You are on page 1of 45

Parkinson's disease

Parkinson's disease (PD), or simply Parkinson's, is a chronic


Parkinson's disease
degenerative disorder of the central nervous system that affects
both the motor system and non-motor systems. The symptoms Other names Parkinson disease,
usually emerge slowly, and as the disease progresses, non- idiopathic or primary
motor symptoms become more common. Early symptoms are parkinsonism,
tremor, rigidity, slowness of movement, and difficulty with hypokinetic rigid
walking. Problems may also arise with cognition, behaviour, syndrome, paralysis
sleep, and sensory systems. Parkinson's disease dementia is agitans, shaking
common in advanced stages.
palsy
The motor symptoms of the disease result from the nerve cell
death in the substantia nigra, a midbrain region that supplies
dopamine to the basal ganglia. The cause of this cell death is
poorly understood but involves the aggregation of the protein
alpha-synuclein into Lewy bodies within the neurons.
Collectively, the main motor symptoms are known as
parkinsonism. Contributing factors include a combination of
genetic and environmental factors. Those with an affected
family member are at an increased risk of getting the disease, Specialty Neurology
with certain genes known to be inheritable risk factors. Symptoms Tremor, rigidity,
Environmental risks include exposure to pesticides and prior
slowness of
head injuries; a history of exposure to trichloroethylene is also
suspected. movement, difficulty
walking[1]
Diagnosis of Parkinson's disease is mainly based on symptoms, Complications Dementia,
usually motor-related. PD typically occurs in people over the depression,
age of 60, of whom about one percent are affected. In those
anxiety,[2] eating
younger than 50, it is termed early-onset PD. The average post-
problems, and sleep
diagnosis life expectancy is 7–15 years. No cure for PD is
known, and treatment aims to mitigate symptoms. Initial problems
treatment typically includes L-DOPA, MAO-B inhibitors, or Usual onset Age over 60[1][3]
dopamine agonists. As the disease progresses, these Causes Unknown[4]
medications become less effective and produce a side effect
marked by involuntary muscle movements. Diet and certain Risk factors Pesticide exposure,
forms of rehabilitation have shown some effectiveness at head injuries[4]
improving symptoms. Surgery to place microelectrodes for Diagnostic Based on
deep brain stimulation has been used to reduce severe motor method symptoms[1]
symptoms where drugs are ineffective. Evidence for treatments
for the nonmovement-related symptoms of PD, such as sleep Differential Dementia with Lewy
disturbances and emotional problems, is less strong. diagnosis bodies, progressive
supranuclear palsy,
The disease is named after English doctor James Parkinson, essential tremor,
who published the first detailed description in An Essay on the antipsychotic use[5]
Shaking Palsy, in 1817. Public awareness campaigns include
Treatment Medications,
World Parkinson's Day and the use of a red tulip symbolizes
surgery[1]
Parkinson's awareness. People with PD who have increased Medication L-DOPA, dopamine
the public's awareness of the condition include boxer agonists[2]
Muhammad Ali and actor Michael J. Fox.
Frequency 6.2 million (2015)[6]
Deaths 117,400 (2015)[7]
Classification
Named after James Parkinson
A progressive neurodegenerative disease,[8][9] Parkinson's
disease (PD) is the most common form of parkinsonism—which encompasses tremor, bradykinesia,
rigidity, and postural instability—and is also called idiopathic parkinsonism, meaning that it has no
identifiable cause.[10][11]

Parkinson's can be classified in other ways. The parkinsonian syndromes include Parkinson's, along with
other conditions.[12] It is included among the Lewy body diseases,[13] and is a synucleinopathy, meaning
that it is characterized by abnormal deposits of alpha-synuclein protein in the brain. The synucleinopathies
include dementia with Lewy bodies, multiple system atrophy, and other rarer conditions.[14][9] Some rare
genetic forms of Parkinson's do not exhibit alpha-synuclein aggregation.[15]

Signs and symptoms


The most recognizable symptoms are movement (motor) related, and include tremor, bradykinesia
(slowness of movement), rigidity, and shuffling/stooped gait.[16] Non-motor symptoms, including
autonomic dysfunction (dysautonomia), neuropsychiatric problems (mood, cognition, behavior or thought
alterations), and sensory (especially altered sense of smell) and sleep difficulties may be present as well.
Patients may have non-motor symptoms that precede the onset of motor symptoms including constipation,
anosmia, and REM Behavior Disorder. Generally, symptoms such as dementia, psychosis, orthostasis, and
more severe falls occur later.[16] Dysphagia can begin at any time during the course of Parkinson's, and
affects more than 80% of patients.[17][18]

Motor

Four motor symptoms are


considered as cardinal signs in
PD: tremor, slowness of
movement (bradykinesia),
rigidity, and postural
instability.[16]

The most common presenting


sign is a coarse, slow tremor of
the hand at rest, which
disappears during voluntary
movement of the affected arm
and in the deeper stages of
sleep.[16] It typically appears in
only one hand, eventually
affecting both hands as the
disease progresses.[16] [19]
Features of Parkinsonian gait (more obvious in the side view).
Frequency of PD tremor is
between 4–6 hertz (cycles per
second). A common characteristic of tremor is pill-rolling, the tendency of the index finger and thumb to
touch and perform together with a circular movement.[16][20] The term derives from the similarity between
the movement of people with PD and the early pharmaceutical technique of manually making pills.[20]

Bradykinesia is due to disturbances in motor planning of movement initiation, and associated with
difficulties along the whole course of the movement process, from planning to initiation to execution of a
movement. Performance of sequential and simultaneous movement is impaired. Bradykinesia is the most
handicapping symptom of Parkinson's disease, presenting as difficulties with everyday tasks such as
dressing, feeding, and bathing. It leads to particular difficulty in carrying out two independent motor
activities at the same time, and can be made worse by emotional stress or concurrent illnesses.
Paradoxically, people with PD can ride a bicycle or climb stairs more easily than walk on the level.
Although most physicians may readily notice bradykinesia, formal assessment requires persons to do
repetitive movements with their fingers and feet.[21]

In parkinsonism, rigidity or hypokinesia can be uniform, known as lead-pipe rigidity, or ratcheted, known
as cogwheel rigidity.[10][16][22][23] The combination of tremor and increased tone is considered to be at the
origin of cogwheel rigidity.[24] Rigidity may be associated with joint pain; such pain being a frequent initial
manifestation of the disease.[16] In early stages of PD, rigidity is asymmetrical and tends to affect the neck
and shoulder muscles before the muscles of the face and extremities.[25] With the progression of the
disease, rigidity typically affects the whole body and reduces the ability to move.

Postural instability is typical in the later stages of the disease, leading to impaired balance and frequent
falls,[26] and secondarily to bone fractures, loss of confidence, and reduced mobility.[27] Instability is absent
in the initial stages, especially in younger people, especially before the development of bilateral
symptoms.[28] Up to 40% of people diagnosed with PD may experience falls, and around 10% may have
falls weekly, with the number of falls being related to the severity of PD.[16]

Other recognized motor signs and symptoms include gait and posture disturbances such as festination (rapid
shuffling steps and a forward-flexed posture when walking with no flexed arm swing). Other common
signs include freezing of gait (brief arrests when the feet seem to get stuck to the floor, especially on turning
or changing direction), a slurred, monotonous, quiet voice, mask-like facial expression, and handwriting
that gets smaller and smaller.[29] Although individuals with PD typically have motor symptoms
predominantly on one side of the body, mask-like facial expression can occur on both sides of the face.[30]

Cognitive

PD causes neuropsychiatric disturbances ranging from mild to severe including disorders of cognition,
mood, behavior, and thought.[16] Cognitive disturbances can occur in the early stages or before diagnosis,
and increase in prevalence with duration of the disease.[16][31] The most common cognitive deficit is
executive dysfunction, which can include problems with planning, cognitive flexibility, abstract thinking,
rule acquisition, inhibiting inappropriate actions, initiating appropriate actions, working memory, and
control of attention.[31][32] Other cognitive difficulties include slowed cognitive processing speed, impaired
recall, and impaired perception and estimation of time.[31][32] Nevertheless, improvement appears when
recall is aided by cues.[31] Visuospatial difficulties are a part of the disease, seen for example when the
individual is asked to perform tests of facial recognition and perception of the orientation of drawn
lines.[31][32]

A person with PD has two to six times the risk of dementia compared with the general population.[16][31]
Up to 78% of people with PD have Parkinson's disease dementia.[33] The prevalence of dementia increases
with age, and to a lesser degree, duration of the disease.[34] Dementia is associated with a reduced quality
of life in people with PD and their caregivers, increased mortality, and a higher probability of needing
nursing home care.[31]

Psychosis

Psychosis can be considered a symptom with a prevalence from 26 to 83%.[35][36] Hallucinations or


delusions occur in about 50% of people with PD over the course of the illness, and may herald the
emergence of dementia. These range from minor hallucinations – sense of passage (something quickly
passing beside the person) or sense of presence (the perception of something or someone standing to the
side or behind the person) – to full blown vivid, formed visual hallucinations and paranoid ideation.
Auditory hallucinations are uncommon in PD, and are rarely described as voices. Psychosis is believed to
be an integral part of the disease. A psychosis with delusions and associated delirium is a recognized
complication of anti-Parkinson drug treatment. Urinary tract infections (frequent in the elderly) and
underlying brain pathology or changes in neurotransmitters or their receptors (e.g., acetylcholine, serotonin)
are thought to play a role in psychosis in PD.[37][38]

Neuropsychiatric

Behavior and mood alterations are more common in PD without cognitive impairment than in the general
population and are usually present in PD with dementia. The most frequent mood difficulties are
depression, apathy, and anxiety.[16] Depression impacts an estimated 20% to 35% of patients, and may
appear at any stage of the disease. It can manifest with symptoms common to the disease process (fatigue,
insomnia, and difficulty with concentration), which makes diagnosis difficult. The imbalance and changes
in dopamine, serotonin, and noradrenergic hormones and functional impairment are causes of depression in
PD-affected people.[35][39] Suicidal ideation is higher than in the general population, but suicidal attempts
themselves are lower.[35][39] Risk factors for depression include disease onset under age 50, being female,
previous history of depression, or severe motor symptoms.[35]

Anxiety has been estimated to have a prevalence in PD-affected people usually around 30–40% and up to
60% has been found.[35][39] Anxiety with PD is complex and consists of symptoms specific to PD.[40]
Anxiety can be higher during motor "off" periods (times when medication is ineffective) and is likely to be
diagnosed after diagnosis due to dysfunction of neurotransmitter pathways.[40] PD-affected people
experience panic attacks more frequently compared with the general population. Both anxiety and
depression have been found to be associated with decreased quality of life.[35][41] Symptoms can range
from mild and episodic to chronic with potential causes being abnormal gamma-aminobutyric acid levels
and embarrassment or fear about symptoms or disease.[35][41] Risk factors for anxiety in PD are disease
onset under age 50, women, and off periods.[35] No standard treatment for PD-associated anxiety exists.[40]

Apathy and anhedonia can be defined as a loss of motivation and an impaired ability to experience
pleasure[42] and are symptoms classically associated with depression, but differ in PD-affected people in
treatment and mechanism. Apathy presents in around 16.5–40%. Symptoms of apathy include reduced
initiative/interests in new activities or the world around them, emotional indifference, and loss of affection
or concern for others.[35] Apathy is associated with deficits in cognitive functions including executive and
verbal memory.[39] Anhedonia occurs in 5–75% of people with PD, depending on the study population
assessed and overlap with apathy.[43]

Impulse-control disorders, including pathological gambling, compulsive sexual behavior, binge eating,
compulsive shopping, and reckless generosity, can be medication-related, particularly orally active
dopamine agonists. The dopamine dysregulation syndrome – with wanting of medication contributing to
overuse – is a rare complication of levodopa use.[44]

Punding, complicated, repetitive, aimless, stereotyped behaviors, is another side effect of anti-Parkinson
medication.

Gastrointestinal

Gastrointestinal issues in Parkinson's disease include constipation, impaired stomach emptying (gastric
dysmotility), and excessive production of saliva can be severe enough to cause discomfort or endanger
health.[45][46] Other upper gastrointestinal symptoms include swallowing impairment (Oropharyngeal
dysphagia) and small intestinal bacterial overgrowth.[47]

Individuals with Parkinson's have alpha-synuclein deposits in the digestive tract as well as the brain.[47]
Constipation is one of the symptoms associated with an increased risk of PD and may precede diagnosis of
PD.[47]

Other

Sleep disorders occur with PD and can be worsened by medications.[16] Symptoms can manifest as
daytime drowsiness (including sudden sleep attacks resembling narcolepsy), disturbances in Rapid eye
movement sleep, or insomnia.[16] REM behavior disorder may begin years before the development of
motor or cognitive elements of PD or dementia with Lewy bodies.[48]

Alterations in the autonomic nervous system can lead to orthostatic hypotension (low blood pressure on
standing), oily skin, excessive sweating, urinary incontinence, and altered sexual function.[16]

Changes in perception may include an impaired sense of smell, disturbed vision, pain, and paresthesia
(tingling and numbness).[16] These symptoms can occur years before diagnosis of the disease.[16]

Causes
None of the proposed risk factors have been conclusively proven.[49] The most frequently replicated
relationships are an increased risk in those exposed to pesticides and a reduced risk in smokers.[49][50]
Research indicates that PD results from a complex interaction between genetic and environmental
factors.[4] Only a small minority of cases can be attributed solely to genetic causes. Moreover, as idiopathic
PD can last and progress over decades, the timing of exposure factor may influence the progression or
severity of certain stages.[51] Statistically significant geographical clusters of Parkinson's cases have been
reported, suggesting a common environmental cause that is either chemical or infectious.[52]

Genetic

Research indicates that PD results from a complex interaction between genetic and environmental
factors.[4] Around 15% of diagnosed individuals have a first-degree relative who has the disease,[10] and 5–
10% have a mutation in genes.[53][54] Harboring one of these gene mutations may not lead to the disease;
susceptibility factors put them at an increased risk, in combination with other factors, which affect age of
onset, severity and progression.[53] At least 11 autosomal dominant and nine autosomal recessive gene
mutations have been implicated in the development of PD. The autosomal dominant genes include SNCA,
PARK3, UCHL1, LRRK2, GIGYF2, HTRA2, EIF4G1, TMEM230, CHCHD2, RIC3, and VPS35.
Autosomal recessive genes include PRKN, PINK1, DJ-1,
ATP13A2, PLA2G6, FBXO7, DNAJC6, SYNJ1, and VPS13C.
Some genes are X-linked or have unknown inheritance pattern;
those include USP24, PARK12, and PARK16. A 22q11 deletion is
known to be associated with PD.[55][54] An autosomal dominant
form has been associated with mutations in the LRP10 gene.[56][57]

About 5% of people with PD have mutations in the GBA1


gene.[58] These mutations are present in fewer than 1% of the
unaffected population. The risk of developing PD is increased 20– Parkin crystal structure
30-fold if these mutations are present. PD associated with these
mutations has the same clinical presentation, but an earlier age of
onset and a more rapid cognitive and motor decline. This gene encodes glucocerebrosidase. Low levels of
this enzyme cause Gaucher's disease.

Alpha-synuclein, a protein encoded by SNCA gene mutations, is the main component of the Lewy bodies
that accumulate in the brains of people with PD.[53] Alpha-synuclein activates ataxia telangiectasia mutated,
a major DNA damage-repair signaling kinase.[59] In addition, alpha-synuclein activates the non-
homologous end joining DNA repair pathway. The aggregation of alpha-synuclein in Lewy bodies appears
to be a link between reduced DNA repair and brain-cell death in PD.[59]

Mutations in some genes, including SNCA, LRRK2, and GBA, have been found to be risk factors for
sporadic (nonfamilial) PD.[53] Mutations in the gene LRRK2 are the most common known cause of familial
and sporadic PD, accounting for around 5% of individuals with a family history of the disease and 3% of
sporadic PD.[60][53] A mutation in GBA presents the greatest genetic risk of developing Parkinsons
disease.[61]

Parkinson-related genes are involved in the function of lysosomes, organelles that digest cellular waste
products. Lysosomal disorders that reduce the ability of cells to break down alpha-synuclein may cause
PD.[62]

Non-genetic

Chemical and medicational

Multi-decade studies have identified an increased likelihood of Parkinson's in association with agricultural
work,[63][64] pesticide (e.g., paraquat[65][66]) exposure, and rural habitation. Moreover, chlorinated solvents
—used in various commercial and industrial application like dry cleaning and degreasing—are also
associated with increased PD risk, particularly trichloroethylene.[63][64]

Carbon disulfide is another risk factor, and has been identified in industrial worker case studies and has
induced parkinsonism in mice.[67] Manganese, which has induced Parkinson's-like pathology in mice, is
also associated with a higher risk. Welders are at a higher PD risk, tentatively from manganese fumes.
Moreover, exposure to suspended particles from traffic fumes is also associated with PD.[64]
Non-genetic Parkinson's risk factors include industrial and agricultural chemicals like carbon disulfide (left) and
traumatic brain injuries (right).

Some medical drugs are also implicated in parkinsonism. Drug-induced parkinsonism is normally reversible
by stopping the offending agent,[15] such as phenothiazines, butyrophenones, metoclopramide, and
Tetrabenazine. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a drug known for causing
irreversible parkinsonism that is commonly used in animal-model research.[15][68][69] Low concentrations
of urate in the blood are associated with an increased risk.[70]

Biological

Traumatic brain injuries (TBIs) are strongly implicated as risk factors for PD,[71] although the association
may be distorted due to recall bias and sampling bias in retrospective studies.[72] Possibly due to the
recurrent head impacts, playing American football for a longer period and at higher levels is associated with
higher PD risk.[73] Veterans are also at a higher risk. Proposed mechanisms by which TBIs could induce or
accelerate Parkinson's include alpha-synuclein accumulation, inflammation, and metabolic
dysregulation.[71]

Vascular parkinsonism is the phenomenon of the presence of Parkinson's disease symptoms combined with
findings of vascular events (such as a cerebral stroke). The damaging of the dopaminergic pathways is
similar in cause for both vascular parkinsonism and idiopathic PD, and so present with similar symptoms.
Differentiation can be made with careful bedside examination, history evaluation, and imaging.[74][15][75]

A possible link exists between PD and Helicobacter pylori infection that can prevent the absorption of
some drugs, including levodopa.[76][77]

Psychological
Behavioral and psychological factors have also been associated with risk of PD. For example, a meta-
analysis found that neuroticism was associated with higher risk of incident PD.[78] Similarly, in a sample of
about a half million individuals found that adults who reported being lonely had a greater risk of developing
PD over a 15 years of follow-up.[79]

Pathophysiology

A Lewy body (stained brown) in a brain cell of the substantia nigra in Parkinson's disease: the brown colour is
positive immunohistochemistry staining for alpha-synuclein (structure at right).

The main pathological characteristics of PD are cell death in the brain's basal ganglia (affecting up to 70%
of the dopamine-secreting neurons in the substantia nigra pars compacta by the end of life).[60] In
Parkinson's disease, alpha-synuclein becomes misfolded and clump together with other alpha-synuclein.
Cells are unable to remove these clumps, and the alpha-synuclein becomes cytotoxic, damaging the
cells.[80][81] These clumps can be seen in neurons under a microscope and are called Lewy bodies. Loss of
neurons is accompanied by the death of astrocytes (star-shaped glial cells) and an increase in the number of
microglia (another type of glial cell) in the substantia nigra.[82] Severity of progression of the parts of the
brain affected by PD can be measured with Braak staging. According to this staging, PD starts in the
medulla and the olfactory bulb before moving to the substantia nigra pars compacta and the rest of the
midbrain/basal forebrain. Movement symptom onset is associated when the disease begins to affect the
substantia nigra pars compacta.[83]

Five major pathways in the brain connect other brain areas to the basal ganglia. These are known as the
motor, oculomotor, associative, limbic, and orbitofrontal circuits. Names indicate the main projection area of
each circuit.[85] All are affected in PD, and their disruption causes movement-, attention- and learning-
related symptoms of the disease.[85] Scientifically, the motor circuit has been examined the most
intensively.[85]

Since 1980, a particular conceptual model of the motor circuit and its alteration with PD has been of
influence although some limitations have been pointed out which have led to modifications.[85] In this
model, the basal ganglia normally exert a constant inhibitory influence on a wide range of motor systems,
preventing them from becoming active at inappropriate times. When a decision is made to perform a
particular action, inhibition is reduced for the required motor system, thereby releasing it for activation.
Dopamine acts to facilitate this release of inhibition, so high levels of dopamine function tend to promote
motor activity, while low levels of dopamine function, such as occur in PD, demand greater exertions of
effort for any given movement. The result of dopamine depletion is to produce hypokinesia, an overall
reduction in motor output.[85] Drugs that are used to treat PD,
conversely, may produce excessive dopamine activity, allowing
motor systems to be activated at inappropriate times and thereby
producing dyskinesias.[85]

Brain cell death

One mechanism causing brain cell death results from abnormal


accumulation of the protein alpha-synuclein bound to ubiquitin in
damaged cells. This insoluble protein accumulates inside neurons
forming inclusions, known as Lewy bodies.[60][86] These bodies
first appear in the olfactory bulb, medulla oblongata and pontine A. Schematic initial
tegmentum; individuals at this stage may be asymptomatic or have progression of Lewy body
early nonmotor symptoms (such as loss of sense of smell or some deposits in the first stages
sleep or automatic dysfunction). As the disease progresses, Lewy of PD, as proposed by
bodies develop in the substantia nigra, areas of the midbrain and Braak and colleagues
basal forebrain, and finally, the neocortex.[60] These brain sites are B. Localization of the area of
the main places of neuronal degeneration in PD, but Lewy bodies significant brain volume
may be protective from cell death (with the abnormal protein reduction in initial PD
sequestered or walled off). Other forms of alpha-synuclein (e.g. compared with a group of
oligomers) that are not aggregated into Lewy bodies and Lewy participants without PD[84]
neurites, may in fact be the toxic forms of the protein.[87][86] In
people with dementia, a generalized presence of Lewy bodies is
common in cortical areas. Neurofibrillary tangles and senile
plaques, characteristic of Alzheimer's disease, are uncommon
unless the person has dementia.[82]

Other mechanisms include proteasomal and lysosomal systems


dysfunction and reduced mitochondrial activity.[87] Iron
accumulation in the substantia nigra is typically observed in
conjunction with the protein inclusions. It may be related to
oxidative stress, protein aggregation, and neuronal death, but the
An illustration of the dopamine
mechanisms are obscure.[88] pathways throughout the brain

Neuroimmune interaction

The neuroimmune interaction is heavily implicated in PD pathology. PD and autoimmune disorders share
genetic variations and molecular pathways. Some autoimmune diseases may even increase one's risk of
developing PD, up to 33% in one study.[89] Autoimmune diseases linked to protein expression profiles of
monocytes and CD4+ T cells are linked to PD. Herpes virus infections can trigger autoimmune reactions to
alpha-synuclein, perhaps through molecular mimicry of viral proteins.[90] Alpha-synuclein, and its
aggregate form, Lewy bodies, can bind to microglia. Microglia can proliferate and be over-activated by
alpha-synuclein binding to MHC receptors on inflammasomes, bringing about a release of proinflammatory
cytokines like IL-1β, IFNγ, and TNFα.[91]

Activated microglia influence the activation of astrocytes, converting their neuroprotective phenotype to a
neurotoxic one. Astrocytes in healthy brains serve to protect neuronal connections. In PD patients,
astrocytes cannot protect the dopaminergic connections in the striatum. Microglia present antigens via
MHC-I and MHC-II to T cells. CD4+ T cells, activated by this process, are able to cross the blood brain
barrier (BBB) and release more proinflammatory cytokines, like interferon-γ (IFNγ), TNFα, and IL-1β.
Mast cell degranulation and subsequent proinflammatory cytokine release is implicated in BBB breakdown
in PD. Another immune cell implicated in PD are peripheral monocytes and have been found in the
substantia nigra of PD patients. These monocytes can lead to more dopaminergic connection breakdown. In
addition, monocytes isolated from PD patients express higher levels of the PD-associated protein, LRRK2,
compared with non-PD individuals via vasodilation.[92] In addition, high levels of pro-inflammatory
cytokines, such as IL-6, can lead to the production of C-reactive protein by the liver, another protein
commonly found in PD patients, that can lead to an increase in peripheral inflammation.[93][94]

Peripheral inflammation can affect the gut-brain axis, an area of the body highly implicated in PD. PD
patients have altered gut microbiota and colon problems years before motor issues arise.[93][94] Alpha-
synuclein is produced in the gut and may migrate via the vagus nerve to the brainstem, and then to the
substantia nigra.[95] Furthermore, the bacteria Proteus mirabilis has been associated with higher levels of
alpha-synuclein and an increase of motor symptoms in PD patients.[96]

Diagnosis
Physician's initial assessment is typically based on medical history and neurological examination.[16] They
assess motor symptoms (bradykinesia, rest tremors, etc.) using clinical diagnostic criteria. The finding of
Lewy bodies in the midbrain on autopsy is usually considered final proof that the person had PD. The
clinical course of the illness over time may diverge from PD, requiring that presentation is periodically
reviewed to confirm the accuracy of the diagnosis.[16][97]

Multiple causes can occur for parkinsonism or diseases that look similar. Stroke, certain medications, and
toxins can cause "secondary parkinsonism" and need to be assessed during visit.[83][97] Parkinson-plus
syndromes, such as progressive supranuclear palsy and multiple system atrophy, must be considered and
ruled out appropriately to begin a different treatment and disease progression (anti-Parkinson's medications
are typically less effective at controlling symptoms in Parkinson-plus syndromes).[16] Faster progression
rates, early cognitive dysfunction or postural instability, minimal tremor, or symmetry at onset may indicate
a Parkinson-plus disease rather than PD itself.[98]

Medical organizations have created diagnostic criteria to ease and standardize the diagnostic process,
especially in the early stages of the disease. The most widely known criteria come from the UK Queen
Square Brain Bank for Neurological Disorders and the U.S. National Institute of Neurological Disorders
and Stroke. The Queen Square Brain Bank criteria require slowness of movement (bradykinesia) plus
either rigidity, resting tremor, or postural instability. Other possible causes of these symptoms need to be
ruled out. Finally, three or more of the following supportive symptoms are required during onset or
evolution: unilateral onset, tremor at rest, progression in time, asymmetry of motor symptoms, response to
levodopa for at least five years, the clinical course of at least ten years and appearance of dyskinesias
induced by the intake of excessive levodopa.[99] Assessment of sudomotor function through
electrochemical skin conductance can be helpful in diagnosing dysautonomia.[100]

When PD diagnoses are checked by autopsy, movement disorders experts are found on average to be
79.6% accurate at initial assessment and 83.9% accurate after refining diagnoses at follow-up examinations.
When clinical diagnoses performed mainly by nonexperts are checked by autopsy, the average accuracy is
73.8%. Overall, 80.6% of PD diagnoses are accurate, and 82.7% of diagnoses using the Brain Bank criteria
are accurate.[101]

Imaging
Computed tomography (CT) scans of people with PD usually appear normal.[102] Magnetic resonance
imaging has become more accurate in diagnosis of the disease over time, specifically through iron-sensitive
T2* and susceptibility weighted imaging sequences at a magnetic field strength of at least 3T, both of
which can demonstrate absence of the characteristic 'swallow tail' imaging pattern in the dorsolateral
substantia nigra.[103] In a meta-analysis, absence of this pattern was highly sensitive and specific for the
disease.[104] A meta-analysis found that neuromelanin-MRI can discriminate individuals with Parkinson's
from healthy subjects.[105] Diffusion MRI has shown potential in distinguishing between PD and
Parkinson-plus syndromes, as well as between PD motor subtypes,[106] though its diagnostic value is still
under investigation.[102] CT and MRI are used to rule out other diseases that can be secondary causes of
parkinsonism, most commonly encephalitis and chronic ischemic insults, as well as less-frequent entities
such as basal ganglia tumors and hydrocephalus.[102]

The metabolic activity of dopamine transporters in the basal ganglia can be directly measured with positron
emission tomography and single-photon emission computed tomography scans. It has shown high
agreement with clinical diagnoses of PD.[107] Reduced dopamine-related activity in the basal ganglia can
help exclude drug-induced Parkinsonism. This finding is nonspecific and can be seen with both PD and
Parkinson-plus disorders.[102] In the United States, DaTSCANs are only FDA approved to distinguish PD
or Parkinsonian syndromes from essential tremor.[108]

Iodine-123-meta-iodobenzylguanidine myocardial scintigraphy can help locate denervation of the muscles


of the heart which can support a PD diagnosis.[83]

Differential diagnosis

Secondary parkinsonism – The multiple causes of parkinsonism can be differentiated through careful
history, physical examination, and appropriate imaging.[15][83][109] This topic is further discussed in the
causes section here.

Other Parkinson-plus syndromes can have similar movement symptoms but have a variety of associated
symptoms. Some of these are also synucleinopathies. Lewy body dementia involves motor symptoms with
early onset of cognitive dysfunction and hallucinations which precede motor symptoms. Alternatively,
multiple systems atrophy or MSA usually has early onset of autonomic dysfunction (such as orthostasis),
and may have autonomic predominance, cerebellar symptom predominance, or Parkinsonian
predominance.[110]

Other Parkinson-plus syndromes involve tau, rather than alpha-synuclein. These include progressive
supranuclear palsy (PSP) and corticobasal syndrome (CBS). PSP predominantly involves rigidity, early
falls, bulbar symptoms, and vertical gaze restriction; it can be associated with frontotemporal dementia
symptoms. CBS involves asymmetric parkinsonism, dystonia, alien limb, and myoclonic jerking.[111]
Presentation timelines and associated symptoms can help differentiate similar movement disorders from
idiopathic Parkinson disease.

Parkinson-plus syndrome – Multiple diseases can be considered part of the Parkinson's plus group,
including corticobasal syndrome, multiple system atrophy, progressive supranuclear palsy, and dementia
with Lewy bodies. Differential diagnosis can be narrowed down with careful history and physical exam
(especially focused on the sequential onset of specific symptoms), progression of the disease, and response
to treatment.[112][109] Some key symptoms:[15][109]

Corticobasal syndrome – levodopa-resistance, myoclonus, dystonia, corticosensory loss,


apraxia, and non-fluent aphasia
Dementia with Lewy bodies – levodopa resistance,
cognitive predominance before motor symptoms, and
fluctuating cognitive symptoms, (visual hallucinations
are common in this disease)
Essential tremor – This can at first look like
parkinsonism, but has key differentiators. In essential
tremor, the tremor gets worse with action (improves in
PD), a lack of other symptoms is common in PD, and
normal DatSCAN is seen.[109][15]
Multiple system atrophy – levodopa resistance, rapidly
progressive, autonomic failure, stridor, present Babinski
sign, cerebellar ataxia, and specific MRI findings
Progressive supranuclear palsy – levodopa resistance,
restrictive vertical gaze, specific MRI findings, and early Hot Cross Bun sign that is
and different postural difficulties commonly found in MRI of multiple
system atrophy
Other conditions that can have similar presentations to PD
include:[113][15]

Arthritis
Creutzfeldt–Jakob disease
Depression
Dystonia
Fragile X-associated tremor/ataxia syndrome
Frontotemporal dementia and parkinsonism linked to chromosome 17
Huntington's disease
Idiopathic basal ganglia calcification
Neurodegeneration with brain iron accumulation
Normal-pressure hydrocephalus
Obsessional slowness
Psychogenic parkinsonism
Wilson's disease

Prevention
Exercise in middle age may reduce the risk of PD later in life.[114] Caffeine appears protective with a
greater decrease in risk occurring with a larger intake of caffeinated beverages such as coffee.[115]

Antioxidants, such as vitamins C and E, have been proposed to protect against the disease, but results of
studies have been contradictory and no positive effect has been shown.[49] The results regarding fat and
fatty acids have been contradictory.[49] Use of nonsteroidal anti-inflammatory drugs (NSAIDs) and calcium
channel blockers may be protective.[4] A 2010 meta-analysis found that NSAIDs (apart from aspirin), have
been associated with at least a 15% (higher in long-term and regular users) reduction in the incidence of the
development of PD.[116] As of 2019 meta-analyses have failed to confirm this link. Multiple studies have
demonstrated a link between the use of ibuprofen and a decreased risk of Parkinson's development.[117]

Management
No cure for Parkinson's disease is known. Medications, surgery, and physical treatment may provide relief,
improve the quality of a person's life, and are much more effective than treatments for other neurological
disorders such as Alzheimer's disease, motor neuron disease, and Parkinson-plus syndromes.[118] The main
families of drugs useful for treating motor symptoms are levodopa always combined with a dopa
decarboxylase inhibitor and with a COMT inhibitor, dopamine agonists, and MAO-B inhibitors. The stage
of the disease and the age at disease onset determine which group is most useful.[118]

Braak staging of PD uses six stages that can identify early, middle, and late stages.[119] The initial stage in
which some disability has already developed and requires pharmacological treatment is followed by later
stages associated with the development of complications related to levodopa usage, and a third stage when
symptoms unrelated to dopamine deficiency or levodopa treatment may predominate.[119]

Treatment in the first stage aims for an optimal trade-off between symptom control and treatment side
effects. The start of levodopa treatment may be postponed by initially using other medications, such as
MAO-B inhibitors and dopamine agonists, instead, in the hope of delaying the onset of complications due
to levodopa use.[120] Levodopa is still the most effective treatment for the motor symptoms of PD and
treatment should be prompt in people when their quality of life is impaired. Levodopa-related dyskinesias
correlate more strongly with duration and severity of the disease than duration of levodopa treatment.[121]

In later stages, the aim is to reduce PD symptoms, while controlling fluctuations in the effect of the
medication. Sudden withdrawals from medication or its overuse must be managed.[120] When oral
medications are inadequate in controlling symptoms, surgery (deep brain stimulation or high-intensity
focused ultrasound[122]), subcutaneous waking-day apomorphine infusion, and enteral dopa pumps may be
useful.[123] Late-stage PD presents challenges requiring a variety of treatments, including those for
psychiatric symptoms particularly depression, orthostatic hypotension, bladder dysfunction, and erectile
dysfunction.[123] In the final stages of the disease, palliative care is provided to improve a person's quality
of life.[124]

A 2020 Cochrane review found no certain evidence that cognitive training is beneficial for people with
Parkinson's disease, dementia or mild cognitive impairment.[125] The findings are based on low certainty
evidence of seven studies.

Medications

Levodopa

Levodopa is usually the first drug of choice when treating


Parkinson's disease and has been the most widely used PD
treatment since the 1980s.[120][126] The motor symptoms of PD are
the result of reduced dopamine production in the brain's basal
ganglia. Dopamine fails to cross the blood–brain barrier so it cannot
be taken as a medicine to boost the brain's depleted levels of
LCE pills, containing Levodopa,
dopamine. A precursor of dopamine, levodopa, can pass through to
carbidopa, and entacapone
the brain where it is readily converted to dopamine. Administration
of levodopa temporarily diminishes the motor symptoms of PD.

Only 5–10% of levodopa crosses the blood–brain barrier. Much of the remainder is metabolized to
dopamine elsewhere in the body, causing a variety of side effects, including nausea, vomiting, and
orthostatic hypotension.[127] Carbidopa and benserazide are dopa decarboxylase inhibitors that fail to cross
the blood–brain barrier and inhibit the conversion of levodopa to dopamine outside the brain, reducing side
effects and improving the availability of levodopa for passage into the brain. One of these drugs is usually
taken along with levodopa and is available combined with levodopa in the same pill.[128]

Prolonged use of levodopa is associated with the development of complications, such as involuntary
movements (dyskinesias) and fluctuations in the impact of the medication.[120] When fluctuations occur, a
person can cycle through phases with good response to medication and reduced PD symptoms (on state),
and phases with poor response to medication and increased PD symptoms (off state).[120][129] Using lower
doses of levodopa may reduce the risk and severity of these levodopa-induced complications.[130] A former
strategy, called "drug holidays", to reduce levodopa-related dyskinesia and fluctuations was to withdraw
levodopa medication for some time[126] which can bring on dangerous side effects such as neuroleptic
malignant syndrome and is discouraged.[120] Most people with PD eventually need levodopa and later
develop levodopa-induced fluctuations and dyskinesias.[120] Adverse effects of levodopa, including
dyskinesias, mistakenly influence patients and providers to delay treatment which reduces potential for
optimal results.

Levodopa by itself is available in oral (tablets and capsules), oral inhalation, and infusion form. Inhaled
levodopa can be used when oral levodopa therapy has reached a point where "off" periods have increased
in length.[131][132][133]

COMT inhibitors

During the course of PD, affected people can experience a


wearing-off phenomenon, where a recurrence of symptoms occurs
after a dose of levodopa, but right before their next dose.[83]
Catechol-O-methyltransferase (COMT) is a protein that degrades
levodopa before it can cross the blood–brain barrier and COMT
inhibitors allow for more levodopa to cross.[135] They are normally
used in the management of later symptoms, but can be used in
conjunction with levodopa/carbidopa when a person is
COMT metabolizes levodopa to 3-O-
experiencing the wearing off-phenomenon with their motor
methyldopa. COMT inhibitors help
symptoms.[83][126] stop this reaction, allowing for more
levodopa to cross the blood–brain
Three COMT inhibitors are used to treat adults with PD and end-
barrier and become dopamine where
of-dose motor fluctuations – opicapone, entacapone, and
it is needed.[134]
tolcapone.[83] Tolcapone has been available for but its usefulness is
limited by possible liver damage complications requiring liver-
function monitoring.[136][15][83][135] Entacapone and opicapone cause little alteration to liver
function.[135][137][138] Licensed preparations of entacapone contain entacapone alone or in combination
with carbidopa and levodopa.[139][15][140] Opicapone is a once-daily COMT inhibitor.[141][83]

Dopamine agonists

Dopamine agonists that bind to dopamine receptors in the brain have similar effects to levodopa.[120] These
were initially used as a complementary therapy to levodopa for individuals experiencing levodopa
complications (on-off fluctuations and dyskinesias); they are mainly used on their own as first therapy for
the motor symptoms of PD with the aim of delaying the initiation of levodopa therapy, thus delaying the
onset of levodopa's complications.[120][142] Dopamine agonists include bromocriptine, pergolide,
pramipexole, ropinirole, piribedil, cabergoline, apomorphine, and lisuride.
Though dopamine agonists are less effective than levodopa at controlling PD motor symptoms, they are
effective enough to manage these symptoms in the first years of treatment.[10] Dyskinesias due to dopamine
agonists are rare in younger people who have PD, but along with other complications, become more
common with older age at onset.[10] Thus, dopamine agonists are the preferred initial treatment for
younger-onset PD, and levodopa is preferred for older-onset PD.[10]

Dopamine agonists produce side effects, including drowsiness, hallucinations, insomnia, nausea, and
constipation.[120][126] Side effects appear with minimal clinically effective doses giving the physician
reason to search for a different drug.[120] Agonists have been related to impulse-control disorders (such as
increased sexual activity, eating, gambling, and shopping) more strongly than other antiparkinson
medications.[143][126]

Apomorphine, a dopamine agonist, may be used to reduce off periods and dyskinesia in late PD.[120] It is
administered only by intermittent injections or continuous subcutaneous infusions.[120] Secondary effects
such as confusion and hallucinations are common, individuals receiving apomorphine treatment should be
closely monitored.[120] Two dopamine agonists administered through skin patches (lisuride and rotigotine)
are useful for people in the initial stages and possibly to control off states in those in advanced states.[8] Due
to an increased risk of cardiac fibrosis with ergot-derived dopamine agonists (bromocriptine, cabergoline,
dihydroergocryptine, lisuride, and pergolide), they should only be considered for adjunct therapy to
levodopa.[126]

MAO-B inhibitors

MAO-B inhibitors (safinamide, selegiline and rasagiline) increase the amount of dopamine in the basal
ganglia by inhibiting the activity of monoamine oxidase B, an enzyme that breaks down dopamine.[120]
They have been found to help alleviate motor symptoms when used as monotherapy (on their own); when
used in conjunction with levodopa, time spent in the off phase is reduced.[144][126] Selegiline has been
shown to delay the need for beginning levodopa, suggesting that it might be neuroprotective and slow the
progression of the disease.[145] An initial study indicated that selegiline in combination with levodopa
increased the risk of death, but this has been refuted.[146]

Common side effects are nausea, dizziness, insomnia, sleepiness, and (in selegiline and rasagiline)
orthostatic hypotension.[145][83] MAO-Bs are known to increase serotonin and cause a potentially
dangerous condition known as serotonin syndrome.[145]

Other drugs

Other drugs such as amantadine may be useful as treatment of motor symptoms, but evidence for use is
lacking.[120][147] Anticholinergics should not be used for dyskinesia or motor fluctuations but may be
considered topically for drooling.[126] A diverse range of symptoms beyond those related to motor function
can be treated pharmaceutically.[148] Examples are the use of quetiapine or clozapine for psychosis,
cholinesterase inhibitors or memantine for dementia, and modafinil for excessive daytime
sleepiness.[148][149][126] In 2016, pimavanserin was approved for the management of PD psychosis.[150]
Doxepin and rasagline may reduce physical fatigue in PD.[151]

Surgery
Treating motor symptoms with surgery was once a common practice but
the discovery of levodopa has decreased the amount of procedures.[152]
Studies have led to great improvements in surgical techniques, so surgery
can be used in people with advanced PD for whom drug therapy is no
longer sufficient.[152] Surgery for PD can be divided in two main groups –
lesional and deep brain stimulation (DBS). Target areas for DBS or lesions
include the thalamus, globus pallidus, or subthalamic nucleus.[152] DBS
involves the implantation of a medical device called a neurostimulator,
which sends electrical impulses to specific parts of the brain. DBS is
recommended for people who have PD with motor fluctuations and tremor
inadequately controlled by medication, or to those who are intolerant to
medication lacking severe neuropsychiatric problems.[153] Other less
common surgical therapies involve intentional formation of lesions to
suppress overactivity of specific subcortical areas. For example,
pallidotomy involves surgical destruction of the globus pallidus to control Placement of an electrode
into the brain: the head is
dyskinesia.[152]
stabilised in a frame for
Four areas of the brain have been treated with neural stimulators in stereotactic surgery.
PD.[154] These are the globus pallidus interna, thalamus, subthalamic
nucleus, and pedunculopontine nucleus. DBS of the globus pallidus interna
improves motor function, while DBS of the thalamic DBS improves tremor, but has little impact on
bradykinesia or rigidity. DBS of the subthalamic nucleus is usually avoided if a history of depression or
neurocognitive impairment is present. DBS of the subthalamic nucleus is associated with a reduction in
medication. Pedunculopontine nucleus DBS remains experimental at present. Generally, DBS is associated
with 30–60% improvement in motor score evaluations.[155]

Rehabilitation

Exercise programs are recommended in people with PD.[114] Some evidence shows that speech or mobility
problems can improve with rehabilitation, although studies are scarce and of low quality.[156][157] Regular
physical exercise with or without physical therapy can be beneficial to maintain and improve mobility,
flexibility, strength, gait speed, and quality of life.[157] When an exercise program is performed under the
supervision of a physiotherapist, more improvements occur in motor symptoms, mental and emotional
functions, daily living activities, and quality of life compared with a self-supervised exercise program at
home.[158] Clinical exercises may be an effective intervention targeting overall well-being of individuals
with Parkinson's. Improvement in motor function and depression may happen.[159]

In improving flexibility and range of motion for people experiencing rigidity, generalized relaxation
techniques such as gentle rocking have been found to decrease excessive muscle tension. Other effective
techniques to promote relaxation include slow rotational movements of the extremities and trunk, rhythmic
initiation, diaphragmatic breathing, and meditation techniques.[160] As for gait and addressing the
challenges associated with the disease such as hypokinesia, shuffling, and decreased arm swing,
physiotherapists have a variety of strategies to improve functional mobility and safety. Areas of interest
concerning gait during rehabilitation programs focus on improving gait speed, the base of support, stride
length, and trunk and arm-swing movement. Strategies include using assistive equipment (pole walking and
treadmill walking), verbal cueing (manual, visual, and auditory), exercises (marching and PNF patterns),
and altering environments (surfaces, inputs, open vs. closed).[161] Strengthening exercises have shown
improvements in strength and motor function for people with primary muscular weakness and weakness
related to inactivity with mild to moderate PD, but reports show an interaction between strength and the
time the medications were taken. Therefore, people with PD should perform exercises 45 minutes to one
hour after medications when they are capable.[162] Deep diaphragmatic breathing exercises are beneficial in
improving chest-wall mobility and vital capacity decreased by a forward flexed posture and respiratory
dysfunctions in advanced PD.[163] Exercise may improve constipation.[45] Whether exercise reduces
physical fatigue in PD remains unclear.[151]

Strength training exercise has been shown to increase manual dexterity in PD patients after exercising with
manual putty. This positively affects everyday life when gripping for PD patients.[164]

The Lee Silverman voice treatment (LSVT) is one of the most widely practiced treatments for speech
disorders associated with PD.[156][165] Speech therapy and specifically LSVT may improve speech.[156]
Occupational therapy (OT) aims to promote health and quality of life by helping people with the disease to
participate in a large percentage of their daily living activities.[156] Few studies have been conducted on the
effectiveness of OT, and their quality is poor, although with some indication that it may improve motor
skills and quality of life for the duration of the therapy.[156][166]

Palliative care

The goal of Palliative care is to improve quality of life for both the patient and family by providing relief
from the symptoms and stress of illnesses.[167] As Parkinson's is uncurable, treatments focus on slowing
decline and improving quality of life and are therefore palliative.[168]

Palliative care should be involved earlier, rather than later, in the disease course.[169][170] Palliative care
specialists can help with physical symptoms, emotional factors such as loss of function and jobs,
depression, fear, and existential concerns.[169][170][171]

Along with offering emotional support to both the affected person and family, palliative care addresses
goals of care. People with PD may have difficult decisions to make as the disease progresses, such as
wishes for feeding tube, noninvasive ventilator or tracheostomy, wishes for or against cardiopulmonary
resuscitation, and when to use hospice care.[168] Palliative-care team members can help answer questions
and guide people with PD on these complex and emotional topics to help them make decisions based on
values.[170][172]

Muscles and nerves that control the digestive process may be affected by PD, resulting in constipation and
gastroparesis (prolonged emptying of stomach contents).[45] A balanced diet, based on periodical nutritional
assessments, is recommended, and should be designed to avoid weight loss or gain and minimize the
consequences of gastrointestinal dysfunction.[45] As the disease advances, swallowing difficulties
(dysphagia) may appear. Using thickening agents for liquid intake and an upright posture when eating may
be useful; both measures reduce the risk of choking. Gastrostomy can be used to deliver food directly into
the stomach.[45]

Levodopa and proteins use the same transportation system in the intestine and the blood–brain barrier,
thereby competing for access.[45] Taking them together results in reduced effectiveness of the drug.[45]
Therefore, when levodopa is introduced, excessive protein consumption is discouraged in favour of a well-
balanced Mediterranean diet. In advanced stages, additional intake of low-protein products such as bread or
pasta is recommended for similar reasons.[45] To minimize interaction with proteins, levodopa should be
taken 30 minutes before meals.[45] At the same time, regimens for PD restrict proteins during breakfast and
lunch, allowing protein intake in the evening.[45]

Prognosis
PD invariably progresses with time. A severity rating method known as the Unified Parkinson's disease
rating scale (UPDRS) is the most commonly used metric for a clinical study. A modified version known as
the MDS-UPDRS is also used. An older scaling method known as the Hoehn and Yahr scale (originally
published in 1967), and a similar scale known as the Modified Hoehn and Yahr scale, have been used. The
Hoehn and Yahr scale defines five basic stages of progression.

Motor symptoms may advance aggressively in the early stages of the disease and more slowly later.
Untreated, individuals are expected to lose independent ambulation after an average of eight years and be
bedridden after 10 years. Medication has improved the prognosis of motor symptoms. In people taking
levodopa, the progression time of symptoms to a stage of high dependency from caregivers may be over 15
years. Predicting what course the disease will take for a given individual is difficult.[173] Age is an
appropriate predictor of disease progression. The rate of motor decline is greater in those with less
impairment at the time of diagnosis, while cognitive impairment is more frequent in those who are over 70
years of age at symptom onset.[87]

No hard correlation between PD progression and PD disability as a whole has been found as of 2023;
however, initial PD disability often mainly concerns the motor symptoms, whereas in later stages it is often
mainly related to the non-motor symptoms of the disease. Therapies exist that can improve or alleviate these
latter symptoms to some extent.[87] [173] As the disease advances, disability is more related to motor
symptoms that are uncontrollable by medication (such as difficulties with swallowing and dysartria, and
gait and balance problems) and to levodopa-induced complications, which appear in up to 50% of
individuals after five years of levodopa usage. Finally, after ten years most people with the disease have
autonomic disturbances, sleep problems, mood alterations and cognitive decline; these symptoms,
especially cognitive decline, greatly increase disability.[87][173]

The life expectancy of people with PD is reduced. Mortality ratios are around twice those of unaffected
people. Cognitive decline and dementia, old age at onset, a more advanced disease state, and presence of
swallowing problems are all mortality risk factors. A disease pattern mainly characterized by tremor as
opposed to rigidity, though, predicts an improved survival. Death from aspiration pneumonia is twice as
common in individuals with PD as in the healthy population.[173]

In 2016, PD resulted in about 211,000 deaths globally, an increase of 161% since 1990.[174] The overall
death rate increased by 19% to 1.81 per 100,000 people during that time.[174]

Epidemiology
PD is the second most common neurodegenerative disorder after Alzheimer's disease and affects
approximately seven million people globally and one million people in the United States.[26][49][175] The
proportion in a population at a given time is about 0.3% in industrialized countries. PD is more common in
the elderly and rates rise from 1% in those over 60 years of age to 4% of the population over 80.[49][129]
The mean age of onset is around 60 years, although 5–10% begin between the ages of 20 and 50 is
classified as young onset PD.[10] Males are affected at a ratio of around 3:2 compared with females.[4] PD
may be less prevalent in those of African and Asian ancestry, although this finding is disputed.[49] The
number of new diagnoses per year of PD is between 8–18 per 100,000 person–years.[49]
China is predicted to have nearly half of the Parkinson's disease population in the world in 2030.[176] By
2040 the number of patients is expected to grow to approximately 14 million people; this growth has been
referred to as the Parkinson's pandemic.[177]

History
Early sources, including an Egyptian papyrus, an ayurvedic medical
treatise, the Bible, and Galen's writings, describe symptoms
resembling those of PD.[180] After Galen, no references
unambiguously related to PD appear until the 17th century.[180] In
the 17th and 18th centuries, Franciscus Sylvius, Hieronymus David
Gaubius, John Hunter and Auguste François Chomel wrote about
elements of the disease.[180][181][182]

In 1817, James Parkinson published his essay reporting six people


with paralysis agitans.[183] An Essay on the Shaking Palsy described
the characteristic resting tremor, abnormal posture and gait, paralysis
and diminished muscle strength, and the way that the disease
progresses over time.[184][185] Early neurologists who made further
additions to the knowledge of the disease include Trousseau,
Gowers, Kinnier Wilson and Erb, and Jean-Martin Charcot, whose
studies between 1868 and 1881 increased the understanding of the
disease.[183] Among other advances, Charcot made the distinction
between rigidity, weakness and bradykinesia.[183] He championed Jean-Martin Charcot made
the renaming of the disease in honor of James Parkinson.[183] contributions to the understanding
of PD and proposed its name
In 1912, Frederic Lewy described microscopic particles in affected honoring James Parkinson
brains, later named Lewy bodies.[183] In 1919, Konstantin Tretiakoff
reported that the substantia nigra was the main cerebral structure
affected, but this finding was rejected until it was confirmed by
further studies published by Rolf Hassler in 1938.[183] The
underlying biochemical changes in the brain were identified in the
1950s, due largely to the work of Arvid Carlsson on the
neurotransmitter dopamine and Oleh Hornykiewicz on its role on
PD.[186] In 1997, alpha-synuclein was found to be the main
component of Lewy bodies by Spillantini, Trojanowski, Goedert,
and others.[86]

Anticholinergics and surgery (lesioning of the corticospinal pathway


or some of the basal ganglia structures) were the only treatments Illustration of (advanced)
Parkinson's disease by William
until the arrival of levodopa, which reduced their use
Richard Gowers, published in
dramatically.[181][187] Levodopa was first synthesized in 1911 by
1886[178][179]
Casimir Funk, but it received little attention until the mid 20th
century.[186] It entered clinical practice in 1967 and brought about a
revolution in the management of PD.[186][188] By the late 1980s
deep brain stimulation introduced by Alim Louis Benabid and colleagues at Grenoble, France, emerged as
a possible treatment.[189]
An illustration of advanced Parkinson's disease by William Richard Gowers was published in 1886 in A
Manual of Diseases of the Nervous System, based on 1879 photographs attributed to Albert
Londe.[178][179] A new image was created in 2020 to represent diversity in levels of severity.[190][191]

Society and culture

Social impact

For some people with PD, masked facial expressions and difficulty moderating facial expressions of
emotion or recognizing other people's facial expressions can impact social well-being.[30] As the condition
progresses, tremor, other motor symptoms, difficulty communicating, or issues with mobility may interfere
with social engagement, causing individuals with PD to feel isolated.[192] Public perception and awareness
of PD symptoms such as shaking, hallucinating, slurring speech, and being off balance is lacking in some
countries and can lead to stigma.[192]

Cost

The costs of PD to society are high; in 2007, the largest share of direct cost came from inpatient care and
nursing homes, while the share coming from medication was substantially lower.[193] Indirect costs are
high, due to reduced productivity and the burden on caregivers.[193] In addition to economic costs, PD
reduces quality of life of those with the disease and their caregivers.[193]

A study based on 2017 data estimated the US economic PD burden at $51.9 billion, including direct
medical costs of $25.4 billion and $26.5 billion in indirect and non-medical costs. The projected total
economic burden surpasses $79 billion by 2037. These findings highlight the need for interventions to
reduce PD incidence, delay disease progression, and alleviate symptom burden that may reduce the future
economic burden of PD.[194]

Advocacy

The birthday of James Parkinson, 11 April, has been designated as World


Parkinson's Day.[183] A red tulip was chosen by international organizations as the
symbol of the disease in 2005; it represents the 'James Parkinson' tulip cultivar,
registered in 1981 by a Dutch horticulturalist.[195]

Advocacy organizations include the National Parkinson Foundation, which has


provided more than $180 million in care, research, and support services since
1982,[196] Parkinson's Disease Foundation, which has distributed more than Parkinson's
$115 million for research and nearly $50 million for education and advocacy awareness logo with
red tulip symbol
programs since its founding in 1957 by William Black;[197][198] the American
Parkinson Disease Association, founded in 1961;[199] and the European
Parkinson's Disease Association, founded in 1992.[200]

Notable cases

In the 21st century, the diagnosis of Parkinson's among notable figures has increased the public's
understanding of the disorder.[201]
Actor Michael J. Fox was diagnosed with PD at 29
years old,[202] and has used his diagnosis to increase
awareness of the disease.[203] To illustrate the effects of
the disease, Fox has appeared without medication in
television roles and before the United States Congress
without medication.[204] The Michael J. Fox
Foundation, which he founded in 2000, has raised over
$2 billion for Parkinson's research.[205] Boxer
Muhammad Ali showed signs of PD when he was 38,
but was undiagnosed until he was 42, and has been
called the "world's most famous Parkinson's
patient".[206] Whether he had PD or parkinsonism
related to boxing is unresolved.[207][208] Cyclist and Actor Michael J. Fox and boxer Muhammad Ali
Olympic medalist Davis Phinney, diagnosed with (center) are pictured in 2002 speaking before the
US Senate to urge increased funding for
Parkinson's at 40, started the Davis Phinney Foundation
Parkinson's research.
in 2004 to support PD research.[209][210]

At the time of his suicide in 2014, Robin Williams, the


American actor and comedian, had been diagnosed with PD[211] but his autopsy revealed dementia with
Lewy bodies.[211][212][213][214][215]

Historical figures have been theorized to have had Parkinson's,[216][217] including English philosopher
Thomas Hobbes,[218][219][220] Adolf Hitler,[221][222][217] and Mao Zedong.[223]

Research
As of 2022, no disease-modifying drugs (drugs that
target the causes or damage) are approved for
Parkinson's, so this is a major focus of Parkinson's
research.[224][225] Active research directions include the
search for new animal models of the disease and studies
of the potential usefulness of gene therapy, stem cell
transplants, and neuroprotective agents.[226] To aid in
earlier diagnosis, research criteria for identifying
prodromal biomarkers of the disease have been
established.[227]
Astronaut Alexander Gerst conducting
The role of the gut–brain axis and the gut flora in PD Parkinson's research aboard the International
are recognized but the mechanism that causes Space Station in 2018
gastrointestinal symptoms is unclear.[228]

Gene therapy

Gene therapy typically involves the use of a noninfectious virus[229] to shuttle genetic material into a part of
the brain. Approaches have involved the expression of growth factors to prevent damage (Neurturin – a
GDNF-family growth factor), and enzymes such as glutamic acid decarboxylase (GAD – the enzyme that
produces GABA), tyrosine hydroxylase (the enzyme that produces L-DOPA) and catechol-O-methyl
transferase (COMT – the enzyme that converts L-DOPA to dopamine). No safety concerns have been
reported but the approaches have largely failed in phase two clinical trials.[226] The delivery of GAD
showed promise in phase two trials in 2011, but while effective at improving motor function, was inferior to
DBS. Follow-up studies in the same cohort have suggested persistent improvement.[230]

Neuroprotective treatments

A vaccine that primes the human immune system to destroy alpha-synuclein, PD01A, entered clinical trials
and a phase one report in 2020 suggested safety and tolerability.[231][232] In 2018, an antibody,
PRX002/RG7935, showed preliminary safety evidence in stage I trials supporting continuation to stage II
trials.[233]

Cell-based therapies

Beginning in the early 1980s, fetal, porcine, carotid or retinal tissues have been used in cell transplants, in
which dissociated cells are injected into the substantia nigra in the hope that they will incorporate
themselves into the brain in a way that replaces the dopamine-producing cells that have been lost.[87] These
sources of tissues have been largely replaced by induced pluripotent stem cell derived dopaminergic
neurons, as this is thought to represent a more feasible source of tissue. Initial evidence showed
mesencephalic dopamine-producing cell transplants being beneficial, but long-term benefit is
undetermined.[234] An additional problem was the excess release of dopamine by the transplanted tissue
resulting in dyskinesia.[234] In 2020, a first in human clinical trial reported the transplantation of induced
pluripotent stem cells into the brain of a person with PD.[235]

Pharmaceutical

Antagonists of adenosine receptors (specifically A2A) have been explored for Parkinson's.[236] Of these,
istradefylline has emerged as the most successful medication and was approved for medical use in the
United States in 2019.[237] It is approved as an add-on treatment to the levodopa/carbidopa regime.[237]

References
1. "Parkinson's Disease Information Page" (https://www.ninds.nih.gov/Disorders/All-Disorders/
Parkinsons-Disease-Information-Page). NINDS. 30 June 2016. Retrieved 18 July 2016.
2. Sveinbjornsdottir S (October 2016). "The clinical symptoms of Parkinson's disease" (https://d
oi.org/10.1111%2Fjnc.13691). Journal of Neurochemistry. 139 (Suppl 1): 318–324.
doi:10.1111/jnc.13691 (https://doi.org/10.1111%2Fjnc.13691). PMID 27401947 (https://pubm
ed.ncbi.nlm.nih.gov/27401947).
3. Carroll WM (2016). International Neurology (https://books.google.com/books?id=mRl6DAAA
QBAJ&pg=PA188). John Wiley & Sons. p. 188. ISBN 978-1118777367. Archived (https://we
b.archive.org/web/20170908154209/https://books.google.com/books?id=mRl6DAAAQBAJ&
pg=PA188) from the original on 8 September 2017.
4. Kalia LV, Lang AE (August 2015). "Parkinson's disease". Lancet. 386 (9996): 896–912.
doi:10.1016/s0140-6736(14)61393-3 (https://doi.org/10.1016%2Fs0140-6736%2814%2961
393-3). PMID 25904081 (https://pubmed.ncbi.nlm.nih.gov/25904081). S2CID 5502904 (http
s://api.semanticscholar.org/CorpusID:5502904).
5. Ferri FF (2010). "Chapter P". Ferri's differential diagnosis: a practical guide to the differential
diagnosis of symptoms, signs, and clinical disorders (2nd ed.). Philadelphia, PA:
Elsevier/Mosby. ISBN 978-0323076999.
6. Vos T, Allen C, Arora M, Barber RM, Bhutta ZA, Brown A, et al. (GBD 2015 Disease and
Injury Incidence and Prevalence Collaborators) (October 2016). "Global, regional, and
national incidence, prevalence, and years lived with disability for 310 diseases and injuries,
1990–2015: a systematic analysis for the Global Burden of Disease Study 2015" (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC5055577). Lancet. 388 (10053): 1545–1602.
doi:10.1016/S0140-6736(16)31678-6 (https://doi.org/10.1016%2FS0140-6736%2816%2931
678-6). PMC 5055577 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5055577).
PMID 27733282 (https://pubmed.ncbi.nlm.nih.gov/27733282).
7. Wang H, Naghavi M, Allen C, Barber RM, Bhutta ZA, Carter A, et al. (GBD 2015 Mortality
and Causes of Death Collaborators) (October 2016). "Global, regional, and national life
expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–
2015: a systematic analysis for the Global Burden of Disease Study 2015" (https://www.ncbi.
nlm.nih.gov/pmc/articles/PMC5388903). Lancet. 388 (10053): 1459–1544.
doi:10.1016/s0140-6736(16)31012-1 (https://doi.org/10.1016%2Fs0140-6736%2816%2931
012-1). PMC 5388903 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5388903).
PMID 27733281 (https://pubmed.ncbi.nlm.nih.gov/27733281).
8. Tolosa E, Katzenschlager R (2007). "Pharmacological management of Parkinson's
disease". In Tolosa E, Jankovic JJ (eds.). Parkinson's disease and movement disorders.
Hagerstwon, MD: Lippincott Williams & Wilkins. pp. 110–145. ISBN 978-0-7817-7881-7.
9. Tulisiak CT, Mercado G, Peelaerts W, Brundin L, Brundin P (2019). "Can infections trigger
alpha-synucleinopathies?" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6857718). Prog
Mol Biol Transl Sci. Progress in Molecular Biology and Translational Science. 168: 299–
322. doi:10.1016/bs.pmbts.2019.06.002 (https://doi.org/10.1016%2Fbs.pmbts.2019.06.002).
ISBN 9780128178744. PMC 6857718 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6857
718). PMID 31699323 (https://pubmed.ncbi.nlm.nih.gov/31699323).
10. Samii A, Nutt JG, Ransom BR (May 2004). "Parkinson's disease" (https://zenodo.org/record/
1259791). Lancet. 363 (9423): 1783–1793. doi:10.1016/S0140-6736(04)16305-8 (https://doi.
org/10.1016%2FS0140-6736%2804%2916305-8). PMID 15172778 (https://pubmed.ncbi.nl
m.nih.gov/15172778). S2CID 35364322 (https://api.semanticscholar.org/CorpusID:3536432
2).
11. Schrag A (2007). "Epidemiology of movement disorders". In Tolosa E, Jankovic JJ (eds.).
Parkinson's disease and movement disorders. Hagerstown, Maryland: Lippincott Williams &
Wilkins. pp. 50–66. ISBN 978-0-7817-7881-7.
12. Levin J, Kurz A, Arzberger T, Giese A, Höglinger GU (February 2016). "The differential
diagnosis and treatment of atypical parkinsonism" (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC4782269). Dtsch Ärztebl Int (Review). 113 (5): 61–69. doi:10.3238/arztebl.2016.0061
(https://doi.org/10.3238%2Farztebl.2016.0061). PMC 4782269 (https://www.ncbi.nlm.nih.go
v/pmc/articles/PMC4782269). PMID 26900156 (https://pubmed.ncbi.nlm.nih.gov/26900156).
13. Kon T, Tomiyama M, Wakabayashi K (February 2020). "Neuropathology of Lewy body
disease: Clinicopathological crosstalk between typical and atypical cases" (https://doi.org/1
0.1111%2Fneup.12597). Neuropathology. 40 (1): 30–39. doi:10.1111/neup.12597 (https://do
i.org/10.1111%2Fneup.12597). PMID 31498507 (https://pubmed.ncbi.nlm.nih.gov/3149850
7). S2CID 201983865 (https://api.semanticscholar.org/CorpusID:201983865).
14. Goedert M, Jakes R, Spillantini MG (2017). "The synucleinopathies: Twenty years on" (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC5345650). J Parkinsons Dis (Review). 7 (s1):
S51–S71. doi:10.3233/JPD-179005 (https://doi.org/10.3233%2FJPD-179005).
PMC 5345650 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5345650). PMID 28282814
(https://pubmed.ncbi.nlm.nih.gov/28282814).
15. Simon RP, Greenberg D, Aminoff MJ (2017). Clinical Neurology (10th ed.). New York:
McGraw-Hill. ISBN 978-1-259-86172-7.
16. Jankovic J (April 2008). "Parkinson's disease: clinical features and diagnosis" (http://jnnp.b
mj.com/content/79/4/368.full). Journal of Neurology, Neurosurgery, and Psychiatry. 79 (4):
368–376. doi:10.1136/jnnp.2007.131045 (https://doi.org/10.1136%2Fjnnp.2007.131045).
PMID 18344392 (https://pubmed.ncbi.nlm.nih.gov/18344392). Archived (https://web.archive.
org/web/20150819084941/http://jnnp.bmj.com/content/79/4/368.full) from the original on 19
August 2015.
17. "Swallowing and Parkinson's Disease | Parkinson's Disease" (https://www.michaeljfox.org/n
ews/swallowing-and-parkinsons-disease). 5 November 2013.
18. Suttrup I, Warnecke T (February 2016). "Dysphagia in Parkinson's Disease". Dysphagia. 31
(1): 24–32. doi:10.1007/s00455-015-9671-9 (https://doi.org/10.1007%2Fs00455-015-9671-
9). PMID 26590572 (https://pubmed.ncbi.nlm.nih.gov/26590572). S2CID 13003301 (https://a
pi.semanticscholar.org/CorpusID:13003301).
19. Connie T, Aderinola TB, Ong TS, Goh MKO, Erfianto B, Purnama B (12 December 2022).
"Pose-Based Gait Analysis for Diagnosis of Parkinson's Disease" (https://doi.org/10.3390%2
Fa15120474). Algorithms. MDPI AG. 15 (12): 474. doi:10.3390/a15120474 (https://doi.org/1
0.3390%2Fa15120474). ISSN 1999-4893 (https://www.worldcat.org/issn/1999-4893).
20. Cooper G, Eichhorn G, Rodnitzky RL (2008). "Parkinson's disease". In Conn PM (ed.).
Neuroscience in medicine. Totowa, NJ: Humana Press. pp. 508–512. ISBN 978-1-60327-
454-8.
21. Lees AJ, Hardy J, Revesz T (June 2009). "Parkinson's disease". Lancet. 373 (9680): 2055–
2066. doi:10.1016/S0140-6736(09)60492-X (https://doi.org/10.1016%2FS0140-6736%280
9%2960492-X). PMID 19524782 (https://pubmed.ncbi.nlm.nih.gov/19524782).
S2CID 42608600 (https://api.semanticscholar.org/CorpusID:42608600).
22. Banich MT, Compton RJ (2011). "Motor control". Cognitive neuroscience. Belmont, CA:
Wadsworth, Cengage learning. pp. 108–144. ISBN 978-0-8400-3298-0.
23. Longmore M, Wilkinson IB, Turmezei T, Cheung CK (4 January 2007). Oxford Handbook of
Clinical Medicine. Oxford University Press. p. 486. ISBN 978-0-19-856837-7.
24. Fung VS, Thompson PD (2007). "Rigidity and spasticity". In Tolosa E, Jankovic E (eds.).
Parkinson's disease and movement disorders. Hagerstown, MD: Lippincott Williams &
Wilkins. pp. 504–513. ISBN 978-0-7817-7881-7.
25. O'Sullivan & Schmitz 2007, pp. 856–857.
26. Yao SC, Hart AD, Terzella MJ (May 2013). "An evidence-based osteopathic approach to
Parkinson disease". Osteopathic Family Physician. 5 (3): 96–101.
doi:10.1016/j.osfp.2013.01.003 (https://doi.org/10.1016%2Fj.osfp.2013.01.003).
27. Hallett M, Poewe W (2008). Therapeutics of Parkinson's Disease and Other Movement
Disorders (https://books.google.com/books?id=fEezGoZ4h7YC&pg=PA417). John Wiley &
Sons. p. 417. ISBN 978-0-470-71400-3. Archived (https://web.archive.org/web/20170908154
209/https://books.google.com/books?id=fEezGoZ4h7YC&pg=PA417) from the original on 8
September 2017.
28. Hoehn MM, Yahr MD (May 1967). "Parkinsonism: onset, progression and mortality" (https://d
oi.org/10.1212%2Fwnl.17.5.427). Neurology. 17 (5): 427–442. doi:10.1212/wnl.17.5.427 (htt
ps://doi.org/10.1212%2Fwnl.17.5.427). PMID 6067254 (https://pubmed.ncbi.nlm.nih.gov/606
7254).
29. Pahwa R, Lyons KE (2003). Handbook of Parkinson's Disease (https://books.google.com/bo
oks?id=gX5mAIk5F6UC&pg=PA76) (Third ed.). CRC Press. p. 76. ISBN 978-0-203-91216-
4. Archived (https://web.archive.org/web/20170908154209/https://books.google.com/books?i
d=gX5mAIk5F6UC&pg=PA76) from the original on 8 September 2017.
30. Prenger MTM, Madray R, Van Hedger K, Anello M, MacDonald PA (2020). "Social
Symptoms of Parkinson's Disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC779058
5). Parkinsons Dis. 2020: 8846544. doi:10.1155/2020/8846544 (https://doi.org/10.1155%2F
2020%2F8846544). PMC 7790585 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC779058
5). PMID 33489081 (https://pubmed.ncbi.nlm.nih.gov/33489081).
31. Caballol N, Martí MJ, Tolosa E (September 2007). "Cognitive dysfunction and dementia in
Parkinson disease". Movement Disorders. 22 (Suppl 17): S358–S366.
doi:10.1002/mds.21677 (https://doi.org/10.1002%2Fmds.21677). PMID 18175397 (https://pu
bmed.ncbi.nlm.nih.gov/18175397). S2CID 3229727 (https://api.semanticscholar.org/CorpusI
D:3229727).
32. Parker KL, Lamichhane D, Caetano MS, Narayanan NS (October 2013). "Executive
dysfunction in Parkinson's disease and timing deficits" (https://www.ncbi.nlm.nih.gov/pmc/art
icles/PMC3813949). Frontiers in Integrative Neuroscience. 7: 75.
doi:10.3389/fnint.2013.00075 (https://doi.org/10.3389%2Ffnint.2013.00075). PMC 3813949
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3813949). PMID 24198770 (https://pubmed.
ncbi.nlm.nih.gov/24198770).
33. Gomperts SN (April 2016). "Lewy Body Dementias: Dementia With Lewy Bodies and
Parkinson Disease Dementia" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5390937).
Continuum (Minneap Minn) (Review). 22 (2 Dementia): 435–463.
doi:10.1212/CON.0000000000000309 (https://doi.org/10.1212%2FCON.000000000000030
9). PMC 5390937 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5390937).
PMID 27042903 (https://pubmed.ncbi.nlm.nih.gov/27042903).
34. Garcia-Ptacek S, Kramberger MG (September 2016). "Parkinson Disease and Dementia".
Journal of Geriatric Psychiatry and Neurology. 29 (5): 261–270.
doi:10.1177/0891988716654985 (https://doi.org/10.1177%2F0891988716654985).
PMID 27502301 (https://pubmed.ncbi.nlm.nih.gov/27502301). S2CID 21279235 (https://api.s
emanticscholar.org/CorpusID:21279235).
35. Han JW, Ahn YD, Kim WS, Shin CM, Jeong SJ, Song YS, et al. (November 2018).
"Psychiatric Manifestation in Patients with Parkinson's Disease" (https://www.ncbi.nlm.nih.g
ov/pmc/articles/PMC6236081). Journal of Korean Medical Science. 33 (47): e300.
doi:10.3346/jkms.2018.33.e300 (https://doi.org/10.3346%2Fjkms.2018.33.e300).
PMC 6236081 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6236081). PMID 30450025
(https://pubmed.ncbi.nlm.nih.gov/30450025).
36. Ffytche DH, Creese B, Politis M, Chaudhuri KR, Weintraub D, Ballard C, Aarsland D
(February 2017). "The psychosis spectrum in Parkinson disease" (https://www.ncbi.nlm.nih.
gov/pmc/articles/PMC5656278). Nature Reviews. Neurology. 13 (2): 81–95.
doi:10.1038/nrneurol.2016.200 (https://doi.org/10.1038%2Fnrneurol.2016.200).
PMC 5656278 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5656278). PMID 28106066
(https://pubmed.ncbi.nlm.nih.gov/28106066).
37. Shergill SS, Walker Z, Le Katona C (October 1998). "A preliminary investigation of laterality
in Parkinson's disease and susceptibility to psychosis" (https://www.ncbi.nlm.nih.gov/pmc/art
icles/PMC2170290). Journal of Neurology, Neurosurgery, and Psychiatry. 65 (4): 610–611.
doi:10.1136/jnnp.65.4.610 (https://doi.org/10.1136%2Fjnnp.65.4.610). PMC 2170290 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC2170290). PMID 9771806 (https://pubmed.ncbi.nl
m.nih.gov/9771806).
38. Friedman JH (November 2010). "Parkinson's disease psychosis 2010: a review article".
Parkinsonism & Related Disorders. 16 (9): 553–560. doi:10.1016/j.parkreldis.2010.05.004 (h
ttps://doi.org/10.1016%2Fj.parkreldis.2010.05.004). PMID 20538500 (https://pubmed.ncbi.nl
m.nih.gov/20538500).
39. Weintraub D, Mamikonyan E (September 2019). "The Neuropsychiatry of Parkinson
Disease: A Perfect Storm" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7015280). The
American Journal of Geriatric Psychiatry. 27 (9): 998–1018. doi:10.1016/j.jagp.2019.03.002
(https://doi.org/10.1016%2Fj.jagp.2019.03.002). PMC 7015280 (https://www.ncbi.nlm.nih.go
v/pmc/articles/PMC7015280). PMID 31006550 (https://pubmed.ncbi.nlm.nih.gov/31006550).
40. Dissanayaka NN (8 March 2021). "Chapter 9: Anxiety in Parkinson's Disease". In Byrne GJ,
Panchana NA (eds.). Anxiety in Older People: Clinical and Research Perspectives.
Cambridge: Cambridge University Press. pp. 139–156. doi:10.1017/9781139087469.009 (ht
tps://doi.org/10.1017%2F9781139087469.009). S2CID 87250745 (https://api.semanticschol
ar.org/CorpusID:87250745).
41. Goetz CG (2010). "New developments in depression, anxiety, compulsiveness, and
hallucinations in Parkinson's disease" (http://doi.wiley.com/10.1002/mds.22636). Movement
Disorders. 25 (S1): S104–S109. doi:10.1002/mds.22636 (https://doi.org/10.1002%2Fmds.22
636). PMID 20187250 (https://pubmed.ncbi.nlm.nih.gov/20187250). S2CID 35420377 (http
s://api.semanticscholar.org/CorpusID:35420377).
42. Husain M, Roiser JP (August 2018). "Neuroscience of apathy and anhedonia: a
transdiagnostic approach" (https://discovery.ucl.ac.uk/id/eprint/10053434/). Nature Reviews.
Neuroscience. 19 (8): 470–484. doi:10.1038/s41583-018-0029-9 (https://doi.org/10.1038%2
Fs41583-018-0029-9). PMID 29946157 (https://pubmed.ncbi.nlm.nih.gov/29946157).
S2CID 49428707 (https://api.semanticscholar.org/CorpusID:49428707).
43. Turner V, Husain M (2022). "Anhedonia in Neurodegenerative Diseases". Current Topics in
Behavioral Neurosciences. 58: 255–277. doi:10.1007/7854_2022_352 (https://doi.org/10.10
07%2F7854_2022_352). ISBN 978-3-031-09682-2. PMID 35435648 (https://pubmed.ncbi.nl
m.nih.gov/35435648).
44. Noyce AJ, Bestwick JP, Silveira-Moriyama L, et al. (December 2012). "Meta-analysis of early
nonmotor features and risk factors for Parkinson disease" (https://www.ncbi.nlm.nih.gov/pmc/
articles/PMC3556649). Annals of Neurology (Review). 72 (6): 893–901.
doi:10.1002/ana.23687 (https://doi.org/10.1002%2Fana.23687). PMC 3556649 (https://www.
ncbi.nlm.nih.gov/pmc/articles/PMC3556649). PMID 23071076 (https://pubmed.ncbi.nlm.nih.
gov/23071076).
45. Barichella M, Cereda E, Pezzoli G (October 2009). "Major nutritional issues in the
management of Parkinson's disease". Movement Disorders. 24 (13): 1881–1892.
doi:10.1002/mds.22705 (https://doi.org/10.1002%2Fmds.22705). hdl:2434/67795 (https://hdl.
handle.net/2434%2F67795). PMID 19691125 (https://pubmed.ncbi.nlm.nih.gov/19691125).
S2CID 23528416 (https://api.semanticscholar.org/CorpusID:23528416).
46. Warnecke T, Schäfer KH, Claus I, Del Tredici K, Jost WH (March 2022). "Gastrointestinal
involvement in Parkinson's disease: pathophysiology, diagnosis, and management" (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC8948218). npj Parkinson's Disease. 8 (1): 31.
doi:10.1038/s41531-022-00295-x (https://doi.org/10.1038%2Fs41531-022-00295-x).
PMC 8948218 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8948218). PMID 35332158
(https://pubmed.ncbi.nlm.nih.gov/35332158).
47. Skjærbæk C, Knudsen K, Horsager J, Borghammer P (January 2021). "Gastrointestinal
Dysfunction in Parkinson's Disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC786679
1). J Clin Med. 10 (3): 493. doi:10.3390/jcm10030493 (https://doi.org/10.3390%2Fjcm10030
493). PMC 7866791 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7866791).
PMID 33572547 (https://pubmed.ncbi.nlm.nih.gov/33572547).
48. Kim YE, Jeon BS (1 January 2014). "Clinical implication of REM sleep behavior disorder in
Parkinson's disease". Journal of Parkinson's Disease. 4 (2): 237–244. doi:10.3233/jpd-
130293 (https://doi.org/10.3233%2Fjpd-130293). PMID 24613864 (https://pubmed.ncbi.nlm.
nih.gov/24613864).
49. de Lau LM, Breteler MM (June 2006). "Epidemiology of Parkinson's disease". The Lancet.
Neurology. 5 (6): 525–535. doi:10.1016/S1474-4422(06)70471-9 (https://doi.org/10.1016%2
FS1474-4422%2806%2970471-9). PMID 16713924 (https://pubmed.ncbi.nlm.nih.gov/16713
924). S2CID 39310242 (https://api.semanticscholar.org/CorpusID:39310242).
50. Barreto GE, Iarkov A, Moran VE (January 2015). "Beneficial effects of nicotine, cotinine and
its metabolites as potential agents for Parkinson's disease" (https://www.ncbi.nlm.nih.gov/pm
c/articles/PMC4288130). Frontiers in Aging Neuroscience. 6: 340.
doi:10.3389/fnagi.2014.00340 (https://doi.org/10.3389%2Ffnagi.2014.00340). PMC 4288130
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4288130). PMID 25620929 (https://pubmed.
ncbi.nlm.nih.gov/25620929).
51. De Miranda BR, Goldman SM, Miller GW, Greenamy T, Dorseye ER (2022). "Preventing
Parkinson's Disease: An Environmental Agenda". Journal of Parkinson's Disease. 12: 45–
68. doi:10.3233/JPD-212922 (https://doi.org/10.3233%2FJPD-212922). PMID 34719434 (htt
ps://pubmed.ncbi.nlm.nih.gov/34719434). S2CID 240235393 (https://api.semanticscholar.or
g/CorpusID:240235393).
52. Kumar A, Calne SM, Schulzer M, Mak E, Wszolek Z, Van Netten C, Tsui JK, Stoessl AJ,
Calne DB (July 2004). "Clustering of Parkinson disease: shared cause or coincidence?".
Arch Neurol. 61 (7): 1057–60. doi:10.1001/archneur.61.7.1057 (https://doi.org/10.1001%2Far
chneur.61.7.1057). PMID 15262735 (https://pubmed.ncbi.nlm.nih.gov/15262735).
53. Lesage S, Brice A (April 2009). "Parkinson's disease: from monogenic forms to genetic
susceptibility factors" (https://doi.org/10.1093%2Fhmg%2Fddp012). Human Molecular
Genetics. 18 (R1): R48–59. doi:10.1093/hmg/ddp012 (https://doi.org/10.1093%2Fhmg%2Fd
dp012). PMID 19297401 (https://pubmed.ncbi.nlm.nih.gov/19297401).
54. Deng H, Wang P, Jankovic J (March 2018). "The genetics of Parkinson disease" (https://pub
med.ncbi.nlm.nih.gov/29288112). Ageing Research Reviews. 42: 72–85.
doi:10.1016/j.arr.2017.12.007 (https://doi.org/10.1016%2Fj.arr.2017.12.007).
PMID 29288112 (https://pubmed.ncbi.nlm.nih.gov/29288112). S2CID 28246244 (https://api.s
emanticscholar.org/CorpusID:28246244).
55. Puschmann A (September 2017). "New Genes Causing Hereditary Parkinson's Disease or
Parkinsonism" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5522513). Current Neurology
and Neuroscience Reports. 17 (9): 66. doi:10.1007/s11910-017-0780-8 (https://doi.org/10.10
07%2Fs11910-017-0780-8). PMC 5522513 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC
5522513). PMID 28733970 (https://pubmed.ncbi.nlm.nih.gov/28733970).
56. Quadri M, Mandemakers W, Grochowska MM, Masius R, Geut H, Fabrizio E, et al. (July
2018). "LRP10 genetic variants in familial Parkinson's disease and dementia with Lewy
bodies: a genome-wide linkage and sequencing study" (https://doi.org/10.1016%2Fs1474-4
422%2818%2930179-0). The Lancet. Neurology. 17 (7): 597–608. doi:10.1016/s1474-
4422(18)30179-0 (https://doi.org/10.1016%2Fs1474-4422%2818%2930179-0).
PMID 29887161 (https://pubmed.ncbi.nlm.nih.gov/29887161). S2CID 47009438 (https://api.s
emanticscholar.org/CorpusID:47009438).
57. Chen Y, Cen Z, Zheng X, Pan Q, Chen X, Zhu L, et al. (June 2019). "LRP10 in autosomal-
dominant Parkinson's disease" (https://pubmed.ncbi.nlm.nih.gov/30964957). Movement
Disorders. 34 (6): 912–916. doi:10.1002/mds.27693 (https://doi.org/10.1002%2Fmds.27693).
PMID 30964957 (https://pubmed.ncbi.nlm.nih.gov/30964957). S2CID 106408549 (https://ap
i.semanticscholar.org/CorpusID:106408549).
58. Stoker TB, Torsney KM, Barker RA (2018). "Pathological mechanisms and clinical aspects
of GBA1 mutation-associated Parkinson's disease.". In Stoker TB, Greenland JC (eds.).
Parkinson's Disease: Pathogenesis and clinical aspects. Brisbane: Codon Publications.
59. Abugable AA, Morris JL, Palminha NM, et al. (September 2019). "DNA repair and
neurological disease: From molecular understanding to the development of diagnostics and
model organisms" (https://doi.org/10.1016%2Fj.dnarep.2019.102669). DNA Repair. 81:
102669. doi:10.1016/j.dnarep.2019.102669 (https://doi.org/10.1016%2Fj.dnarep.2019.1026
69). PMID 31331820 (https://pubmed.ncbi.nlm.nih.gov/31331820).
60. Davie CA (2008). "A review of Parkinson's disease" (https://doi.org/10.1093%2Fbmb%2Fldn
013). British Medical Bulletin. 86 (1): 109–127. doi:10.1093/bmb/ldn013 (https://doi.org/10.10
93%2Fbmb%2Fldn013). PMID 18398010 (https://pubmed.ncbi.nlm.nih.gov/18398010).
61. Kalia LV, Lang AE (August 2015). "Parkinson's disease". Lancet. 386 (9996): 896–912.
doi:10.1016/S0140-6736(14)61393-3 (https://doi.org/10.1016%2FS0140-6736%2814%2961
393-3). PMID 25904081 (https://pubmed.ncbi.nlm.nih.gov/25904081). S2CID 5502904 (http
s://api.semanticscholar.org/CorpusID:5502904).
62. Gan-Or Z, Dion PA, Rouleau GA (2 September 2015). "Genetic perspective on the role of the
autophagy-lysosome pathway in Parkinson disease" (https://www.ncbi.nlm.nih.gov/pmc/artic
les/PMC4590678). Autophagy. 11 (9): 1443–1457. doi:10.1080/15548627.2015.1067364 (ht
tps://doi.org/10.1080%2F15548627.2015.1067364). PMC 4590678 (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC4590678). PMID 26207393 (https://pubmed.ncbi.nlm.nih.gov/262073
93).
63. Dorsey ER, Zafar M, Lettenberger SE, et al. (2023). "Trichloroethylene: An Invisible Cause of
Parkinson's Disease?" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10041423). J
Parkinsons Dis. 13 (2): 203–218. doi:10.3233/JPD-225047 (https://doi.org/10.3233%2FJPD-
225047). PMC 10041423 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10041423).
PMID 36938742 (https://pubmed.ncbi.nlm.nih.gov/36938742).
64. Simon DK, Tanner CM, Brundin P (2020). "Parkinson Disease Epidemiology, Pathology,
Genetics, and Pathophysiology". Clinical Geriatric Medicine. 36 (1): 1–12.
doi:10.1016/j.cger.2019.08.002 (https://doi.org/10.1016%2Fj.cger.2019.08.002).
PMID 31733690 (https://pubmed.ncbi.nlm.nih.gov/31733690). S2CID 201961979 (https://ap
i.semanticscholar.org/CorpusID:201961979).
65. Sharma, Priyanshu; Mittal, Payal (2023). "Paraquat (herbicide) as a cause of Parkinson's
Disease" (https://www.sciencedirect.com/science/article/abs/pii/S1353802023010118).
Parkinsonism & Related Disorders. Elsevier BV: 105932.
doi:10.1016/j.parkreldis.2023.105932 (https://doi.org/10.1016%2Fj.parkreldis.2023.105932).
ISSN 1353-8020 (https://www.worldcat.org/issn/1353-8020).
66. Vellingiri, Balachandar; Chandrasekhar, Mamatha; Sri Sabari, S.; Gopalakrishnan, Abilash
Valsala; Narayanasamy, Arul; Venkatesan, Dhivya; Iyer, Mahalaxmi; Kesari, Kavindra; Dey,
Abhijit (2022). "Neurotoxicity of pesticides – A link to neurodegeneration". Ecotoxicology and
Environmental Safety. Elsevier BV. 243: 113972. doi:10.1016/j.ecoenv.2022.113972 (https://
doi.org/10.1016%2Fj.ecoenv.2022.113972). ISSN 0147-6513 (https://www.worldcat.org/issn/
0147-6513).
67. Zu L (2023). "Chronic carbon disulfide exposure induces parkinsonian pathology via a-
synuclein aggregation and necrosome complex interaction". iScience. 26 (10): 1–16.
PMID 37731606 (https://pubmed.ncbi.nlm.nih.gov/37731606).
68. Langston JW (6 March 2017). "The MPTP Story" (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC5345642). Journal of Parkinson's Disease. 7 (s1): S11–S19. doi:10.3233/JPD-179006
(https://doi.org/10.3233%2FJPD-179006). PMC 5345642 (https://www.ncbi.nlm.nih.gov/pmc/
articles/PMC5345642). PMID 28282815 (https://pubmed.ncbi.nlm.nih.gov/28282815).
69. Song L, Xu MB, Zhou XL, Zhang DP, Zhang SL, Zheng GQ (2017). "A Preclinical Systematic
Review of Ginsenoside-Rg1 in Experimental Parkinson's Disease" (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC5366755). Oxidative Medicine and Cellular Longevity. 2017:
2163053. doi:10.1155/2017/2163053 (https://doi.org/10.1155%2F2017%2F2163053).
PMC 5366755 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5366755). PMID 28386306
(https://pubmed.ncbi.nlm.nih.gov/28386306).
70. Chahine LM, Stern MB, Chen-Plotkin A (January 2014). "Blood-based biomarkers for
Parkinson's disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4070332).
Parkinsonism & Related Disorders. 20 (Suppl 1): S99–103. doi:10.1016/S1353-
8020(13)70025-7 (https://doi.org/10.1016%2FS1353-8020%2813%2970025-7).
PMC 4070332 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4070332). PMID 24262199
(https://pubmed.ncbi.nlm.nih.gov/24262199).
71. Delic V, Beck KD, Pang KC, Citron BA (2020). "Biological links between traumatic brain
injury and Parkinson's disease" (https://doi.org/10.1186%2Fs40478-020-00924-7). Acta
Neuropathologica Communications. 8 (1): 1–16. doi:10.1186/s40478-020-00924-7 (https://d
oi.org/10.1186%2Fs40478-020-00924-7). PMID 32264976 (https://pubmed.ncbi.nlm.nih.gov/
32264976).
72. Bellou V, Belbasis L, Tzoulaki I, Evangelou E, Ioannidis J (2015). "Environmental risk
factors and Parkinson's disease: An umbrella review of meta-analyses". Parkinsonism
Related Disorders. 23: 1–9. doi:10.1016/j.parkreldis.2015.12.008 (https://doi.org/10.1016%2
Fj.parkreldis.2015.12.008). hdl:10044/1/31820 (https://hdl.handle.net/10044%2F1%2F3182
0). PMID 26739246 (https://pubmed.ncbi.nlm.nih.gov/26739246).
73. Bruce HJ, Tripodis Y, McClean M, et al. (2023). "American Football Play and Parkinson
Disease Among Men" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10422187). JAMA
Network Open. 6 (8): e2328644. doi:10.1001/jamanetworkopen.2023.28644 (https://doi.org/1
0.1001%2Fjamanetworkopen.2023.28644). PMC 10422187 (https://www.ncbi.nlm.nih.gov/p
mc/articles/PMC10422187). PMID 37566412 (https://pubmed.ncbi.nlm.nih.gov/37566412).
74. Gupta D, Kuruvilla A (December 2011). "Vascular parkinsonism: what makes it different?" (ht
tps://pmj.bmj.com/content/87/1034/829). Postgraduate Medical Journal. 87 (1034): 829–836.
doi:10.1136/postgradmedj-2011-130051 (https://doi.org/10.1136%2Fpostgradmedj-2011-13
0051). PMID 22121251 (https://pubmed.ncbi.nlm.nih.gov/22121251). S2CID 29227069 (http
s://api.semanticscholar.org/CorpusID:29227069).
75. Miguel-Puga A, Villafuerte G, Salas-Pacheco J, Arias-Carrión O (22 September 2017).
"Therapeutic Interventions for Vascular Parkinsonism: A Systematic Review and Meta-
analysis" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5614922). Frontiers in Neurology.
8: 481. doi:10.3389/fneur.2017.00481 (https://doi.org/10.3389%2Ffneur.2017.00481).
PMC 5614922 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5614922). PMID 29018399
(https://pubmed.ncbi.nlm.nih.gov/29018399).
76. Çamcı G, Oğuz S (April 2016). "Association between Parkinson's Disease and Helicobacter
Pylori" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4828559). Journal of Clinical
Neurology. 12 (2): 147–150. doi:10.3988/jcn.2016.12.2.147 (https://doi.org/10.3988%2Fjcn.2
016.12.2.147). PMC 4828559 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4828559).
PMID 26932258 (https://pubmed.ncbi.nlm.nih.gov/26932258).
77. McGee DJ, Lu XH, Disbrow EA (2018). "Stomaching the Possibility of a Pathogenic Role for
Helicobacter pylori in Parkinson's Disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC
6130334). Journal of Parkinson's Disease. 8 (3): 367–374. doi:10.3233/JPD-181327 (https://
doi.org/10.3233%2FJPD-181327). PMC 6130334 (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC6130334). PMID 29966206 (https://pubmed.ncbi.nlm.nih.gov/29966206).
78. Terracciano, A; Aschwanden, D; Stephan, Y; Cerasa, A; Passamonti, L; Toschi, N; Sutin, AR
(August 2021). "Neuroticism and Risk of Parkinson's Disease: A Meta-Analysis" (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC8376751). Movement Disorders. 36 (8): 1863–1870.
doi:10.1002/mds.28575 (https://doi.org/10.1002%2Fmds.28575). PMC 8376751 (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC8376751). PMID 33817817 (https://pubmed.ncbi.nlm.ni
h.gov/33817817).
79. Terracciano, Antonio; Luchetti, Martina; Karakose, Selin; Stephan, Yannick; Sutin, Angelina
R. (2 October 2023). "Loneliness and Risk of Parkinson Disease" (https://jamanetwork.com/j
ournals/jamaneurology/fullarticle/2809774). JAMA Neurology. 80 (11): 1138.
doi:10.1001/jamaneurol.2023.3382 (https://doi.org/10.1001%2Fjamaneurol.2023.3382).
ISSN 2168-6149 (https://www.worldcat.org/issn/2168-6149). PMC 10546293 (https://www.nc
bi.nlm.nih.gov/pmc/articles/PMC10546293). PMID 37782489 (https://pubmed.ncbi.nlm.nih.g
ov/37782489).
80. Villar-Piqué A, Lopes da Fonseca T, Outeiro TF (October 2016). "Structure, function and
toxicity of alpha-synuclein: the Bermuda triangle in synucleinopathies" (https://doi.org/10.11
11%2Fjnc.13249). Journal of Neurochemistry. 139 (Suppl 1): 240–255.
doi:10.1111/jnc.13249 (https://doi.org/10.1111%2Fjnc.13249). PMID 26190401 (https://pubm
ed.ncbi.nlm.nih.gov/26190401). S2CID 11420411 (https://api.semanticscholar.org/CorpusID:
11420411).
81. Burré J, Sharma M, Südhof TC (March 2018). "Cell Biology and Pathophysiology of α-
Synuclein" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5519445). Cold Spring Harbor
Perspectives in Medicine. 8 (3): a024091. doi:10.1101/cshperspect.a024091 (https://doi.org/
10.1101%2Fcshperspect.a024091). PMC 5519445 (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC5519445). PMID 28108534 (https://pubmed.ncbi.nlm.nih.gov/28108534).
82. Dickson DV (2007). "Neuropathology of movement disorders". In Tolosa E, Jankovic JJ
(eds.). Parkinson's disease and movement disorders. Hagerstown, MD: Lippincott Williams
& Wilkins. pp. 271–283. ISBN 978-0-7817-7881-7.
83. Armstrong MJ, Okun MS (February 2020). "Diagnosis and Treatment of Parkinson Disease:
A Review". JAMA. 323 (6): 548–560. doi:10.1001/jama.2019.22360 (https://doi.org/10.100
1%2Fjama.2019.22360). PMID 32044947 (https://pubmed.ncbi.nlm.nih.gov/32044947).
S2CID 211079287 (https://api.semanticscholar.org/CorpusID:211079287).
84. Jubault T, Brambati SM, Degroot C, Kullmann B, Strafella AP, Lafontaine AL, Chouinard S,
Monchi O (December 2009). Gendelman HE (ed.). "Regional brain stem atrophy in
idiopathic Parkinson's disease detected by anatomical MRI" (https://www.ncbi.nlm.nih.gov/p
mc/articles/PMC2784293). PLOS ONE. 4 (12): e8247. Bibcode:2009PLoSO...4.8247J (http
s://ui.adsabs.harvard.edu/abs/2009PLoSO...4.8247J). doi:10.1371/journal.pone.0008247 (ht
tps://doi.org/10.1371%2Fjournal.pone.0008247). PMC 2784293 (https://www.ncbi.nlm.nih.go
v/pmc/articles/PMC2784293). PMID 20011063 (https://pubmed.ncbi.nlm.nih.gov/20011063).
85. Obeso JA, Rodríguez-Oroz MC, Benitez-Temino B, Blesa FJ, Guridi J, Marin C, Rodriguez M
(2008). "Functional organization of the basal ganglia: therapeutic implications for
Parkinson's disease". Movement Disorders. 23 (Suppl 3): S548–S559.
doi:10.1002/mds.22062 (https://doi.org/10.1002%2Fmds.22062). PMID 18781672 (https://pu
bmed.ncbi.nlm.nih.gov/18781672). S2CID 13186083 (https://api.semanticscholar.org/Corpu
sID:13186083).
86. Schulz-Schaeffer WJ (August 2010). "The synaptic pathology of alpha-synuclein
aggregation in dementia with Lewy bodies, Parkinson's disease and Parkinson's disease
dementia" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2892607). Acta
Neuropathologica. 120 (2): 131–143. doi:10.1007/s00401-010-0711-0 (https://doi.org/10.100
7%2Fs00401-010-0711-0). PMC 2892607 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2
892607). PMID 20563819 (https://pubmed.ncbi.nlm.nih.gov/20563819).
87. Obeso JA, Rodriguez-Oroz MC, Goetz CG, et al. (June 2010). "Missing pieces in the
Parkinson's disease puzzle". Nature Medicine. 16 (6): 653–661. doi:10.1038/nm.2165 (http
s://doi.org/10.1038%2Fnm.2165). PMID 20495568 (https://pubmed.ncbi.nlm.nih.gov/204955
68). S2CID 3146438 (https://api.semanticscholar.org/CorpusID:3146438).
88. Hirsch EC (December 2009). "Iron transport in Parkinson's disease". Parkinsonism &
Related Disorders. 15 (Suppl 3): S209–S211. doi:10.1016/S1353-8020(09)70816-8 (https://
doi.org/10.1016%2FS1353-8020%2809%2970816-8). PMID 20082992 (https://pubmed.ncb
i.nlm.nih.gov/20082992).
89. Li X, Sundquist J, Sundquist K (23 December 2011). "Subsequent Risks of Parkinson
Disease in Patients with Autoimmune and Related Disorders: A Nationwide Epidemiological
Study from Sweden" (https://dx.doi.org/10.1159/000333222). Neurodegenerative Diseases.
10 (1–4): 277–284. doi:10.1159/000333222 (https://doi.org/10.1159%2F000333222).
ISSN 1660-2854 (https://www.worldcat.org/issn/1660-2854). PMID 22205172 (https://pubme
d.ncbi.nlm.nih.gov/22205172). S2CID 39874367 (https://api.semanticscholar.org/CorpusID:3
9874367).
90. Lai SW, Lin CH, Lin HF, Lin CL, Lin CC, Liao KF (February 2017). "Herpes zoster correlates
with increased risk of Parkinson's disease in older people: A population-based cohort study
in Taiwan" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5319504). Medicine (Baltimore).
96 (7): e6075. doi:10.1097/MD.0000000000006075 (https://doi.org/10.1097%2FMD.000000
0000006075). PMC 5319504 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5319504).
PMID 28207515 (https://pubmed.ncbi.nlm.nih.gov/28207515).
91. Tan EK, Chao YX, West A, Chan LL, Poewe W, Jankovic J (June 2020). "Parkinson disease
and the immune system - associations, mechanisms and therapeutics". Nat Rev Neurol. 16
(6): 303–318. doi:10.1038/s41582-020-0344-4 (https://doi.org/10.1038%2Fs41582-020-0344
-4). ISSN 1759-4758 (https://www.worldcat.org/issn/1759-4758). PMID 32332985 (https://pu
bmed.ncbi.nlm.nih.gov/32332985). S2CID 216111568 (https://api.semanticscholar.org/Corp
usID:216111568).
92. Raj T, Rothamel K, Mostafavi S, Ye C, Lee MN, Replogle JM, et al. (May 2014). "Polarization
of the effects of autoimmune and neurodegenerative risk alleles in leukocytes" (https://www.
ncbi.nlm.nih.gov/pmc/articles/PMC4910825). Science. 344 (6183): 519–23.
Bibcode:2014Sci...344..519R (https://ui.adsabs.harvard.edu/abs/2014Sci...344..519R).
doi:10.1126/science.1249547 (https://doi.org/10.1126%2Fscience.1249547). PMC 4910825
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4910825). PMID 24786080 (https://pubmed.
ncbi.nlm.nih.gov/24786080).
93. Du G, Dong W, Yang Q, Yu X, Ma J, Gu W, Huang Y (2020). "Altered Gut Microbiota Related
to Inflammatory Responses in Patients With Huntington's Disease" (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC7933529). Front Immunol. 11: 603594.
doi:10.3389/fimmu.2020.603594 (https://doi.org/10.3389%2Ffimmu.2020.603594).
ISSN 1664-3224 (https://www.worldcat.org/issn/1664-3224). PMC 7933529 (https://www.ncb
i.nlm.nih.gov/pmc/articles/PMC7933529). PMID 33679692 (https://pubmed.ncbi.nlm.nih.gov/
33679692).
94. Gamborg M, Hvid LG, Dalgas U, Langeskov-Christensen M (3 October 2021). "Review for
"Parkinson's disease and intensive exercise therapy — An updated systematic review and
meta‐analysis" " (https://doi.org/10.1111%2Fane.13579%2Fv1%2Freview2). Acta
Neurologica Scandinavica. doi:10.1111/ane.13579/v1/review2 (https://doi.org/10.1111%2Fa
ne.13579%2Fv1%2Freview2). ISSN 1600-0404 (https://www.worldcat.org/issn/1600-0404).
95. Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, et al. (August 2019).
"Transneuronal Propagation of Pathologic α-Synuclein from the Gut to the Brain Models
Parkinson's Disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6706297). Neuron.
103 (4): 627–641.e7. doi:10.1016/j.neuron.2019.05.035 (https://doi.org/10.1016%2Fj.neuron.
2019.05.035). PMC 6706297 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6706297).
PMID 31255487 (https://pubmed.ncbi.nlm.nih.gov/31255487).
96. Choi JG, Kim N, Ju IG, Eo H, Lim SM, Jang SE, et al. (January 2018). "Oral administration of
Proteus mirabilis damages dopaminergic neurons and motor functions in mice" (https://www.
ncbi.nlm.nih.gov/pmc/articles/PMC5775305). Scientific Reports. 8 (1): 1275.
Bibcode:2018NatSR...8.1275C (https://ui.adsabs.harvard.edu/abs/2018NatSR...8.1275C).
doi:10.1038/s41598-018-19646-x (https://doi.org/10.1038%2Fs41598-018-19646-x).
PMC 5775305 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5775305). PMID 29352191
(https://pubmed.ncbi.nlm.nih.gov/29352191).
97. The National Collaborating Centre for Chronic Conditions, ed. (2006). "Diagnosing
Parkinson's Disease" (http://guidance.nice.org.uk/CG35/Guidance/pdf/English). Parkinson's
Disease. London: Royal College of Physicians. pp. 29–47. ISBN 978-1-86016-283-1.
Archived (https://web.archive.org/web/20100924153546/http://guidance.nice.org.uk/CG35/G
uidance/pdf/English) from the original on 24 September 2010.
98. Poewe W, Wenning G (November 2002). "The differential diagnosis of Parkinson's disease".
European Journal of Neurology. 9 (Suppl 3): 23–30. doi:10.1046/j.1468-1331.9.s3.3.x (http
s://doi.org/10.1046%2Fj.1468-1331.9.s3.3.x). PMID 12464118 (https://pubmed.ncbi.nlm.nih.
gov/12464118).
99. Gibb WR, Lees AJ (June 1988). "The relevance of the Lewy body to the pathogenesis of
idiopathic Parkinson's disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1033142).
Journal of Neurology, Neurosurgery, and Psychiatry. 51 (6): 745–752.
doi:10.1136/jnnp.51.6.745 (https://doi.org/10.1136%2Fjnnp.51.6.745). PMC 1033142 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC1033142). PMID 2841426 (https://pubmed.ncbi.nl
m.nih.gov/2841426).
100. Mustafa HI, Fessel JP, Barwise J, Shannon JR, Raj SR, Diedrich A, et al. (January 2012).
"Dysautonomia: perioperative implications" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC
3296831). Anesthesiology. 116 (1): 205–215. doi:10.1097/ALN.0b013e31823db712 (https://
doi.org/10.1097%2FALN.0b013e31823db712). PMC 3296831 (https://www.ncbi.nlm.nih.go
v/pmc/articles/PMC3296831). PMID 22143168 (https://pubmed.ncbi.nlm.nih.gov/22143168).
101. Rizzo G, Copetti M, Arcuti S, Martino D, Fontana A, Logroscino G (February 2016).
"Accuracy of clinical diagnosis of Parkinson disease: A systematic review and meta-
analysis". Neurology. 86 (6): 566–576. doi:10.1212/WNL.0000000000002350 (https://doi.or
g/10.1212%2FWNL.0000000000002350). PMID 26764028 (https://pubmed.ncbi.nlm.nih.go
v/26764028). S2CID 207110404 (https://api.semanticscholar.org/CorpusID:207110404).
102. Brooks DJ (April 2010). "Imaging approaches to Parkinson disease" (https://doi.org/10.296
7%2Fjnumed.108.059998). Journal of Nuclear Medicine. 51 (4): 596–609.
doi:10.2967/jnumed.108.059998 (https://doi.org/10.2967%2Fjnumed.108.059998).
PMID 20351351 (https://pubmed.ncbi.nlm.nih.gov/20351351).
103. Schwarz ST, Afzal M, Morgan PS, Bajaj N, Gowland PA, Auer DP (2014). "The 'swallow tail'
appearance of the healthy nigrosome – a new accurate test of Parkinson's disease: a case-
control and retrospective cross-sectional MRI study at 3T" (https://www.ncbi.nlm.nih.gov/pm
c/articles/PMC3977922). PLOS ONE. 9 (4): e93814. Bibcode:2014PLoSO...993814S (http
s://ui.adsabs.harvard.edu/abs/2014PLoSO...993814S). doi:10.1371/journal.pone.0093814
(https://doi.org/10.1371%2Fjournal.pone.0093814). PMC 3977922 (https://www.ncbi.nlm.nih.
gov/pmc/articles/PMC3977922). PMID 24710392 (https://pubmed.ncbi.nlm.nih.gov/2471039
2).
104. Mahlknecht P, Krismer F, Poewe W, Seppi K (April 2017). "Meta-analysis of dorsolateral
nigral hyperintensity on magnetic resonance imaging as a marker for Parkinson's disease".
Movement Disorders. 32 (4): 619–623. doi:10.1002/mds.26932 (https://doi.org/10.1002%2F
mds.26932). PMID 28151553 (https://pubmed.ncbi.nlm.nih.gov/28151553). S2CID 7730034
(https://api.semanticscholar.org/CorpusID:7730034).
105. Cho SJ, Bae YJ, Kim JM, et al. (September 2020). "Diagnostic performance of
neuromelanin-sensitive magnetic resonance imaging for patients with Parkinson's disease
and factor analysis for its heterogeneity: a systematic review and meta-analysis". European
Radiology. 30 (10): 1268–1280. doi:10.1007/s00330-020-07240-7 (https://doi.org/10.1007%
2Fs00330-020-07240-7). PMID 32886201 (https://pubmed.ncbi.nlm.nih.gov/32886201).
S2CID 221478854 (https://api.semanticscholar.org/CorpusID:221478854).
106. Boonstra JT, Michielse S, Temel Y, Hoogland G, Jahanshahi A (February 2021).
"Neuroimaging Detectable Differences between Parkinson's Disease Motor Subtypes: A
Systematic Review" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7853198). Movement
Disorders Clinical Practice. 8 (2): 175–192. doi:10.1002/mdc3.13107 (https://doi.org/10.100
2%2Fmdc3.13107). PMC 7853198 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC785319
8). PMID 33553487 (https://pubmed.ncbi.nlm.nih.gov/33553487).
107. Suwijn SR, van Boheemen CJ, de Haan RJ, Tissingh G, Booij J, de Bie RM (2015). "The
diagnostic accuracy of dopamine transporter SPECT imaging to detect nigrostriatal cell loss
in patients with Parkinson's disease or clinically uncertain parkinsonism: a systematic
review" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4385258). EJNMMI Research. 5: 12.
doi:10.1186/s13550-015-0087-1 (https://doi.org/10.1186%2Fs13550-015-0087-1).
PMC 4385258 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4385258). PMID 25853018
(https://pubmed.ncbi.nlm.nih.gov/25853018).
108. "DaTSCAN Approval Letter" (https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/0
22454Orig1s000Lbl.pdf) (PDF). FDA.gov. Food and Drug Administration. Retrieved
22 March 2019.
109. Stoker TB, Greenland JC, Barker RA (December 2018). The Differential Diagnosis of
Parkinson's Disease (https://pubmed.ncbi.nlm.nih.gov/30702835/). Codon Publications.
pp. 109–128. doi:10.15586/codonpublications.parkinsonsdisease.2018.ch6 (https://doi.org/1
0.15586%2Fcodonpublications.parkinsonsdisease.2018.ch6). ISBN 978-0-9944381-6-4.
PMID 30702835 (https://pubmed.ncbi.nlm.nih.gov/30702835). S2CID 80908095 (https://api.s
emanticscholar.org/CorpusID:80908095).
110. McCann H, Stevens CH, Cartwright H, Halliday GM (January 2014). "α-Synucleinopathy
phenotypes" (http://handle.unsw.edu.au/1959.4/53593). Parkinsonism & Related Disorders.
20 (Suppl 1): S62–S67. doi:10.1016/S1353-8020(13)70017-8 (https://doi.org/10.1016%2FS
1353-8020%2813%2970017-8). hdl:1959.4/53593 (https://hdl.handle.net/1959.4%2F53593).
PMID 24262191 (https://pubmed.ncbi.nlm.nih.gov/24262191).
111. Ganguly J, Jog M (5 November 2020). "Tauopathy and Movement Disorders-Unveiling the
Chameleons and Mimics" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7674803).
Frontiers in Neurology. 11: 599384. doi:10.3389/fneur.2020.599384 (https://doi.org/10.338
9%2Ffneur.2020.599384). PMC 7674803 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC76
74803). PMID 33250855 (https://pubmed.ncbi.nlm.nih.gov/33250855).
112. Levin J, Kurz A, Arzberger T, Giese A, Höglinger GU (February 2016). "The Differential
Diagnosis and Treatment of Atypical Parkinsonism" (https://www.ncbi.nlm.nih.gov/pmc/articl
es/PMC4782269). Deutsches Ärzteblatt International. 113 (5): 61–69.
doi:10.3238/arztebl.2016.0061 (https://doi.org/10.3238%2Farztebl.2016.0061).
PMC 4782269 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4782269). PMID 26900156
(https://pubmed.ncbi.nlm.nih.gov/26900156).
113. Greenland J, Stoker TB (2018). Parkinson's Disease: Pathogenesis and Clinical Aspects.
Codon Publications. pp. 109–128. ISBN 978-0-9944381-6-4.
114. Ahlskog JE (July 2011). "Does vigorous exercise have a neuroprotective effect in Parkinson
disease?" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3136051). Neurology. 77 (3):
288–294. doi:10.1212/wnl.0b013e318225ab66 (https://doi.org/10.1212%2Fwnl.0b013e3182
25ab66). PMC 3136051 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3136051).
PMID 21768599 (https://pubmed.ncbi.nlm.nih.gov/21768599).
115. Costa J, Lunet N, Santos C, Santos J, Vaz-Carneiro A (2010). "Caffeine exposure and the
risk of Parkinson's disease: a systematic review and meta-analysis of observational studies"
(https://doi.org/10.3233%2FJAD-2010-091525). Journal of Alzheimer's Disease. 20 (Suppl
1): S221–238. doi:10.3233/JAD-2010-091525 (https://doi.org/10.3233%2FJAD-2010-09152
5). PMID 20182023 (https://pubmed.ncbi.nlm.nih.gov/20182023).
116. Gagne JJ, Power MC (March 2010). "Anti-inflammatory drugs and risk of Parkinson disease:
a meta-analysis" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2848103). Neurology. 74
(12): 995–1002. doi:10.1212/WNL.0b013e3181d5a4a3 (https://doi.org/10.1212%2FWNL.0b
013e3181d5a4a3). PMC 2848103 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC284810
3). PMID 20308684 (https://pubmed.ncbi.nlm.nih.gov/20308684).
117. Elkouzi A, Vedam-Mai V, Eisinger RS, Okun MS (April 2019). "Emerging therapies in
Parkinson disease – repurposed drugs and new approaches" (https://www.ncbi.nlm.nih.gov/
pmc/articles/PMC7758837). Nat Rev Neurol. 15 (4): 204–223. doi:10.1038/s41582-019-
0155-7 (https://doi.org/10.1038%2Fs41582-019-0155-7). PMC 7758837 (https://www.ncbi.nl
m.nih.gov/pmc/articles/PMC7758837). PMID 30867588 (https://pubmed.ncbi.nlm.nih.gov/30
867588).
118. Connolly BS, Lang AE (30 April 2014). "Pharmacological treatment of Parkinson disease: a
review". JAMA. 311 (16): 1670–1683. doi:10.1001/jama.2014.3654 (https://doi.org/10.1001%
2Fjama.2014.3654). PMID 24756517 (https://pubmed.ncbi.nlm.nih.gov/24756517).
S2CID 205058847 (https://api.semanticscholar.org/CorpusID:205058847).
119. Olanow CW, Stocchi F, Lang AE (2011). "The non-motor and non-dopaminergic features of
PD". Parkinson's Disease: Non-Motor and Non-Dopaminergic Features. Wiley-Blackwell.
ISBN 978-1405191852. OCLC 743205140 (https://www.worldcat.org/oclc/743205140).
120. The National Collaborating Centre for Chronic Conditions, ed. (2006). "Symptomatic
pharmacological therapy in Parkinson's disease" (https://web.archive.org/web/20100924153
546/http://guidance.nice.org.uk/CG35/Guidance/pdf/English). Parkinson's Disease
(Technical report). London: Royal College of Physicians. pp. 59–100. ISBN 978-1-86016-
283-1. Archived from the original (http://guidance.nice.org.uk/CG35/Guidance/pdf/English)
on 24 September 2010. Retrieved 14 March 2023.
121. Zhang J, Tan LC (2016). "Revisiting the Medical Management of Parkinson's Disease:
Levodopa versus Dopamine Agonist" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC48765
91). Current Neuropharmacology. 14 (4): 356–363.
doi:10.2174/1570159X14666151208114634 (https://doi.org/10.2174%2F1570159X1466615
1208114634). PMC 4876591 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4876591).
PMID 26644151 (https://pubmed.ncbi.nlm.nih.gov/26644151).
122. Moosa S, Martínez-Fernández R, Elias WJ, Del Alamo M, Eisenberg HM, Fishman PS
(September 2019). "The role of high-intensity focused ultrasound as a symptomatic
treatment for Parkinson's disease". Mov Disord. 34 (9): 1243–1251. doi:10.1002/mds.27779
(https://doi.org/10.1002%2Fmds.27779). ISSN 0885-3185 (https://www.worldcat.org/issn/088
5-3185). PMID 31291491 (https://pubmed.ncbi.nlm.nih.gov/31291491). S2CID 195879250
(https://api.semanticscholar.org/CorpusID:195879250).
123. Pedrosa DJ, Timmermann L (2013). "Review: management of Parkinson's disease" (https://
www.ncbi.nlm.nih.gov/pmc/articles/PMC3592512). Neuropsychiatric Disease and Treatment
(Review). 9: 321–340. doi:10.2147/NDT.S32302 (https://doi.org/10.2147%2FNDT.S32302).
PMC 3592512 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3592512). PMID 23487540
(https://pubmed.ncbi.nlm.nih.gov/23487540).
124. The National Collaborating Centre for Chronic Conditions, ed. (2006). "Palliative care in
Parkinson's disease" (http://guidance.nice.org.uk/CG35/Guidance/pdf/English). Parkinson's
Disease. London: Royal College of Physicians. pp. 147–151. ISBN 978-1-86016-283-1.
Archived (https://web.archive.org/web/20100924153546/http://guidance.nice.org.uk/CG35/G
uidance/pdf/English) from the original on 24 September 2010.
125. Orgeta V, McDonald KR, Poliakoff E, Hindle JV, Clare L, Leroi I (February 2020). "Cognitive
training interventions for dementia and mild cognitive impairment in Parkinson's disease" (ht
tps://www.ncbi.nlm.nih.gov/pmc/articles/PMC7043362). The Cochrane Database of
Systematic Reviews. 2020 (2): CD011961. doi:10.1002/14651858.cd011961.pub2 (https://d
oi.org/10.1002%2F14651858.cd011961.pub2). PMC 7043362 (https://www.ncbi.nlm.nih.go
v/pmc/articles/PMC7043362). PMID 32101639 (https://pubmed.ncbi.nlm.nih.gov/32101639).
126. "Recommendations Parkinson's disease in adults Guidance NICE" (https://www.nice.org.uk/
guidance/ng71/chapter/Recommendations). NICE. 2006. Retrieved 16 March 2023.
127. Maria N (2017). Levodopa pharmacokinetics – from stomach to brain: A study on patients
with Parkinson's disease. Linköping: Linköping University Electronic Press. p. 10.
ISBN 978-9176855577. OCLC 993068595 (https://www.worldcat.org/oclc/993068595).
128. Oertel WH (13 March 2017). "Recent advances in treating Parkinson's disease" (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC5357034). F1000Research (Review). 6: 260.
doi:10.12688/f1000research.10100.1 (https://doi.org/10.12688%2Ff1000research.10100.1).
PMC 5357034 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5357034). PMID 28357055
(https://pubmed.ncbi.nlm.nih.gov/28357055).
129. Binde et al. 2018, p. 1989.
130. Aquino CC, Fox SH (January 2015). "Clinical spectrum of levodopa-induced complications".
Movement Disorders. 30 (1): 80–89. doi:10.1002/mds.26125 (https://doi.org/10.1002%2Fmd
s.26125). PMID 25488260 (https://pubmed.ncbi.nlm.nih.gov/25488260). S2CID 22301199 (h
ttps://api.semanticscholar.org/CorpusID:22301199).
131. Yan Z, Wang W, Tao X, Cheng W, Zuo G, Chen Z (30 July 2022). "High-dose versus low-
dose inhaled levodopa (CVT-301) in patients with Parkinson disease for the treatment of
OFF episodes: a meta-analysis of randomized controlled trials" (https://doi.org/10.1007/s100
72-022-06298-z). Neurological Sciences. 43 (11): 6233–6241. doi:10.1007/s10072-022-
06298-z (https://doi.org/10.1007%2Fs10072-022-06298-z). PMID 35907110 (https://pubmed.
ncbi.nlm.nih.gov/35907110). S2CID 251162631 (https://api.semanticscholar.org/CorpusID:2
51162631). Retrieved 14 March 2023.
132. "Parkinson's disease" (https://www.mayoclinic.org/diseases-conditions/parkinsons-disease/
diagnosis-treatment/drc-20376062). Mayo Clinic. 8 July 2022. Retrieved 14 March 2023.
133. Paik J (June 2020). "Levodopa Inhalation Powder: A Review in Parkinson's Disease".
Drugs. 80 (8): 821–828. doi:10.1007/s40265-020-01307-x (https://doi.org/10.1007%2Fs4026
5-020-01307-x). PMID 32319076 (https://pubmed.ncbi.nlm.nih.gov/32319076).
S2CID 216033034 (https://api.semanticscholar.org/CorpusID:216033034).
134. Tambasco N, Romoli M, Calabresi P (2018). "Levodopa in Parkinson's Disease: Current
Status and Future Developments" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6187751).
Current Neuropharmacology. 16 (8): 1239–1252.
doi:10.2174/1570159X15666170510143821 (https://doi.org/10.2174%2F1570159X1566617
0510143821). PMC 6187751 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6187751).
PMID 28494719 (https://pubmed.ncbi.nlm.nih.gov/28494719).
135. Akhtar MJ, Yar MS, Grover G, Nath R (January 2020). "Neurological and psychiatric
management using COMT inhibitors: A review" (https://doi.org/10.1016%2Fj.bioorg.2019.10
3418). Bioorganic Chemistry. 94: 103418. doi:10.1016/j.bioorg.2019.103418 (https://doi.org/
10.1016%2Fj.bioorg.2019.103418). PMID 31708229 (https://pubmed.ncbi.nlm.nih.gov/3170
8229).
136. "Tasmar 100 mg Tablets – Summary of Product Characteristics (SmPC) – (emc)" (https://we
b.archive.org/web/20200806041659/https://www.medicines.org.uk/emc/product/3902).
www.medicines.org.uk. Archived from the original (https://www.medicines.org.uk/emc/produc
t/3902) on 6 August 2020. Retrieved 7 January 2021.
137. Scott LJ (September 2016). "Opicapone: A Review in Parkinson's Disease". Drugs. 76 (13):
1293–1300. doi:10.1007/s40265-016-0623-y (https://doi.org/10.1007%2Fs40265-016-0623-
y). PMID 27498199 (https://pubmed.ncbi.nlm.nih.gov/27498199). S2CID 5787752 (https://ap
i.semanticscholar.org/CorpusID:5787752).
138. Watkins P (2000). "COMT inhibitors and liver toxicity" (https://pubmed.ncbi.nlm.nih.gov/1114
7510). Neurology. 55 (11 Suppl 4): S51–52, discussion S53–56. PMID 11147510 (https://pu
bmed.ncbi.nlm.nih.gov/11147510).
139. "Comtess 200 mg film-coated Tablets – Summary of Product Characteristics (SmPC) –
(emc)" (https://www.medicines.org.uk/emc/product/1632). www.medicines.org.uk. Retrieved
7 January 2021.
140. "Stalevo 150 mg/37.5 mg/200 mg Film-coated Tablets – Summary of Product Characteristics
(SmPC) – (emc)" (https://www.medicines.org.uk/emc/product/6517/smpc).
www.medicines.org.uk. Retrieved 7 January 2021.
141. "Ongentys 50 mg hard capsules – Summary of Product Characteristics (SmPC) – (emc)" (htt
ps://www.medicines.org.uk/emc/product/7386). www.medicines.org.uk. Retrieved 7 January
2021.
142. Goldenberg MM (October 2008). "Medical management of Parkinson's disease" (https://ww
w.ncbi.nlm.nih.gov/pmc/articles/PMC2730785). P & T. 33 (10): 590–606. PMC 2730785 (http
s://www.ncbi.nlm.nih.gov/pmc/articles/PMC2730785). PMID 19750042 (https://pubmed.ncbi.
nlm.nih.gov/19750042).
143. Ceravolo R, Frosini D, Rossi C, Bonuccelli U (December 2009). "Impulse control disorders
in Parkinson's disease: definition, epidemiology, risk factors, neurobiology and
management". Parkinsonism & Related Disorders. 15 (Suppl 4): S111–S115.
doi:10.1016/S1353-8020(09)70847-8 (https://doi.org/10.1016%2FS1353-8020%2809%2970
847-8). PMID 20123548 (https://pubmed.ncbi.nlm.nih.gov/20123548).
144. Binde et al. 2018, p. 1924.
145. Alborghetti M, Nicoletti F (2019). "Different Generations of Type-B Monoamine Oxidase
Inhibitors in Parkinson's Disease: From Bench to Bedside" (https://www.ncbi.nlm.nih.gov/pm
c/articles/PMC7052841). Current Neuropharmacology. 17 (9): 861–873.
doi:10.2174/1570159X16666180830100754 (https://doi.org/10.2174%2F1570159X1666618
0830100754). PMC 7052841 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7052841).
PMID 30160213 (https://pubmed.ncbi.nlm.nih.gov/30160213).
146. Ives NJ, Stowe RL, Marro J, Counsell C, Macleod A, Clarke CE, et al. (September 2004).
"Monoamine oxidase type B inhibitors in early Parkinson's disease: meta-analysis of 17
randomised trials involving 3525 patients" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5
16655). BMJ. 329 (7466): 593. doi:10.1136/bmj.38184.606169.AE (https://doi.org/10.1136%
2Fbmj.38184.606169.AE). PMC 516655 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC516
655). PMID 15310558 (https://pubmed.ncbi.nlm.nih.gov/15310558).
147. Crosby N, Deane KH, Clarke CE (2003). "Amantadine in Parkinson's disease" (https://www.
ncbi.nlm.nih.gov/pmc/articles/PMC8715353). The Cochrane Database of Systematic
Reviews. 2010 (1): CD003468. doi:10.1002/14651858.CD003468 (https://doi.org/10.1002%
2F14651858.CD003468). PMC 8715353 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC87
15353). PMID 12535476 (https://pubmed.ncbi.nlm.nih.gov/12535476).
148. The National Collaborating Centre for Chronic Conditions, ed. (2006). "Non-motor features
of Parkinson's disease" (http://guidance.nice.org.uk/CG35/Guidance/pdf/English).
Parkinson's Disease. London: Royal College of Physicians. pp. 113–133. ISBN 978-1-
86016-283-1. Archived (https://web.archive.org/web/20100924153546/http://guidance.nice.o
rg.uk/CG35/Guidance/pdf/English) from the original on 24 September 2010.
149. Hasnain M, Vieweg WV, Baron MS, Beatty-Brooks M, Fernandez A, Pandurangi AK (July
2009). "Pharmacological management of psychosis in elderly patients with parkinsonism".
The American Journal of Medicine. 122 (7): 614–622. doi:10.1016/j.amjmed.2009.01.025 (htt
ps://doi.org/10.1016%2Fj.amjmed.2009.01.025). PMID 19559160 (https://pubmed.ncbi.nlm.n
ih.gov/19559160).
150. "FDA approves first drug to treat hallucinations and delusions associated with Parkinson's
disease" (https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm498442.ht
m). www.fda.gov (Press release). 29 April 2016. Retrieved 12 October 2018.
151. Elbers RG, Verhoef J, van Wegen EE, Berendse HW, Kwakkel G (October 2015).
"Interventions for fatigue in Parkinson's disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC9240814). The Cochrane Database of Systematic Reviews (Review). 2015 (10):
CD010925. doi:10.1002/14651858.CD010925.pub2 (https://doi.org/10.1002%2F14651858.
CD010925.pub2). PMC 9240814 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9240814).
PMID 26447539 (https://pubmed.ncbi.nlm.nih.gov/26447539).
152. The National Collaborating Centre for Chronic Conditions, ed. (2006). "Surgery for
Parkinson's disease" (http://guidance.nice.org.uk/CG35/Guidance/pdf/English). Parkinson's
Disease. London: Royal College of Physicians. pp. 101–111. ISBN 978-1-86016-283-1.
Archived (https://web.archive.org/web/20100924153546/http://guidance.nice.org.uk/CG35/G
uidance/pdf/English) from the original on 24 September 2010.
153. Bronstein JM, Tagliati M, Alterman RL, et al. (February 2011). "Deep brain stimulation for
Parkinson disease: an expert consensus and review of key issues" (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC4523130). Archives of Neurology. 68 (2): 165.
doi:10.1001/archneurol.2010.260 (https://doi.org/10.1001%2Farchneurol.2010.260).
PMC 4523130 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4523130). PMID 20937936
(https://pubmed.ncbi.nlm.nih.gov/20937936).
154. Dallapiazza RF, Vloo PD, Fomenko A, et al. (2018). "Considerations for Patient and Target
Selection in Deep Brain Stimulation surgery for Parkinson's disease". In Stoker TB,
Greenland JC (eds.). Parkinson's disease: Pathogenesis and clinical aspects. Brisbane:
Codon Publications.
155. Stoker TB, Greenland JC, Dallapiazza RF, De Vloo P, Fomenko A, Lee DJ, Hamani C,
Munhoz RP, Hodaie M, Lozano AM, Fasano A, Kalia SK (2018), Stoker TB (ed.),
"Considerations for Patient and Target Selection in Deep Brain Stimulation Surgery for
Parkinson's Disease" (http://www.ncbi.nlm.nih.gov/books/NBK536714/), Parkinson's
Disease: Pathogenesis and Clinical Aspects, Brisbane (AU): Codon Publications,
ISBN 978-0-9944381-6-4, PMID 30702838 (https://pubmed.ncbi.nlm.nih.gov/30702838),
retrieved 19 November 2021
156. The National Collaborating Centre for Chronic Conditions, ed. (2006). "Other key
interventions" (http://guidance.nice.org.uk/CG35/Guidance/pdf/English). Parkinson's
Disease. London: Royal College of Physicians. pp. 135–146. ISBN 978-1-86016-283-1.
Archived (https://web.archive.org/web/20100924153546/http://guidance.nice.org.uk/CG35/G
uidance/pdf/English) from the original on 24 September 2010.
157. Goodwin VA, Richards SH, Taylor RS, Taylor AH, Campbell JL (April 2008). "The
effectiveness of exercise interventions for people with Parkinson's disease: a systematic
review and meta-analysis". Movement Disorders. 23 (5): 631–640. doi:10.1002/mds.21922
(https://doi.org/10.1002%2Fmds.21922). hdl:10871/17451 (https://hdl.handle.net/10871%2F
17451). PMID 18181210 (https://pubmed.ncbi.nlm.nih.gov/18181210). S2CID 3808899 (http
s://api.semanticscholar.org/CorpusID:3808899).
158. Dereli EE, Yaliman A (April 2010). "Comparison of the effects of a physiotherapist-
supervised exercise programme and a self-supervised exercise programme on quality of life
in patients with Parkinson's disease". Clinical Rehabilitation. 24 (4): 352–362.
doi:10.1177/0269215509358933 (https://doi.org/10.1177%2F0269215509358933).
PMID 20360152 (https://pubmed.ncbi.nlm.nih.gov/20360152). S2CID 10947269 (https://api.s
emanticscholar.org/CorpusID:10947269).
159. Jin X, Wang L, Liu S, Zhu L, Loprinzi PD, Fan X (December 2019). "The Impact of Mind-
body Exercises on Motor Function, Depressive Symptoms, and Quality of Life in Parkinson's
Disease: A Systematic Review and Meta-analysis" (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC6981975). International Journal of Environmental Research and Public Health. 17 (1):
31. doi:10.3390/ijerph17010031 (https://doi.org/10.3390%2Fijerph17010031).
PMC 6981975 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6981975). PMID 31861456
(https://pubmed.ncbi.nlm.nih.gov/31861456).
160. O'Sullivan & Schmitz 2007, pp. 873, 876.
161. O'Sullivan & Schmitz 2007, p. 879.
162. O'Sullivan & Schmitz 2007, p. 877.
163. O'Sullivan & Schmitz 2007, p. 880.
164. Ramazzina I, Bernazzoli B, Costantino C (31 March 2017). "Systematic review on strength
training in Parkinson's disease: an unsolved question" (https://www.ncbi.nlm.nih.gov/pmc/art
icles/PMC5384725). Clinical Interventions in Aging. 12: 619–628. doi:10.2147/CIA.S131903
(https://doi.org/10.2147%2FCIA.S131903). PMC 5384725 (https://www.ncbi.nlm.nih.gov/pm
c/articles/PMC5384725). PMID 28408811 (https://pubmed.ncbi.nlm.nih.gov/28408811).
165. Fox CM, Ramig LO, Ciucci MR, Sapir S, McFarland DH, Farley BG (November 2006). "The
science and practice of LSVT/LOUD: neural plasticity-principled approach to treating
individuals with Parkinson disease and other neurological disorders". Seminars in Speech
and Language. 27 (4): 283–299. doi:10.1055/s-2006-955118 (https://doi.org/10.1055%2Fs-2
006-955118). PMID 17117354 (https://pubmed.ncbi.nlm.nih.gov/17117354).
S2CID 260320960 (https://api.semanticscholar.org/CorpusID:260320960).
166. Dixon L, Duncan D, Johnson P, et al. (July 2007). "Occupational therapy for patients with
Parkinson's disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6991932). The
Cochrane Database of Systematic Reviews. 2007 (3): CD002813.
doi:10.1002/14651858.CD002813.pub2 (https://doi.org/10.1002%2F14651858.CD002813.p
ub2). PMC 6991932 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6991932).
PMID 17636709 (https://pubmed.ncbi.nlm.nih.gov/17636709).
167. Ferrell B, Connor SR, Cordes A, et al. (June 2007). "The national agenda for quality
palliative care: the National Consensus Project and the National Quality Forum" (https://doi.
org/10.1016%2Fj.jpainsymman.2007.02.024). Journal of Pain and Symptom Management.
33 (6): 737–744. doi:10.1016/j.jpainsymman.2007.02.024 (https://doi.org/10.1016%2Fj.jpain
symman.2007.02.024). PMID 17531914 (https://pubmed.ncbi.nlm.nih.gov/17531914).
168. Lorenzl S, Nübling G, Perrar KM, Voltz R (2013). "Palliative treatment of chronic neurologic
disorders". Ethical and Legal Issues in Neurology. Handbook of Clinical Neurology.
Vol. 118. pp. 133–139. doi:10.1016/B978-0-444-53501-6.00010-X (https://doi.org/10.1016%
2FB978-0-444-53501-6.00010-X). ISBN 978-0444535016. PMID 24182372 (https://pubmed.
ncbi.nlm.nih.gov/24182372).
169. Ghoche R (December 2012). "The conceptual framework of palliative care applied to
advanced Parkinson's disease". Parkinsonism & Related Disorders. 18 (Suppl 3): S2–5.
doi:10.1016/j.parkreldis.2012.06.012 (https://doi.org/10.1016%2Fj.parkreldis.2012.06.012).
PMID 22771241 (https://pubmed.ncbi.nlm.nih.gov/22771241).
170. Wilcox SK (January 2010). "Extending palliative care to patients with Parkinson's disease".
British Journal of Hospital Medicine. 71 (1): 26–30. doi:10.12968/hmed.2010.71.1.45969 (htt
ps://doi.org/10.12968%2Fhmed.2010.71.1.45969). PMID 20081638 (https://pubmed.ncbi.nl
m.nih.gov/20081638).
171. Moens K, Higginson IJ, Harding R (October 2014). "Are there differences in the prevalence
of palliative care-related problems in people living with advanced cancer and eight non-
cancer conditions? A systematic review" (https://doi.org/10.1016%2Fj.jpainsymman.2013.11.
009). Journal of Pain and Symptom Management. 48 (4): 660–677.
doi:10.1016/j.jpainsymman.2013.11.009 (https://doi.org/10.1016%2Fj.jpainsymman.2013.1
1.009). PMID 24801658 (https://pubmed.ncbi.nlm.nih.gov/24801658).
172. Casey G (August 2013). "Parkinson's disease: a long and difficult journey". Nursing New
Zealand. 19 (7): 20–24. PMID 24195263 (https://pubmed.ncbi.nlm.nih.gov/24195263).
173. Poewe W (December 2006). "The natural history of Parkinson's disease". Journal of
Neurology. 253 (Suppl 7): vii2–vii6. doi:10.1007/s00415-006-7002-7 (https://doi.org/10.100
7%2Fs00415-006-7002-7). PMID 17131223 (https://pubmed.ncbi.nlm.nih.gov/17131223).
S2CID 35082340 (https://api.semanticscholar.org/CorpusID:35082340).
174. Feigin VL, Nichols E, Alam T, et al. (May 2019). "Global, regional, and national burden of
neurological disorders, 1990–2016: a systematic analysis for the Global Burden of Disease
Study 2016" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6459001). Lancet Neurol. 18
(5): 459–480. doi:10.1016/S1474-4422(18)30499-X (https://doi.org/10.1016%2FS1474-442
2%2818%2930499-X). PMC 6459001 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6459
001). PMID 30879893 (https://pubmed.ncbi.nlm.nih.gov/30879893).
175. Mhyre TR, Boyd JT, Hamill RW, Maguire-Zeiss KA (2012). "Parkinson's Disease". Protein
Aggregation and Fibrillogenesis in Cerebral and Systemic Amyloid Disease. Subcellular
Biochemistry. Vol. 65. pp. 389–455. doi:10.1007/978-94-007-5416-4_16 (https://doi.org/10.1
007%2F978-94-007-5416-4_16). ISBN 978-94-007-5415-7. PMC 4372387 (https://www.ncb
i.nlm.nih.gov/pmc/articles/PMC4372387). PMID 23225012 (https://pubmed.ncbi.nlm.nih.gov/
23225012).
176. Li G, Ma J, Cui S, He Y, Xiao Q, Liu J, Chen S (31 July 2019). "Parkinson's disease in
China: a forty-year growing track of bedside work" (https://www.ncbi.nlm.nih.gov/pmc/article
s/PMC6668186). Translational Neurodegeneration. 8 (1): 22. doi:10.1186/s40035-019-0162-
z (https://doi.org/10.1186%2Fs40035-019-0162-z). PMC 6668186 (https://www.ncbi.nlm.nih.
gov/pmc/articles/PMC6668186). PMID 31384434 (https://pubmed.ncbi.nlm.nih.gov/3138443
4).
177. Dorsey ER, Sherer T, Okun MS, Bloem BR (2018). "The Emerging Evidence of the
Parkinson Pandemic" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6311367). Journal of
Parkinson's Disease. 8 (s1): S3–S8. doi:10.3233/JPD-181474 (https://doi.org/10.3233%2FJ
PD-181474). PMC 6311367 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6311367).
PMID 30584159 (https://pubmed.ncbi.nlm.nih.gov/30584159).
178. "Art and Medicine Bibliography, Paul de Saint-Léger" (http://www.artandmedicine.com/bibli
o/authors/french/SaintLeger.html). Art and Medicine. Retrieved 29 April 2023.
179. Lewis PA, Plun-Favreau H, Rowley M, Spillane J (March 2020). "Pierre D. and the First
Photographs of Parkinson's Disease" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC71550
99). Movement Disorders. 35 (3): 389–391. doi:10.1002/MDS.27965 (https://doi.org/10.100
2%2FMDS.27965). PMC 7155099 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC715509
9). PMID 31975439 (https://pubmed.ncbi.nlm.nih.gov/31975439).
180. García Ruiz PJ (December 2004). "Prehistoria de la enfermedad de Parkinson" (https://ww
w.researchgate.net/publication/8159672) [Prehistory of Parkinson's disease]. Neurologia (in
Spanish). 19 (10): 735–737. PMID 15568171 (https://pubmed.ncbi.nlm.nih.gov/15568171)..
The article mistakenly refers to Job 34:19 instead of Job 33:19.
181. Lanska DJ (2009). "Chapter 33 the history of movement disorders". History of Neurology.
Handbook of Clinical Neurology. Vol. 95. pp. 501–546. doi:10.1016/S0072-9752(08)02133-7
(https://doi.org/10.1016%2FS0072-9752%2808%2902133-7). ISBN 978-0444520098.
PMID 19892136 (https://pubmed.ncbi.nlm.nih.gov/19892136).
182. Koehler PJ, Keyser A (September 1997). "Tremor in Latin texts of Dutch physicians: 16th–
18th centuries". Movement Disorders. 12 (5): 798–806. doi:10.1002/mds.870120531 (https://
doi.org/10.1002%2Fmds.870120531). PMID 9380070 (https://pubmed.ncbi.nlm.nih.gov/938
0070). S2CID 310819 (https://api.semanticscholar.org/CorpusID:310819).
183. Lees AJ (September 2007). "Unresolved issues relating to the shaking palsy on the
celebration of James Parkinson's 250th birthday". Movement Disorders. 22 (Suppl 17):
S327–S334. doi:10.1002/mds.21684 (https://doi.org/10.1002%2Fmds.21684).
PMID 18175393 (https://pubmed.ncbi.nlm.nih.gov/18175393). S2CID 9471754 (https://api.se
manticscholar.org/CorpusID:9471754).
184. Parkinson J (1817). An Essay on the Shaking Palsy (http://www.gutenberg.org/files/23777/2
3777-h/23777-h.htm). London: Whittingham and Roland for Sherwood, Neely, and Jones.
Archived (https://web.archive.org/web/20150924210730/http://www.gutenberg.org/files/2377
7/23777-h/23777-h.htm) from the original on 24 September 2015.
185. Louis ED (November 1997). "The shaking palsy, the first forty-five years: a journey through
the British literature". Movement Disorders. 12 (6): 1068–1072. doi:10.1002/mds.870120638
(https://doi.org/10.1002%2Fmds.870120638). PMID 9399240 (https://pubmed.ncbi.nlm.nih.g
ov/9399240). S2CID 34630080 (https://api.semanticscholar.org/CorpusID:34630080).
186. Fahn S (2008). "The history of dopamine and levodopa in the treatment of Parkinson's
disease". Movement Disorders. 23 (Suppl 3): S497–508. doi:10.1002/mds.22028 (https://doi.
org/10.1002%2Fmds.22028). PMID 18781671 (https://pubmed.ncbi.nlm.nih.gov/18781671).
S2CID 45572523 (https://api.semanticscholar.org/CorpusID:45572523).
187. Guridi J, Lozano AM (November 1997). "A brief history of pallidotomy". Neurosurgery. 41 (5):
1169–1180, discussion 1180–1183. doi:10.1097/00006123-199711000-00029 (https://doi.or
g/10.1097%2F00006123-199711000-00029). PMID 9361073 (https://pubmed.ncbi.nlm.nih.g
ov/9361073).
188. Hornykiewicz O (2002). "L-DOPA: from a biologically inactive amino acid to a successful
therapeutic agent". Amino Acids. 23 (1–3): 65–70. doi:10.1007/s00726-001-0111-9 (https://d
oi.org/10.1007%2Fs00726-001-0111-9). PMID 12373520 (https://pubmed.ncbi.nlm.nih.gov/1
2373520). S2CID 25117208 (https://api.semanticscholar.org/CorpusID:25117208).
189. Coffey RJ (March 2009). "Deep brain stimulation devices: a brief technical history and
review". Artificial Organs. 33 (3): 208–220. doi:10.1111/j.1525-1594.2008.00620.x (https://do
i.org/10.1111%2Fj.1525-1594.2008.00620.x). PMID 18684199 (https://pubmed.ncbi.nlm.nih.
gov/18684199).
190. "UF neurologists create a new image of Parkinson disease" (https://mbi.ufl.edu/2020/07/27/u
f-neurologists-create-a-new-image-of-parkinson-disease/). McKnight Brain Institute,
University of Florida. 27 July 2020. Retrieved 17 November 2023.
191. Armstrong MJ, Okun MS (November 2020). "Time for a New Image of Parkinson Disease".
JAMA Neurol. 77 (11): 1345–1346. doi:10.1001/jamaneurol.2020.2412 (https://doi.org/10.10
01%2Fjamaneurol.2020.2412). PMID 32716484 (https://pubmed.ncbi.nlm.nih.gov/3271648
4). S2CID 220796667 (https://api.semanticscholar.org/CorpusID:220796667).
192. Crooks S, Carter G, Wilson CB, Wynne L, Stark P, Doumas M, Rodger M, O'Shea E, Mitchell
G (2023). "Exploring public perceptions and awareness of Parkinson's disease: A scoping
review" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10503766). PLOS ONE. 18 (9):
e0291357. Bibcode:2023PLoSO..1891357C (https://ui.adsabs.harvard.edu/abs/2023PLoS
O..1891357C). doi:10.1371/journal.pone.0291357 (https://doi.org/10.1371%2Fjournal.pone.
0291357). PMC 10503766 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10503766).
PMID 37713383 (https://pubmed.ncbi.nlm.nih.gov/37713383).
193. Findley LJ (September 2007). "The economic impact of Parkinson's disease". Parkinsonism
& Related Disorders. 13 (Suppl): S8–S12. doi:10.1016/j.parkreldis.2007.06.003 (https://doi.o
rg/10.1016%2Fj.parkreldis.2007.06.003). PMID 17702630 (https://pubmed.ncbi.nlm.nih.gov/
17702630).
194. Yang W, Hamilton JL, Kopil C, et al. (2020). "Current and projected future economic burden
of Parkinson's disease in the U.S" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7347582).
npj Parkinson's Disease. 6: 15. doi:10.1038/s41531-020-0117-1 (https://doi.org/10.1038%2F
s41531-020-0117-1). PMC 7347582 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC734758
2). PMID 32665974 (https://pubmed.ncbi.nlm.nih.gov/32665974). Material was copied from
this source, which is available under a Creative Commons Attribution 4.0 International
License (https://creativecommons.org/licenses/by/4.0/).
195. "Parkinson's – 'the shaking palsy' " (https://web.archive.org/web/20110514151652/http://ww
w.gsk.com/infocus/parkinsons.htm). GlaxoSmithKline. 1 April 2009. Archived from the
original (http://www.gsk.com/infocus/parkinsons.htm) on 14 May 2011.
196. "National Parkinson Foundation – Mission" (https://web.archive.org/web/20101221103201/h
ttp://parkinson.org/About-Us/Mission). Archived from the original (http://www.parkinson.org/A
bout-Us/Mission) on 21 December 2010. Retrieved 28 March 2011.
197. "Education: Joy in Giving" (https://web.archive.org/web/20110220012106/http://www.time.co
m/time/magazine/article/0,9171,828597,00.html). Time. 18 January 1960. Archived from the
original (http://www.time.com/time/magazine/article/0,9171,828597,00.html) on 20 February
2011. Retrieved 2 April 2011.
198. "About PDF" (https://web.archive.org/web/20110515204903/http://www.pdf.org/en/about_pd
f). Parkinson's Disease Foundation. Archived from the original (http://www.pdf.org/en/about_
pdf) on 15 May 2011. Retrieved 24 July 2016.
199. "American Parkinson Disease Association: Home" (https://web.archive.org/web/201205101
65933/http://www.apdaparkinson.org/userND/index.asp). American Parkinson Disease
Association. Archived from the original (http://www.apdaparkinson.org/userND/index.asp) on
10 May 2012. Retrieved 9 August 2010.
200. "About EPDA" (http://www.epda.eu.com/about-us/). European Parkinson's Disease
Association. 2010. Archived (https://web.archive.org/web/20100815232300/http://www.epda.
eu.com/about-us/) from the original on 15 August 2010. Retrieved 9 August 2010.
201. "Notable Figures with Parkinson's" (https://www.parkinson.org/understanding-parkinsons/sta
tistics/notable-figures). Parkinson's Foundation. Retrieved 22 November 2023.
202. "Michael's Story" (https://www.michaeljfox.org/michaels-story). The Michael J. Fox
Foundation for Parkinson's Research. Retrieved 7 May 2023.
203. Davis P (3 May 2007). "Michael J. Fox" (https://web.archive.org/web/20110425013526/http://
www.time.com/time/specials/2007/time100/article/0,28804,1595326_1615754_1615882,00.
html). The Time 100. Time. Archived from the original (http://www.time.com/time/specials/200
7/time100/article/0,28804,1595326_1615754_1615882,00.html) on 25 April 2011. Retrieved
2 April 2011.
204. Brockes E (11 April 2009). " 'It's the gift that keeps on taking' " (https://www.theguardian.com/l
ifeandstyle/2009/apr/11/michael-j-fox-parkinsons). The Guardian. Archived (https://web.archi
ve.org/web/20131008000425/http://www.theguardian.com/lifeandstyle/2009/apr/11/michael-j
-fox-parkinsons) from the original on 8 October 2013. Retrieved 25 October 2010.
205. Burleson, Nate; Breen, Kerry (9 November 2023). "Michael J. Fox talks funding
breakthrough research for Parkinson's disease" (https://www.cbsnews.com/news/michael-j-f
ox-parkinsons-research-funded-by-his-foundation/). CBS News. Retrieved 23 November
2023.
206. Brey RL (April 2006). "Muhammad Ali's Message: Keep Moving Forward" (https://web.archiv
e.org/web/20110927022505/http://www.aan.com/elibrary/neurologynow/?event=home.show
Article&id=ovid.com%3A%2Fbib%2Fovftdb%2F01222928-200602020-00003). Neurology
Now. 2 (2): 8. doi:10.1097/01222928-200602020-00003 (https://doi.org/10.1097%2F012229
28-200602020-00003). Archived from the original (http://www.aan.com/elibrary/neurologyno
w/?event=home.showArticle&id=ovid.com:/bib/ovftdb/01222928-200602020-00003) on 27
September 2011. Retrieved 22 August 2020.
207. Matthews W (April 2006). "Ali's Fighting Spirit". Neurology Now. 2 (2): 10–23.
doi:10.1097/01222928-200602020-00004 (https://doi.org/10.1097%2F01222928-20060202
0-00004). S2CID 181104230 (https://api.semanticscholar.org/CorpusID:181104230).
208. Tauber P (17 July 1988). "Ali: Still Magic" (https://www.nytimes.com/1988/07/17/magazine/al
i-still-magic.html). The New York Times. Archived (https://web.archive.org/web/2016111715
1827/http://www.nytimes.com/1988/07/17/magazine/ali-still-magic.html) from the original on
17 November 2016. Retrieved 2 April 2011.
209. Macur J (26 March 2008). "For the Phinney Family, a Dream and a Challenge" (https://www.
nytimes.com/2008/03/26/sports/othersports/26cycling.html). The New York Times. Archived
(https://web.archive.org/web/20141106025145/http://www.nytimes.com/2008/03/26/sports/ot
hersports/26cycling.html) from the original on 6 November 2014. Retrieved 25 May 2013.
"About 1.5 million Americans have received a diagnosis of Parkinson's disease, but only 5
to 10 percent learn of it before age 40, according to the National Parkinson Foundation.
Davis Phinney was among the few."
210. "Who We Are" (http://davisphinneyfoundation.org/dpf/who-we-are/). Davis Phinney
Foundation. Archived (https://web.archive.org/web/20120111054225/http://davisphinneyfoun
dation.org/dpf/who-we-are/) from the original on 11 January 2012. Retrieved 18 January
2012.
211. Gallman S (4 November 2015). "Robin Williams' widow speaks: Depression didn't kill my
husband" (http://www.cnn.com/2015/11/03/health/robin-williams-widow-susan-williams/inde
x.html). CNN. Archived (https://web.archive.org/web/20151104002632/http://www.cnn.com/2
015/11/03/health/robin-williams-widow-susan-williams/index.html) from the original on 4
November 2015. Retrieved 6 April 2018.
212. Williams SS (September 2016). "The terrorist inside my husband's brain" (http://n.neurology.
org/content/87/13/1308.long). Neurology. 87 (13): 1308–1311.
doi:10.1212/WNL.0000000000003162 (https://doi.org/10.1212%2FWNL.000000000000316
2). PMID 27672165 (https://pubmed.ncbi.nlm.nih.gov/27672165).
213. Robbins R (30 September 2016). "How Lewy body dementia gripped Robin Williams" (http
s://www.scientificamerican.com/article/how-lewy-body-dementia-gripped-robin-williams1/).
Scientific American. Retrieved 9 April 2018.
214. "LBDA Clarifies Autopsy Report on Comedian, Robin Williams" (https://web.archive.org/we
b/20200812093153/https://www.lbda.org/content/lbda-clarifies-autopsy-report-comedian-robi
n-williams). Lewy Body Dementia Association. 10 November 2014. Archived from the
original (https://www.lbda.org/content/lbda-clarifies-autopsy-report-comedian-robin-williams)
on 12 August 2020. Retrieved 19 April 2018.
215. McKeith IG (6 November 2015). "Robin Williams had dementia with Lewy bodies – so, what
is it and why has it been eclipsed by Alzheimer's?" (https://theconversation.com/robin-willia
ms-had-dementia-with-lewy-bodies-so-what-is-it-and-why-has-it-been-eclipsed-by-alzheime
rs-50221). The Conversation. Archived (https://web.archive.org/web/20161104010452/http
s://theconversation.com/robin-williams-had-dementia-with-lewy-bodies-so-what-is-it-and-wh
y-has-it-been-eclipsed-by-alzheimers-50221) from the original on 4 November 2016.
Retrieved 6 April 2018.
216. Luca A, Nicoletti A, Mostile G, Zappia M (2018). "The Parkinsonian Personality: More Than
Just a "Trait" ". Frontiers in Neurology. 9 (1191): 1–2. PMID 30697187 (https://pubmed.ncbi.n
lm.nih.gov/30697187).
217. Gerstenbrand F, Karamat E (2007). "Adolf Hitler's Parkinson's disease and an attempt to
analyse his personality structure". European Journal of Neurology. 6 (2): 121–127.
doi:10.1111/j.1468-1331.1999.tb00003.x (https://doi.org/10.1111%2Fj.1468-1331.1999.tb00
003.x). PMID 10053222 (https://pubmed.ncbi.nlm.nih.gov/10053222). S2CID 30118747 (http
s://api.semanticscholar.org/CorpusID:30118747).
218. McCrum, Robert (20 November 2017). "The 100 best nonfiction books: No 94 – Leviathan
by Thomas Hobbes (1651)" (https://www.theguardian.com/books/2017/nov/20/the-100-best-
nonfiction-books-no-94-leviathan-thomas-hobbes-1651). The Guardian. Retrieved
23 November 2023.
219. Kinsley, Michael (21 April 2014). "Have You Lost Your Mind?" (https://www.newyorker.com/m
agazine/2014/04/28/have-you-lost-your-mind). The New Yorker. Retrieved 23 November
2023.
220. Raudino F (2011). "The Parkinson disease before James Parkinson". History of Neurology.
33: 945–949.
221. Gupta R, Kim C, Agarwal N, Lieber B, Monaco EA (2015). "Understanding the Influence of
Parkinson Disease on Adolf Hitler's Decision-Making during World War II". World
Neurosurgery. 84 (5): 1447–1452. doi:10.1016/j.wneu.2015.06.014 (https://doi.org/10.1016%
2Fj.wneu.2015.06.014). PMID 26093359 (https://pubmed.ncbi.nlm.nih.gov/26093359).
222. Boettcher L, Bonney P, Smitherman A, Sughrue M (2015). "Hitler's parkinsonism".
Neurosurgical Focus. 39 (1): E8. doi:10.3171/2015.4.FOCUS1563 (https://doi.org/10.3171%
2F2015.4.FOCUS1563). S2CID 19721269 (https://api.semanticscholar.org/CorpusID:19721
269).
223. Glass, Andrew (9 September 2016). "Mao Zedong dies in Beijing at age 82, Sept. 9, 1976"
(https://www.politico.com/story/2016/09/mao-zedong-dies-in-beijing-at-age-82-sept-9-1976-2
27742). Politico. Retrieved 30 October 2023.
224. Mari Z, Mestre TA (2022). "The Disease Modification Conundrum in Parkinson's Disease:
Failures and Hopes" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8920063). Frontiers in
Aging Neuroscience. 14: 810860. doi:10.3389/fnagi.2022.810860 (https://doi.org/10.3389%2
Ffnagi.2022.810860). PMC 8920063 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC89200
63). PMID 35296034 (https://pubmed.ncbi.nlm.nih.gov/35296034).
225. McFarthing K, Rafaloff G, Baptista M, Mursaleen L, Fuest R, Wyse RK, Stott SR (24 May
2022). "Parkinson's Disease Drug Therapies in the Clinical Trial Pipeline: 2022 Update" (htt
ps://www.ncbi.nlm.nih.gov/pmc/articles/PMC9198738). Journal of Parkinson's Disease. 12
(4): 1073–1082. doi:10.3233/JPD-229002 (https://doi.org/10.3233%2FJPD-229002).
PMC 9198738 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9198738). PMID 35527571
(https://pubmed.ncbi.nlm.nih.gov/35527571).
226. Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, et al. (March 2017).
"Parkinson disease". Nature Reviews. Disease Primers. 3 (1): 17013.
doi:10.1038/nrdp.2017.13 (https://doi.org/10.1038%2Fnrdp.2017.13). PMID 28332488 (http
s://pubmed.ncbi.nlm.nih.gov/28332488). S2CID 11605091 (https://api.semanticscholar.org/C
orpusID:11605091).
227. Heinzel S, Berg D, Gasser T, Chen H, Yao C, Postuma RB (October 2019). "Update of the
MDS research criteria for prodromal Parkinson's disease" (https://doi.org/10.1002%2Fmds.2
7802). Movement Disorders. 34 (10): 1464–1470. doi:10.1002/mds.27802 (https://doi.org/10.
1002%2Fmds.27802). PMID 31412427 (https://pubmed.ncbi.nlm.nih.gov/31412427).
S2CID 199663713 (https://api.semanticscholar.org/CorpusID:199663713).
228. Menozzi E, Macnaughtan J, Schapira AH (December 2021). "The gut-brain axis and
Parkinson disease: clinical and pathogenetic relevance" (https://www.ncbi.nlm.nih.gov/pmc/
articles/PMC8078923). Ann Med. 53 (1): 611–625. doi:10.1080/07853890.2021.1890330 (ht
tps://doi.org/10.1080%2F07853890.2021.1890330). PMC 8078923 (https://www.ncbi.nlm.ni
h.gov/pmc/articles/PMC8078923). PMID 33860738 (https://pubmed.ncbi.nlm.nih.gov/338607
38).
229. Sudhakar V, Richardson RM (January 2019). "Gene Therapy for Neurodegenerative
Diseases" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6361055). Neurotherapeutics. 16
(1): 166–175. doi:10.1007/s13311-018-00694-0 (https://doi.org/10.1007%2Fs13311-018-006
94-0). PMC 6361055 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6361055).
PMID 30542906 (https://pubmed.ncbi.nlm.nih.gov/30542906).
230. Hitti FL, Yang AI, Gonzalez-Alegre P, Baltuch GH (September 2019). "Human gene therapy
approaches for the treatment of Parkinson's disease: An overview of current and completed
clinical trials". Parkinsonism & Related Disorders. 66: 16–24.
doi:10.1016/j.parkreldis.2019.07.018 (https://doi.org/10.1016%2Fj.parkreldis.2019.07.018).
PMID 31324556 (https://pubmed.ncbi.nlm.nih.gov/31324556). S2CID 198132349 (https://ap
i.semanticscholar.org/CorpusID:198132349).
231. Volc D, Poewe W, Kutzelnigg A, et al. (July 2020). "Safety and immunogenicity of the α-
synuclein active immunotherapeutic PD01A in patients with Parkinson's disease: a
randomised, single-blinded, phase 1 trial". The Lancet. Neurology. 19 (7): 591–600.
doi:10.1016/S1474-4422(20)30136-8 (https://doi.org/10.1016%2FS1474-4422%2820%2930
136-8). PMID 32562684 (https://pubmed.ncbi.nlm.nih.gov/32562684). S2CID 219947651 (htt
ps://api.semanticscholar.org/CorpusID:219947651).
232. "World's first Parkinson's vaccine is trialled" (https://www.newscientist.com/article/mg214286
82.000-worlds-first-parkinsons-vaccine-is-trialled.html). New Scientist. London. 7 June 2012.
Archived (https://web.archive.org/web/20150423080305/http://www.newscientist.com/article/
mg21428682.000-worlds-first-parkinsons-vaccine-is-trialled.html) from the original on 23
April 2015.
233. Jankovic J, Goodman I, Safirstein B, et al. (October 2018). "Safety and Tolerability of Multiple
Ascending Doses of PRX002/RG7935, an Anti-α-Synuclein Monoclonal Antibody, in
Patients With Parkinson Disease: A Randomized Clinical Trial" (https://www.ncbi.nlm.nih.go
v/pmc/articles/PMC6233845). JAMA Neurology. 75 (10): 1206–1214.
doi:10.1001/jamaneurol.2018.1487 (https://doi.org/10.1001%2Fjamaneurol.2018.1487).
PMC 6233845 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6233845). PMID 29913017
(https://pubmed.ncbi.nlm.nih.gov/29913017).
234. Henchcliffe C, Parmar M (2018). "Repairing the Brain: Cell Replacement Using Stem Cell-
Based Technologies" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6311366). Journal of
Parkinson's Disease. 8 (s1): S131–S137. doi:10.3233/JPD-181488 (https://doi.org/10.323
3%2FJPD-181488). PMC 6311366 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC631136
6). PMID 30584166 (https://pubmed.ncbi.nlm.nih.gov/30584166).
235. Schweitzer JS, Song B, Herrington TM, et al. (May 2020). "Personalized iPSC-Derived
Dopamine Progenitor Cells for Parkinson's Disease" (https://www.ncbi.nlm.nih.gov/pmc/artic
les/PMC7288982). The New England Journal of Medicine. 382 (20): 1926–1932.
doi:10.1056/NEJMoa1915872 (https://doi.org/10.1056%2FNEJMoa1915872).
PMC 7288982 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7288982). PMID 32402162
(https://pubmed.ncbi.nlm.nih.gov/32402162).
236. Jenner P (2014). "An overview of adenosine A2A receptor antagonists in Parkinson's
disease". International Review of Neurobiology. 119: 71–86. doi:10.1016/B978-0-12-
801022-8.00003-9 (https://doi.org/10.1016%2FB978-0-12-801022-8.00003-9). ISBN 978-
0128010228. ISSN 2162-5514 (https://www.worldcat.org/issn/2162-5514). PMID 25175961
(https://pubmed.ncbi.nlm.nih.gov/25175961).
237. Office of the Commissioner (20 February 2020). "FDA approves new add-on drug to treat off
episodes in adults with Parkinson's disease" (http://www.fda.gov/news-events/press-announ
cements/fda-approves-new-add-drug-treat-episodes-adults-parkinsons-disease). FDA.
Retrieved 23 February 2020.

Book sources
O'Sullivan SB, Schmitz TJ (2007). "Parkinson's Disease". Physical Rehabilitation (5th ed.).
Philadelphia: F.A. Davis.

Journal sources
Binde CD, Tvete IF, Gåsemyr J, Natvig B, Klemp M (21 May 2018). "A multiple treatment
comparison meta-analysis of monoamine oxidase type B inhibitors for Parkinson's disease"
(https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6089809). British Journal of Clinical
Pharmacology. 84 (9): 1917–1927. doi:10.1111/bcp.13651 (https://doi.org/10.1111%2Fbcp.1
3651). PMC 6089809 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6089809).
PMID 29847694 (https://pubmed.ncbi.nlm.nih.gov/29847694).

External links
Parkinson's disease (https://curlie.org/Health/Conditions_and_Diseases/Neurological_Disor
ders/Parkinson%27s_Disease/) at Curlie

Retrieved from "https://en.wikipedia.org/w/index.php?title=Parkinson%27s_disease&oldid=1187632010"

You might also like