in Parkinson's disease is not dopamine-mediated.

15 : PET in LRRK2 mutations: comparison to sporadic Parkinson's disease and evidence for presymptomatic compensation.

16 : The absolute number of nerve cells in substantia nigra in normal subjects and in patients with Parkinson's disease estimated with an unbiased stereological method.

17 : Dopaminergic innervation of the human striatum in Parkinson's disease.

18 : Ageing and Parkinson's disease: substantia nigra regional selectivity.

19 : Degeneration of the centrémedian-parafascicular complex in Parkinson's disease.

20 : Parkinson's disease is associated with hippocampal atrophy.

21 : Pael-R is accumulated in Lewy bodies of Parkinson's disease.

22 : Synphilin-1 is present in Lewy bodies in Parkinson's disease.

23 : The Lewy body in Parkinson's disease: molecules implicated in the formation and degradation of alpha-synuclein aggregates.

24 : Lewy bodies in the amygdala: increase of alpha-synuclein aggregates in neurodegenerative diseases with tau-based inclusions.

25 : Parkinson disease subtypes.

26 : Pharmacological promotion of inclusion formation: a therapeutic approach for Huntington's and Parkinson's diseases.

27 : 100 years of Lewy pathology.

28 : Invited Article: Nervous system pathology in sporadic Parkinson disease.

29 : A critical evaluation of the Braak staging scheme for Parkinson's disease.

30 : Neuropathology of sporadic Parkinson's disease: evaluation and changes of concepts.

31 : Diagnosis and treatment of Parkinson disease: molecules to medicine.

32 : The progression of Parkinson disease: a hypothesis.

33 : Ubiquitin pathways in neurodegenerative disease.

34 : The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson's disease.

35 : Cell death mechanisms in Parkinson's disease.

36 : Apoptosis in Parkinson's disease: signals for neuronal degradation.

37 : Microtubule: a common target for parkin and Parkinson's disease toxins.

38 : Mechanisms of Disease: astrocytes in neurodegenerative disease.

39 : The role of early life environmental risk factors in Parkinson disease: what is the evidence?

40 : Developmental pesticide models of the Parkinson disease phenotype.

41 : The role of alpha-synuclein in Parkinson's disease: insights from animal models.

42 : Synaptic failure andα-synuclein.

43 : New genetic insights into Parkinson's disease.

44 : A critical review of the development and importance of proteinaceous aggregates in animal models of Parkinson's disease: new insights into Lewy body formation.

45 : Alpha-synuclein in Lewy bodies.

46 : Alpha-synuclein propagation: New insights from animal models.

47 : Pathologicalα-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice.

48 : Intracerebral inoculation of pathologicalα-synuclein initiates a rapidly progressive neurodegenerativeα-synucleinopathy in mice.

49 : Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease.

50 : Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation.

51 : Alpha-synuclein, especially the Parkinson's disease-associated mutants, forms pore-like annular and tubular protofibrils.

52 : Pathogenesis-targeted therapeutic strategies in Parkinson's disease.

53 : Poly(ADP-ribose) drives pathologicα-synuclein neurodegeneration in Parkinson's disease.

54 : Exploring Braak's Hypothesis of Parkinson's Disease.

55 : Revisiting protein aggregation as pathogenic in sporadic Parkinson and Alzheimer diseases.

56 : Molecular events underlying Parkinson's disease - an interwoven tapestry.

57 : Lysosomal impairment in Parkinson's disease.

58 : Autophagy and apoptosis dysfunction in neurodegenerative disorders.

59 : Autophagy and Alpha-Synuclein: Relevance to Parkinson's Disease and Related Synucleopathies.

60 : Parkin, PINK1, and DJ-1 form a ubiquitin E3 ligase complex promoting unfolded protein degradation.

61 : Deficiency of ATP13A2 leads to lysosomal dysfunction,α-synuclein accumulation, and neurotoxicity.

62 : Toxin-induced models of Parkinson's disease.

63 : MPTP as a mitochondrial neurotoxic model of Parkinson's disease.

64 : Neurotoxin-induced ER stress in mouse dopaminergic neurons involves downregulation of TRPC1 and inhibition of AKT/mTOR signaling.

65 : Mitochondrial complex I deficiency in Parkinson's disease.

66 : Biomedicine. Parkinson's--divergent causes, convergent mechanisms.

67 : Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2.

68 : Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor.

69 : Molecular basis of programmed cell death involved in neurodegeneration.

70 : Mechanism of toxicity in rotenone models of Parkinson's disease.

71 : A heterozygous effect for PINK1 mutations in Parkinson's disease?

72 : Mitochondria in the aetiology and pathogenesis of Parkinson's disease.

73 : Challenging conventional wisdom: the etiologic role of dopamine oxidative stress in Parkinson's disease.

74 : Neuroinflammatory processes in Parkinson's disease.

75 : S-nitrosylated protein-disulphide isomerase links protein misfolding to neurodegeneration.

76 : Brain iron homeostasis and neurodegenerative disease.

77 : Individual dopaminergic neurons show raised iron levels in Parkinson disease.

78 : Iron dysregulation in movement disorders.

79 : Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export.

80 : Prevention and restoration of lactacystin-induced nigrostriatal dopamine neuron degeneration by novel brain-permeable iron chelators.

81 : Neuroinflammation in Parkinson's disease: a target for neuroprotection?

82 : Pathogenesis of Parkinson's disease.

83 : Cyclooxygenase-2 is instrumental in Parkinson's disease neurodegeneration.

84 : Microglial activation and dopamine terminal loss in early Parkinson's disease.

85 : Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease.

86 : The prevalence of Parkinson's disease: a systematic review and meta-analysis.

87 : Global, regional, and national burden of Parkinson's disease, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016.

88 : Trends in Mortality From Parkinson Disease in the United States, 1999-2019.

89 : Epidemiology of Parkinson's disease.

90 : Parkinson disease male-to-female ratios increase with age: French nationwide study and meta-analysis.

91 : Meta-analysis of early nonmotor features and risk factors for Parkinson disease.

92 : Parkinson's disease: evidence for environmental risk factors.

93 : Smoking duration, intensity, and risk of Parkinson disease.

94 : Pooled analysis of tobacco use and risk of Parkinson disease.

95 : Caffeine intake, smoking, and risk of Parkinson disease in men and women.

96 : Tobacco smoking and the risk of Parkinson disease: A 65-year follow-up of 30,000 male British doctors.

97 : Smoking, nicotine and Parkinson's disease.

98 : Are Parkinson disease patients protected from some but not all cancers?

99 : Impulse control disorders and pathological gambling in patients with Parkinson disease.

100 : Parkinson disease and smoking revisited: ease of quitting is an early sign of the disease.

101 : Relationship between impulsive sensation seeking traits, smoking, alcohol and caffeine intake, and Parkinson's disease.

102 : Risky behaviors and Parkinson disease: A mendelian randomization study.

103 : Caffeine and risk of Parkinson's disease in a large cohort of men and women.

104 : Caffeine exposure and the risk of Parkinson's disease: a systematic review and meta-analysis of observational studies.

105 : Physical activity and risk of Parkinson's disease: a prospective cohort study.

106 : Physical activity and the risk of Parkinson disease.

107 : Recreational physical activity and risk of Parkinson's disease.

108 : Physical activities and future risk of Parkinson disease.

109 : Does vigorous exercise have a neuroprotective effect in Parkinson disease?

110 : Physical activity and prodromal features of Parkinson disease.

111 : Association of Physical Activity, Including Amount and Maintenance, With All-Cause Mortality in Parkinson Disease.

112 : Association of Levels of Physical Activity With Risk of Parkinson Disease: A Systematic Review and Meta-analysis.

113 : NSAID use and the risk of Parkinson's disease: systematic review and meta-analysis of observational studies.

114 : Anti-inflammatory drugs and risk of Parkinson disease: a meta-analysis.

115 : Use of ibuprofen and risk of Parkinson disease.

116 : Non-steroidal anti-inflammatory drugs as disease-modifying agents for Parkinson's disease: evidence from observational studies.

117 : Statin Use and the Risk of Parkinson's Disease: An Updated Meta-Analysis.

118 : Statin use and risk of Parkinson's disease: A meta-analysis.

119 : Confounding of the association between statins and Parkinson disease: systematic review and meta-analysis.

120 : Statins may facilitate Parkinson's disease: Insight gained from a large, national claims database.

121 : Effects of statins on dopamine loss and prognosis in Parkinson's disease.

122 : Enhancing glycolysis attenuates Parkinson's disease progression in models and clinical databases.

123 : Association of Glycolysis-Enhancingα-1 Blockers With Risk of Developing Parkinson Disease.

124 : Depression and subsequent risk of Parkinson disease: A nationwide cohort study.

125 : Risk of Parkinson disease after depression: a nationwide population-based study.

126 : Depression and the subsequent risk of Parkinson's disease in the NIH-AARP Diet and Health Study.

127 : Occurrence of depression and anxiety prior to Parkinson's disease.

128 : Use of antidepressants and the risk of Parkinson's disease: a prospective study.

129 : A systematic review of depression and mental illness preceding Parkinson's disease.

130 : Constipation preceding Parkinson's disease: a systematic review and meta-analysis.

131 : Pesticide exposure and risk for Parkinson's disease.

132 : Chemical exposures and Parkinson's disease: a population-based case-control study.

133 : Parkinson's disease and residential exposure to maneb and paraquat from agricultural applications in the central valley of California.

134 : Pesticides and risk of Parkinson disease: a population-based case-control study.

135 : Professional exposure to pesticides and Parkinson disease.

136 : Persistent organochlorine pesticides in serum and risk of Parkinson disease.

137 : Exposure to pesticides or solvents and risk of Parkinson disease.

138 : Association of NO2 and Other Air Pollution Exposures With the Risk of Parkinson Disease.

139 : Dairy foods intake and risk of Parkinson's disease: a dose-response meta-analysis of prospective cohort studies.

140 : The epidemiology of Parkinson's disease: risk factors and prevention.

141 : Head injury and risk of Parkinson disease: a systematic review and meta-analysis.

142 : Traumatic brain injury in later life increases risk for Parkinson disease.

143 : Mild TBI and risk of Parkinson disease: A Chronic Effects of Neurotrauma Consortium Study.

144 : Vitamin D status and Parkinson's disease: a systematic review and meta-analysis.

145 : Vitamin D from different sources is inversely associated with Parkinson disease.

146 : Serum 25-hydroxyvitamin D concentrations in Mid-adulthood and Parkinson's disease risk.

147 : Midlife migraine and late-life parkinsonism: AGES-Reykjavik study.

148 : Metal emissions and urban incident Parkinson disease: a community health study of Medicare beneficiaries by using geographic information systems.

149 : Solvent exposures and Parkinson disease risk in twins.

150 : Plantation work and risk of Parkinson disease in a population-based longitudinal study.

151 : Parkinson's disease risks associated with dietary iron, manganese, and other nutrient intakes.

152 : Body mass index and the risk of Parkinson disease.

153 : Association between diabetes and subsequent Parkinson disease: A record-linkage cohort study.

154 : Metabolic syndrome and risk of Parkinson disease: A nationwide cohort study.

155 : Type 2 Diabetes as a Determinant of Parkinson's Disease Risk and Progression.

156 : Education and occupations preceding Parkinson disease: a population-based case-control study.

157 : Mortality from Amyotrophic Lateral Sclerosis and Parkinson's Disease Among Different Occupation Groups - United States, 1985-2011.

158 : Meta-analysis of the relationship between Parkinson disease and melanoma.

159 : Shared predispositions of parkinsonism and cancer: a population-based pedigree-linked study.

160 : The role of alpha-synuclein in melanin synthesis in melanoma and dopaminergic neuronal cells.

161 : Cutaneous malignant melanoma and Parkinson disease: Common pathways?

162 : Parkinson Disease and Melanoma: Confirming and Reexamining an Association.

163 : The genetics of Parkinson's disease: progress and therapeutic implications.

164 : Risk of Parkinson's disease among first-degree relatives: A community-based study.

165 : Parkinson disease in twins: an etiologic study.

166 : Parkinson disease, 10 years after its genetic revolution: multiple clues to a complex disorder.

167 : Genetic analysis of Mendelian mutations in a large UK population-based Parkinson's disease study.

168 : Frequency of known mutations in early-onset Parkinson disease: implication for genetic counseling: the consortium on risk for early onset Parkinson disease study.

169 : Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease.

170 : The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia.

171 : Type and frequency of mutations in the LRRK2 gene in familial and sporadic Parkinson's disease*.

172 : Mapping of a gene for Parkinson's disease to chromosome 4q21-q23.

173 : Mutation in the alpha-synuclein gene identified in families with Parkinson's disease.

174 : Clinical genetic analysis of Parkinson's disease in the Contursi kindred.

175 : Clinical features of parkinsonian patients with the alpha-synuclein (G209A) mutation.

176 : Genotype-phenotype relations for the Parkinson's disease genes SNCA, LRRK2, VPS35: MDSGene systematic review.

177 : Clinical heterogeneity of alpha-synuclein gene duplication in Parkinson's disease.

178 : Phenotypic variation in a large Swedish pedigree due to SNCA duplication and triplication.

179 : Association between cardiac denervation and parkinsonism caused by alpha-synuclein gene triplication.

180 : Evidence for a common pathway linking neurodegenerative diseases.

181 : Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease.

182 : Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson's disease.

183 : Genome-wide association study reveals genetic risk underlying Parkinson's disease.

184 : Imputation of sequence variants for identification of genetic risks for Parkinson's disease: a meta-analysis of genome-wide association studies.

185 : A comprehensive analysis of SNCA-related genetic risk in sporadic parkinson disease.

186 : A new locus for Parkinson's disease (PARK8) maps to chromosome 12p11.2-q13.1.

187 : Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease.

188 : Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology.

189 : The importance of LRRK2 mutations in Parkinson disease.

190 : Clinical and pathologic features of families with LRRK2-associated Parkinson's disease.

191 : Genetic analysis of LRRK2 mutations in patients with Parkinson disease.

192 : How much does dardarin contribute to Parkinson's disease?

193 : G2019S LRRK2 mutation in French and North African families with Parkinson's disease.

194 : Frequency of LRRK2 mutations in early- and late-onset Parkinson disease.

195 : A common LRRK2 mutation in idiopathic Parkinson's disease.

196 : LRRK2 mutations in Parkinson disease.

197 : Prevalence of the LRRK2 G2019S mutation in a UK community based idiopathic Parkinson's disease cohort.

198 : LRRK2 exon 41 mutations in sporadic Parkinson disease in Europeans.

199 : Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a case-control study.

200 : Genetic screening for a single common LRRK2 mutation in familial Parkinson's disease.

201 : A frequent LRRK2 gene mutation associated with autosomal dominant Parkinson's disease.

202 : LRRK2 mutations in Spanish patients with Parkinson disease: frequency, clinical features, and incomplete penetrance.

203 : LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews.

204 : Evaluation of LRRK2 G2019S penetrance: relevance for genetic counseling in Parkinson disease.

205 : Recurrent LRRK2 (Park8) mutations in early-onset Parkinson's disease.

206 : Parkinson's disease and LRRK2: frequency of a common mutation in U.S. movement disorder clinics.

207 : Familial Parkinson's disease: clinical and genetic analysis of four Basque families.

208 : Progression in the LRRK2-Asssociated Parkinson Disease Population.

209 : Cancer outcomes among Parkinson's disease patients with leucine rich repeat kinase 2 mutations, idiopathic Parkinson's disease patients, and nonaffected controls.

210 : The LRRK2 G2019S mutation is associated with Parkinson disease and concomitant non-skin cancers.

211 : Higher frequency of certain cancers in LRRK2 G2019S mutation carriers with Parkinson disease: a pooled analysis.

212 : Exploring cancer in LRRK2 mutation carriers and idiopathic Parkinson's disease.

213 : Lrrk2 and Lewy body disease.

214 : Corticobasal syndrome and primary progressive aphasia as manifestations of LRRK2 gene mutations.

215 : LRRK2 mutations on Crete: R1441H associated with PD evolving to PSP.

216 : Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease.

217 : Association between early-onset Parkinson's disease and mutations in the parkin gene.

218 : How much phenotypic variation can be attributed to parkin genotype?

219 : Parkin disease: a phenotypic study of a large case series.

220 : Parkin disease: a clinicopathologic entity?

221 : Next-generation phenotyping using the parkin example: time to catch up with genetics.

222 : A multidisciplinary study of patients with early-onset PD with and without parkin mutations.

223 : Positron emission tomographic analysis of the nigrostriatal dopaminergic system in familial parkinsonism associated with mutations in the parkin gene.

224 : Recent advances in research on Parkinson disease: synuclein and parkin.

225 : Lewy body Parkinson's disease in a large pedigree with 77 Parkin mutation carriers.

226 : Heterozygous parkin point mutations are as common in control subjects as in Parkinson's patients.

227 : Parkin gene variations and parkinsonism: association does not imply causation.

228 : A comprehensive analysis of deletions, multiplications, and copy number variations in PARK2.

229 : Heterogeneous phenotype in a family with compound heterozygous parkin gene mutations.

230 : Hereditary early-onset Parkinson's disease caused by mutations in PINK1.

231 : Analysis of the PINK1 gene in a large cohort of cases with Parkinson disease.

232 : PINK1 mutations are associated with sporadic early-onset parkinsonism.

233 : PARK6-linked autosomal recessive early-onset parkinsonism in Asian populations.

234 : Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, and phenotypes.

235 : PINK1 mutations and parkinsonism.

236 : PINK1-linked parkinsonism is associated with Lewy body pathology.

237 : Absence of Lewy pathology associated with PINK1 homozygous mutation.

238 : PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling.

239 : Mitochondrial dysfunction and oxidative stress in Parkinson's disease.

240 : Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism.

241 : Early-onset Parkinson's disease caused by a compound heterozygous DJ-1 mutation.

242 : Genetics of Parkinson disease and essential tremor.

243 : F-box only protein 7 gene in parkinsonian-pyramidal disease.

244 : The VPS35 gene and Parkinson's disease.

245 : Genetics and genomics of Parkinson's disease.

246 : The role of ATP13A2 in Parkinson's disease: Clinical phenotypes and molecular mechanisms.

247 : LRP10 genetic variants in familial Parkinson's disease and dementia with Lewy bodies: a genome-wide linkage and sequencing study.

248 : Multicenter analysis of glucocerebrosidase mutations in Parkinson's disease.

249 : GBA-associated PD presents with nonmotor characteristics.

250 : The link between the GBA gene and parkinsonism.

251 : Reduced glucocerebrosidase is associated with increasedα-synuclein in sporadic Parkinson's disease.

252 : A Large-Scale Full GBA1 Gene Screening in Parkinson's Disease in the Netherlands.

253 : Differential effects of severe vs mild GBA mutations on Parkinson disease.

254 : Increased yield of full GBA sequencing in Ashkenazi Jews with Parkinson's disease.

255 : Large-scale screening of the Gaucher's disease-related glucocerebrosidase gene in Europeans with Parkinson's disease.

256 : Penetrance of Glucocerebrosidase (GBA) Mutations in Parkinson's Disease: A Kin Cohort Study.

257 : Penetrance of Parkinson disease in glucocerebrosidase gene mutation carriers.

258 : Comparison of Parkinson risk in Ashkenazi Jewish patients with Gaucher disease and GBA heterozygotes.

259 : A clinical and family history study of Parkinson's disease in heterozygous glucocerebrosidase mutation carriers.

260 : The incidence of Parkinsonism in patients with type 1 Gaucher disease: data from the ICGG Gaucher Registry.

261 : Genetic movement disorders in patients of jewish ancestry.

262 : Cognitive performance of GBA mutation carriers with early-onset PD: the CORE-PD study.

263 : Clinical and biochemical differences in patients having Parkinson disease with vs without GBA mutations.

264 : GBA Variants are associated with a distinct pattern of cognitive deficits in Parkinson's disease.

265 : Specifically neuropathic Gaucher's mutations accelerate cognitive decline in Parkinson's.

266 : Survival and dementia in GBA-associated Parkinson's disease: The mutation matters.

267 : Association of GBA Genotype With Motor and Functional Decline in Patients With Newly Diagnosed Parkinson Disease.

268 : Gene-wise association of variants in four lysosomal storage disorder genes in neuropathologically confirmed Lewy body disease.

269 : Excessive burden of lysosomal storage disorder gene variants in Parkinson's disease.

270 : Emerging links between pediatric lysosomal storage diseases and adult parkinsonism.

271 : Arylsulfatase A, a genetic modifier of Parkinson's disease, is anα-synuclein chaperone.

272 : SMPD1 mutations, activity, andα-synuclein accumulation in Parkinson's disease.

273 : The contribution of Niemann-Pick SMPD1 mutations to Parkinson disease in Ashkenazi Jews.

274 : Systematic Genetic Analysis of the SMPD1 Gene in Chinese Patients with Parkinson's Disease.

275 : The emerging role of SMPD1 mutations in Parkinson's disease: Implications for future studies.

276 : The emerging role of SMPD1 mutations in Parkinson's disease: Implications for future studies.

277 : Genetic testing in Parkinson disease: promises and pitfalls.

278 : Canadian guideline for Parkinson disease.